ArticlePDF Available

Abstract

Obesity is associated with increased risk for chronic diseases, and affects both developed and developing nations. Yogurt is a nutrient-dense food that may benefit individuals with lactose intolerance, constipation and diarrheal diseases, hypertension, cardiovascular diseases, diabetes and certain types of cancer. Emerging evidence suggests that yogurt consumption might also improve the health of obese individuals. Obesity is often accompanied by chronic, low-grade inflammation perpetuated by adipose tissue and the gut. In the gut, obesity-associated dysregulation of microbiota and impaired gut barrier function may increase endotoxin exposure. Intestinal barrier function can be compromised by pathogens, inflammatory cytokines, endocannabinoids, diet, exercise, and gastrointestinal peptides. Yogurt consumption may improve gut health and reduce chronic inflammation by enhancing innate and adaptive immune responses, intestinal barrier function, lipid profiles, and by regulating appetite. While this evidence suggests that yogurt consumption is beneficial for obese individuals, randomized-controlled trials are needed to further support this hypothesis.
This article was downloaded by: [University of Wisconsin - Madison]
On: 20 April 2015, At: 06:45
Publisher: Taylor & Francis
Informa Ltd Registered in England and Wales Registered Number: 1072954 Registered office: Mortimer House,
37-41 Mortimer Street, London W1T 3JH, UK
Click for updates
Critical Reviews in Food Science and Nutrition
Publication details, including instructions for authors and subscription information:
http://www.tandfonline.com/loi/bfsn20
Evidence for the Effects of Yogurt on Gut Health and
Obesity
Ruisong Peia, Derek A. Martina, Diana M. DiMarcoa & Bradley W. Bollinga
a Department of Nutritional Sciences, University of Connecticut, Storrs, CT.
Accepted author version posted online: 15 Apr 2015.
To cite this article: Ruisong Pei, Derek A. Martin, Diana M. DiMarco & Bradley W. Bolling (2015): Evidence for the Effects of
Yogurt on Gut Health and Obesity, Critical Reviews in Food Science and Nutrition, DOI: 10.1080/10408398.2014.883356
To link to this article: http://dx.doi.org/10.1080/10408398.2014.883356
Disclaimer: This is a version of an unedited manuscript that has been accepted for publication. As a service
to authors and researchers we are providing this version of the accepted manuscript (AM). Copyediting,
typesetting, and review of the resulting proof will be undertaken on this manuscript before final publication of
the Version of Record (VoR). During production and pre-press, errors may be discovered which could affect the
content, and all legal disclaimers that apply to the journal relate to this version also.
PLEASE SCROLL DOWN FOR ARTICLE
Taylor & Francis makes every effort to ensure the accuracy of all the information (the “Content”) contained
in the publications on our platform. However, Taylor & Francis, our agents, and our licensors make no
representations or warranties whatsoever as to the accuracy, completeness, or suitability for any purpose of the
Content. Any opinions and views expressed in this publication are the opinions and views of the authors, and
are not the views of or endorsed by Taylor & Francis. The accuracy of the Content should not be relied upon and
should be independently verified with primary sources of information. Taylor and Francis shall not be liable for
any losses, actions, claims, proceedings, demands, costs, expenses, damages, and other liabilities whatsoever
or howsoever caused arising directly or indirectly in connection with, in relation to or arising out of the use of
the Content.
This article may be used for research, teaching, and private study purposes. Any substantial or systematic
reproduction, redistribution, reselling, loan, sub-licensing, systematic supply, or distribution in any
form to anyone is expressly forbidden. Terms & Conditions of access and use can be found at http://
www.tandfonline.com/page/terms-and-conditions
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
1
Title: Evidence for the effects of yogurt on gut health and obesity.
Authors: Ruisong Pei1, Derek A. Martin1, Diana M. DiMarco1, and Bradley W. Bolling1*
Affiliations: 1Department of Nutritional Sciences, University of Connecticut, 3624 Horsebarn
Rd Extension, Unit 4017, Storrs, CT 06269-4017,
*Corresponding Author (Tel: 860-486-2180; Fax: 860-486-3674; E-mail:
bradley.bolling@uconn.edu)
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
2
Abstract:
Obesity is associated with increased risk for chronic diseases, and affects both developed and
developing nations. Yogurt is a nutrient-dense food that may benefit individuals with lactose
intolerance, constipation and diarrheal diseases, hypertension, cardiovascular diseases, diabetes
and certain types of cancer. Emerging evidence suggests that yogurt consumption might also
improve the health of obese individuals. Obesity is often accompanied by chronic, low-grade
inflammation perpetuated by adipose tissue and the gut. In the gut, obesity-associated
dysregulation of microbiota and impaired gut barrier function may increase endotoxin exposure.
Intestinal barrier function can be compromised by pathogens, inflammatory cytokines,
endocannabinoids, diet, exercise, and gastrointestinal peptides. Yogurt consumption may
improve gut health and reduce chronic inflammation by enhancing innate and adaptive immune
responses, intestinal barrier function, lipid profiles, and by regulating appetite. While this
evidence suggests that yogurt consumption is beneficial for obese individuals, randomized-
controlled trials are needed to further support this hypothesis.
Keywords: yogurt; obesity; inflammation; intestine; chronic disease; bioactives
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
3
INTRODUCTION
Yogurt has been consumed for centuries. As early as 1908, Metchnikoff ascribed the
prolonged life of the Bulgarians to consumption of sour milk fermented by lactic acid bacteria
(O'Sullivan et al., 1992). Yogurt is a milk product fermented by L. bulgaricus, S. thermophilus
and L. acidophilus (CODEX STAN 243-2003). In addition to these Lactic acid bacteria (LAB),
other strains of Lactobacillus and Bifidobacterium are commonly used as yogurt starter cultures
(Desobry-Banon et al., 1999). Yogurts may also be enriched in other probiotic strains that
convey additional health benefits beyond those of traditional yogurt cultures (Shah, 2007).
The global rise in obesity is an increasing health concern. The causes of obesity and
approaches needed to reduce obesity are multifactorial in nature (Holes-Lewis et al., 2013).
Effective social, behavioral, and dietary interventions are needed to mitigate the adverse effects
of obesity on personal health outcomes (Wadden et al., 2012). Obesity impairs gut health, which
may be a potential target for therapeutic dietary interventions (Tilg and Kaser, 2011). Yogurt is
rich with potential bioactive components and emerging evidence points toward the efficacy of
yogurt and its components to improve gut health in obesity.
YOGURT BIOACTIVES
Nutrients
Dairy products are rich in high-quality proteins, calcium, potassium, phosphorus,
magnesium, zinc and B vitamins (Table 1) (Buttriss, 1997). Fermentation can improve the
nutrient content of dairy products. For example, some bacteria synthesize B vitamins. S.
thermophilus can produce folate during yogurt fermentation, and certain inoculations can
increase folate levels 6-fold (Crittenden et al., 2003). Yogurt also contains conjugated linoleic
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
4
acid (CLA), a derivative of linoleic acid (Aneja and Murthi, 1990; Shahani and Chandan, 1979).
CLA may improve body composition by increasing lean body mass while decreasing fat mass,
and has immunostimulatory and anticarcinogenic effects (Park et al., 1997; Whigham et al.,
2000).
Fermentation also improves the digestibility of milk proteins. LAB proteolytic enzymes
and peptidases increase free amino acids in yogurt (Gorbach, 1990). Upon digestion, yogurt had
smaller clots of curd than milk, which facilitated digestive enzyme activity (Breslaw and Kleyn,
1973). In addition, the viscous texture of yogurt might decrease the gastric emptying rate, which
increases duration of the enzymatic hydrolysis (Gaudichon et al., 1994; Shahani and Chandan,
1979).
Yogurt is also considered to be a good source of minerals. Dairy products are a good
source of calcium, not just because of the abundance of calcium but also because of the high
absorbability of calcium from yogurt. The presence of lactose, phosphopeptides, and amino acids
derived from casein in dairy products facilitates the absorption of calcium by promoting its
active transport or passive diffusion (Gueguen and Pointillart, 2000). However, intervention
studies have not demonstrated greater bioavailability of dairy calcium than supplemental calcium
(Recker et al., 1988; Sheikh et al., 1987; Zhao et al., 2005). In contrast, dairy calcium was more
effective than supplementary calcium in reducing weight and fat in energy-restricted adults
(Zemel et al., 2000; Zemel et al., 2004). Although there is no evidence showing that yogurt
serves as a better source of calcium than milk or other dairy products, yogurt has the advantage
of being well tolerated by lactase-deficient individuals (Smith et al., 1985).
Other bioactives
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
5
Dairy products contain bioactive proteins, such as immunoglobulins, α-lactoglobulin, β-
lactoglobulin, lactoferrin, and phosphopeptides, which may regulate immune response, modulate
blood pressure, and facilitate mineral absorption (Ebringer et al., 2008). Bacterial hydrolysis of
milk protein can yield oligopeptides with additional biological activities. For instance, some
peptides (e.g. Val-Pro-Pro and Ile-Pro-Pro) have hypotensive effects via inhibiting angiotensin-
converting-enzyme (Nakamura et al., 2009). A pentapeptide hydrolyzed from casein, Ile-Ile-Ala-
Glu-Lys, has hypocholesterolemic effects in vitro (Morikawa et al., 2007). Other effects of
bioactive peptides such as antithrombotic, antioxidant, antimicrobial and antifungal activities
have also been reported (Ebringer et al., 2008).
Dairy products also contain various bioactive lipids and oligosaccharides. Phospho- and
sphingolipids may reduce blood cholesterol, enhance brain function, and inhibit colon cancer
(Ebringer et al., 2008; Rombaut et al., 2005). Some short chain fatty acids in dairy products such
as butyric acid (C4:0), caprylic acid (C8:0), capric acid (C10:0), lauric acid (C12:0) have
anticarcinogenic, antiviral and antibacterial activities (Ebringer et al., 2008). In addition, dairy
products contain some oligosaccharides such as lactulose, which could serve as prebiotics to
support the growth of commensal bacteria (Marconi et al., 2004).
