You are on page 1of 20

Critical Reviews in Food Science and Nutrition

ISSN: 1040-8398 (Print) 1549-7852 (Online) Journal homepage: https://www.tandfonline.com/loi/bfsn20

The impact of probiotics, prebiotics, and synbiotics


on the biochemical, clinical, and immunological
markers, as well as on the gut microbiota of obese
hosts

Tatiane Ferreira da Silva, Sabrina Neves Casarotti, Gislane Lelis Vilela de


Oliveira & Ana Lúcia Barretto Penna

To cite this article: Tatiane Ferreira da Silva, Sabrina Neves Casarotti, Gislane Lelis Vilela de
Oliveira & Ana Lúcia Barretto Penna (2020): The impact of probiotics, prebiotics, and synbiotics
on the biochemical, clinical, and immunological markers, as well as on the gut microbiota of obese
hosts, Critical Reviews in Food Science and Nutrition, DOI: 10.1080/10408398.2020.1733483

To link to this article: https://doi.org/10.1080/10408398.2020.1733483

Published online: 10 Mar 2020.

Submit your article to this journal

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=bfsn20
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION
https://doi.org/10.1080/10408398.2020.1733483

REVIEW

The impact of probiotics, prebiotics, and synbiotics on the biochemical, clinical,


and immunological markers, as well as on the gut microbiota of obese hosts
cia Barretto
Tatiane Ferreira da Silvaa, Sabrina Neves Casarottib, Gislane Lelis Vilela de Oliveirac, and Ana Lu
Pennaa
a
Departamento de Engenharia e Tecnologia de Alimentos, Universidade Estadual Paulista (UNESP), S~ao Jose do Rio Preto, Brazil; bInstituto
de Ci^encias Naturais e Exatas, Universidade Federal de Rondonopolis (UFR), Rondonopolis, Brazil; cBrigham and Women’s Hospital, Harvard
Medical School, Boston, Massachusetts, USA

ABSTRACT KEYWORDS
Obesity is currently considered a global epidemic and it leads to several alterations on the human functional food; dysbiosis;
body and its metabolism. There are evidences showing that the intestinal microbiota can influence obesity; intestinal health
on the pathogenesis of obesity. Microbiota plays a vital role not only in the digestion and absorp-
tion of nutrients, but also in the homeostatic maintenance of host immunity, metabolism, and gut
barrier. Its dietary alteration is an important target in the treatment of obesity. Emerging evidence
suggests that modifying the composition of the gut microbiota through probiotic, prebiotic, and
synbiotic supplementation may be a viable adjuvant treatment option for obese individuals. In
this review, the impact of probiotics, prebiotics, and synbiotics on the anthropometric profile, bio-
chemical regulation, clinical, and immunological markers, as well as on the gut microbiota of
obese hosts is described. It also emphasizes how changes in the composition and/or metabolic
activity of the gut microbiota through the administration of nutrients with probiotic, prebiotic, or
synbiotic properties can modulate the host’s gene expression and metabolism, and thereby posi-
tively influence on the host’s adipose tissue development and related metabolic disorders. The
beneficial effects on the host’s metabolism promoted by prebiotics, probiotics, and synbiotics
have been successfully demonstrated by several studies. However, further investigation is needed
to fully explain the cellular mechanisms of action of probiotics and prebiotics on human health,
and also to elucidate the relationship between microbiota and obesity etiology, using well-
designed, long-term, and large-scale clinical interventions.

Introduction highly dynamic metabolism (Delgado and Tamashiro 2018).


Due to the complexity of obesity, a single solution against
According to the literature, over the last 40 years,
this growing epidemic has not been disclosed in studies
overweight, obesity, and its complications have stood out
published in scientific circles. Hence, individualized cautious
extraordinarily. The period goes from 1975 to 2016, when
procedures should be studied in order to prevent and treat
there was a dramatic increase in obesity according to data
compiled by the World Health Organization (WHO). overweight and obesity (Borgeraas et al. 2018).
The data describe about 1.9 billion people aged 18 years or It is also known that obesity is directly related to other
older who are at least overweight (body mass index or complications, including metabolic syndrome, hypertension,
BMI > 25 kg/m2); among them, around 650 million are diag- type 2 diabetes, dyslipidemia, coronary artery disease, stroke,
nosed with obesity (BMI > 30 kg/m2). Thus, 3 years ago, 39% asthma, arthritis, sleep apnea, some types of cancer, and
and 13% of people worldwide were overweight or obese, inflammation-related pathologies (Torres-Fuentes et al. 2015;
respectively. Overweight and obesity are increasingly worrying Seganfredo et al. 2017). Thus, there is a growing demand for
in both low- and middle-income countries, especially in urban scientific studies with nutritional strategies to promote
areas (WHO 2018). This expansion is expressed by the set of health (Bailey and Holscher 2018).
biological, behavioral, and environmental processes that still The disproportionate relationship between overeating and
need further studies (Rastelli, Knauf, and Cani 2018). energy expenditures is one of the reasons of energy imbal-
According to the WHO, the diagnosis of obesity is body ance known as obesity. After the ingestion of food, each
fat that poses a health risk (WHO 2018). Thus, there is an organism performs different weight-regulating mechanisms,
accumulation of adipose cells (adipocytes), which signal the such as peripheral and central signs of hunger and satiety,
increase or the deficiency of total body energy. The adipo- and response of the gastrointestinal tract organs; these
cyte set builds adipose tissue, an endocrine organ with mechanisms are known to be individualized. Obesity is also

CONTACT Ana Lucia B. Penna ana.lb.penna@unesp.br Department of Food Engineering and Technology, Universidade Estadual Paulista (UNESP), Rua
Cristov~ao Colombo, 2265. 15054-000 S~ao Jose do Rio Preto, Brazil.
ß 2020 Taylor & Francis Group, LLC
2 T. F. DA SILVA ET AL.

characterized by complex genetic interactions among genes, and synbiotics on the biochemical, clinical, and immuno-
environmental factors (diet, food components) and lifestyle logical markers, as well as on the gut microbiota of obese
(sedentary or not), as well as imbalance of the intestinal hosts. Additionally, it aimed to clarify questions about cur-
microbiota, which negatively affects this process (Gomes rent knowledge, to suggest future studies and dietary inter-
et al. 2017; Kobyliak et al. 2018). ventions to improve obese patients’ health.
Studies have shown that obesity and its complications are
accompanied by changes in the intestinal microbiota (Al-
Assal et al. 2018). Nonetheless, appetite can be reduced and Effects of probiotics, prebiotics, and synbiotics on
clinical parameters of obesity
satiety increased, according to the product generated from
the bacterial fermentation process and by modulation of bile The literature has shown that eating habits positively correl-
acid metabolism; an increase in energy expenditure modu- ate with the human intestinal microbiota, since dietary
lated by the microbiota can occur and thus inhibit diet- changes can alter its diversity and composition. Short-term
induced obesity (Cani and Delzenne 2009; Watanabe et al. changes in diet profile can change the gut microbiota and
2012; Sayin et al. 2013). could be a key alternative in the treatment of obesity. One
There are three ways in which the intestinal microbiota of the ways to do so is by consuming probiotics, prebiotics,
can modulate the mechanisms of obesity: i) effect on energy and synbiotics aiming at improving the host’s health (Al-
extraction and absorption - the fermentation of indigestible Assal et al. 2018; John et al. 2018). Many studies are avail-
fibers increases energy extraction from the diet, while the able in the literature.
microbiota also facilitates nutrient absorption; ii) induction Dewulf et al. (2013) and Salazar et al. (2015) had been
or prevention metabolic endotoxemia through changes in pioneers in testing the impact of inulin-type fructans (ITF)
intestinal permeability - obese microbiota can increase intes- as prebiotic in obese women and their effects in gut micro-
tinal inflammation, intestinal permeability and subsequently biota composition and metabolic activity. Dewulf et al.
systemic inflammation by increasing the tumor necrosis fac- (2013) reported that prebiotics were correlated with bacter-
tor alpha (TNF-a) and interleukin 1beta (IL-1b) production ial-related metabolites (phosphatidylcholine, lactate, hippur-
by peritoneal macrophages; iii) regulation and secretion of ate) and with serum lipopolysaccharide (LPS) levels, despite
intestinal metabolites – the short chain fatty acids (SCFAs) a lack of significant effect on body weight. Salazar et al.
produced by intestinal microbiota activate specific receptors (2015) observed that prebiotic consumption resulted in sig-
for nutrients that stimulate the intestinal release of the sati- nificantly lower total SCFAs, acetate and propionate, which
ety-inducing hormones peptide YY and glucagon-like pep- positively correlated with BMI, fasting insulinemia and
tide-1 (GLP-1), by decreasing systemic inflammation or by homeostasis model assessment (HOMA) (p < 0.05), com-
signaling to the brain. SCFAs also target adipocytes, where pared to the placebo group after the treatment period.
they increase lipolysis and leptin release. Additionally, these Bernini et al. (2016) observed that a daily intake (80 mL)
mechanisms of action can work simultaneously or be con- of the probiotic Bifidobacterium lactis HN019 correlated
nected (Mulders et al. 2018). negatively with the side effects of the metabolic syndrome
The intestinal microbial modification through feeding (MS) and the cardiovascular risks. They observed that BMI,
mainly occurs with the ingestion of probiotic, prebiotics, total cholesterol (TC), and low-density lipoprotein choles-
and synbiotics. Thus, the influence on the intestinal micro- terol (LDL) decreased compared with the control group after
biota through feeding becomes attractive in the fight the consumption of probiotic fermented milk.
against obesity. Probiotics are “live microorganisms that, In other study, Madjd et al. (2016) selected 89 overweight
when administered in adequate amounts, confer a health and obese women who had the habit of eating traditional
benefit on the host” (Hill et al. 2014). The benefits can be low-fat (LF) yogurt and they were randomly asked to con-
either directly or through interactions with other microor- sume probiotic (PY) or LF yogurt. Compared to baseline, in
ganisms (John et al. 2018). It is important to stand out that both groups, there were lower anthropometric measures and
the potential beneficial effects are strain and not species positive changes in cardiometabolic risk characteristics.
specific. Prebiotics are generally carbohydrate-based, select- Compared to LF group, the PY group stood out by decreas-
ively fermented by the host’s microbiota, and are noted for ing TC, LDL-cholesterol, 2-h postprandial glycemia, homeo-
their beneficial effect on health (Gibson et al. 2017). The stasis model assessment of insulin resistance (HOMA-IR),
synbiotic term is used when combining probiotic strains and fasting insulin concentration. Hence, in an intervention
and prebiotic substrates (John et al. 2018). In this sense, focused on weight loss, the ingestion of PY improved the
probiotic strains, prebiotics, and synbiotics are used in lipid profiles and insulin sensitivity.
food production or used as supplements to positively mod- Gomes et al. (2017) recruited overweight or obese women
ify the host’s microbiota, and thus possibly aid the clinical who were divided into 2 groups: one who received a pro-
treatment against obesity and malnutrition (Dror et al. biotic mix (PM - Lactobacillus acidophilus and casei;
2017). The gut microbiota of obese individuals and the Lactococcus lactis; B. bifidum and B. lactis; 2  1010 CFU/
impact of probiotics and prebiotics on the hosts are sum- day) and the other received placebo. Both groups received a
marized in Figure 1. dietary prescription. The PM group had a greater reduction
This review focused on highlighting the current state of in waist circumference, waist-height ratio, plasma polyunsat-
knowledge on the potential role of prebiotics, probiotics, urated fatty acids, and increased glutathione peroxidase
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 3

Figure 1. The impact of probiotics, prebiotics, synbiotics and obesity on host’s gut microbiota. AMPs -Antimicrobial proteins, DC - Dendritic cell, GLP-1 - Glucagon-
like peptide-1, GLP-2 - Glucagon-like peptide-2, IFNc - Interferon gamma, IL-10 - Interleukin – 10, IL-17 - Interleukin - 17, LPS - Lipopolysaccharide, M cells -
Microfold cells, PYY - Peptide YY, SCFAs - Short-chain fatty acids, sIgA - Secretory Ig A, TGF-b, Transforming growth factor b, Th1 - T helper 1 cells, Th17 - T helper
17 cells, and ZO-1 - Zonula occludens-1.

activity. In this sense, the probiotic supplementation had a by Nikbakht et al. (2018). None of the groups showed posi-
positive effect when compared to the dietary prescrip- tive results; however, when FBG is highly unbalanced
tion alone. (7 mmol/L), the use of probiotics has proved to be effect-
The effect of probiotic and synbiotic consumption on ive, and that multispecies probiotics may have more impact
fasting blood glucose (FBG) levels in adults was evaluated on FBG than single species.
4 T. F. DA SILVA ET AL.