Microorganisms
S. thermophilus and L. bulgaricus are the most frequent microorganisms used to produce
yogurt. In the United States, some yogurts have additional L. acidophilus, B. bifidum, B. lactis, L.
casei, and/or L. rhamnosus content, among others, and are branded as probiotic yogurts. The
most basic definition of probiotics is, live microorganisms which when administered in
adequate amounts confer a health benefit on the host (Pineiro and Embarek, 2002). However,
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
6
others have proposed that probiotics must originate from humans, be viable through the
gastrointestinal tract, adhere to the intestinal wall to facilitate colonization, produce
antimicrobials, and provide a demonstrable health effect (Guarner et al., 2005).
It is commonly thought that 105 - 107 CFU/mL living probiotic bacteria are needed to
confer a health benefit to the host (Schillinger, 1999; Vélez et al., 2007). Yogurt culture content
ranges from 104 to 108 CFU/g/strain (Dunlap et al., 2009). In the US, yogurt can be certified with
a live and active culture seal from the National Yogurt Association if it contains 108 CFU/g at
the time of manufacture (National-Yogurt-Association, 2008). While the viability of yogurt
microorganisms may be enumerated at manufacture, viability declines throughout the shelf-life
of products. For example, L. acidophilus, a culture commonly added to yogurt post-fermentation,
is relatively unstable in yogurt. This is likely due to hydrogen peroxide produced by L.
bulgaricus during yogurt production (Gilliland and Speck, 1977). A survey of yogurts in
Columbia found poor survival and inconsistent labeling of strains (Vélez et al., 2007). In a study
of yogurts of European origin, bacterial counts in some products were as low as 104 CFU per
gram per strain by the sell-by date (Schillinger, 1999). Temperature fluctuations may also reduce
viability of yogurt probiotics. After 6 h at room temperature, reductions of 9-46.2% were seen in
the CFU count for L. GG, L. johnsonii, and L. acidophilus (Scharl et al., 2010). Thus, it is
expected that the amount of traditional and probiotic strains present in yogurt varies considerably
by manufacturer, storage conditions, and time of consumption. Despite this, yogurt cultures may
not need to be viable to confer a health benefit. For example, a preparation of mixed DNA from
various probiotic strains inhibited colitis in IL-10-/- mice (Jijon et al., 2004). Conventional yogurt
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
7
LAB improve lactose digestion, despite poor viability and the inability to survive the digestive
process (Martini et al., 1991).
OBESITY, YOGURT AND CHRONIC DISEASE RISK
Obesity is an abnormal or excessive accumulation of fat that poses a risk to health. A
person with a body mass index (BMI) between 25 and 30 is classified as overweight and a BMI
greater than 30 is obese. The International Obesity Task Force estimates that at least 1.1 billion
adults are overweight, with 312 million of those obese (Haslam and James, 2005). In the US,
nearly 70% of adults are classified as overweight or obese (Flegal et al., 2010). Obesity is a
major risk factor for a number of chronic diseases such as diabetes, cardiovascular disease
(CVD) and certain cancers. Furthermore, obese adults are projected to lose 7 years of life
expectancy (Peeters et al., 2003). The morbidity associated with obesity accounts for 2-7% of
health care costs in the developed world (Hossain et al., 2007). Morbidities attributed to obesity
include CVD, type 2 diabetes, hypertension, cancer, chronic inflammation, and compromised gut
health. A limited number of yogurt intervention studies relevant to obesity and chronic disease
risk have demonstrated positive outcomes on lipid profiles and chronic inflammation (Table 2).
Cardiovascular disease
CVD is one of the leading causes of death and premature mortality. Ischaemic heart
diseases and stroke account for nearly one in four deaths worldwide (Lozano et al., 2012).
Visceral obesity has a critical role in the development of CVD (Grundy, 2007). Mathieu et al.
reviewed how inflammation linked obesity and CVD (Mathieu et al., 2010). Briefly, excessive
accumulation of fat in the adipose tissue leads to macrophage infiltration and elevated production
of proinflammatory cytokines, which contribute to the development of atherosclerosis. In
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
8
addition, obesity is related to atherogenic dyslipidemia characterized by increased levels of
triglyceride (TG), small dense low-density lipoprotein (sdLDL) particles, as well as decreased
level of high-density lipoprotein cholesterol (HDL-C) (Tenenbaum and Fisman, 2012). Obesity
can also directly affect the structure and functions of the cardiovascular system. Obese
individuals have increased cardiac output which can lead to left ventricular hypertrophy and
other structural abnormalities (Lavie et al., 2009). Obesity also causes left atrial enlargement due
to increased circulating blood volume and abnormal left ventricular diastolic filling (Lavie et al.,
2009). These abnormalities compromise cardiovascular function and increase CVD risk for
obese individuals (Lavie et al., 2009).
Several recent expert reviews have summarized the potential benefits of dairy
consumption on CVD risk. Van Meijl et al. reviewed the physiological effects of dairy
consumption on metabolic syndrome and concluded that dairy calcium and protein had important
roles in reducing metabolic syndrome risk (Van Meijl et al., 2008). In a prospective, matched
case-control study using serum milk fat biomarkers, it was found that biomarkers of milk fat
were inversely associated with the first myocardial infarction in Swedish women after
multivariable adjustment for confounders (OR 0.74, 95% CI: 0.58, 0.94); moreover, reported
intake of fermented milk products were inversely related to the first myocardial infarction (P <
0.05 for trend) (Warensjö et al., 2010). German et al. reviewed the effects of dairy foods and
dairy fats on CVD risk (German et al., 2009). They suggested that although dairy products
contributed to saturated fat intake, there was no consistent association between dairy
consumption and risk of CVD (German et al., 2009). Similarly, another group examined the
influence of milk fat containing dairy foods and CVD health and concluded that dairy
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
9
consumption did not increase the risk of CVD, coronary heart disease or stroke, regardless of
milk fat levels (Huth and Park, 2012). Most research on dairy and CVD risk have not evaluated
yogurt specifically. Given the differences in nutrient and bioactive content between yogurt and
other dairy products, more attention is needed on this specific product category.
Conventional yogurt consumption may improve lipid profiles in healthy and
hypercholesterolemic adults. The effects of conventional yogurt, yogurt with L. acidophilus and
B. lactis, or no yogurt on blood lipids were evaluated in healthy Iranian women (n = 90)
(Sadrzadeh-Yeganeh et al., 2010). Consumption of 300 g/d of conventional and probiotic yogurt
for 6 wk reduced the total cholesterol and total:HDL cholesterol ratio relative to the control
group (Sadrzadeh-Yeganeh et al., 2010). The probiotic yogurt-consuming group also
experienced an 8.8% increase in HDL cholesterol (Sadrzadeh-Yeganeh et al., 2010). Probiotic or
prebiotic containing yogurt may also further improve lipid profiles in adults. A randomized
cross-over study of 40 hypercholesterolemic US adults consuming 200 g yogurt with L.
acidophilus L1 for 4 wk reduced serum cholesterol by 3.2% relative to a yogurt prepared with an
L. acidophilus strain with poor viability and low in vitro cholesterol-lowering activity (Anderson
and Gilliland, 1999). In a cross-over study of 29 healthy women, which included
hypercholesterolemic individuals, 300 g of yogurt with L. acidophilus and B. longum for 7
weeks increased serum HDL cholesterol by 0.3 mmol/L relative to a control yogurt without these
strains (Kießling et al., 2002). In a parallel study of 33 normocholesterolemic women,
consumption of 100 g/d yogurt for 2 wk and 200 g/d for another 2 wk reduced the LDL/HDL
cholesterol ratio in healthy women, with no differences from the probiotic culture L. casei
containing yogurt (Fabian and Elmadfa, 2006). Consumption of 375 mL of yogurt containing L.
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
10
acidophilus and fructo-oligosaccharides for 3 wk lowered serum total cholesterol, LDL-
cholesterol, and the LDL/HDL-ratio in 30 healthy men relative to conventional yogurt
(Schaafsma et al., 1998).
Hypertension
Hypertension is associated with vascular disease mortality, CVD, and renal diseases
(Chobanian et al., 2003; Lewington et al., 2002). Hypertension in US adults increased from
23.9% in 1988-1994 to 29% in 2007-2008 based on data from the National Health and Nutrition
Examination Survey (NHANES) (Egan et al., 2010). Previous studies have indicated a strong
relationship between obesity and hypertension. Cross-sectional studies indicate that more than
85% of hypertensive individuals have a BMI of over 25 kg/m2 (Kastarinen et al., 2000). Several
mechanisms are involved in the pathogenesis of obesity-related hypertension. The sympathetic
nervous system, the renin-angiotensin system (RAS), and aldosterone contribute to the
development of hypertension in obesity (Rahmouni et al., 2005). Long-term over-activation of
sympathetic nervous system which is found in obesity could raise arterial pressure by inducing
peripheral vasoconstriction and increasing sodium reabsorption in the renal tubules (Rahmouni et
al., 2005). Adipose RAS is activated in obesity; animal models of visceral obesity suggest that
adipose RAS contributes to obesity-associated hypertension (Massiéra et al., 2001). Plasma
aldosterone levels were elevated in obese hypertensive patients (Goodfriend and Calhoun, 2004);
on the other hand, an aldosterone antagonist was found to inhibit the development of high blood
pressure in dietary-induced obese dog models (De Paula et al., 2004).
Therefore, given the need for dietary strategies to mitigate hypertension, the
antihypertensive effects of dairy consumption have been investigated. The Dietary Approaches
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
11
to Stop Hypertension (DASH) trial showed that a diet rich in fruits and vegetables lowered blood
pressure and that additional inclusion of low-fat dairy products with reduced saturated and total
fat further augmented these blood pressure-lowering effects (Appel et al., 1997). A recent review
and meta-analysis of five cohort studies involving nearly 45,000 subjects revealed an inverse
association between dairy consumption and development of elevated blood pressure, defined as
130 mm Hg systolic and/or 84 mm Hg diastolic blood pressure (RR 0.87, 0.81-0.94 95% CI)
(Ralston et al., 2012). Another meta-analysis of prospective cohort studies similarly reported that
increased consumption of 200 g/d of low-fat dairy products reduced the risk of hypertension (RR
0.96, 95% CI, 0.93-0.99) (Soedamah-Muthu et al., 2012). Based on these data, low-fat dairy
consumption appears protective against hypertension in adults, but well-designed randomized,
controlled trials (RCTs) are needed to confirm if yogurt is also antihypertensive.
Cancer
In 2010, 8 million people died from cancer globally, accounting for 15.1% of all deaths
worldwide (Lozano et al., 2012). Overweight and obesity are estimated to contribute to 14% of
all cancer deaths in men and 20% of deaths in women (Calle et al., 2003). Meta-analyses indicate
that higher BMI is associated with an increased incidence of endometrial, colorectal, and
postmenopausal breast cancer (Larsson and Wolk, 2007; Moghaddam et al., 2007; Reeves et al.,
2007). It is hypothesized that obesity disturbs the physiological function of adipose tissue, which
leads to insulin resistance, chronic inflammation, and dysregulation of adipokine secretion,
factors contributing to the promotion and progression of cancer (Van Kruijsdijk et al., 2009).
Accumulating evidence indicates potential beneficial effects of yogurt consumption on cancers.
A prospective study involving 82,220 Swedish individuals found that the risk for bladder cancer
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
12
was lowest in individuals consuming the highest levels of sour milk and yogurt (RR 0.62, 95%
CI, 0.46-0.85; 2 servings/d vs. 0 serving/d) (Larsson et al., 2008). In another prospective study
in an Italian cohort involving 45,241 volunteers, after adjusting for energy, simple sugar,
calcium, fiber, animal fat, alcohol and red meat intake, body mass index, smoking, education,
and physical activity, the hazard ratio for colorectal cancer in the highest versus lowest tertile of
yogurt intake was 0.65 (95% CI, 0.48-0.89) (Pala et al., 2011). Animal studies support the
beneficial effects of yogurt. For example, LABs from yogurt were shown to effectively inhibit
the genotoxic effects of heterocyclic aromatic amines on rats (Zsivkovits et al., 2003). L.
acidophilus isolated from yogurt reduced tumor growth rate and increased lymphocyte
proliferation in a mouse model of breast cancer (Maroof et al., 2012). A potential mechanism for
reduced cancer risk is lower fecal mutagenicity, as demonstrated by consumption of yogurt with
B. lactis by elderly individuals (Matsumoto et al., 2001). Given these promising results for
yogurt intake and reduced risk for bladder and colon cancers, further work is warranted to
evaluate if yogurt is similarly protective against other cancers.
Diabetes
The worldwide prevalence of diabetes in adults was estimated at 6.4% in 2010, and is
projected to increase to 7.7% by 2030 (Shaw et al., 2010). Excess weight may contribute to 90%
of type 2 diabetes cases (Hossain et al., 2007). More than 197 million people worldwide have
impaired glucose tolerance attributed to obesity or metabolic syndrome (Hossain et al., 2007).
Many studies have illustrated the mechanisms linking obesity and type 2 diabetes. Adipose tissue
has a pivotal role in type 2 diabetes by releasing non-esterified fatty acids (NEFAs), hormones,
and various proinflammatory cytokines (Shoelson et al., 2006). Overabundant intracellular
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
13
NEFAs inhibit key enzymes involved in glucose metabolism (Kahn et al., 2006). Furthermore,
the corresponding intracellular fatty acid metabolites activate the serine/threonine kinase cascade
which disturbs the insulin signaling pathway (Shulman, 2000). To compensate for insulin
resistance, the pancreatic β-cells secrete more insulin, eventually causing endoplasmic reticulum
stress and protein misfolding which lead to β-cell apoptosis (Muoio and Newgard, 2008).
Dairy consumption may reduce risk of type 2 diabetes. For example, an 8 yr prospective
cohort study of 82,076 postmenopausal women demonstrated that low-fat dairy products were
inversely associated with the risk of type 2 diabetes (RR 0.65; 95% CI: 0.44-0.96 for the highest
quintile of intake) (Margolis et al., 2011). A recent meta-analysis of cohort studies showed that
the adjusted relative risk of type 2 diabetes for highest versus lowest quartiles of dairy intake was
0.86 (95% CI, 0.79-0.92) (Tong et al., 2011). A subgroup analysis revealed a relative risk of 0.83
(95% CI, 0.74-0.93) for the intake of yogurt (Tong et al., 2011). A newer prospective study
including 340,234 subjects did not find an association between total dairy products and diabetes.
However, in the dairy subtype analysis, a higher combined intake of fermented dairy products
(cheese, yogurt and thick fermented milk) was inversely associated with diabetes (HR, 0.88; 95%
CI, 0.79-0.99) (Sluijs et al., 2012). In another smaller-sized prospective study, fermented dairy
intake was inversely associated with fasting plasma glucose and HbA1c, although no significant
association between intake and incidence of diabetes was found (Sluijs et al., 2012). Although
epidemiological studies support the beneficial effects of yogurt consumption on reduced type 2
diabetes risk, RCTs are needed to confirm the causal effects of dairy consumption on improved
diabetes outcomes.
OBESITY, YOGURT AND CHRONIC INFLAMMATION
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
14
The anti-inflammatory effects of low-fat dairy products have been well documented
(Sakamoto et al., 2001; Schiffrin et al., 2009; Yang and Sheu, 2012). Inflammation is
characterized by redness, swelling, heat, and pain and is typically resolved shortly after the insult
or stimuli are removed (Hotamisligil, 2006). In contrast, obesity-associated chronic inflammation
is unresolved, low-grade inflammation that originates from metabolic cells (e.g. adipocytes) in
response to excessive nutrient intake (Gregor and Hotamisligil, 2011). Overactive metabolic
signals induce the activation of proinflammatory pathways, which cause low-level induction of
cytokines in metabolic tissues; these inflammatory signals recruit immune cells into metabolic
tissues and disrupt the normal metabolic cell functions (Gregor and Hotamisligil, 2011).
Obesity leads to increased levels of inflammatory biomarkers in a variety of tissues
(Table 3). For example, protein kinases such as JNK and inhibitor of κ kinase (IKK) induce the
expression of proinflammatory cytokines (Solinas and Karin, 2010). Obese women had a
significantly higher amount of phosphorylated (active form) JNK in omental fat compared with
lean women (Bashan et al., 2007). In rodents, Hirosumi et al. observed significant increases in
total JNK activity in liver, muscle and adipose tissues of both dietary and genetic (ob/ob) obesity
models (Hirosumi et al., 2002). Increased activation of JNK and NF-κB pathways were also
detected in the hypothalamus of high-fat-fed mice, accompanied by increased secretion of
proinflammatory cytokines (De Souza et al., 2005). Elevated NF-κB and IKK activities were
found in the livers of both genetic and diet-induced obese mice (Cai et al., 2005). In high-fat-fed
mice, increased IKK activity and downstream products of NF-κB pathway were observed in
lysates of the thoracic aorta (Kim et al., 2007). Therefore, the metabolic and inflammatory
consequences of obesity affect a wide variety of tissues. Animal and a limited number of human
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
15
studies indicate a potential role for dairy or yogurt consumption to mitigate chronic inflammation
associated with obesity, as detailed below.
Increased infiltration of immune cells into metabolic tissues
Obesity increases the infiltration of immune cells into various metabolic tissues.
Macrophages infiltrate adipose tissue in obese individuals and are responsible for nearly all
adipose-derived TNF-α expression (Weisberg et al., 2003). Similarly, obesity leads to increased
inflammatory macrophages in visceral adipose tissue (Curat et al., 2006). Macrophage-derived
proinflammatory cytokines can subsequently initiate insulin resistance and compromise β-cells
(Solinas and Karin, 2010).
Animal models of obesity corroborate the infiltration of macrophages and other
immunocytes. Macrophages and microphages were increased in white adipose tissue in both
genetic and high-fat diet-induced models of obese mice (Xu et al., 2003). Ehses et al. observed
increased islet-associated macrophages in high-fat-fed mice and db/db obese mice (Ehses et al.,
2007). Diet-induced obese mice had increased accumulation of T cells in adipose tissue relative
to lean mice (Wu et al., 2007). Likewise, natural killer T (NKT) cells infiltrated visceral adipose
tissue in high-fat-fed mice (Ohmura et al., 2010). In the same model, depletion of NKT cells
ameliorated visceral adipose tissue inflammation (Ohmura et al., 2010).
Yogurt and LAB can modulate the immune response through cytokine production.
However, studies have not focused on the role of yogurt or dairy on obesity-associated
immunocyte dysregulation. In diet-induced obese mice, compared to the high calcium diet, a
nonfat dry milk-supplemented diet reduced weight gain and associated adipose tissue
inflammation as shown by decreased mRNA abundance of (monocyte chemoattractant protein)
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
16
MCP-1, TNF-α, and IL-6; this suggested that some active components in dairy other than
calcium could modulate the immune response (Thomas et al., 2012).