In that same year, the potential probiotic benefits were lipid levels in overweight or obese subjects. Their search
published; Borgeraas et al. (2018) recruited 957 subjects, resulted in 6162 articles; however, only 12 articles met the
either overweight or obese, and a high reduction in body inclusion criteria for meta-analysis. These articles included a
weight, BMI, and fat percentage was observed in comparison total of 767 samples, with 391 treatments and 376 controls.
with the placebo group; however, the effect level was low. The results showed that obese or overweight participants
Its effect was not significant at the fat mass level either. In receiving probiotic supplementation had significantly larger
this sense, the effects of probiotic supplementation on reductions in TC and LDL-cholesterol concentrations than
anthropometric measurements should be further explored in the control subjects. Even though the mode of action
long-term research. has not yet been understood, it has been postulated that
The positive modification of the intestinal microbiota probiotics can help the elimination of cholesterol and bile
through the intake of prebiotics has been reported by acid by changes in cholesterol esters pathways and lipopro-
Kellow, Coughlan, and Reid (2014), who evaluated the tein transporters, without affecting the synthesis of hepatic
impact of a prebiotic supplementation through 26 random- cholesterol.
ized clinical trials involving 831 participants with metabolic A systematic review and meta-analysis of clinical trials of
abnormalities. The authors found evidence supporting synbiotic supplementation in obesity treatment was recently
a dietary intake of prebiotics for appetite regulation and published by Hadi et al. (2020). In the study, it was identi-
reduction of circulating postprandial glucose and insulin fied 12057 articles, 7992 were screened, 50 full-text articles
concentrations; however, there is insufficient evidence at were accessed, and 23 articles were included in meta-
present to recommend dietary prebiotics for reducing total analysis. The data analyses revealed that symbiotic supple-
energy intake, body weight, peptide YY, glucagon-like mentation might have modest effects on body weight and
peptide-1 (GLP-1) concentrations, gastric emptying times, waist circumference through modulation of gut microbiota
insulin sensitivity, lipids, inflammatory markers, and composition.
immune function. Such contradiction may be because the Additional studies related to the impact of probiotic,
research was short-term (3-8 weeks). Therefore, longer-term prebiotic, or synbiotic on the human obese gut microbiota
(12-17 weeks), more participants, and larger-scale studies are and on clinical parameters of obesity are summarized
important and necessary to evaluate the modification of the in Table 1. Some studies using animal models are shown on
intestinal microbiota and its primary metabolic outcome. Table 2. According to the aforementioned studies, the food
The clinical study performed by Beserra et al. (2015) intake of probiotic and prebiotic for positive modification
demonstrated that a dietary intake of prebiotics decreased of the intestinal microbiome is a tool that stands out in
plasma cholesterol and LDL-cholesterol levels in the overall the fight against obesity and leads to reductions in
analysis, besides decreasing triglyceride levels and increased anthropometric measurements when compared to the pla-
HDL-cholesterol levels in diabetic research, while a synbiotic cebo. Although such findings are promising, more research
food intake decreased levels of insulin and triglycerides. is fundamental to establish the ideal formulation and to
The results support that a dietary intake of prebiotics and verify the benefits regarding the time of consumption, in
synbiotics can be considered a positive alternative regarding order to evaluate if obese and overweight patients would
complications related to obesity, such as dyslipidemia and benefit from intestinal microbiome modification, in addition
insulin resistance. to assessing the durability of these effects.
The food intake of probiotic and prebiotic impact on
serum lipid profile and insulin resistance in 84 participants
Effects of probiotics, prebiotics, and synbiotics on
with nonalcoholic fatty liver disease (NAFLD) was studied
satiety hormones regulation
as follows: (i) probiotic capsules (B. longum and L. acidoph-
ilus: 2  107 CFU/d) and placebo of prebiotic (maltodextrin The gut-brain axis is a complex network composed of
powder); (ii) sachet of prebiotic (inulin HP: 10 g/d) and pla- numerous systems, including the enteric nervous system
cebo of probiotic (fat- and lactose-free milk capsules); (iii) (ENS), the central nervous system, the autonomic nervous
probiotic and prebiotic; and (iv) placebo of probiotic and system, the sympathetic, and the parasympathetic branches,
prebiotic. Probiotic was able to decrease BMI and weight in the neuroendocrine and the immunological systems, which
all treatments in comparison to the placebo. The serum lev- are important for metabolic homeostasis (Bliss and
els of HDL- and LDL-cholesterol differed significantly in the Whiteside 2018). Several neurons and neurotransmitters,
probiotic (i) and the pro- and prebiotic (iii) groups when which are part of the enteric nervous system, are responsible
compared to the placebo group. Food intake of probiotic for the communication between brain and gut; they send
and prebiotic may be beneficial to enhance the serum qual- information to the brain through spinal and vagal afferent
ity of lipid profile and insulin resistance markers in NAFLD nerves (Rastelli, Knauf, and Cani 2018).
participants (Javadi et al. 2017). Changes in the communication between the gut and the
Several clinical studies dedicated to evaluating the effect brain axis are closely related to the difficulty in controlling
of probiotic supplementation on clinical parameters of obes- the appetite and to a disturbed perception of satiety (Lam
ity. In the study of Yan et al. (2019), a comprehensive et al. 2017). Food ingestion and body’s energy homeostasis
search across multiple databases was performed to identify are modulated by gut hormones released along the intestinal
studies that focused on the effects of probiotics on blood tract by enteroendocrine cells (EECs) and their concentrations
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 5

Table 1. Characteristics of human intervention studies with probiotics, prebiotics and synbiotics on microbiota.
Description Duration (weeks) Subjects Outcomes References
Prebiotic interventions
A double blind, placebo- 12 N ¼ 30, obese women. ITF treatment " Bifidobacterium and Dewulf et al. (2013)
controlled intervention study Faecalibacterium prausnitzii and #
with ITF prebiotics (inulin/ serum LPS levels. ITF also #
oligofructose 50/50 mix) or Bacteroides intestinalis, Bacteroides
placebo (maltodextrin), vulgatus and Propionibacterium, and
16 g/day. slight # in fat mass, plasma lactate
and phosphatidylcholine levels.
Randomized, double blind, 12 N ¼ 30, obese women. Population of Bifidobacterium longum, Salazar et al. (2015)
parallel, placebo-controlled Bifidobacterium pseudocatenulatum
trial, randomly assigned to and Bifidobacterium adolescentis " at
groups that received either the end of the treatment in the
16 g/day ITF or prebiotic group; B. longum
maltodextrin (placebo). negatively correlated with serum
LPS endotoxin.
Treatment: GOS, 15 g. 12 N ¼ 44, overweight or GOS " abundance of Bifidobacterium Canfora et al. (2017)
obese, prediabetic, species in feces by 5-fold. Microbial
BMI ¼ 28-40 kg/m2. richness or diversity in fecal samples
were not affected, nor was the
insulin sensitivity or related
substrate and energy metabolism in
overweight or obese
prediabetic consumers.
Treatments: i) oligofructose- 16 N ¼ 42, children, 7-12 OI treatment # BW z-score, # BF%, and Nicolucci et al. (2017)
enriched inulin (OI; 8 g/day); years old, overweight # percent trunk fat, # serum
ii) maltodextrin placebo or obese (> 85th triglycerides compared to placebo, #
(isocaloric dose). percentile of BMI) but IL-6, " Bifidobacterium spp. and #
otherwise healthy. Bacteroides vulgatus.
Treatments: i) 2 doses/day of 12 N ¼ 125, overweight and WP # BF. Hunger, desire to eat and Reimer et al. (2017)
prebiotic bar with Inulin-type obese, healthy, BMI > prospective food consumption were
fructans (ITF, 6 g oligofructose 25 kg/m2. all lower with all interventions
þ 2 g inulin); ii) protein bar compared to the control. ITF and
(5 g WP); iii) combination bar ITF þ WP " Bifidobacterium sp.
(8 g ITF þ 5 g WP).
Treatment: Oligofructose (8 g/day 36 N ¼ 14, NASH, NAS  5. Oligofructose supplementation Bomhof et al. (2018)
for 12 wks þ 16 g/day for improved liver steatosis and overall
24 wks). NAS score. Oligofructose "
Bifidobacterium and # Clostridium
cluster XI and I.
Treatments: HFM: (i) 24 g inulin 2 days of trial twice N ¼ 14, overweight to Inulin supplementation " fat oxidation Van Der Beek et al.
of which 0.5 g was U-13C- and 5 days of obese, male, " plasma free fatty acids in the EPP (2018)
inulin; (ii) 24 g washout period BMI ¼ 25–35 kg/m2. (0-3 h), # plasma glucose # Insulin
maltodextrin placebo. in between. and " SCFA.
A double-blind, placebo- 4 N ¼ 43, with high risk for Bread with barley beta glucans # total Velikonja et al.
controlled, randomized clinical metabolic syndrome plasma cholesterol, # waist (2019)
trial with bread containing 6 g development or with circumference, " propionic acid and
of barley beta glucans or diagnosed # acetic acid in control group. A
equal bread but without metabolic syndrome. significant # Clostridium leptum and
beta glucans Collinsella aerofaciens, and #
Agathobacter rectalis after BGB
intervention. Beta glucan changed
the composition of gut microbiota,
lowering the diversity and richness
of the microbial populations.
Probiotic interventions
Treatments: two groups received 12 N ¼ 87 subjects, Probiotic group # abdominal visceral Kadooka et al.
200 g/day of FM: (i) FM BMI ¼ 24.2-30.7 Kg/m2. and # subcutaneous fat areas, # BW (2010)
containing 5  1010 CFU/100 g and # BMI, and " serum adiponectin
L. gasseri SBT2055 LG2055: (ii) levels compared to placebo.
FM without LG2055.
Lactobacillus casei Shirota, 12 N ¼ 28 patients, Gut permeability of MetS patients was Leber et al. (2012)
6.5  109 CFU/dose. overweight and obese, increased significantly compared
MetS (13 patients/ with controls. L. casei Shirota
probiotic group and 15/ administration did not have any
control group) and 10 influence in the LBP and sCD14
healthy controls levels compared with
were included. healthy controls.
Capsules of Lactobacillus gasseri 12 N ¼ 62, obese, BMI > The administration of BNR17 in Jung et al. (2013)
BNR17, 1  1010 CFU/dose. 23 kg/m2. capsules reduced weight, waist, and
hip circumferences; however, there
were no significant differences
between the two groups.
Treatment: chewable tablets 4 N ¼ 24 Chinese subjects LcZ significantly altered the Zhang et al. (2014)
with 10.6 log CFU L. casei composition of intestinal microbiota
Zhang (LcZ) once daily and the gut microbiota diversity.
(continued)
6 T. F. DA SILVA ET AL.

Table 1. Continued.
Description Duration (weeks) Subjects Outcomes References
LcZ " Prevotella, Lactobacillus,
Faecalibacterium, Propionibacterium,
Bifidobacterium and an unidentified
genus from Bacteroidaceae and
Lachnospiraceae, and # Clostridium,
Phascolarctobacterium, Serratia,
Enterococcus, Shigella
and Shewanella.
Probiotic intervention: 200 g/day 8 N ¼ 75, overweight and Probiotic intervention # CRP, # TNF-a Zarrati et al. (2014)
of yogurt containing L. obese individuals with/ and # IL-17 when compared to
acidophilus La5, B. lactis Bb12, without WLD. baseline in treated group.
and L. casei DN001 (1  107 Probiotic þ WLD resulted in
CFU/mL). differences in thymus-specific
isoform of the retinoic acid
receptor-related orphan receptor
gamma (ROR-ct) gene expression, in
peripheral blood mononuclear cells.
Probiotic þ WLD had synergistic
impacts on immune inflammatory
markers in obesity.
Treatments 200 g/day of FM: i) 12 N ¼ 210, healthy adults Probiotic FM # abdominal visceral fat Kadooka et al. (2013)
106 CFU/100 g L. gasseri with large visceral fat areas, # BMI, # waist and hip
SBT2055 (LG2055); ii) 107 areas (80.2-187.8 cm2). circumferences and # BFM.
CFU/100 g L. gasseri
SBT2055(LG2055); iii) FM no
probiotic (control).
Treatments: i) hypocaloric diet þ 3 N ¼ 40, obese, MetS. Probiotic cheese # plasma triglycerides, Sharafedtinov
50 g/day of probiotic cheese arterial BP and BMI and risk of et al. (2013)
containing Lactobacillus metabolic syndrome.
plantarum TENSIA (DSM
21380); ii) hypocaloric diet þ
50 g/day of probiotic-
free cheese.
Treatment: Bifidobacterium breve 12 N ¼ 44, BMI ¼ 24-30 kg/m2, Probiotic # BFM and improved blood Minami et al. (2015)
B-3, 50  109 CFU/dose. overweight, healthy. parameters related to liver functions
and inflammation, such as
c-glutamyltranspeptidase and high-
sensitivity CRP.
Treatments: i) Probiotic powder 12 N ¼ 41, BMI ¼ 25-30 kg/m2, Fasting plasma glucose, HbA1c, fasting Higashikawa
with heat-killed Pediococcus overweight, healthy. insulin, HOMA-IR and serum lipid et al. (2016)
pentosaceus LP28, 100  109 levels did not change by intaking
CFU/dose; ii) alive Pediococcus neither living LP28 nor heat-
pentosaceus LP28, 100  109 killed LP28.
CFU/dose.
Treatments: i) high-dose 12 N ¼ 81, obese, The high-dose probiotic # metabolic ska et al. (2018)
Szulin
probiotic (1  1010 CFU/twice postmenopausal female. endotoxemia, # LPS, # HOMA-IR,
a day); ii) low-dose probiotic and improved insulin resistance
(2.5  109 CFU/twice a day); index in the obese
iii) placebo. Probiotic postmenopausal women.
intervention: B. bifidum W23,
B. lactis W51, B. lactis W52, L.
acidophilus W37, L. brevis
W63, L. casei W56, L. salivarius
W24, Lactococcus lactis W19,
and Lc. lactis W58.1.
A randomized, double-blind, 8 N ¼ 44 (22 men and 22 Probiotic yogurt # blood glucose level Rezazadeh et al. (2019)
placebo-controlled study with women), MetS patients, and VCAM-1. Probiotic yogurt
300 g/d of probiotic yogurt 20 to 65 years. promoted significant changes in
containing Lactobacillus plasminogen activator inhibitor 1,
bulgaricus, Streptococcus VCAM-1, insulin, HOMA-IR, and
thermophilus, 6.45  106 CFU/ insulin sensitivity were observed
g L. acidophilus La5 and after intervention compared
4.94  106 CFU/g B. lactis with baseline.
Bb12 or regular
yogurt (placebo).
A randomized, crossover, 12 N ¼ 53 adults with MetS. L. reuteri V3401 # IL-6 and VCAM-1 Tenorio-Jimenez
placebo-controlled, single- and " Verrucomicrobia phylum and et al. (2019)
center trial with a capsule Akkermansia genus.
containing either the
probiotic L. reuteri V3401
(5  109 CFU) or a placebo.
(continued)
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 7