Yogurt and its associated cultures also have immunostimulatory effects in healthy
individuals. Consumption of yogurt containing L. bulgaricus and S. thermophilus increased
production of IFN-γ by T cells in young adults (Halpern et al., 1991). IFN-γ regulates the
induction of pro-inflammatory cytokines and the activation of macrophages and natural killer
cells. LAB directly stimulates human lymphocyte IFN-γ in vitro (De Simone et al., 1986). An
observational retrospective study showed that supplementation with yogurt containing L.
rhamnosus increased the CD4 count in a group of people living with HIV (Irvine et al., 2010).
Consumption of fermented milk containing L. acidophilus significantly increased the
phagocytosis of E. coli in adults (Schiffrin et al., 1997). Likewise, fermented milk with L. casei,
L. acidophilus, or a mixture of both increased phagocytic lymphocytic activities in Swiss mice
(Perdigon et al., 1995). Oral administration of L. acidophilus alone improved immunoreactivity
of peripheral blood leukocytes and peritoneal phagocytes and enhanced serum antibody response
to orally and systemically administrated antigens in mice (Gill et al., 2000). Since yogurt
consumption in obese individuals does not produce pro-inflammatory effects (Labonté et al.,
2013), further work is needed to identify how yogurt modulates immune cells in obesity, and
whether these effects are localized to the gut or have broader activities at metabolic tissues.
OBESITY, YOGURT, AND INTESTINAL BARRIER FUNCTION
The chronic inflammation associated with obesity may be exacerbated by impaired
intestinal barrier function. Leptin-deficient and hyperleptinemic obese mice have increased
intestinal permeability, modified distribution of junction proteins in the intestinal mucosa, as
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
17
well as increased circulating levels of inflammatory cytokines compared with lean control mice
(Brun et al., 2007). Diet-induced obese mice fed high-fat diets had increased intestinal
permeability assessed by gavage of fluorescent-dextran, increased plasma LPS levels, and
reduced expression of genes for tight junction proteins (Cani et al., 2008). Obese women had
increased paracellular permeability measured by lactulose excretion relative to lean women
(Teixeira et al., 2012). Intestinal paracellular permeability was correlated with waist
circumference and HOMA values (Teixeira et al., 2012). Likewise, intestinal barrier function
was more strongly correlated with central adiposity than BMI in overweight adults (Gummesson
et al., 2011). Dysregulation of intestinal barrier function may be attributed to dysregulation of
gut microbiota, endotoxin exposure, the mucus bilayer, secretory immunoglobulin A (sIgA),
antimicrobial peptides, and tight junction proteins. Emerging evidence supports the ability of
yogurt consumption to modulate these functions, as discussed below.
Dysregulation of gut microbiota
The intestine is essential for nutrient absorption and host defense. Gut microbiota
facilitate these functions by fermenting non-digestible nutrients, vitamin synthesis, and
participating in host defense (Salzman et al., 2007). Favorable gut microbiota may compete with
pathogens for space and nutrients and produce anti-microbial compounds such as bacteriocins
and lactic acids (O'Hara and Shanahan, 2006). Gut microbiota also contribute to energy
homeostasis and fat storage. Interestingly, germ-free mice were protected against diet induced
obesity (Bäckhed et al., 2007). On the other hand, conventionalization of germ-free mice with a
normal microbiota harvested from the cecum of conventionally raised mice caused a 60%
increase in body fat within 14 d despite reduced food consumption (Bäckhed et al., 2004). The
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
18
authors proposed that the gut microbiota helped to absorb monosaccharides from the lumen
which further induced de novo hepatic lipogenesis (Bäckhed et al., 2004). Colonization of germ-
free mice with microbiota from obese mice induced a more significant increase in total body fat
than colonization with microbiota from lean mice (Turnbaugh et al., 2006). This suggested that
the composition of gut microbiota affects the development of obesity. In both mice and humans,
Bacteroidetes and Firmicutes are the major species comprising the microbiota (Bäckhed, 2009).
Obese adults have a lower proportion of Bacteroidetes to Firmicutes than lean, although this
ratio can be improved with weight loss from energy restriction (Ley et al., 2006).
Conventional yogurt cultures have limited viability in the gut and a limited ability to
influence the composition of the gut microbiota. Adults consuming yogurt with S. thermophilus
and L. bulgaricus had less than 103 CFU/g of these cultures in feces (Del Campo et al., 2005). In
another study, participants consumed 125 g of a commercial yogurt twice per day for one week,
providing 108 CFU of S. thermophilus and L. bulgaricus (Elli et al., 2006). S. thermophilus was
not present in feces, although L. bulgaricus was present in about 70% of the fecal samples
provided on days 2 and 7 of the yogurt-consumption period. However, the levels of L. bulgaricus
detected on average did not exceed the 105 CFU/g minimum deemed necessary to exert
beneficial effects (Elli et al., 2006). Another study providing a higher dose of yogurt cultures
(375 g yogurt, 108 CFU/g) for two weeks, reported a median value of approximately 104 CFU
each of S. thermophilus and L. bulgaricus per gram of feces (Mater et al., 2005). Although
yogurt cultures have apparently low viability through the entire gastrointestinal tract, more
information is needed about their small intestine viability.
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
19
Certain probiotic strains may have improved viability in the gut relative to S.
thermophilus and L. bulgaricus. Healthy adults that consumed 230 mL yogurt with additional L.
acidophilus and B. bifidum at 107 CFU/g daily for 10 days had decreased aerobic bacteria and
increased anaerobic bacteria in fecal samples (Chen et al., 1999). Additionally, the bifidus to
coliform ratio favorably increased and B. bifidum was measurable for up to 8 days after
consumption (Chen et al., 1999). In contrast, L. acidophilus, S. thermophilus, and L. bulgaricus,
were not detectable in feces (Chen et al., 1999).
McNulty et al (2011) investigated the effect of B. animalis subsp. lactis on the gut
microbiota of mice and humans. Healthy pairs of monozygotic twins consumed a fermented milk
product (FMP) with L. bulgaricus and B. animalis subsp. lactis or no product daily for seven
weeks. Fecal samples analyzed before, during, and after the intervention did not show a
statistically significant change in the microbiota composition (McNulty et al., 2011).
Additionally, the FMP cultures did not persist in the microbiota longer than two weeks after
ceasing its consumption. In the same study, human-gut-derived bacterial strains and FMP strains
were transplanted into germ-free mice. Similar to humans, the humanized intestinal microbiota
was not drastically altered by FMP, but genes related to carbohydrate metabolism were up-
regulated by FMP consumption (McNulty et al., 2011).
Animal models suggest that yogurt-induced improvements in intestinal permeability are
associated with changes to gut microbiota. In Wistar rat pups, consumption of yogurt with L.
casei counteracted acute gastroenteritis-induced barrier dysfunction (Isolauri et al., 1993). In
atopic dermatitis (AD) patients with increased intestinal permeability, 4 wk consumption of
yogurt with B. lactis, L. bulgaricus, and S. thermophilus increased polyamine-producing
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
20
bacterial species which was associated with improved intestinal barrier function (Matsumoto et
al., 2007).
Cultures used in yogurt may also be modified to improve their viability in the gut by
protecting cultures from stomach acid. For example, yogurt with encapsulated or free L.
acidophilus ATCC 4356 was subjected to a simulated human digestive system (Ortakci and Sert,
2012). Encapsulated L. acidophilus had improved viability up to 2 h of incubation in artificial
human gastric juice.
Thus, conventional yogurt cultures have low to no viability in the gut. Probiotic or
encapsulated strains may have greater viability, and their metabolic effects or competition with
coliforms in the intestine are apparent. These studies suggest that strains may not need to adhere
to the intestinal epithelium and proliferate in order to exert the desired health effects. If this is the
case, it suggests that consistent and prolonged probiotic consumption may be needed to achieve
measurable health benefits from these strains.
Contribution of bacterial endotoxin to chronic inflammation
Gut microbiota contribute to systemic low-grade inflammation by increasing the
exposure to proinflammatory bacterial products, especially the Gram-negative-derived LPS
among others (Okamura et al., 2001; Rallabhandi et al., 2006). LPS typically consists of a
hydrophobic domain known as lipid A, a non-repeating core oligosaccharide, and a distal
polysaccharide (Raetz and Whitfield, 2002). LPS initiates inflammatory signaling through LPS
binding protein (LBP), CD14, Toll-like receptor-4 (TLR-4) and MD-2. LBP is thought to extract
LPS and subsequently deliver it to CD14 or lipoprotein; the former may lead to the activation of
target cells while the latter may result in the clearance by liver (Van Bossuyt et al., 1988). CD14
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
21
serves as a pattern-recognition receptor in proinflammatory signaling which can be stimulated by
various ligands (Park et al., 2009; Pugin et al., 1994). MD-2 physically associates with TLR-4 on
the cell surface and acts as co-receptor with TLR-4 for the detection of LPS (Manco et al., 2010).
Once activated by LPS, TLR-4 undergoes oligomerization and recruits its two adaptor protein
pairs, TRAM-TRIF and MAL-MyD88, ultimately activating the NF-κB pathway (Manco et al.,
2010). Human and animal studies have shown LPS as a strong inducer of proinflammatory
cytokines such asIL-6 and TNF in most tissues including adipocytes (Andreasen et al., 2010;
Cani et al., 2007; Creely et al., 2007; Kemna et al., 2005; Stoll et al., 2004).
Overweight and obese adults have increased endotoxin exposure (Sun et al., 2010). Acute
and chronic fat consumption is associated with increased exposure to endotoxin. A cross-
sectional study of 201 healthy French men reported that total energy and fat, but not
carbohydrate or protein were correlated with plasma LPS (Amar et al., 2008). These observations
were confirmed in mice and indicated fat was more efficient in facilitating translocation of LPS
into circulation than carbohydrate (Amar et al., 2008). In addition, a single high-fat meal can
induce postprandial endotoxemia and inflammation (Erridge et al., 2007; Ghanim et al., 2010;
Laugerette et al., 2011). Obesity-associated dysregulation of gut microbiota may also increase
endotoxin exposure (Cani et al., 2007; Cani et al., 2008).
Preliminary studies in elderly individuals have demonstrated that yogurt consumption
inhibits markers of endotoxin exposure in elderly individuals (Schiffrin et al., 2009). Elderly
individuals (n = 23) with small-intestinal bacterial overgrowth consumed 300 g/d of yogurt with
109 CFU L. johnsonii La1 for 4 wk. By the end of the trial, yogurt consumption decreased
plasma levels of LBP and sCD14, LPS pattern recognition receptors in elderly with small-
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
22
intestinal bacterial overgrowth (Schiffrin et al., 2009). Furthermore, yogurt consumption also
reduced plasma endotoxin in healthy elderly participants (Schiffrin et al., 2009). Small intestinal
bacterial overgrowth may affect 41% of obese individuals (Jouet et al., 2011). Therefore, further
studies are warranted to evaluate the ability of yogurt to reduce endotoxin exposure in obese
individuals.
Mucus bilayer
The mucus bilayer is produced by goblet cells and separates gut microbiota from
endothelial cells. This bilayer is formed by a mesh-like structure of mucins, high molecular
weight glycoproteins with increased hydration capacity due to negative surface charges
(Dharmani et al., 2009). Mucins lubricate and maintain the hydrated layer of the epithelium, as
well as create a permeable unstirred, gel-like layer that facilitates nutrient exchange (Dharmani et
al., 2009). The mucus bilayer is essential to the innate host defense. The outer mucus layer
provides space and nutrients for the residence of commensal microflora which might inhibit the
growth and invasion of pathogens; the inner layer is impervious to bacteria and acts like a
protective barrier for the epithelium (Kim and Ho, 2010; Turner, 2009). The goblet cells also
produce intestinal trefoil factor and resistin-like molecule-β, proteins that strengthen the barrier
by stabilizing the mucin polymers or regulating mucin secretions (Dharmani et al., 2009).
Additionally, the mucus layer contains other defensive components such as secretory IgA and
antimicrobial peptides.
Probiotics associated with yogurt may stimulate the production of intestinal mucins and
improve host defense. For example, L. plantarum 299 v incubated with HT-29 intestinal
epithelial cells increased mucin mRNA expression and inhibited the adherence of an attaching
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
23
and effecting pathogenic E. coli. in vitro (Mack et al., 1999). Similarly, in Wistar rats, 7 d
consumption of Lactobacilli, Bifidobacteria, and Streptococci increased basal luminal mucin
content by 60% (Caballero-Franco et al., 2007). Whey peptides derived from α- and β- caseins
also increased mucin secretion in HT29-MTX cells (Martínez-Maqueda et al., 2012). Thus, this
emerging evidence suggests that dairy products or their associated probiotics could be of benefit
to the mucus bilayer. A recent diet-induced obese mice model showed that obesity was
associated with decreased mucus layer thickness due to the decreased level of Akkermansia
muciniphila, a mucus layer resident that played an essential role in mucus turnover (Everard et
al., 2013). Thus, it appears worthwhile to further investigate the effects of yogurt on the obese-
compromised mucus layer.
Secretory IgA (sIgA)
sIgA is the major effector of the mucosa-associated lymphoid tissue (MALT) and
protects against commensal bacterial penetration of the lumen (Brandtzaeg et al., 1999). MALT
consists of lymphocytes such as T cells and B cells, as well as plasma cells and macrophages,
which are stimulated by antigens. sIgA, dimeric or polymeric IgA, is produced by plasma cells in
the intestinal mucosa and is the predominant antibody class in the intestinal lumen (Woof and
Ken, 2006; Macpherson and Uhr, 2004). Interstitial sIgA inhibits pathogens and toxins by 1)
preventing the adhesion and entry of pathogens and toxins by interfering with epithelial receptor
recognition, 2) binding pathogens and promoting their clearance, or 3) inhibiting virus
production (Corthésy, 2007). Low serum IgA may indicate compromised immune function,
while high serum IgA is associated with chronic inflammation, central adiposity, and advanced
age (Gonzalez-Quintela et al., 2008).
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
24
Yogurt consumption appears to modulate the gut immune function by increasing sIgA.
For example, consumption of yogurt with L. acidophilus by 30 healthy adults increased total
serum IgA and the production of specific serum IgA against an attenuated strain of S.
typhimurium (Link-Amster et al., 1994). Consuming 400 mL yogurt with L. acidophilus daily for
4 wk reduced H. pylori and increased the serum IgA level in 38 infected children (Yang and
Sheu, 2012). Rodent studies also support the IgA-promoting effects of yogurt. In a mouse model,
orally administrated LAB alone and in yogurt increased the intestinal IgA producing cells and
IgA (Perdigon et al., 1995). Furthermore, a 7 d yogurt treatment partially prevented the infection
of S. typhimurium and inhibited intestinal carcinomas induced by 1-2-dimethylhydrazine
(Perdigon et al., 1995). Similarly, mice fed yogurt for 4 wk had increased serum IgA after a S.
typhimurium challenge, relative to the milk-treated control group (Puri et al., 1996). Thus, both
animal and human studies have demonstrated induction of sIgA defenses following yogurt
consumption, which may improve immunity.
Antimicrobial peptides
Defensins are antimicrobial peptides secreted by Paneth cells located in the crypts of the
small intestinal mucosa (Porter et al., 2002). Defensins have bactericidal activity against various
Gram-positive and Gram-negative bacteria (Salzman et al., 2007). These peptides facilitate
bacterial membrane collapse through electrostatic and hydrophobic interactions (Zasloff, 2002).
Paneth cell function and defensin levels are compromised in obese individuals, which may be
explained by activated unfolded protein response in the intestine (Hodin et al., 2011). In healthy
women, consumption of 200 mL yogurt with or without B. lactis Bb12 for 3 wk did not alter
fecal β-defensin-2, although both treatments increased fecal sIgA from baseline (Kabeerdoss et
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
25
al., 2011). However, the probiotic Lactobacillus and E.coli Nissle 1917 increased β-defensin 2
expression in Caco-2 cells (Schlee et al., 2008; Schlee et al., 2007). Further work is needed to
determine if dietary approaches are feasible to overcome obesity-compromised defensin
production.
Mucosal cells and tight junctions
The innermost layer of the intestine is a monolayer of enterocytes, endocrine cells,
microfold cells, G cells, and Paneth cells (Scaldaferri et al., 2012). Enterocytes are the most
abundant cells and are connected by apical junctions, which are mainly adherent or tight
junctions (Hartsock and Nelson, 2008). Adherent junctions consist of the transmembrane protein
E-cadherin and the catenin family members, including p120-catenin, β-catenin, and α-catenin
(Hartsock and Nelson, 2008). Adherent junctions initiate and stabilize cell-cell adhesion, regulate
the actin cytoskeleton, and contribute to intracellular signaling (Hartsock and Nelson, 2008).
Tight junctions are composed of occludins, claudins, and junction adhesion molecules (JAM),
transmembrane proteins that are linked to the cytoskeleton through zonula occludens (ZO)
scaffolding proteins (Hartsock and Nelson, 2008). Tight junctions are the primary barrier to
intestinal intercellular space, but are not impermeable. The paracellular pathway is selective to
ions and other small molecules, and depends on the cell type (Tsukita et al., 2001).
Increased plasma endotoxin levels suggest that obese individuals have compromised
intestinal barrier function (Sun et al., 2010). Compromised intestinal barrier function is proposed
to contribute to chronic inflammation in obesity by initiating inflammation through endotoxin
exposure (Cani et al., 2007). The perturbation of proinflammatory cytokines, gastrointestinal
peptides, and endocannabinoids associated with obesity can compromise tight junctions (Bluher
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
26
et al., 2006; Cluny et al., 2012; Côté et al., 2007). In rodents, glucagon-like peptide (GLP)-2
protects barrier function, while melatonin can increase permeability (Cameron and Perdue, 2005;
Cameron et al., 2003; Sommansson et al., 2013). IFN-γ (Bruewer et al., 2005; Clark et al., 2005;
Yang et al., 2002; Youakim and Ahdieh, 1999), TNF-α (Al-Sadi et al., 2009; Li et al., 2008;
Mankertz et al., 2000; Schmitz et al., 1999), and IL-6 (Al-Sadi and Ma, 2007; Yang et al., 2003)
can disrupt barrier function. In contrast, IL-10 (Madsen et al., 1997; Oshima et al., 2001),
transforming growth factor (TGF)-β (Howe et al., 2005), and IL-17 (Kinugasa et al., 2000)
improve barrier function in human T84 colonic epithelial cells. Obese ob/ob mice had improved
barrier function and lower plasma LPS when treated with a CB receptor 1 antagonist (Muccioli
et al., 2010).
Yogurt and its associated probiotics may improve intestinal barrier function by
maintaining the expression of tight junction proteins. Yogurt with B. lactis prevented the
increase in intestinal permeability induced by partial restraint stress in rats, and restored occludin
and JAM-A expression (Agostini et al., 2012). In addition, calcium, which is rich in yogurt,
plays a critical role in tight junction biogenesis and supplementation of calcium was shown to be
able to inhibit alteration in tight junction function in a diabetic rat model (Leal et al., 2010; Stuart
et al., 1994). Therefore, these rodent studies suggest that dairy calcium or probiotic yogurt could
be beneficial for maintaining function of tight junctions.
OTHER POTENTIAL BENEFITS OF YOGURT CONSUMPTION ON GUT HEALTH
Increased yogurt consumption has the potential to improve intestinal health, ameliorate
lactose intolerance, prevent constipation and diarrheal diseases, decrease allergies in vulnerable
populations, and reduce the risks of colon cancer and inflammatory bowel diseases (Adolfsson et
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
27
al., 2004; Parvez et al., 2006). The mechanisms for these actions are not fully described, but may
include modulating gut pH, inhibiting the proliferation and adhesion of pathogenic bacteria,
secreting antibacterial substances, and regulating immune function.