Table 1. Continued.
Description Duration (weeks) Subjects Outcomes References
Synbiotic interventions
Dietary supplement containing 13 N ¼ 40, obese. Phyto-supplement # BW, # BFM, and # Doria et al. (2013)
phloridzin, isoflavones, waist, thigh and buttock
Lactobacillus casei, circumference compared to the
Lactobacillus acidophilus, control group.
7.5  108 CFU/dose.
Treatment: 2 capsules/day of LPR 12 N ¼ 125, BMI ¼ 29-41 kg/ Lactobacillus rhamnosus CGMCC1.3724 Sanchez et al. (2013)
(10 mg L. rhamnosus m2, overweight and formulation helps obese women to
CGMCC1.3724 providing obese, healthy. achieve sustainable weight loss.
1.62  108 CFU, 300 mg of
oligofructose þ inulin mixture
[70:30, v/v] and 3 mg of
magnesium stearate).
Treatments: i) probiotics (5  109 8 N ¼ 50, female, BMI > Probiotics and BTS, or BTS alone did Lee et al. (2014)
CFU S. thermophilus KCTC 25 kg/m2 and waist not affect metabolic endotoxemia
11870BP, L. plantarum KCTC circumference > 85 cm. and intestinal permeability
10782BP, L. acidophilus KCTC compared to baseline. B. breve in
11906BP, L. rhamnosus KCTC obese patients # endotoxin
12202BP, B. lactis KCTC concentrations, weight, BMI, fat
11904BP, B. longum KCTC mass, " Firmicutes phylum and S.
12200BP, and B. breve KCTC thermophilus, and # intestinal
12201BP) þ herbal medicine permeability.
BTS; ii) BTS þ placebo.
Treatments: (i) probiotic group 12 N ¼ 120, nondiabetic Probiotic group # BW, # BF%, # BFM, # Jung et al. (2015)
consumed 2 g probiotic and overweight. ox-LDL, # Lp-PLA2 and " LDL
powder twice a day particle size.
containing 0.1 g of L. curvatus
HY7601, 0.1 g of L. plantarum
KY1032 (each at 2.5  109
CFU), 1.24 g of crystalline
cellulose, 0.5 g of lactose, and
0.06 g of blueberry-flavoring
agent; (ii) placebo group
consumed the same amount
of powder without
any probiotics.
Treatments: i) Microcrystalline 24 N ¼ 225, BMI ¼ 28-34.9 kg/ LU þ B420 and B420 $ weight. Stenman et al. (2016)
cellulose (MC), 12 g/day m2, overweight and LU þ B420 # BFM, LU alone and
(control); ii) LU, 12 g/day; iii) obese, healthy. B420 alone $ BFM. B420 and
B. animalis ssp. lactis 420 LU þ B420 # energy intake
(B420), 1  1010 CFU/ compared to placebo. LU þ B420 #
dose þ MC; B. animalis ssp. blood zonulin levels and # hsCRP
lactis 420, 1  1010 were associated with corresponding
CFU/dose þ LU. # trunk fat mass. Probiotic
treatment with or without dietary
fiber $ BFM. B420 and LU þ B420
#waist circumference and # food
intake; LU alone had no effect on
the measured outcomes.
Treatments: (i) synbiotic group 9 N ¼ 70, non-obese Synbiotic # FBG # HbA1c, $ Ebrahimi et al. (2017)
(500 mg/day) ¼ probiotics patients with T2D. lipid profiles.
(Lactobacillus sp and
Bifidobacterium sp,
Streptococcus thermophilus),
prebiotics (FOS) and B group
vitamins (1 mg), lactose
(0.5 mg), maltodextrin,
magnesium saturate; (ii)
placebo group consumed the
same amount of product
without any probiotics
or prebiotics.
Treatment: i) symbiotic capsule 12 N ¼ 20, male and female Synbiotic treatment $ body mass, Sergeev et al. (2020).
with 69 mg or 15  109 CFU overweight/obese, BMI, body fat mass, body fat
of Lactobacillus acidophilus BMI ¼ 33.5 kg/m2. percentage, body lean mass, and
DDS-1, Bifidobacterium lactis bone mineral content. Synbiotic
UABla-12, Bifidobacterium supplementation " abundance and
longum UABl-14, and richness gut microbiota associated
Bifidobacterium bifidum UABb- with positive health effects,
10 þ prebiotic component at especially Bifidobacterium and
a dose of 5.5 g/d (2.75 g GOS Lactobacillus. Bacteroides was the
and the remainder simple most abundant genus in both
(continued)
8 T. F. DA SILVA ET AL.

Table 1. Continued.
Description Duration (weeks) Subjects Outcomes References
sugars); ii) placebo groups. Prevotella and Gardnerella
supplement was similar in genera were significantly decreased
appearance and energy after the synbiotic intervention.
content as the
synbiotic supplement.
The strains were not informed. AA – arachidonic acid; BF% – body fat percentage; BFM – body fat mass; BMI – body mass index; BP – blood pressure; BTS –
Bofu-tsusho-san; BW – body weight; CFU – colony forming units; CRP – C-reactive protein; DHA – docosahexaenoic acid; EPP – early postprandial phase; FBG –
fasting blood glucose; FM – fermented milk; FOS – fructooligosaccharide; GOS – galactooligosaccharide; HbA1c – glycated hemoglobin; HFM – high-fat
milkshake; HOMA-IR – homeostatic model assessment for insulin resistance; hsCRP – high-sensitivity C-reactive protein; IL – interleukin; ITF – Inulin-type
fructans; LBP – lipopolysaccharide-binding protein; LC-PUFA – long chain poly unsaturated fatty acids; LDL – low-density lipoprotein Lp-PLA2 – lipoprotein-
associated; LPS – lipopolysaccharide; MC – Microcrystalline cellulose; MetS – metabolic syndrome; ox-LDL – oxidized low-density lipoprotein; LU – Litesse
Ultra Polydextrose; NASH – non-alcoholic steatohepatitis; NAS – non-alcoholic fatty liver activity score; PBMCs – peripheral blood mononuclear cells; sCD14 –
soluble CD14; SCFA – short-chain fatty acids, and T2D – type 2 Diabetes; TNF-a – tumor necrosis factor – alfa; VCAM-1 - vascular cell adhesion molecule cell;
WLD – weight-loss diet; WP – whey protein.

vary after each meal. Among these hormones, there is ghrelin, 2013). Chen et al. (2018) reported that the probiotic L. mali
which stimulates appetite, and leptin, glucagon-like peptide APS1 (5  108 CFU in saline through daily gavage) increased
one and two (GLP-1 and 2), peptide YY (PYY), cholecysto- the excretion of SCFAs and concomitantly increased the
kinin (CCK), and oxyntomodulin (OXM), which regulate expression of PYY and PP in mice receiving a HDF
food ingestion and control body weight (Delgado and for 6 weeks.
Tamashiro 2018). Besides stimulating the production of hormones through
A large number of studies have been showing that gut SCFAs production, some bacteria can secret some neuro-
microbiota can affect behavior and core brain mechanisms, chemicals such as ghrelin, leptin, PYY, and neuropeptide Y,
including the production and release of hormones involved which may have an effect on the vagus nerve activity and,
in regulating appetite and satiety (Bliss and Whiteside 2018; consequently, the host’s eating pattern (Nie, Luo, and Lin
Cani and Delzenne 2009). For such reasons, shifting intes- 2018). In this context, the search for probiotic strains with
tinal microbiota composition into a more favorable profile this characteristic is desirable considering that they could be
regarding the regulation of such hormones is an alternative used in functional food specifically for the control of
for the control of obesity, appetite, and their related meta- weight gain.
bolic comorbidities (Falcinelli et al. 2018). In this context, Regarding human trials, the consumption of a synbiotic
probiotics and prebiotics can be used successfully for this capsule (Protexin Balance, Probiotics International Limited,
purpose. Even though other benefits coming from the con- Somerset, United Kingdom), containing the following pro-
sumption of such products have been extensively explored, biotic strains: L. casei PXN 37, L. rhamnosus PXN 54, L.
the therapeutic potential of prebiotic and probiotic con- acidophilus PXN 35, L. bulgaricus PXN 35, S. thermophilus
sumption on food intake regulation has started to gain PXN 66, B. breve PXN 25, and B. longum PXN 30, at
attention in the last two decades. 2  108 CFU and 125 mg of fructooligosaccharides (FOS)
The major mechanism of action by which probiotics and significantly increased GLP-1 and PYY amounts in patients
prebiotics exert control on appetite involves the production with a metabolic syndrome (BMI  25 kg/m2) (Rabiei
of SCFAs (Dao and Clement 2018; Torres-Fuentes et al. et al. 2018).
2017). Some probiotic strains have the capacity to produce Although there is evidence that shows the effect of
SCFAs (Chen et al. 2018) whilst prebiotics can be converted probiotic bacteria and prebiotics on gut hormone secretion,
into SCFAs by the gut microbiota. The main SCFAs pro- further studies linking the probiotic and prebiotic consump-
duced through bacterial fermentation are acetate, propion- tion with appetite are desirable. In this line, Forssten et al.
ate, and butyrate, which will vary according to the (2013) detected a trend for reduced food consumption
carbohydrate type, microbiota diversity, composition, activ- among mice groups receiving fermented milk (2.5 mL/day)
ity, and gut transit time (Byrne et al. 2015). through a single oral gavage with the probiotic strains
Some early in vitro studies showed that SCFAs have cru- L. rhamnosus MUH 142 and L. bulgaricus MUH 192;
cial functions in the gut-brain axis in the host, leading to such tendency was attributed to the high levels of hippuric
the stimulation of the secretion of satiety hormones (Karaki acid in the probiotic fermented milk when compared to the
et al. 2008; Tolhurst et al. 2012). These findings encouraged treatment without any probiotic strains.
further studies with animal and human subjects, even Another trial carried out by Sanchez et al. (2017) with
though at the present moment data from animal studies are obese subjects demonstrated that a symbiotic capsule (con-
more abundant compared to human trials, which makes it taining 1.62  108 CFU of L. rhamnosus CGMCC1.3724,
difficult to establish a definitive explanation of the mecha- 210 mg of oligofructose, and 90 mg of inulin) administered
nisms through which SCFAs control appetite. for 12 weeks positively affected the appetite sensation and
The administration of a probiotic mixture VSL#3 (5 mg/ eating patterns, as it contributed to an increased sense of
kg body weight) to mice receiving a high-fat diet (HFD) for satiety in women. These outcomes support the assumption
8 weeks increased the GLP-1 secretion and has been related that appetite regulation and associated behaviors in the
with the increase in production of butyrate (Yadav et al. obesity treatment can be influenced by the gut-brain axis.
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 9

Table 2. Characteristics of animal intervention studies with probiotics, prebiotics and synbiotics on microbiota.
Description Duration (weeks) Subjects Outcomes References
Prebiotic interventions
Treatments: i) standard diet 14 N ¼ 8 per group, wild-type HFD resulted in " Gram-negative gut Cani et al. (2007b)
(control); ii) HFD containing mice, Murine model. microbes and # Gram-positive
49.5% fat (g/100 g of total dry bacteria (Eubacterium rectal,
diet) corresponding to 72% of Clostridium coccoides group and
the total energy content; iii) a Bifidobacterium). The LPS receptor
mix of HFD þ OFS; protein CD14 system was driving
iv) HFD þ NFDF. the metabolic effects of the HFD in
the wild-type mice.
Treatments: i) control diet; ii) 5 N ¼ 10/group, ob/ob mice Treated animals showed # plasma LPS Cani et al. (2009a)
prebiotic oligofructose and iii) (C57BL/6 background; and cytokines, and # hepatic
control Jackson Laboratory, Bar expression of inflammatory and
(microcrystalline cellulose). Harbor, Maine, USA). oxidative stress markers. # intestinal
permeability and " tight-junction
integrity compared to controls and
" endocannabinoid signaling.
Treatments: i) HFD (60% fat and 8 N ¼ 40; 10-week-old C57BL/ Prebiotic treatment " Reg3g expression Everard et al. (2014)
20% carbohydrates), PRE 6J mice. (by 50-fold), " intectin and
(oligofructose, 0.3 g per improved intestinal homeostasis.
mouse/day), ii) HFD þ PRE, The mice microbiome was greatly
and iii) CT groups. dominated by the phyla Firmicutes
and Bacteroidetes. HFD treatment
profoundly affected the caecal
Firmicutes/Bacteroidetes ratio, and
the gut microbiota modulations
induced by prebiotics counteracted
HFD-induced inflammation.
Treatments: i) HFS ad libitum; ii) 10 N ¼ 104; 12-week-old virgin The prebiotic intake during pregnancy Paul, Bomhof, Vogel,
HFS þ 10% wt/wt OFS ad female Sprague-Dawley and lactation improves maternal and Reimer (2016)
libitum; iii) weight matched to rats. Rats (N ¼ 90) were metabolism in diet-induced obese
the OFS group with WM; iv) fed a high-fat/sucrose diet rats (HFS) (# maternal energy
lean reference group was (Diet #102412, Dyets, Inc) intake, # gestational weight gain),
maintained on control AIN- ad libitum for 10 weeks to and prevents increased adiposity in
93 diet. induce obesity. The 42 rats dams and their offspring.
with the greatest weight
gain were allocated to one
of three groups
throughout gestation
and lactation.
Treatments: i) LFD; ii) HFD; iii) 22.5 N ¼ 15; C57BL/6J male mice. Polymannuronic acid # diet-induced Liu, Wang, Shi, Wang,
HFD þ 150 mg BW gain and blood TAG levels (P < Xue, and
polymannuronic acid/kg 005) and " glucose tolerance. PM # Tang (2017)
body weight. LPS in blood and ameliorated local
inflammation in the colon and the
epididymal adipose tissue. PM also
had a profound impact on the
microbial composition in the gut
microbiome and resulted in a
distinct microbiome structure. PM "
abundance of probiotic
Lactobacillus reuteri.
Probiotic interventions
Probiotic treatment: B. 7 N ¼ 4 groups (n  6 mice The probiotic CECT 7765 " insulin Cano, Santacruz, Trejo,
pseudocatenulatum per group), male wild-type resistance and glucose tolerance, IL- and Sanz (2013)
CECT 7765. C57BL-6 mice. HFD B6 4 levels, # serum concentrations of
obese mice. leptin, IL-6, MCP-1 and recovered
the macrophage and dendritic cell
functions, cytokine secretion and
ability to induce T-cell activation
and proliferation.
Probiotic treatment: 1  108 13 N ¼ 4 groups (7 or 8 mice Probiotic group # weight gain, " Kim, Park, Kim, Kim,
CFU/day of L. rhamnosus per group), male wild-type insulin sensitivity and " serum and Hyun, (2013)
GG (LGG). C57BL/6J mice. B6 adiponectin, which promotes an
HFD mice. anti-inflammatory state in
macrophage in adipose tissues.
Probiotic treatment: L. reuteri 12 N ¼ 10-30 mice per group Probiotic treated animals presented " Poutahidis et al. (2013)
ATCC 6475. (each experiment included serum anti-inflammatory IL-10. The
5-15 animals per group IL-10-deficient mice fed with
with two replications). Western diet and probiotic became
Western fast food-diet obese and failed to respond for
Swiss mice. probiotic treatment. Slim mice fed
with Western diet þ L. reuteri ATCC
(continued)
10 T. F. DA SILVA ET AL.