Lactose intolerance
In lactase-deficient individuals, lactose enters the colon and is fermented by colonic
bacteria. The colonic metabolites of lactose include short-chain fatty acids which, together with
electrolytes, introduce an osmotic load that can cause diarrhea and discomfort (Lomer et al.,
2008). In a cross-sectional study, subjects with self-perceived lactose intolerance had a
significantly lower calcium intake from dairy foods and reported higher rate of physician-
diagnosed diabetes and hypertension (Nicklas et al., 2011). Early studies indicated that subjects
with lactase deficiency had better digestion and absorption of lactose from yogurt than the
lactose in milk (Kolars et al., 1984). After ingestion of around 18 g of lactose in water, milk, or
yogurt, subjects receiving yogurt had only one third of the hydrogen excretion, an indicator of
undigested lactose, compared with those receiving lactose in water or milk (Kolars et al., 1984).
Furthermore, the consumption of yogurt led to fewer symptoms of diarrhea and flatulence
relative to milk (Kolars et al., 1984).
Diarrhea
Diarrhea is the leading cause of morbidity and death of children in developing countries
(Boschi-Pinto et al., 2008). Emerging evidence suggests that consumption of yogurt and its
related probiotic cultures prevent or treat diarrhea. In a double-blind, placebo-controlled trial,
infants that received formula with B. bifidum and S. thermophilus reduced the incidence of acute
diarrhea and rotavirus shedding (Saavedra et al., 1994). A meta-analysis of RCTs published from
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
28
1966 to 2000 suggested that Lactobacillus supplementation (108 to 1011 CFU daily) safely
reduced the frequency and duration of acute infectious diarrhea in children (Van Niel et al.,
2002). Moreover, a more recent meta-analysis of RCTs showed that administration of
Lactobacillus through capsules or fermented milk during antibiotic treatment significantly
reduced the risk of developing antibiotic-associated diarrhea (RR 0.35, 0.19-0.67 95% CI) (Kale-
Pradhan et al., 2010). However, the risk reduction was only significant in adults after subgroup
analysis (Kale-Pradhan et al., 2010).
H. pylori infection
Consumption of yogurt with L. gasseri for 8 wk significantly suppressed H. pylori-
induced gastric mucosal inflammation in the elderly (Sakamoto et al., 2001). In children affected
by H. pylori, yogurt consumption decreased serum IL-6 level after 4 wk (Yang and Sheu, 2012).
Inhibition of Colitis
The prevalence of the inflammatory bowel diseases (IBD) Crohns disease (CD) and
ulcerative colitis (UC) is increasing in industrialized nations, and although the cause(s) are
unknown, they likely result from an aberrant immune response to intestinal microbiota (Chaves
et al., 2011). Probiotics administered to murine models of IBD improve disease outcomes; this
has been reviewed elsewhere (Claes et al., 2011). Yogurt consumption also inhibits experimental
IBD in mice. Consumption of yogurt with eight L. bulgaricus strains and two S. thermophilus
strains decreased mortality rate and prevented intestinal inflammation and tissue damage in mice
with trinitrobenzene sulfonic acid (TNBS)-induced intestinal inflammation (Chaves et al., 2011).
Yogurt consumption prevented an increase in colonic CD4+ and CD8+ T cell numbers, decreased
TLR-4 positive cells at 14 days, but not 3 or 7 days post TNBS administration (Chaves et al.,
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
29
2011). Yogurt without added probiotic strains inhibited TNBS-induced colitis in mice, increased
the number of IgA producing cells, and decreased CD8+ T cells two wk after TNBS
administration (Gobbato et al., 2008).
Clinical studies have mixed outcomes for the probiotic treatment of IBD and are strain-
dependent (Hedin et al., 2007; Jonkers et al., 2012; Kato et al., 2004; Lorea Baroja et al., 2007;
Miele et al., 2009; Sood et al., 2009). Clinical studies have not used conventional yogurt as an
intervention for IBD, despite self-reported benefits of yogurt reported by IBD patients (Cohen et
al., 2013). Consumption of yogurt with L rhamnosus GR-1 and L. reuteri RC-14 improved
markers of inflammation in monocytes from 20 patients with IBD, including increasing
CD4+CD25 high T cells (Lorea Baroja et al., 2007). Yogurt could be an effective delivery vehicle
for probiotic strains for treatment of IBD. However, more work is needed to identify clinically-
significant probiotic strains for inhibiting colonic inflammation.
Appetite control
Obesity is a result of positive energy balance. Some studies have demonstrated that
yogurt might help reduce energy intake by suppressing appetite. For example, consumption of
yogurt either in semisolid or liquid form led to lower hunger and higher fullness feeling,
compared with a fruit drink or dairy fruit drink (Tsuchiya et al., 2006). Similarly, subjects felt
higher satiety after consumption of yogurt as evidenced by rating of hunger, appetite, desire to
eat, and fullness, compared with ingestion of chocolate bars (Chapelot and Payen, 2010). Yogurt
consumption also suppressed appetite rating and reduced subsequent food intake or delayed
subsequent eating, compared with isovolumetric water (Dougkas et al., 2012; Douglas et al.,
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
30
2013). Therefore, yogurt consumption may provide a further benefit of appetite-suppression,
although the molecular mechanisms for this effect remain uncharacterized.
CONCLUSION
Chronic inflammation is a hallmark of obesity and partially explains the increased risk of
chronic disease in obese individuals. Altered gut microbiota and impaired intestinal barrier
function contribute to the chronic inflammation associated with obesity. Animal models and a
limited number of clinical studies demonstrate that dairy and yogurt consumption reduce chronic
inflammation. New evidence from animal studies indicates that the beneficial effects of yogurt
consumption might also derive from its effects on intestinal barrier function. However, there is
no clinical evidence for the effects of yogurt consumption on inflammation and gut barrier
function in the obese population. The benefits of yogurt for lactose intolerance are well-
established, and emerging evidence supports the ability of yogurt to modulate the gut immunity
and barrier function. Yogurt consumption is beneficial for intestinal health by restoring normal
gut microbiota and suppressing inflammation. Further studies are needed to isolate the effects of
conventional yogurt and yogurt fortified with probiotics, considering that yogurt is a vehicle for
nutrients and other bioactive components. Research investigating the effects of yogurt
consumption on inflammation and intestinal barrier function in the obese population may yield
further insight to the mechanism(s) for its anti-inflammatory effects.
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
31
References
Adolfsson, O., Meydani, S. N. and Russell, R. M. (2004). Yogurt and gut function. Am. J. Clin.
Nutr. 80: 245-256.
Agostini, S., Goubern, M., Tondereau, V., Salvador-Cartier, C., Bezirard, V., Lévèque, M., et al.
(2012). A marketed fermented dairy product containing Bifidobacterium lactis CNCM I-
2494 suppresses gut hypersensitivity and colonic barrier disruption induced by acute stress in
rats. Neurogastroenterol. Motil. 24: 376-e172.
Al-Sadi, R., Boivin, M. and Ma, T. (2009). Mechanism of cytokine modulation of epithelial tight
junction barrier. Front. Biosci. 14: 2765-2778.
Al-Sadi, R. M. and Ma, T. Y. (2007). IL-1β causes an increase in intestinal epithelial tight
junction permeability. J. Immunol. 178: 4641-4649.
Amar, J., Burcelin, R., Ruidavets, J. B., Cani, P. D., Fauvel, J., Alessi, M. C., et al. (2008).
Energy intake is associated with endotoxemia in apparently healthy men. Am. J. Clin. Nutr.
87: 1219-1223.
Anderson, J. W. and Gilliland, S. E. (1999). Effect of fermented milk (yogurt) containing
Lactobacillus acidophilus L1 on serum cholesterol in hypercholesterolemic humans. J. Am.
Coll. Nutr. 18: 43-50.
Andreasen, A. S., Larsen, N., Pedersen-Skovsgaard, T., Berg, R. M. G., Mller, K., Svendsen, K.
D., et al. (2010). Effects of Lactobacillus acidophilus NCFM on insulin sensitivity and the
systemic inflammatory response in human subjects. Br. J. Nutr. 104: 1831-1838.
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
32
Aneja, R. P. and Murthi, T. N. (1990). Conjugated linoleic acid contents of Indian curds and
ghee. Indian J. Dairy Sci. 43: 231-238.
Appel, L. J., Moore, T. J., Obarzanek, E., Vollmer, W. M., Svetkey, L. P., Sacks, F. M., et al.
(1997). A clinical trial of the effects of dietary patterns on blood pressure. N. Engl. J. Med.
336: 1117-1124.
Bäckhed, F. (2009). Changes in intestinal microflora in obesity: Cause or consequence? J.
Pediatr. Gastroenterol. Nutr. 48: S56-S57.
Bäckhed, F., Ding, H., Wang, T., Hooper, L. V., Gou, Y. K., Nagy, A., et al. (2004). The gut
microbiota as an environmental factor that regulates fat storage. Proc. Natl. Acad. Sci. U. S.
A. 101: 15718-15723.
Bäckhed, F., Manchester, J. K., Semenkovich, C. F. and Gordon, J. I. (2007). Mechanisms
underlying the resistance to diet-induced obesity in germ-free mice. Proc. Natl. Acad. Sci. U.
S. A. 104: 979-984.
Bashan, N., Dorfman, K., Tarnovscki, T., Harman-Boehm, I., Liberty, I. F., Her, M. B., et al.
(2007). Mitogen-activated protein kinases, inhibitory-κB kinase, and insulin signaling in
human omental versus subcutaneous adipose tissue in obesity. Endocrinology 148: 2955-
2962.
Bluher, M., Engeli, S., Kloting, N., Berndt, J., Fasshauer, M., Batkai, S., et al. (2006).
Dysregulation of the peripheral and adipose tissue endocannabinoid system in human
abdominal obesity. Diabetes 55: 3053-3060.
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
33
Boschi-Pinto, C., Velebit, L. and Shibuya, K. (2008). Estimating child mortality due to diarrhoea
in developing countries. Bull. World Health Organ. 86: 710-717.
Brake, D. K., Smith, E. O. B., Mersmann, H., Smith, C. W. and Robker, R. L. (2006). ICAM-1
expression in adipose tissue: Effects of diet-induced obesity in mice. Am. J. Physiol. Cell
Physiol. 291: C1232-C1239.
Brandtzaeg, P., Baekkevold, E. S., Farstad, I. N., Jahnsen, F. L., Johansen, F. E., Nilsen, E. M.,
et al. (1999). Regional specialization in the mucosal immune system: What happens in the
microcompartments? Immunol. Today 20: 141-151.
Breslaw, E. S. and Kleyn, D. H. (1973). In vitro digestibility of protein in yogurt at various
stages of processing. J. Food Sci. 38: 1016-1021.
Bruewer, M., Utech, M., Ivanov, A. I., Hopkins, A. M., Parkos, C. A. and Nusrat, A. (2005).
Interferon-γ induces internalization of epithelial tight junction proteins via a
macropinocytosis-like process. FASEB J. 19: 923-933.
Brun, P., Castagliuolo, I., Di Leo, V., Buda, A., Pinzani, M., Palù, G., et al. (2007). Increased
intestinal permeability in obese mice: New evidence in the pathogenesis of nonalcoholic
steatohepatitis. Am. J. Physiol. Gastrointest. Liver Physiol. 292: G518-G525.
Buttriss, J. (1997). Nutritional properties of fermented milk products. Int. J. Dairy Technol. 50:
21-27.
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
34
Caballero-Franco, C., Keller, K., De Simone, C. and Chadee, K. (2007). The VSL#3 probiotic
formula induces mucin gene expression and secretion in colonic epithelial cells. Am. J.
Physiol. Gastrointest. Liver Physiol. 292: G315-G322.
Cai, D., Yuan, M., Frantz, D. F., Melendez, P. A., Hansen, L., Lee, J., et al. (2005). Local and
systemic insulin resistance resulting from hepatic activation of IKK-β and NF-κB. Nat. Med.
11: 183-190.
Calle, E. E., Rodriguez, C., Walker-Thurmond, K. and Thun, M. J. (2003). Overweight, obesity,
and mortality from cancer in a prospectively studied cohort of U.S. Adults. N. Engl. J. Med.
348: 1625-1638.
Cameron, H. L. and Perdue, M. H. (2005). Stress impairs murine intestinal barrier function:
Improvement by glucagon-like peptide-2. J. Pharmacol. Exp. Ther. 314: 214-220.
Cameron, H. L., Yang, P. C. and Perdue, M. H. (2003). Glucagon-like peptide-2-enhanced
barrier function reduces pathophysiology in a model of food allergy. Am. J. Physiol.
Gastrointest. Liver Physiol. 284: G905-G912.
Cani, P. D., Amar, J., Iglesias, M. A., Poggi, M., Knauf, C., Bastelica, D., et al. (2007).
Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 56: 1761-1772.
Cani, P. D., Bibiloni, R., Knauf, C., Waget, A., Neyrinck, A. M., Delzenne, N. M., et al. (2008).
Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat
diet-induced obesity and diabetes in mice. Diabetes 57: 1470-1481.
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
35
Chapelot, D. and Payen, F. (2010). Comparison of the effects of a liquid yogurt and chocolate
bars on satiety: A multidimensional approach. Br. J. Nutr. 103: 760-767.
Chaves, S., Perdigon, G. and De Moreno De Leblanc, A. (2011). Yoghurt consumption regulates
the immune cells implicated in acute intestinal inflammation and prevents the recurrence of
the inflammatory process in a mouse model. J. Food Prot. 74: 801-811.
Chen, R. M., Wu, J. J., Lee, S. C., Huang, A. H. and Wu, H. M. (1999). Increase of intestinal
Bifidobacterium and suppression of coliform bacteria with short-term yogurt ingestion. J.
Dairy Sci. 82: 2308-2314.
Chobanian, A. V., Bakris, G. L., Black, H. R., Cushman, W. C., Green, L. A., Izzo Jr, J. L., et al.
(2003). The Seventh Report of the Joint National Committee on Prevention, Detection,
Evaluation, and Treatment of High Blood Pressure: The JNC 7 Report. JAMA. 289: 2560-
2572.
Claes, I. J. J., De Keersmaecker, S. C. J., Vanderleyden, J. and Lebeer, S. (2011). Lessons from
probiotic-host interaction studies in murine models of experimental colitis. Mol. Nutr. Food
Res. 55: 1441-1453.
Clark, E., Hoare, C., Tanianis-Hughes, J., Carlson, G. L. and Warhurst, G. (2005). Interferon γ
induces translocation of commensal Escherichia coli across gut epithelial cells via a lipid
raft-mediated process. Gastroenterology 128: 1258-1267.
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
36
Cluny, N. L., Reimer, R. A. and Sharkey, K. A. (2012). Cannabinoid signalling regulates
inflammation and energy balance: The importance of the brain-gut axis. Brain Behav.
Immun. 26: 691-698.
Cohen, A. B., Lee, D., Long, M. D., Kappelman, M. D., Martin, C. F., Sandler, R. S., et al.
(2013). Dietary patterns and self-reported associations of diet with symptoms of
inflammatory bowel disease. Dig. Dis. Sci. 58: 1322-1328.
Corthésy, B. (2007). Roundtrip ticket for secretory IgA: Role in mucosal homeostasis? J.
Immunol. 178: 27-32.
Côté, M., Matias, I., Lemieux, I., Petrosino, S., Alméras, N., Després, J. P., et al. (2007).
Circulating endocannabinoid levels, abdominal adiposity and related cardiometabolic risk
factors in obese men. Int. J. Obes (Lond). 31: 692-699.
Creely, S. J., McTernan, P. G., Kusminski, C. M., Fisher, F. M., Da Silva, N. F., Khanolkar, M.,
et al. (2007). Lipopolysaccharide activates an innate immune system response in human
adipose tissue in obesity and type 2 diabetes. Am. J. Physiol. Endocrinol. Metab. 292: E740-
E747.
Crittenden, R. G., Martinez, N. R. and Playne, M. J. (2003). Synthesis and utilisation of folate by
yoghurt starter cultures and probiotic bacteria. Int. J. Food Microbiol. 80: 217-222.
Curat, C. A., Wegner, V., Sengenès, C., Miranville, A., Tonus, C., Busse, R., et al. (2006).
Macrophages in human visceral adipose tissue: Increased accumulation in obesity and a
source of resistin and visfatin. Diabetologia 49: 744-747.
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
37
De Paula, R. B., Da Silva, A. A. and Hall, J. E. (2004). Aldosterone Antagonism Attenuates
Obesity-Induced Hypertension and Glomerular Hyperfiltration. Hypertension 43: 41-47.
De Simone, C., Bianchi Salvadori, B. and Negri, R. (1986). The adjuvant effect of yogurt on
production of gamma-interferon by Con A-stimulated human peripheral blood lymphocytes.
Nutr. Rep. Int. 33: 419-433.
De Souza, C. T., Araujo, E. P., Bordin, S., Ashimine, R., Zollner, R. L., Boschero, A. C., et al.
(2005). Consumption of a fat-rich diet activates a proinflammatory response and induces
insulin resistance in the hypothalamus. Endocrinology 146: 4192-4199.
Del Campo, R., Bravo, D., Cantón, R., Ruiz-Garbajosa, P., García-Albiach, R., Montesi-Libois,
A., et al. (2005). Scarce evidence of yogurt lactic acid bacteria in human feces after daily
yogurt consumption by healthy volunteers. Appl. Environ. Microbiol. 71: 547-549.
Desobry-Banon, S., Vetier, N. and Hardy, J. (1999). Health benefits of yogurt consumption. A
review. Int. J. Food Prop. 2: 1-12.
Dharmani, P., Srivastava, V., Kissoon-Singh, V. and Chadee, K. (2009). Role of intestinal
mucins in innate host defense mechanisms against pathogens. J. Innate Immun. 1: 123-135.
Dougkas, A., Minihane, A. M., Givens, D. I., Reynolds, C. K. and Yaqoob, P. (2012).
Differential effects of dairy snacks on appetite, but not overall energy intake. Br. J. Nutr.
108: 2274-2285.
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
38
Douglas, S. M., Ortinau, L. C., Hoertel, H. A. and Leidy, H. J. (2013). Low, moderate, or high
protein yogurt snacks on appetite control and subsequent eating in healthy women. Appetite
60: 117-122.
Dunlap, B. S., Yu, H. and Elitsur, Y. (2009). The probiotic content of commercial yogurts in
West Virginia. Clin. Pediatr. (Phila). 48: 522-527.
Ebringer, L., Ferenčík, M. and Krajčovič, J. (2008). Beneficial health effects of milk and
fermented dairy products - Review. Folia Microbiol. 53: 378-394.
Egan, B. M., Zhao, Y. and Axon, R. N. (2010). US trends in prevalence, awareness, treatment,
and control of hypertension, 1988-2008. JAMA. 303: 2043-2050.
Ehses, J. A., Perren, A., Eppler, E., Ribaux, P., Pospisilik, J. A., Maor-Cahn, R., et al. (2007).
Increased number of islet-associated macrophages in type 2 diabetes. Diabetes 56: 2356-
2370.
Elli, M., Callegari, M. L., Ferrari, S., Bessi, E., Cattivelli, D., Soldi, S., et al. (2006). Survival of
yogurt bacteria in the human gut. Appl. Environ. Microbiol. 72: 5113-5117.