Table 2. Continued.
Description Duration (weeks) Subjects Outcomes References
6475 had " Tregs and # IL-17A in
lymphoid tissues. Probiotic benefits
depend on interactions with
immune cells in the gut mucosa
through Tregs cell and oral
tolerance induction.
Probiotic treatment: 1  108 12 N ¼ 4 groups (n ¼ 10 mice Probiotic ameliorates glucose Toral et al. (2014)
CFU/day of L. per group), male C57BL/6J metabolism: # glycaemia, insulin
coryniformis CECT5711. mice. Obesity induced by resistance, LPS plasma levels, TNF-a,
HFD in B6 mice. IL-10, JNK, and MCP-1 genes
expression in the liver, and " mRNA
of the anti-inflammatory
adiponectin in adipose tissue.
Treatment with L. rhamnosus 7 N ¼ 4 groups (5-15 mice per Probiotic administration # F4/80þ Alard et al. (2016)
LMG S-28148 (1  109 CFU) group), male C57BL/6J macrophage accumulation, and
and B. animalis subsp. lactis obese mice. MCP-1 expression. In treated obese
LMG P-28149 (1  109 CFU). mice # TNF-a, IL-1a, IL-6, and IL-17
The animals received: i) gene expression.
LFD þ probiotic; ii) only LFD;
iii) HFD þ probiotic; iv)
only HFD.
Mice were fed a HFD for 6 3 N ¼ 16, 7-week-old male Normal diet þ Lactobacillus mali APS1 Chen et al. (2018)
weeks, followed by C57BL/6 J mice. " BW loss and # caloric intake, and
treatments: i) NDS; ii) normal # fat accumulation in obese mice.
diet þ Lactobacillus mali APS1 Lactobacillus mali APS1 ameliorated
(5  108 CFU/day) (NDAPS1). hepatic steatosis in preexisting
obese mice after dieting,
significantly " abundance of
Lactobacillus mali and # abundance
of the Streptococcaceae family and
Anaerotruncus genus; it also
manipulated the gut microbiome’s
obesity-associated metabolites,
followed by regulation of lipid
metabolism, enhancement of energy
expenditure and inhibition
of appetite.
Rats were subdivided into 4 12 N ¼ 12, obese rats. The probiotic supplementation in Wanchai et al. (2018).
groups: i) normal diet; ii) group iv # metabolic endotoxemia,
normal diet plus probiotics (L. compared to group iii. The probiotic
paracasei HII01 - 1  108 CFU/ administration # NF-jB
mL); iii) high-fat diet, and; iv) phosphorylation, # JNK protein, and
high-fat diet plus probiotics # kidney inflammation. The
(L. paracasei HII01 - 1  108 probiotic administration $ COX-2
CFU/mL). levels in renal tissue, # TNF-a, # IL-
1b, # IL-6 and # MCP-1.
Synbiotic interventions
Treatments: i) control, ii) 10% 8 N ¼ 10 rats per group, male Oligofructose " fasting portal plasma Bomhof, Saha, Reid,
oligofructose, iii) Sprague-Dawley rats. Diet- GLP-1, " PYY, while probiotic " GLP- Paul, and
Bifidobacterium Bb-12, and iv) induced obese. 2 and ghrelin levels. Reimer (2014)
oligofructose þ Bb-12.
Treatments: i) AIN-93G 8 N ¼ 48, high-fat WSD. Early life synbiotics protected mice Mischke et al. (2018)
diet þ prebiotics (scGOS/ against WSD-induced excessive fat
lcFOS); ii) AIN-93G accumulation throughout life. Adult
diet þ synbiotics (scGOS/lcFOS insulin sensitivity and dyslipidaemia
with 109 CFU/g were improved and most
Bifidobacterium breve M-16 V); pronounced changes in gene
iii) AIN-93G diet þ a non- expression were observed in the
active control component ileum. In early life and adulthood,
(REF) and AIN-93G diet þ a there were changes in faecal
non-active control component microbiota composition, and "
(CTRL): 2% w/w maltodextrin, abundance of Bifidobacterium. The
until post-natal day 42. synbiotics-modified microbiota at
Thereafter CT, PRE, and SYN PN42 is not enough to transfer
animals received a WSD (40% long-lasting protection of metabolic
energy from fat) as a health status when transplanted in
challenge until sacrifice germ-free recipients.
on PN98.
AIN-93G diet –American Institute of Nutrition dietary standard 93; BW – body weight; COX-2 – cyclooxygenase-2; CFU – colony forming units; CTRL – control
component; F4/80þ macrophage – macrophage specific genes F4/80þ; GLP-1 – glucagon-like peptide-1; GLP-2 – glucagon-like peptide-2; HFD – high-fat diet;
HFS – high-fat-sucrose diet; IL – interleukin; JNK – c-Jun N-terminal kinase; LFD – low-fat and low-sucrose diet; LPS – lipopolysaccharide; MCP-1 – monocyte
chemotactic protein-1; mRNA – messenger ribonucleic acid; MSG – monosodium glutamate; NDS – normal diet þ saline; NF-jB – nuclear factor kappa-B; NFDF
– non-fermentable dietary fiber; OFS – oligofructose; PN42 – post-natal day 42; PN98 – post-natal day 98; PRE – prebiotic supplementation; Reg3g – regenerat-
ing islet-derived 3-gamma; PYY – peptide YY; SYN – synbiotic supplementation; TAG – triacylglycerol; TNF-a – tumor necrosis factor – alfa; Tregs cell – regula-
tory T cells; WM – limited oligofructose þ fat þ sucrose diet; WSD – Western-style diet.
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 11

The above-mentioned studies successfully proved that Besides the effect of SCFAs on hormone regulation, as
probiotics and prebiotics influence on satiety hormones and previously described, butyrate in particular has exceptional
food ingestion. Nevertheless, the results concerning their ability regarding immunoregulatory effects, such as leuko-
effects on these parameters are still discrepant. For instance, cytes activity, by recruiting them to inflammation sites, and
Jones et al. (2018) reported no significant shift in gut hor- the secretion of TNF, IL-2, IL-6, IL-10, eicosanoids, and spe-
mones as a consequence of receiving VSL#3 probiotic (three cific chemokines. Moreover, SCFAs are recognized for their
packets/day) in a double-blind, randomized placebo-con- ability to suppress the progression of metabolic diseases
trolled trial conducted with 19 obese Latin adolescents. (Vinolo et al. 2011).
Another randomized placebo-controlled trial with over- Studies performed with cell lines, by using adipocytes or
weight or obese individuals (n ¼ 20) aimed at determining macrophages, both in HFD models and in human clinical
the effects of the daily consumption of a prebiotic powdered trials, reported that the administration of probiotics pro-
ingredient mixture for 4 weeks (4 g inulin, blueberry extract, motes an anti-inflammatory state, decreasing the leptin/
and 2.5 g oat b-glucan) or a placebo product. Glucose, insu- resistin secretion, increasing M2 macrophages in adipose tis-
lin, glycosylated hemoglobin, HOMA-IR, triglycerides, and sue and anti-inflammatory molecules, such as IL-10 and adi-
cholesterol (TC, LDL and HDL), SCFAs produced by gut ponectin (Kobyliak et al. 2016).
microbiota, plasma satiety hormones (ghrelin and PYY), and In the last decade, there has been significant increase in
satiety perception were assessed. There was an increased studies evaluating the interaction between foods (including
sense of satiety in the group that received the prebiotic mix- probiotics and prebiotics), microbes, and their metabolites
ture; however, there was no significant difference in the with the mucosal and systemic immune systems (Clemente,
plasma satiety hormones (PYY and ghrelin) concentration Manasson, and Scher 2018; Han and Xiao 2020), by using
between both groups (Rebello et al. 2015). human or animal models.
Further research using human subjects is essential to An important randomized clinical trial investigated the
thoroughly explain the molecular mechanisms through effects of prebiotics on 42 overweight or obese children.
which SCFAs produce these effects and to make it feasible They received oligofructose-enriched inulin or maltodextrin
to use these compounds in the clinical management of obes- placebo. The group that received prebiotics experienced an
ity (Byrne et al. 2015). Furthermore, it is noteworthy that increase in Bifidobacterium species and a decrease in CRP
and IL-6 inflammatory cytokine, in comparison to the pla-
the effect of prebiotics and probiotics on appetite regulation
cebo group. However, there were no differences in LPS lev-
will depend on intrinsic characteristics of the hosts, includ-
els between the two evaluated groups (Nicolucci et al. 2017).
ing genetics, physiology, and lifestyle, as well as on prebiotic
Another randomized clinical trial assessed the effect of
and probiotic type and dose.
administering probiotics with prebiotics (synbiotics) in 225
participants. The synbiotics preparation combined B. ani-
Probiotics, prebiotics, and synbiotics on malis subsp. lactis 420 (109 CFU/per day) with ultra-poly-
inflammatory markers dextrose, an undigested glucose polymer that favors the
Bifidobacteria proliferation in culture systems (Probert et al.
The association between intestinal dysbiosis and obesity 2004). In contrast, another randomized controlled clinical
could be explained by the metabolic endotoxemia propos- trial using synbiotics to treat overweight men and women
ition which connects alterations in the gut microbiota to observed no significant differences in inflammatory markers,
systemic inflammation and metabolic dysfunctions (Torres such as serum levels of CRP, LPS-binding protein (LBP),
et al. 2018; Sroka-Oleksiak et al. 2020). In animal and and adiponectin. The intervention with two capsules/day
human models, obesity and metabolic dysfunctions have containing 10 mg of a L. rhamnosus CGMCC1.3724
been correlated to an increased barrier permeability that (1.62  108 CFU) and 300 mg oligofructose and inulin
induces the bacterial translocation of endotoxins, particularly resulted in weight loss, besides decreased fat mass and leptin
of the Gram-negative bacterial membrane component, the levels in obese women (Sanchez et al. 2014). Furthermore, a
lipopolysaccharides (LPS), which activates innate immune significant decrease in TNF and IL-6 was observed. B. lactis
receptors and promote metabolic endotoxemia (Torres et al. HN019 demonstrated potential to reduce obesity, minimiz-
2018). Additionally, LPS has been identified as a trigger for ing cardiovascular risks in patients with MetS (Bernini
insulin resistance, because it affects the secretion of proin- et al. 2018).
flammatory cytokines. In response to the LPS, the activated Recently, Depommier et al. (2019) conducted a random-
toll-like receptor-4 (TLR-4) induces the inflammatory tran- ized, double-blinded, placebo-controlled clinical trial to
scription nuclear factor kappa B (NF-jB) in gastrointestinal evaluate the effect of Akkermansia muciniphila supplementa-
epithelial cells. The induced NF-jB can trigger a systemic tion in 32 overweight, obese, and insulin resistant patients.
inflammation, classical activation of M1 macrophages, and Individuals were enrolled to receive daily oral administration
infiltration of the visceral adipose tissue, thus contributing of placebo, alive A. muciniphila or pasteurized A. mucini-
to an inflammatory state, with increased TNF-a, interleukin phila (1010 CFU/per day), for three months. The supplemen-
(IL)1b, IL-6, leptin, resistin, and C-reactive protein (CRP), tation was considered safe and well tolerated. Compared to
and subsequent resistance against insulin (Neyrinck et al. control group, pasteurized A. muciniphila reduced the levels
2016; Torres et al. 2018, Zhi et al. 2019). of relevant blood markers of hepatic and renal dysfunction
12 T. F. DA SILVA ET AL.

and inflammation (reduced LPS plasma levels, improved The intestinal microbiota represents a strategic tool for
insulin sensitivity, decreased insulinemia and plasma total reducing metabolic endotoxemia, which has been linked
cholesterol). Moreover, after three months pasteurized to inflammatory states during metabolic dysfunctions.
A. muciniphila improved metabolic parameters (reduced In obesity, the intestinal microbiota modulates energy
body weight, fat mass, and hip circumference) when regulation and the fermentation of dietary fibers not
compared with placebo group. digested by humans, with the production of SCFAs (Bailey
The probiotic L. gasseri SBT2055 (LG2055) have been and Holscher 2018).
associated to anti-obesity effects in obese C57BL/6 mice, The role of diet, prebiotics, and the intestinal microbiota
induced through their diet (B6 mice) during a 24-week in metabolic endotoxemia is under investigation. Research
treatment. Authors reported that LG2055 significantly concerning the different types of dietary habits on the gut
suppressed the inflammatory gene expression, including the microbiota, and their relationship with systemic inflamma-
monocyte chemoattractant protein-1 (MCP-1) and the C-C tion is needed in order to determine the real role of the
chemokine receptor type 2 (CCR2) in adipose tissues. They efficacy of diet interventions to suppress metabolic endotox-
hypothesized that the effects against obesity of L. gasseri emia, inflammation, and metabolic dysfunctions (Bailey and
were associated with the modulation of immune system and Holscher 2018).
anti-inflammatory responses (Miyoshi et al. 2014). Currently, studies have focused on the role of the intes-
Another study investigated the impacts of administering tinal microbiota in obesity, metabolic syndrome, and T2D.
L. rhamnosus, L. acidophilus, and B. bifidum to HFD Swiss However, further studies in animal models are required to
mice, for five weeks (Bagarolli et al. 2017). The HFD led to explore the metabolites-immune system-gut barrier axis in
leaky gut, LPS translocation, and systemic inflammation. By order to investigate how probiotic metabolites interact
administering probiotics to the obese animals, they could with the mucosal immune system, and the impact of this
recover the TLR-4 expression in muscle and liver to the interaction on intestinal permeability, which is involved
baseline, besides suppressing the TLR4 activation, c-Jun in metabolic endotoxemia and obesity. In addition, clinical
N-terminal kinase (JNK), and the insulin receptor phosphor- trials are still scarce, and the number of subjects, strains
ylation. In the same way, TNF and IL-6 expressions were combination, doses, and administration duration need to be
significantly decreased in the treated mice’s livers and better established.
muscles. The LPS levels in portal circulation, involved in
metabolic endotoxemia, were decreased in the probiotic
Effect of probiotics, prebiotics, and synbiotics on obese
group. Furthermore, molecules involved in bacterial
GI microbiota
translocation and gut inflammation, nucleotide-binding
oligomerization domain-containing protein 1 (NOD1), clus- The collective genomes of the human gut microorganisms
ter of differentiation 14 (CD14) decreased in the probiotic and their effect on both health and diseases have been the
group. Also, the probiotic administration decreased F4/80þ focus of considerable studies. However, a deep understand-
macrophage infiltration in adipose tissues and crown-like ing of the role of the gut microbiota in the beginning of dis-
structures, prevalent in obese animals. The zonulin (ZO-1) eases is still under investigation (Rastelli, Knauf, and Cani
and occludin proteins, involved in intestinal permeability 2018; Clemente, Manasson, and Scher 2018).
integrity, were partially reestablished in the treated obese Studies have shown which microbes are normally present
animals (Bagarolli et al. 2017). and how the relationship among them affects health and dis-
In HFD B6 mice, the administration of 5  109 CFU/day eases, besides their role in metabolism, immune system, and
of L. curvatus HY7601 and L. plantarum KY1032, resulted human health (Patterson et al. 2016; Hugon et al. 2017).
in weight loss and reduced fat mass, leptin, and insulin lev- Although most of the studies conducted on the human gut
els, and gene expression of inflammatory TNF, IL-6, IL-1b microbiota concentrated on bacterial diversity, other prokar-
and MCP-1 in adipose tissues. These probiotic strains were yotes (including archaea) and eukaryotic populations also
reported to decrease adipogenesis in 3T3-L1 cell lines (Park participate in other relationships in the gut ecosystem. This
et al. 2013). Another study in HFD B6 mice, through oral microbiota is not fully characterized due to technical restric-
gavage with 5  108 CFU/day of Bacteroides uniformis CECT tions (Hugon et al. 2017).
7771, promoted a decrease in their body weight, serum con- The GI microbiota is susceptible to adaptations and
centrations of glucose, insulin and leptin, and enhanced transformations, with important effects throughout the
their oral tolerance to glucose and immune responses, such human body, due to its ability to produce expressive bio-
as TNF secretion by macrophages and dendritic cells in active metabolites, such as exopolysaccharides, SCFAs, con-
response to LPS stimulation. Obesity interferes in effective jugated fatty acids, and neuroactive metabolites, such as
immune responses and the probiotic administration reestab- serotonin and c-aminobutyric acid (GABA), which are
lishes the capacity of dendritic cells to activate naïve T lym- transported via the circulatory system throughout the body
phocytes in obese mice (Gauffin-Cano et al. 2012). (Ross et al. 2010; Patterson et al. 2016). The GI microbiota
Metabolic endotoxemia, induced by bacterial translocation performs several critical roles in adults, such as ecology pro-
and increased LPS plasma levels, is frequently correlated to tection, metabolism of dietary components, immune system
inflammation in adipose and hepatic tissues (Cani et al. modulation, intestinal motility, cholesterol metabolism, and
2007a; Toral et al. 2014). enterohepatic cycling of bile acids (Ross et al. 2010; Foxx-
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 13

Orenstein and Chey 2012). Thus, the intestinal microbiota composition at the phylum level if compared to the genus
can affect the human health, though in particular situations; and species levels and may be associated with changes in
imbalanced changes in microbiota profile (dysbiosis) can metabolic functions (Bliss and Whiteside 2018).
happen and may pose the risk of developing diseases or syn- The genera Anaerostipes, Bacteroides, Campylobacter,
dromes, including obesity and its related comorbidities, Dialister, Parabacteroides, Porphyromonas, Ruminococcus,
which have been reported in both animals and humans (Al- and Staphylococcus were prevalent in individuals with the
Assal et al. 2018; Makki et al. 2018; Rastelli, Knauf, and obese phenotype and low bacterial richness. They are char-
Cani 2018). The gut microbiota is mostly composed of strict acterized by insulin resistance, more intense general adipos-
anaerobes, facultative anaerobes, and aerobes; its compos- ity and dyslipidemia, and a more significant inflammatory
ition varies widely among individuals because of a natural phenotype when compared with lean individuals, who pre-
selection at both microbial and host levels (Al-Assal et al. sent high bacterial richness. Additionally, species of
2018; Nie, Luo, and Lin 2018). Akkermansia, Alistipes, Bifidobacterium, Butyrivibrio,
Among Prokaryotes, most of the bacteria species har- Coprococcus, Faecalibacterium, Lactobacillus, and
vested from the gut belong to the phyla Firmicutes, Methanobrevibacter were more prevalent in individuals with
Bacteroidetes, Actinobacteria, and Proteobacteria; the dom- high bacterial richness and the lean phenotype.
inant species belong to the families Bacillaceae, Furthermore, the production of metabolites with possible
Enterobacteriaceae, Corynebacteriaceae, and Bacteroidaceae harmful effects on the host’s health (such as procarcinogens)
(Hugon et al. 2015). Genera from Archaea are also present, was significantly higher in low gene count participants, and
such as Methanobrevibacter (Ross et al. 2010). Nevertheless, including modules for b-glucuronide degradation, aromatic
when considered at the level of bacterial species, the vari- amino acids degradation, and dissimilatory nitrate reduction
ation in the composition of interindividual microbial com- (Le Chatelier et al. 2013; Al-Assal et al. 2018). Hence, the
munities is considerably greater than that observed at the metabolism of the intestinal microbiota and the metabolites
phylum level. Moreover, many studies have shown that sev-
produced are also a subject to be further studied.
eral species considered to dominate the healthy gut micro- There are evidences showing that the loss of biodiversity
biota belong to the genera Bacteroides, Bifidobacterium,
can be related to many diseases. People with intestinal dis-
Clostridium, Eubacterium, Fusobacterium, Lactobacillus,
eases, such as inflammatory bowel disorder (IBD), present a
Peptococcus, Peptostreptococcus, Ruminococcus, and
lower diversity of species in their intestines, and the propor-
Veillonella (Hugon et al. 2017).
tion of the different types of bacteria in the microbiota dif-
The gut microbial community is incredibly dynamic, with
fers from that of healthy people. Additionally, elderly
some autochthonous members (permanent residents), and
patients with inflammation and obese individuals also pre-
allochthonous ones (transient residents), deriving from the
sent low richness of gut microbiota (Le Chatelier
hosts’ diet, water, and environment resources. Additionally,
et al. 2013).
diet plays a crucial role in the composition, diversity, and
Studies by Mulders et al. (2018) reported that HFD feed-
richness of the intestinal microbiota (Ross et al. 2010). The
gut microbes, their composition and function, and the host- ing extensively modifies the composition of gut microbiota.
microbe interactions are modulated by food type, quality, It is reported that A. muciniphila, Bifidobacterium spp., B.
and origin. Moreover, a diet rich in sugar and fat, and poor uniformis, and C. coccoides can prevent obesity, based on
in dietary fibers can contribute to the reduction of specific their associations with glycaemic control, leptin levels, and
bacteria in the gut (Makki et al. 2018). adiposity. In contrast, Bilophila wadsworthia and
Some research has demonstrated that obesity and related Oscillibacter spp. are indicated to contribute to the compli-
diseases are linked to alterations in gut microbiota. Indeed, cations of obesity. Moreover, increased intestinal and serum
a higher Firmicutes/Bacteroidetes ratio has been reported in inflammation are linked to relative abundance of A. mucini-
both obese humans and animals (Le Chatelier et al. 2013). phila, whereas multiple correlations between A. muciniphila,
Several systems linking energy metabolism and gut micro- Bifidobacterium spp. and decreased inflammation have
biota in the host have been postulated, including the diet been reported.
energy, regulation of fat metabolism, and synthesis of gut The relationship between obesity and intestinal micro-
peptides involved in energy homeostasis (Cani and Delzenne biota is well-known; however, more studies are necessary to
2009). However, disagreement exists regarding the focus on the capacity of the intestinal microbiota and its
Firmicute/Bacteroidetes proportion to characterize the obese metabolites to target ENS, in order to suggest new
phenotype, since more recent studies have not confirmed approaches for the treatment of obesity and its related
this result. Differences may be associated with changes in comorbidities: intestinal dysmotility disorder, increased food
metabolic functions at the genus and species levels com- intake, and T2D. Additionally, the influence of ENS on glu-
pared to the phylum level (Bliss and Whiteside 2018). cose absorption is not completely understood (Rastelli,
Angelakis et al. (2012) reported that the relationship Knauf, and Cani 2018).
between microbiota and obesity is probably more sophisti- HFD can affect the gut microbiome and intestinal physi-
cated than Firmicutes/Bacteroidetes proportion at a phylum- ology under certain conditions which lead to metabolic dys-
level. This suggests that differences in weight or BMI may functions (Bailey and Holscher 2018). Conversely, several
not be universally related to the intestinal microbiota studies have shown that probiotics and prebiotics play a
14 T. F. DA SILVA ET AL.

crucial role in restoring the balance of the intestinal micro- adipose tissues; it can differentially regulate genes involved
biota of obese human (Table 1) and animals (Table 2). in gut integrity and apoptosis, which may be a system cap-
Bifidobacterium and Lactobacillus species are the most able of decreasing gut inflammation. Moreover, butyrate
studied ones to recover the GI balance. Bifidobacterium spe- exerts a trophic effect on the intestine, resulting in intestinal
cies longum, animalis, bifidum and breve and Lactobacillus morphology changes, such as increased villus height and
species acidophilus, plantarum, rhamnosus, fermentum, reu- crypt depth and thickened mucosal layer, which contribute
teri, bulgaricus, lactis, casei, paracasei, curvatus and sakei are to strengthen the intestinal barrier function, essential for
generally recognized as safe (GRAS) for consumption by preventing the development of metabolic diseases. Butyrate
humans by the United States Food and Drug and propionate activate intestinal gluconeogenesis, thereby
Administration (FDA) and have qualified presumption of promoting glucose control and metabolic benefits reflected
safety (QPS) status by the European Food Safety Authority by body weight, while acetate reduces appetite by activating
– EFSA (Ricci et al. 2017, FDA 2020). the tricarboxylic acid cycle to alter the expression profile of
More recently, A. muciniphila has gained special interest the hypothalamic appetite-regulating neuropeptide.
to recover intestinal dysbiosis. A. muciniphila is a strict Moreover, acetate can be converted into butyrate by enzyme
anaerobe isolated from human feces which uses the mucin butyryl-CoA producer microbes, such as Faecalibacterium
as the sole sources of carbon and nitrogen elements. A. prausnitzii, Anaerostipes spp., Eubacterium spp., and
muciniphila is known to have an essential role in improving Roseburia spp., by using a mechanism known as cross-feed-
the host metabolic functions and immune responses. The ing or cross-talk, when one species produces metabolites
restoration of A. muciniphila has also been shown to that are utilized by another species. (Bailey and Holscher
improve glucose tolerance and insulin action, and low abun- 2018; Luu and Visekruna 2019; Mulders et al. 2018; Zhi
dance of this bacterium was reported for obese individuals et al. 2019).
(Zhi et al. 2019). The beneficial effects of A. muciniphila Diet and particularly dietary fiber have a crucial effect on
include provision of nutrients (acetate and propionate) for the gut microbiota. There is evidence supporting that a
the growth of other bacteria and reduction of intestinal per- high-fiber diet presents many health benefits, such as
meability via the upregulation of mucin. In contrast, the decreased incidence of coronary artery disease, diabetes, and
genera Oscillibacter and Flavonifractor increase inflammation obesity. Higher microbial richness in terms of both tax-
via mechanisms that are poorly understood (Mulders et al. onomy and gene expression has been associated with fiber-
2018; Zhang et al. 2019). rich, plant-based diets; in contrast, low microbial richness
In a recent clinical study, the effects of daily oral supple- has been associated with higher insulin resistance, adiposity,
mentation of 1010 bacteria either alive or pasteurized A. inflammation, and dyslipidemia (Le Chatelier et al. 2013;
muciniphila for 3 months was evaluated in overweight and Bailey and Holscher 2018).
overweight/obese insulin resistant women volunteers Additionally, an increase in the relative abundance of
(N ¼ 32). It reduced the levels of liver dysfunction and Bifidobacterium, Eubacterium rectale, and C. coccoides was
inflammation while the overall gut microbiome structure shown with inulin supplementation, while decreased the
was unaffected (Depommier et al. 2019). relative abundance of Roseburia (Mulders et al. 2018);
The effectiveness of different bacteria strains in clinical in contrast, oligofructose has been linked to increases in
trials testing animals and humans are summarized in A. muciniphilia, F. prausnitzii, Bifidobacterium, and
Table 1. The outcomes of prebiotics and synbiotics on the Lactobacillus (Cani et al. 2009b). Studies have demonstrated
biochemical markers and gut microbiota have also been that prebiotics, such as oligofructose and inulin, increase the
evaluated. Recently, Crovesy et al. (2017) reviewed the concentration of SCFAs, such as acetate and propionate,
impact of Lactobacillus on body fat and weight in have been reported to have a beneficial effect on host
overweight patients. They suggested that possible modes of metabolism and appetite; they improve gut-barrier function,
action include the anti-inflammatory ability of some strains bring about weight-loss, and reduce food intake, by improv-
and the modulation of lipid metabolism parallel to increases ing gut nutrient-sensing mechanisms; they also decrease
in Bifidobacterium spp. systemic inflammation and leptin resistance. The SCFAs
The use of many natural products containing dietetic promote energy metabolism through their effects on
fibers and probiotics has been potentially related to receptor mediated pathways of cellular metabolism. These
improvements in intestinal microbiota. Such modulation effects are associated with changes in the gut microbiota
may occur through the regulation of cytokines activity, acti- composition (Bauer, Hamr, and Duca 2016; Mulders et al.
vation of nuclear receptor proteins, or production of SCFAs 2018; Zhi et al. 2019).
(Daguet et al. 2016). Additionally, the concentrations of fecal Using an in vitro batch culture fermentation, Henrique-
SCFAs (acetate, butyrate, and propionate, in a molar ratio of Bana et al. (2020) evaluated the effect of synbiotic supple-
approximately 60:20:20) are indicative of intestinal health. ments containing short chain FOS (1% w/v) combined with
Other SCFAs such as formate and valerate (pentanoate) as either B. lactis Bb12 or B. lactis HN019 (106 CFU/mL).
well isobutyate and isovalerate (branched-chain fatty acids) Maltodextrin (1% w/v), FOS (1% w/v) and the probiotic
have been detected at much lower levels in the gut of strains were also tested individually. FOS increased the
humans and rodents. Studies demonstrated that butyrate Atopobium cluster while maltodextrin increased the
promotes fatty acid oxidation and thermogenesis in brown Clostridium coccoides-Eubacterium rectale group. Additionally,
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 15

maltodextrin, FOS and synbiotics increased the production of ORCID


acetate and butyrate, compared to the negative control and
Ana L
ucia Barretto Penna http://orcid.org/0000-0001-6715-9276
probiotics individually.
In healthy individuals, the SCFAs homeostasis requires
high abundance of SCFAs-producing bacteria in the gut References
lumen and a continuous intake of non-digestible carbohy- Alard, J., V. Lehrter, M. Rhimi, I. Mangin, V. Peucelle, A.-L. Abraham,
drates such as dietary fiber. In contrast, recent studies have M. Mariadassou, E. Maguin, A.-J. Waligora-Dupriet, B. Pot, et al.
highlighted that restricted gut microbiota composition and 2016. Beneficial metabolic effects of selected probiotics on
atypical production of SCFAs have been associated with diet-induced obesity and insulin resistance in mice are associated
several autoimmune and inflammatory disorders, such as pro- with improvement of dysbiotic gut microbiota. Environmental
Microbiology 18 (5):1484–97. doi: 10.1111/1462-2920.13181.
gressive intestinal inflammation and colitis-associated carcino- Al-Assal, K., A. C. Martinez, R. S. Torrinhas, C. Cardinelli, and D.
genesis, multiple sclerosis, type 1 diabetes and rheumatoid Waitzberg. 2018. Gut microbiota and obesity. Clinical Nutrition
arthritis. Furthermore, SCFAs modulate the metabolic status Experimental 20 (3):60–4. doi: 10.1016/j.yclnex.2018.03.001.
of T cells, thereby impacting on epigenetic modifications and Angelakis, E., F. Armougom, M. Million, and D. Raoult. 2012.
T cell function (Luu and Visekruna 2019). The relationship between gut microbiota and weight gain in
humans. Future Microbiology 7 (1):91–109. doi: 10.2217/fmb.11.142.
In a clinical study, Salazar et al. (2015) treated 30 obese
Bagarolli, R. A., N. Tobar, A. G. Oliveira, T. G. Ara ujo, B. M.
women with 16 g/day inulin-type fructans (N ¼ 15) or malto- Carvalho, G. Z. Rocha, J. F. Vecina, K. Calisto, D. Guadagnini, P. O.
dextrin (N ¼ 15) for 3 months and determined variations on Prada, et al. 2017. Probiotics modulate gut microbiota and improve
fecal SCFAs concentration and explored associations between insulin sensitivity in DIO mice. The Journal of Nutritional
Bifidobacterium species, SCFAs and host biological markers of Biochemistry 50 (12):16–25. doi: 10.1016/j.jnutbio.2017.08.006.
Bailey, M. A., and H. D. Holscher. 2018. Microbiome-mediated effects
metabolism. Changes in B. bifidum and B. adolescentis were
of the Mediterranean diet on inflammation. Advances in Nutrition
inversely associated with fat mass percentage whereas B. breve 9 (3):193–206. doi: 10.1093/advances/nmy013.
negatively correlated with total cholesterol and LDL cholesterol. Bauer, P. V., S. C. Hamr, and F. A. Duca. 2016. Regulation of energy
Moreover B. longum negatively correlated with serum LPS. balance by a gut–brain axis and involvement of the gut microbiota.
Further investigations are desirable to deeply understand Cellular and Molecular Life Sciences 73 (4):737–55. doi: 10.1007/
s00018-015-2083-z.
the mode of action of probiotics and prebiotics on the
Bernini, L. J., A. N. C. Sim~ao, D. F. Alfieri, M. A. B. Lozovoy, N. L.
human health, using long-term and large-scale clinical inter- Mari, C. H. B. de Souza, I. Dichi, and G. N. Costa. 2016. Beneficial
ventions to elucidate how changes in the intestinal micro- effects of Bifidobacterium lactis on lipid profile and cytokines
biota is linked to obesity development. in patients with metabolic syndrome: A randomized trial. Effects
of probiotics on metabolic syndrome. Nutrition 32 (6):716–9. doi:
10.1016/j.nut.2015.11.001.
Final remarks and futures studies Bernini, L. J., A. N. C. Sim~ao, C. H. B. Souza, D. F. Alfieri, L. G.
Segura, G. N. Costa, and I. Dichi. 2018. Effect of Bifidobacterium
Accumulated data about the impact of probiotic, prebiotic lactis HN019 on inflammatory markers and oxidative stress in sub-
and synbiotic food or supplements on obesity diseases are jects with and without the metabolic syndrome. British Journal of
encouraging. Many studies have successfully demonstrated Nutrition 120 (6):645–52. doi: 10.1017/S0007114518001861.
Beserra, B. T. S., R. Fernandes, V. A. Rosario, M. C. Mocellin, M. G. F.
several beneficial effects on the host’s metabolism: enhanced
Kuntz, and E. B. S. M. Trindade. 2015. A systematic review and
metabolism of dietary components, weight loss, decrease in meta-analysis of the prebiotics and synbiotics effects on glycaemia,
BMI and adiposity, enhanced body composition, hormone insulin concentrations and lipid parameters in adult patients with
regulation, decreased pro-inflammatory markers, improved overweight or obesity. Clinical Nutrition 34 (5):845–58. doi: 10.1016/
biochemical markers, and ecology protection. j.clnu.2014.10.004.
Bliss, E. S., and E. Whiteside. 2018. The gut-brain axis, the human gut
Additionally, these foods or supplements play a crucial role in
microbiota and their integration in the development of obesity.
composition, diversity, and richness of gut microbiota profile, Frontiers in Physiology 9 (7):1–27. doi: 10.3389/fphys.2018.00900.
consequently, reducing the incidence of diseases, such as obesity Bomhof, M. R., J. A. Parnell, H. R. Ramay, P. Crotty, K. P. Rioux,
and its comorbidities. Moreover, there is a need to extend the C. S. Probert, … R. A. Reimer. 2018. Histological improvement of
current knowledge about the role of bacteria involved in the non-alcoholic steatohepatitis with a prebiotic: A pilot clinical trial.
pathogenesis of obesity and how particular bacterial groups can European Journal of Nutrition 58 (4): 1735–1745. doi: 10.1007/
s00394-018-1721-2.
present therapeutic potential to avoid or treat obesity. Bomhof, M. R., D. C. Saha, D. T. Reid, H. A. Paul, and R. A. Reimer.
2014. Combined effects of oligofructose and Bifidobacterium ani-
malis on gut microbiota and glycemia in obese rats. Obesity 22 (3):
Disclosure statement 763–71. doi: 10.1002/oby.20632.
The authors declare that they have no conflict of interest. Borgeraas, H., L. K. Johnson, J. Skattebu, J. K. Hertel, and J.
Hjelmesaeth. 2018. Effects of probiotics on body weight, body mass
index, fat mass and fat percentage in subjects with overweight or
Funding obesity: A systematic review and meta-analysis of randomized con-
trolled trials. Obesity Reviews 19 (2):219–32. doi: 10.1111/obr.12626.
This study was sponsored by The Brazilian National Council for Byrne, C. S., E. S. Chambers, D. J. Morrison, and G. Frost. 2015. The
Scientific and Technological Development (CNPq - Project 307155/ role of short chain fatty acids in appetite regulation and energy
2015-3). The authors would like to thank Vivian R. Diamantino for homeostasis. International Journal of Obesity 39 (9):1331–8. doi: 10.
preparing Figure 1. 1038/ijo.2015.84.
16 T. F. DA SILVA ET AL.

Canfora, E. E., C. M. van der Beek, G. D.A. Hermes, G. H. Goossens, reducing body weight and fat mass in overweight women. Current
J. W.E. Jocken, J. J. Holst, H. M. van Eijk, K. Venema, H. Smidt, Topics in Nutraceutical Research 11 (1–2):21–8.
E. G. Zoetendal, et al. 2017. Supplementation of diet with galacto- Dror, T., Y. Dickstein, G. Dubourg, and M. Paul. 2017. Microbiota
oligosaccharides increases bifidobacteria, but not insulin sensitivity, manipulation for weight change. Microbial Pathogenesis 106 (5):
in obese prediabetic individuals. Gastroenterology 153 (1):87–97. doi: 146–61. doi: 10.1016/j.micpath.2016.01.002.
10.1053/j.gastro.2017.03.051. Ebrahimi, Z. S., E. Nasli-Esfahani, A. Nadjarzade, and H. Mozaffari-
Cani, P. D., J. Amar, M. A. Iglesias, M. Poggi, C. Knauf, D. Bastelica, Khosravi. 2017. Effect of symbiotic supplementation on glycemic
A. M. Neyrinck, F. Fava, K. M. Tuohy, C. Chabo, et al. 2007a. control, lipid profiles and microalbuminuria in patients with non-
Metabolic endotoxemia initiates obesity and insulin resistance. obese type 2 diabetes: A randomized, double-blind, clinical trial.
Diabetes 56 (7):1761–72. doi: 10.2337/db06-1491. Journal of Diabetes and Metabolic Disorders 16 (23):1–10.
Cani, P. D., A. M. Neyrinck, F. Fava, C. Knauf, R. G. Burcelin, K. M. Everard, A., V. Lazarevic, N. Gaïa, M. Johansson, M. Ståhlman, F.
Tuohy, G. R. Gibson, and N. M. Delzenne. 2007b. Selective increases Backhed, N. M. Delzenne, J. Schrenzel, P. François, and P. D. Cani.
of bifidobacteria in gut microflora improve high-fat-diet-induced 2014. Microbiome of prebiotic-treated mice reveals novel targets
diabetes in mice through a mechanism associated with endotoxae- involved in host response during obesity. The ISME Journal 8 (10):
mia. Diabetologia 50 (11):2374–83. doi: 10.1007/s00125-007-0791-0. 2116–30. doi: 10.1038/ismej.2014.45.
Cani, P. D., and N. M. Delzenne. 2009. The role of the gut microbiota Falcinelli, S., A. Rodiles, A. Hatef, S. Picchietti, L. Cossignani, D. L.
in energy metabolism and metabolic disease. Current Pharmaceutical Merrifield, S. Unniappan, and O. Carnevali. 2018. Influence of pro-
Design 15 (13):1546–58. doi: 10.2174/138161209788168164. biotics administration on gut microbiota core: A review on the
Cani, P. D., E. Lecourt, E. M. Dewulf, F. M. Sohet, B. D. Pachikian, D. effects on appetite control, glucose, and lipid metabolism. Journal of
Naslain, F. De Backer, A. M. Neyrinck, and N. M. Delzenne. 2009a. Clinical Gastroenterology 52 (11):S50–S56. doi: 10.1097/MCG.
Gut microbiota fermentation of prebiotics increases satietogenic and 0000000000001064.
incretin gut peptide production with consequences for appetite sen- FDA. 2020. GRAS notice. Accessed January 30, 2020. https://www.
sation and glucose response after a meal. The American Journal of accessdata.fda.gov/scripts/fdcc/?set=GRASNotices.
Clinical Nutrition 90 (5):1236–43. doi: 10.3945/ajcn.2009.28095. Forssten, S. D., M. Z. Korczy nska, R. M.L. Zwijsen, W. H. Noordman,
Cani, P. D., S. Possemiers, T. Van de Wiele, Y. Guiot, A. Everard, O. M. Madetoja, and A. C. Ouwehand. 2013. Changes in satiety hor-
Rottier, L. Geurts, D. Naslain, A. Neyrinck, D. M. Lambert, et al. mone concentrations and feed intake in rats in response to lactic acid
2009b. Changes in gut microbiota control inflammation in obese bacteria. Appetite 71 (12):16–21. doi: 10.1016/j.appet.2013.06.093.
mice through a mechanism involving GLP-2-driven improvement of Foxx-Orenstein, A. E., and W. D. Chey. 2012. Manipulation of the gut
gut permeability. Gut 58 (8):1091–103. doi: 10.1136/gut.2008.165886. microbiota as a novel treatment strategy for gastrointestinal disor-
Cano, P. G., A. Santacruz, F. M. Trejo, and Y. Sanz. 2013. ders. The American Journal of Gastroenterology Supplements 1 (1):
Bifidobacterium CECT 7765 improves metabolic and immunological 41–6. doi: 10.1038/ajgsup.2012.8.
alterations associated with obesity in high-fat diet-fed mice. Obesity Gauffin-Cano, P., A. Santacruz, A.  Moya, and Y. Sanz. 2012.
21 (11):2310–21. doi: 10.1002/oby.20330. Bacteroides uniformis CECT 7771 ameliorates metabolic and
Chen, Y. T., N. S. Yang, Y. C. Lin, S. T. Ho, K. Y. Li, J. S. Lin, … immunological dysfunction in mice with high-fat-diet induced obes-
M. J. Chen. 2018. A combination of Lactobacillus mali APS1 and ity. PloS One. 7 (7):1–16. doi: 10.1371/journal.pone.0041079.
dieting improved the efficacy of obesity treatment via manipulating Gibson, R. A., D. Barclay, H. Marshall, J. Moulin, J. C. Maire, and M.
gut microbiome in mice. Nature Scientific Report 8 (6153):1–14. Makrides. 2009. Safety of supplementing infant formula with long-
Clemente, J. C., J. Manasson, and J. U. Scher. 2018. The role of the gut chain polyunsaturated fatty acids and Bifidobacterium lactis in term
microbiome in systemic inflammatory disease. British Medical infants: A randomised controlled trial. British Journal of Nutrition
Journal 360:j5145. doi: 10.1136/bmj.j5145. 101 (11):1706–13. doi: 10.1017/S0007114508084080.
Crovesy, L., M. Ostrowski, D. M. T. P. Ferreira, E. L. Rosado, and M. Gibson, G. R., R. Hutkins, M. E. Sanders, S. L. Prescott, R. A. Reimer,
Soares-Mota. 2017. Effect of Lactobacillus on body weight and body S. J. Salminen, … G. Reid. 2017. Expert consensus document: The
fat in overweight subjects: A systematic review of randomized con- International Scientific Association for Probiotics and Prebiotics
trolled clinical trials. International Journal of Obesity 41 (11): (ISAPP) Consensus Statement on the Definition and Scope of
1607–14. doi: 10.1038/ijo.2017.161. Prebiotics. Nature Reviews Gastroenterology & Hepatology 4 (8):
Daguet, D,. I. Pinheiro, A. Verhelst, S. Possemiers, and M. Marzorati. 491–502. doi: 10.1038/nrgastro.2017.75.
2016. Arabinogalactan and fructooligosaccharides improve the gut Gomes, A. C., R. G. M. Sousa, P. B. Botelho, T. L. N. Gomes, P. O.
barrier function in distinct areas of the colon in the Simulator of Prada, and J. F. Mota. 2017. The additional effects of a probiotic
the Human Intestinal Microbial Ecosystem. Journal of Functional mix on abdominal adiposity and antioxidant status: A double-blind,
Foods 20:369–79. doi: 10.1016/j.jff.2015.11.005. randomized trial. Obesity 25 (1):30–8. doi: 10.1002/oby.21671.
Dao, M. C., and K. Clement. 2018. Gut microbiota and obesity: Hadi, A., K. Alizadeh, H. Hajianfar, H. Mohammadi, and M.
Concepts relevant to clinical care. European Journal of Internal Miraghajani. 2020. Efficacy of synbiotic supplementation in obesity
Medicine 48 (2):18–24. doi: 10.1016/j.ejim.2017.10.005. treatment: A systematic review and meta-analysis of clinical trials.
Delgado, G. T. C., and W. M. S. C. Tamashiro. 2018. Role of prebiotics Critical Reviews in Food Science and Nutrition 60 (4):584–96. doi:
in regulation of microbiota and prevention of obesity. Food Research 10.1080/10408398.2018.1545218.
International 113 (7):183–8. doi: 10.1016/j.foodres.2018.07.013. Han, Y., and H. Xiao. 2020. Whole food-based approaches to modulat-
Depommier, C., A. Everard, C. Druart, H. Plovier, M. Van Hul, S. ing gut microbiota and associated diseases. Annual Review of Food
Vieira-Silva, G. Falony, J. Raes, D. Maiter, N. M. Delzenne, et al. Science and Technology 11 (1):14.1–.25. doi: 10.1146/annurev-food-
2019. Supplemenation with Akkermansia muciniphila in overweight 111519-014337.
and obese human volunteers: A proof-of-concept exploratory study. Henrique-Bana, F. C., X. Wang, G. N. Costa, W. A. Spinosa, L. H.S.
Nature Medicine 25 (7):1096–103. doi: 10.1038/s41591-019-0495-2. Miglioranza, E. Scorletti, P. C. Calder, C. D. Byrne, and G. R.
Dewulf, E. M., P. D. Cani, S. P. Claus, S. Fuentes, P. G. Puylaert, A. M. Gibson. 2020. In vitro effects of Bifidobacterium lactis-based synbiot-
Neyrinck, L. B. Bindels, W. M. de Vos, G. R. Gibson, J. P. Thissen, ics on human faecal bacteria. Food Research International 128:
et al. 2013. Insight into the prebiotic concept: Lessons from an 108776. doi: 10.1016/j.foodres.2019.108776.
exploratory, double blind intervention study with inulin-type fruc- Higashikawa, F., M. Noda, T. Awaya, N. Danshiitsoodol, Y. Matoba, T.
tans in obese women. Gut 62 (8):1112–21., doi: 10.1136/gutjnl-2012- Kumagai, and M. Sugiyama. 2016. Antiobesity effect of Pediococcus
303304. pentosaceus LP28 on overweight subjects: A randomized, double-
Doria, E., E. Buonocore, A. Michelotti, V. Nobile, and F. Marzatico. blind, placebo-controlled clinical trial. European Journal of Clinical
2013. Evaluation of a phyto-supplement efficacy as adjuvant in Nutrition 70 (5):582–7. doi: 10.1038/ejcn.2016.17.
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 17

Hill, C., F. Guarner, G. Reid, G. R. Gibson, D. J. Merenstein, B. Pot, L. Leber, B., N. J. Tripolt, D. Blattl, M. Eder, T. C. Wascher, T. R. Pieber,
Morelli, R. B. Canani, H. J. Flint, S. Salminen, et al. 2014. The R. Stauber, H. Sourij, K. Oettl, and V. Stadlbauer. 2012. The influ-
International Scientific Association for Probiotics and Prebiotics ence of probiotic supplementation on gut permeability in patients
consensus statement on the scope and appropriate use of the term with metabolic syndrome: An open label, randomized pilot study.
probiotic. Nature Reviews Gastroenterology & Hepatology 11 (8): European Journal of Clinical Nutrition 66 (10):1110–5. doi: 10.1038/
506–14., doi: 10.1038/nrgastro.2014.66. ejcn.2012.103.
Hugon, P., J. C. Lagier, P. Colson, F. Bittar, and D. Raoult. 2017. Lee, S. J., S. Bose, J. G. Seo, W. S. Chung, C. Y. Lim, and H. Kim.
Repertoire of human gut microbes. Microbial Pathogenesis 106: 2014. The effects of co-administration of probiotics with herbal
103–12. doi: 10.1016/j.micpath.2016.06.020. medicine on obesity, metabolic endotoxemia and dysbiosis: A
Javadi, L., M. Ghavami, M. Khoshbaten, A. Safaiyan, A. Barzegari, and randomized double-blind controlled clinical trial. Clinical Nutrition
B. P. Gargari. 2017. The potential role of probiotics or/and prebiotic 33 (6):973–81. doi: 10.1016/j.clnu.2013.12.006.
on serum lipid profile and insulin resistance in alcoholic fatty liver Liu, F., X. Wang, H. Shi, Y. Wang, C. Xue, and Q. J. Tang. 2017.
disease: A double blind randomized clinical trial. Crescent Journal of Polymannuronic acid ameliorated obesity and inflammation associ-
Medical and Biological Sciences 4 (3):131–8. ated with a high-fat and high-sucrose diet by modulating the gut
John, G. K., L. Wang, J. Nanavati, C. Twose, R. Singh, and G. Mullin. microbiome in a murine model. British Journal of Nutrition 117 (9):
2018. Dietary alteration of the gut microbiome and its impact on 1332–42. doi: 10.1017/S0007114517000964.
weight and fat mass: A systematic review and meta-analysis. Genes 9 Luu, M., and A. Visekruna. 2019. Short-chain fatty acids: Bacterial
(3):167–19. doi: 10.3390/genes9030167. messengers modulating the immunometabolism of T cells. European
Jones, R. B., T. L. Alderete, A. A. Martin, B. A. Geary, D. H. Hwang, Journal of Immunology 49 (6):842–8. doi: 10.1002/eji.201848009.
S. L. Palmer, and M. I. Goran. 2018. Probiotic supplementation Madjd, A., M. A. Taylor, N. Mousavi, A. Delavari, R. Malekzadeh, I. A.
increases obesity with no detectable effects on liver fat or gut micro- Macdonald, and H. R. Farshchi. 2016. Comparison of the effect of
biota in obese Hispanic adolescents: A 16-week, randomized, pla- daily consumption of probiotic compared with low-fat conventional
cebo-controlled trial. Pediatric Obesity 13 (11):705–14. doi: 10.1111/ yogurt on weight loss in healthy obese women following an energy-
ijpo.12273. restricted diet: A randomized controlled trial. The American Journal
Jung, S. P., K. M. Lee, J. H. Kang, S. I. Yun, H. O. Park, Y. Moon, and of Clinical Nutrition 103 (2):323–9. doi: 10.3945/ajcn.115.120170.
J. Y. Kim. 2013. Effect of Lactobacillus gasseri BNR17 on overweight Makki, K., E. C. Deehan, J. Walter, and F. B€ackhed. 2018. The impact
and obese adults: A randomized, double-blind clinical trial. Korean of dietary fiber on gut microbiota in host health and disease. Cell
Journal of Family Medicine 34 (2):80–9. doi: 10.4082/kjfm.2013.34.2.80. Host & Microbe 23 (6):705–15. doi: 10.1016/j.chom.2018.05.012.
Jung, S., Y. J. Lee, M. Kim, M. Kim, J. H. Kwak, J. W. Lee, Y. T. Ahn, Minami, J., S. Kondo, N. Yanagisawa, T. Odamaki, J. Z. Xiao, F. Abe,
J. H. Sim, and J. H. Lee. 2015. Supplementation with two probiotic … T. Shimoda. 2015. Oral administration of Bifidobacterium breve
B-3 modifies metabolic functions in adults with obese tendencies in a
strains, Lactobacillus curvatus HY7601 and Lactobacillus plantarum
randomised controlled trial. Journal of Nutrition Science 4 (e17):1–7.
KY1032, reduced body adiposity and Lp-PLA2 activity in overweight
Mischke, M., T. Arora, S. Tims, E. Engels, N. Sommer, K. van Limpt,
subjects. Journal of Functional Foods 19 (10):744–52. doi: 10.1016/j.
A. Baars, R. Oozeer, A. Oosting, F. B€ackhed, et al. 2018. Specific
jff.2015.10.006.
synbiotics in early life protect against diet-induced obesity in adult
Kadooka, Y., M. Sato, K. Imaizumi, A. Ogawa, K. Ikuyama, Y. Akai,
mice. Diabetes, Obesity and Metabolism 20 (6):1408–18. doi: 10.
M. Okano, M. Kagoshima, and T. Tsuchida. 2010. Regulation of
1111/dom.13240.
abdominal adiposity by probiotics (Lactobacillus gasseri SBT2055) in
Miyoshi, M., A. Ogawa, S. Higurashi, and Y. Kadooka. 2014. Anti-
adults with obese tendencies in a randomized controlled trial.
obesity effect of Lactobacillus gasseri SBT2055 accompanied by
European Journal of Clinical Nutrition 64 (6):636–43. doi: 10.1038/
inhibition of pro-inflammatory gene expression in the visceral adi-
ejcn.2010.19. pose tissue in diet-induced obese mice. European Journal of
Kadooka, Y., M. Sato, A. Ogawa, M. Miyoshi, H. Uenishi, H. Ogawa, Nutrition 53 (2):599–606. doi: 10.1007/s00394-013-0568-9.
K. Ikuyama, M. Kagoshima, and T. Tsuchida. 2013. Effect of Mulders, R. J., K. C. G. de Git, E. Schele, S. L. Dickson, Y. Sanz, and
Lactobacillus gasseri SBT2055 in fermented milk on abdominal adi- R. A. H. Adan. 2018. Microbiota in obesity: Interactions with enter-
posity in adults in a randomised controlled trial. British Journal of oendocrine, immune and central nervous systems. Obesity Reviews
Nutrition 110 (9):1696–703. doi: 10.1017/S0007114513001037. 19 (4):435–51. doi: 10.1111/obr.12661.
Karaki, S., H. Tazoe, H. Hayashi, H. Kashiwabara, K. Tooyama, Y. Neyrinck, A. M., V. L. Sch€ uppel, T. Lockett, D. Haller, and N. M.
Suzuki, and A. Kuwahara. 2008. Expression of the short-chain fatty Delzenne. 2016. Microbiome and metabolic disorders related to obesity:
acid receptor, GPR43, in the human colon. Journal of Molecular Which lessons to learn from experimental models?. Trends in Food
Histology 39 (2):135–42. doi: 10.1007/s10735-007-9145-y. Science & Technology 57 (8):256–64. doi: 10.1016/j.tifs.2016.08.012.
Kellow, N. J., M. T. Coughlan, and C. M. Reid. 2014. Metabolic bene- Nicolucci, A. C., M. P. Hume, I. Martınez, S. Mayengbam, J. Walter,
fits of dietary prebiotics in human subjects: A systematic review of and R. A. Reimer. 2017. Prebiotics reduce body fat and alter intes-
randomised controlled trials. British Journal of Nutrition 111 (7): tinal microbiota in children who are overweight or with obesity.
1147–61. doi: 10.1017/S0007114513003607. Gastroenterology 153 (3):711–22. doi: 10.1053/j.gastro.2017.05.055.
Kobyliak, N., C. Conte, G. Cammarota, A. P. Haley, I. Styriak, L. Nie, Y., F. Luo, and Q. Lin. 2018. Dietary nutrition and gut microflora:
Gaspar, J. Fusek, L. Rodrigo, and P. Kruzliak. 2016. Probiotics in A promising target for treating diseases. Trends in Food Science &
prevention and treatment of obesity: A critical view. Nutrition & Technology 75 (3):72–80. doi: 10.1016/j.tifs.2018.03.002.
Metabolism 13 (14):14–3. doi: 10.1186/s12986-016-0067-0. Nikbakht, E., S. Khalesi, I. Singh, L. T. Williams, N. P. West, and N.
Kobyliak, N., T. Falalyeyeva, N. Boyko, O. Tsyryuk, T. Beregova, and Colson. 2018. Effect of probiotics and synbiotics on blood glucose: A
L. Ostapchenko. 2018. Probiotics and nutraceuticals as a new fron- systematic review and meta - analysis of controlled trials. European
tier in obesity prevention and management. Diabetes Research and Journal of Nutrition 57 (1):95–106. doi: 10.1007/s00394-016-1300-3.
Clinical Practice 141 (5):190–9. doi: 10.1016/j.diabres.2018.05.005. Park, D. Y., Y. T. Ahn, S. H. Park, C. S. Huh, S. R. Yoo, R. Yu, et al.
Lam, Y. Y., S. Maguire, T. Palacios, and I. D. Caterson. 2017. Are the 2013. Supplementation of Lactobacillus curvatus HY7601 and
gut bacteria telling us to eat or not to eat? reviewing the role of gut Lactobacillus plantarum KY1032 in diet-induced obese mice is asso-
microbiota in the etiology, disease progression and treatment of eat- ciated with gut microbial changes and reduction in obesity. PloS
ing disorders. Nutrients 9 (6):602–15. doi: 10.3390/nu9060602. One. 8 (7):1–12. doi: 10.1371/journal.pone.0059470.
Le Chatelier, E., T. Nielsen, J. Qin, E. Prifti, F. Hildebrand, G. Falony, Patterson, E., P. M. Ryan, J. F. Cryan, T. G. Dinan, R. P. Ross, G. F.
M. Almeida, M. Arumugam, J.-M. Batto, S. Kennedy, et al. 2013. Fitzgerald, and C. Stanton. 2016. Gut microbiota, obesity and dia-
Richness of human gut microbiome correlates with metabolic betes. Postgraduate Medical Journal 92 (1087):286–300. doi: 10.1136/
markers. Nature 500 (7464):541–6. doi: 10.1038/nature12506. postgradmedj-2015-133285.
18 T. F. DA SILVA ET AL.

Paul, H. A., M. R. Bomhof, H. J. Vogel, and R. A. Reimer. 2016. Diet- and weight loss in obesity. Nutrients 12 (1):222. doi: 10.3390/
induced changes in maternal gut microbiota and metabolomic pro- nu12010222.
files influence programming of offspring obesity risk in rats. Sharafedtinov, K. K., O. A. Plotnikova, R. I. Alexeeva, T. B. Sentsova,
Scientific Reports 12 (6):1–14. doi: 10.1038/srep20683. E. Songisepp, J. Stsepetova, I. Smidt, and M. Mikelsaar. 2013.
Poutahidis, T., M. Kleinewietfeld, C. Smillie, T. Levkovich, A. Perrotta, Hypocaloric diet supplemented with probiotic cheese improves body
S. Bhela, B. J. Varian, Y. M. Ibrahim, J. R. Lakritz, S. M. Kearney, mass index and blood pressure indices of obese hypertensive
et al. 2013. Microbial reprogramming inhibits Western diet-associated patients - a randomized double-blind placebo-controlled pilot study.
obesity. PloS One. 8 (7):e68596–11. doi: 10.1371/journal.pone.0068596. Nutrition Journal 12 (1):138. doi: 10.1186/1475-2891-12-138.
Probert, H. M., J. H. A. Apajalahti, N. Rautonen, J. Stowell, and G. R. Sroka-Oleksiak, A., A. Mlodzinska, M. Bulanda, D. Salamon, P. Major,
Gibson. 2004. Polydextrose, lactitol, and fructo-oligosaccharide fer- M. Stanek, and T. Gosiewski. 2020. Metagenomic analysis of duo-
mentation by colonic bacteria in a three-stage continuous culture denal microbiota reveals a potential biomarker of dysbiosis in the
system. Applied and Environmental Microbiology 70 (8):4505–11. course of obesity and type 2 diabetes: A pilot study. Journal of
doi: 10.1128/AEM.70.8.4505-4511.2004. Clinical Medicine 9:369. doi: 10.3390/jcm9020369.
Rabiei, S., M. Hedayati, B. Rashidkhani, N. Saadat, and R. Stenman, L. K., M. J. Lehtinen, N. Meland, J. E. Christensen, N.
Shakerhossini. 2018. The effects of synbiotic supplementation on Yeung, M. T. Saarinen, M. Courtney, R. Burcelin, M.-L. L€ahdeaho,
body mass index, metabolic and inflammatory biomarkers, and J. Linros, et al. 2016. Probiotic with or without fiber controls body
appetite in patients with metabolic syndrome: A triple-blind fat mass, associated with serum zonulin, in overweight and obese
randomized controlled trial. Journal of Dietary Supplements 1 (2:6): adults-randomized controlled trial. EBioMedicine 13 (10):190–200.
1–13. doi: 10.1080/19390211.2018.1455788. doi: 10.1016/j.ebiom.2016.10.036.
Rastelli, M., C. Knauf, and P. D. Cani. 2018. Gut microbes and health: nska, M., I. Łoniewski, S. Van Hemert, M. Sobieska, and P.
Szuli
A focus on the mechanisms linking microbes, obesity, and related Bogda nski. 2018. Dose-dependent effects of multispecies probiotic
disorders. Obesity 26 (5):792–800. doi: 10.1002/oby.22175. supplementation on the lipopolysaccharide (LPS) level and cardiome-
Rebello, C. J., J. Burton, M. Heiman, and F. L. Greenway. 2015. tabolic profile in obese postmenopausal women: A 12-week random-
Gastrointestinal microbiome modulator improves glucose tolerance ized clinical trial. Nutrients 10 (6):1–16. doi: 10.3390/nu10060773.
in overweight and obese subjects: A randomized controlled pilot Tenorio-Jimenez, C., M. J. Martınez-Ramırez, I. Del Castillo-Codes, C.
trial. Journal of Diabetes and Its Complications 29 (8):1272–6. doi: Arraiza-Irigoyen, M. Tercero-Lozano, J. Camacho, N. Chueca, F.
10.1016/j.jdiacomp.2015.08.023. Garcıa, J. Olza, J. Plaza-Dıaz, et al. 2019. Lactobacillus reuteri V3401
Reimer, R. A., H. J. Willis, J. M. Tunnicliffe, H. Park, K. L. Madsen, and reduces inflammatory biomarkers and modifies the gastrointestinal
A. Soto-Vaca. 2017. Inulin-type fructans and whey protein both modu- microbiome in adults with metabolic syndrome: The PROSIR Study.
late appetite but only fructans alter gut microbiota in adults with over- Nutrients 11 (8):1761., doi: 10.3390/nu11081761.
Tolhurst, G., H. Heffron, Y. S. Lam, H. E. Parker, A. M. Habib, E.
weight/obesity: A randomized controlled trial. Molecular Nutrition &
Diakogiannaki, J. Cameron, J. Grosse, F. Reimann, and F. M.
Food Research 61 (11):1700484. doi: 10.1002/mnfr.201700484.
Gribble. 2012. Short-chain fatty acids stimulate Glucagon-like
Rezazadeh, L., B. P. Gargari, M. A. Jafarabadi, and B. Alipour. 2019.
Peptide-1 secretion via the G-protein-coupled receptor FFAR2.
Effects of probiotic yogurt on glycemic indexes and endothelial dys-
Diabetes 61 (2):364–71. doi: 10.2337/db11-1019.
function markers in patients with metabolic syndrome. Nutrition 62:
Toral, M., M. Gomez-Guzman, R. Jimenez, M. Romero, M. Sanchez,
162–8. doi: 10.1016/j.nut.2018.12.011.
M. P. Utrilla, N. Garrido-Mesa, M. E. Rodrıguez-Cabezas, M.
Ricci, A., A. Allende, D. Bolton, M. Chemaly, R. Davies, R. Girones,
Olivares, J. Galvez, et al. 2014. The probiotic Lactobacillus corynifor-
et al. 2017. Scientific opinion on the update of the list of QPS-rec-
mis CECT5711 reduces the vascular pro-oxidant and pro-inflamma-
ommended biological agents intentionally added to food or feed as
tory status in obese mice. Clinical Science 127 (1):33–45. doi: 10.
notified to EFSA. EFSA Journal 15 (3):4664. doi: 10.2903/j.efsa.2017.
1042/CS20130339.
4884. Torres, S., E. Fabersani, A. Marquez, and P. Gauffin-Cano. 2018.
Ross, P., S. Mills, C. Hill, G.F. Fitzgerald, and C. Stanton. 2010. Adipose tissue inflammation and metabolic syndrome. The proactive
Specific metabolite production by gut microbiota as a basis for pro- role of probiotics. European Journal of Nutrition. doi: 10.1007/
biotic function. International Dairy Journal 20 (4):269–76. doi: 10. s00394-018-1790-2.
1016/j.idairyj.2009.12.003. Torres-Fuentes, C., H. Schellekens, T. G. Dinan, and J. F. Cryan. 2017.
Salazar, N., E. M. Dewulf, A. M. Neyrinck, L. B. Bindels, P. D. Cani, J. The microbiota–gut–brain axis in obesity. The Lancet Gastroenterology
Mahillon, W. M. de Vos, J. P. Thissen, M. Gueimonde, C. G. de los & Hepatology 2 (10):747–56. doi: 10.1016/S2468-1253(17)30147-4.
Reyes-Gavilan, et al. 2015. Inulin-type fructans modulate intestinal Torres-Fuentes, C., H. Schellekens, T. G. Dinan, and J. F. Cryan. 2015.
Bifidobacterium species populations and decrease fecal short-chain A natural solution for obesity: Bioactives for the prevention and
fatty acids in obese women. Clinical Nutrition 34 (3):501–7., doi: 10. treatment of weight gain. A review. Nutritional Neuroscience 18 (2):
1016/j.clnu.2014.06.001. 49–65. doi: 10.1179/1476830513Y.0000000099.
Sanchez, M., C. Darimont, V. Drapeau, S. Emady-Azar, M. Lepage, E. van der Beek, C. M., E. E. Canfora, A. M. Kip, S. H.M. Gorissen,
Rezzonico, C. Ngom-Bru, B. Berger, L. Philippe, C. Ammon-Zuffrey, S. W.M. Olde Damink, H. M. van Eijk, J. J. Holst, E. E. Blaak,
et al. 2014. Effect of Lactobacillus rhamnosus CGMCC1.3724 supple- C. H.C. Dejong, and K. Lenaerts. 2018. The prebiotic inulin
mentation on weight loss and maintenance in obese men and improves substrate metabolism and promotes short chain fatty acid
women. British Journal of Nutrition 111 (8):1507–19. doi: 10.1017/ production in overweight to obese men. Metabolism Clinical
S0007114513003875. Metabolism 87 (10):25–35. doi: 10.1016/j.metabol.2018.06.009.
Sayin, S. I., A. Wahlstr€ om, J. Felin, S. J€antti, H.-U. Marschall, K. Velikonja, A., L. Lipoglavsek, M. Zorec, R. Orel, and G. Avgustin.
Bamberg, B. Angelin, T. Hy€ otyl€ainen, M. Oresic, and F. B€ackhed. 2019. Alterations in gut microbiota composition and metabolic
2013. Gut microbiota regulates bile acid metabolism by reducing the parameters after dietary intervention with barley beta glucans in
levels of tauro-beta-muricholic acid, a naturally occurring FXR antag- patients with high risk for metabolic syndrome development.
onist. Cell Metabolism 17 (2):225–35. doi: 10.1016/j.cmet.2013.01.003. Anaerobe 55:67–77. doi: 10.1016/j.anaerobe.2018.11.002.
Seganfredo, F. B., C. A. Blume, M. Moehlecke, A. Giongo, D. S. Vinolo, M. A. R., H. G. Rodrigues, R. T. Nachbar, and R. Curi. 2011.
Casagrande, J. V. N. Spolidoro, A. V. Padoin, B. D. Schaan, and Regulation of inflammation by short chain fatty acids. Nutrients 3
C. C. Mottin. 2017. Weight-loss interventions and gut microbiota (10):858–76. doi: 10.3390/nu3100858.
changes in overweight and obese patients: A systematic review. Wanchai, K., S. Yasom, W. Tunapong, T. Chunchai, S.
Obesity Reviews 18 (8):832–51. doi: 10.1111/obr.12541. Eaimworawuthikul, P. Thiennimitr, C. Chaiyasut, A. Pongchaidecha,
Sergeev, I. N., T. Aljutaily, G. Walton, and E. Huarte. 2020. Effects of V. Chatsudthipong, S. Chattipakorn, et al. 2018. Probiotic
synbiotic supplement on human gut microbiota, body composition Lactobacillus paracasei HII01 protects rats against obese-insulin
CRITICAL REVIEWS IN FOOD SCIENCE AND NUTRITION 19

resistance-induced kidney injury and impaired renal organic anion overweight or obese subjects: A meta-analysis. Food & Function 10
transporter 3 function. Clinical Science 132 (14):1545–63. doi: 10. (3):1747–59. doi: 10.1039/C8FO02163E.
1042/CS20180148. Zarrati, M., E. Salehi, K. Nourijelyani, V. Mofid, M. J. H. Zadeh, F.
Watanabe, M., K. Morimoto, S. M. Houten, N. Kaneko-Iwasaki, T. Najafi, Z. Ghaflati, K. Bidad, M. Chamari, M. Karimi, et al. 2014.
Sugizaki, Y. Horai, C. Mataki, H. Sato, K. Murahashi, E. Arita, et al. Effects of probiotic yogurt on fat distribution and gene expression
2012. Bile acid binding resin improves metabolic control through of proinflammatory factors in peripheral blood mononuclear cells in
the induction of energy expenditure. Plos One. 7 (8):e38286–9. doi: overweight and obese people with or without weight-loss diet.
10.1371/journal.pone.0038286.
Journal of the American College of Nutrition 33 (6):417–25. doi: 10.
WHO (World Health Organization). 2018. Obesity and overweight.
Fact sheet No 311. Geneva, Switzerland. http://www.who.int/media- 1080/07315724.2013.874937.
centre/factsheets/fs311/en/. Zhang, T., Q. Li, L. Cheng, H. Buch, and F. Zhang. 2019. Akkermansia
Yadav, H., J. H. Lee, J. Lloyd, P. Walter, and S. G. Rane. 2013. muciniphila is a promising probiotic. Microbial Biotechnology 12 (6):
Beneficial metabolic effects of a probiotic via butyrate-induced GLP- 1109–25. doi: 10.1111/1751-7915.13410.
1 hormone secretion. Journal of Biological Chemistry 288 (35): Zhi, C., J. Huang, J. Wang, H. Cao, Y. Bai, J. Guo, and Z. Su. 2019.
25088–97. doi: 10.1074/jbc.M113.452516. Connection between gut microbiome and the development of obes-
Yan, S., Z. Tian, M. Li, B. Li, and W. Cui. 2019. Effects of probiotic ity. European Journal of Clinical Microbiology & Infectious Diseases
supplementation on the regulation of blood lipid levels in 38:1987–98. doi: 10.1007/s10096-019-03623-x.

You might also like