Erridge, C., Attina, T., Spickett, C. M. and Webb, D. J. (2007). A high-fat meal induces low-
grade endotoxemia: Evidence of a novel mechanism of postprandial inflammation. Am. J.
Clin. Nutr. 86: 1286-1292.
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
39
Everard, A., Belzer, C., Geurts, L., Ouwerkerk, J. P., Druart, C., Bindels, L. B., et al. (2013).
Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced
obesity. Proc. Natl. Acad. Sci. U. S. A. 110: 9066-9071.
Fabian, E. and Elmadfa, I. (2006). Influence of daily consumption of probiotic and conventional
yoghurt on the plasma lipid profile in young healthy women. Ann. Nutr. Metab. 50: 387-393.
Flegal, K. M., Carroll, M. D., Ogden, C. L. and Curtin, L. R. (2010). Prevalence and trends in
obesity among US adults, 1999-2008. JAMA. 303: 235-241.
Gaudichon, C., Roos, N., Mahe, S., Sick, H., Bouley, C. and Tome, D. (1994). Gastric emptying
regulates the kinetics of nitrogen absorption from 15N- labeled milk and 15N-labeled yogurt
in miniature pigs. J. Nutr. 124: 1970-1977.
German, J. B., Gibson, R. A., Krauss, R. M., Nestel, P., Lamarche, B., Van Staveren, W. A., et
al. (2009). A reappraisal of the impact of dairy foods and milk fat on cardiovascular disease
risk. Eur. J. Nutr. 48: 191-203.
Ghanim, H., Sia, C. L., Upadhyay, M., Korzeniewski, K., Viswanathan, P., Abuaysheh, S., et al.
(2010). Orange juice neutralizes the proinflammatory effect of a high-fat, high-carbohydrate
meal and prevents endotoxin increase and toll-like receptor expression. Am. J. Clin. Nutr. 91:
940-949.
Gill, H. S., Rutherfurd, K. J., Prasad, J. and Gopal, P. K. (2000). Enhancement of natural and
acquired immunity by Lactobacillus rhamnosus (HN001), Lactobacillus acidophilus
(HN017) and Bifidobacterium lactis (HN019). Br. J. Nutr. 83: 167-176.
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
40
Gilliland, S. E. and Speck, M. L. (1977). Instability of Lactobacillus acidophilus in Yogurt. J.
Dairy Sci. 60: 1394-1398.
Gobbato, N., Rachid, M. and Perdigón, G. (2008). Anti-inflammatory effect of yoghurt in an
experimental inflammatory bowel disease in mouse. J. Dairy Res. 75: 497-504.
Gonzalez-Quintela, A., Alende, R., Gude, F., Campos, J., Rey, J., Meijide, L. M., et al. (2008).
Serum levels of immunoglobulins (IgG, IgA, IgM) in a general adult population and their
relationship with alcohol consumption, smoking and common metabolic abnormalities. Clin
Exp Immunol 151: 42-50.
Goodfriend, T. L. and Calhoun, D. A. (2004). Resistant Hypertension, Obesity, Sleep Apnea, and
Aldosterone: Theory and Therapy. Hypertension 43: 518-524.
Gorbach, S. L. (1990). Lactic acid bacteria and human health. Ann. Med. 22: 37-41.
Gregor, M. F. and Hotamisligil, G. S. (2011). Inflammatory mechanisms in obesity. Annu. Rev.
Immunol. 29: 415-445.
Grundy, S. M. (2007). Metabolic syndrome: A multiplex cardiovascular risk factor. J. Clin.
Endocrinol. Metab. 92: 399-404.
Guarner, F., Perdigon, G., Corthier, G., Salminen, S., Koletzko, B. and Morelli, L. (2005).
Should yoghurt cultures be considered probiotic? Br. J. Nutr. 93: 783-786.
Gueguen, L. and Pointillart, A. (2000). The bioavailability of dietary calcium. J. Am. Coll. Nutr.
19: 119S-136S.
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
41
Gummesson, A., Carlsson, L. M. S., Storlien, L. H., Bäckhed, F., Lundin, P., Löfgren, L., et al.
(2011). Intestinal permeability is associated with visceral adiposity in healthy women.
Obesity 19: 2280-2282.
Halpern, G. M., Vruwink, K. G., Van De Water, J., Keen, C. L. and Gershwin, M. E. (1991).
Influence of long-term yoghurt consumption in young adults. Int. J. Immunother. 7: 205-210.
Hartsock, A. and Nelson, W. J. (2008). Adherens and tight junctions: Structure, function and
connections to the actin cytoskeleton. Biochim. Biophys. Acta 1778: 660-669.
Haslam, D. W. and James, W. P. T. (2005). Obesity. Lancet 366: 1197-1209.
Hedin, C., Whelan, K. and Lindsay, J. O. (2007). Evidence for the use of probiotics and
prebiotics in inflammatory bowel disease: A review of clinical trials. Proc. Nutr. Soc. 66:
307-315.
Herder, C., Schneitler, S., Rathmann, W., Haastert, B., Schneitler, H., Winkler, H., et al. (2007).
Low-grade inflammation, obesity, and insulin resistance in adolescents. J. Clin. Endocrinol.
Metab. 92: 4569-4574.
Hirosumi, J., Tuncman, G., Chang, L., Görgün, C. Z., Uysal, K. T., Maeda, K., et al. (2002). A
central, role for JNK in obesity and insulin resistance. Nature 420: 333-336.
Hodin, C. M., Verdam, F. J., Grootjans, J., Rensen, S. S., Verheyen, F. K., Dejong, C. H. C., et
al. (2011). Reduced Paneth cell antimicrobial protein levels correlate with activation of the
unfolded protein response in the gut of obese individuals. J. Pathol. 225: 276-284.
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
42
Holes-Lewis, K. A., Malcolm, R. and O'Neil, P. M. (2013). Pharmacotherapy of obesity: Clinical
treatments and considerations. Am. J. Med. Sci. 345: 284-288.
Hossain, P., Kawar, B. and El Nahas, M. (2007). Obesity and diabetes in the developing world -
A growing challenge. N. Engl. J. Med. 356: 213-215.
Hotamisligil, G. S. (2006). Inflammation and metabolic disorders. Nature 444: 860-867.
Hotamisligil, G. S., Arner, P., Caro, J. F., Atkinson, R. L. and Spiegelman, B. M. (1995).
Increased adipose tissue expression of tumor necrosis factor-α in human obesity and insulin
resistance. J. Clin. Invest. 95: 2409-2415.
Howe, K. L., Reardon, C., Wang, A., Nazli, A. and McKay, D. M. (2005). Transforming growth
factor-β regulation of epithelial tight junction proteins enhances barrier function and blocks
enterohemorrhagic Escherichia coli O157:H7-induced increased permeability. Am. J. Pathol.
167: 1587-1597.
Huth, P. J. and Park, K. M. (2012). Influence of dairy product and milk fat consumption on
cardiovascular disease risk: a review of the evidence. Adv. Nutr. 3: 266-285.
Irvine, S. L., Hummelen, R., Hekmat, S., W. N. Looman, C., Habbema, J. D. F. and Reid, G.
(2010). Probiotic yogurt consumption is associated with an increase of CD4 count among
people living with HIV/AIDS. J. Clin. Gastroenterol. 44: e201-e205.
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
43
Isolauri, E., Kaila, M., Arvola, T., Majamaa, H., Rantala, I., Virtanen, E., et al. (1993). Diet
during rotavirus enteritis affects jejunal permeability to macromolecules in suckling rats.
Pediatr. Res. 33: 548-553.
Jijon, H., Backer, J., Diaz, H., Yeung, H., Thiel, D., McKaigney, C., et al. (2004). DNA from
Probiotic Bacteria Modulates Murine and Human Epithelial and Immune Function.
Gastroenterology 126: 1358-1373.
Jonkers, D., Penders, J., Masclee, A. and Pierik, M. (2012). Probiotics in the management of
inflammatory bowel disease: A systematic review of intervention studies in adult patients.
Drugs 72: 803-823.
Jouet, P., Coffin, B. and Sabate, J. M. (2011). Small intestinal bacterial overgrowth in patients
with morbid obesity. Dig. Dis. Sci. 56: 615-616.
Kabeerdoss, J., Shobana Devi, R., Regina Mary, R., Prabhavathi, D., Vidya, R., Mechenro, J., et
al. (2011). Effect of yoghurt containing Bifidobacterium lactis Bb12® on faecal excretion of
secretory immunoglobulin A and human beta-defensin 2 in healthy adult volunteers. Nutr. J.
10.
Kahn, S. E., Hull, R. L. and Utzschneider, K. M. (2006). Mechanisms linking obesity to insulin
resistance and type 2 diabetes. Nature 444: 840-846.
Kale-Pradhan, P. B., Jassal, H. K. and Wilhelm, S. M. (2010). Role of Lactobacillus in the
prevention of antibiotic-associated diarrhea: A meta-analysis. Pharmacotherapy 30: 119-126.
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
44
Kastarinen, M. J., Nissinen, A. M., Vartiainen, E. A., Jousilahti, P. J., Korhonen, H. J., Puska, P.
M., et al. (2000). Blood pressure levels and obesity trends in hypertensive and normotensive
Finnish population from 1982 to 1997. J. Hypertens. 18: 255-262.
Kato, K., Mizuno, S., Umesaki, Y., Ishii, Y., Sugitani, M., Imaoka, A., et al. (2004). Randomized
placebo-controlled trial assessing the effect of bifidobacteria-fermented milk on active
ulcerative colitis. Aliment. Pharmacol. Ther. 20: 1133-1141.
Kemna, E., Pickkers, P., Nemeth, E., Van Der Hoeven, H. and Swinkels, D. (2005). Time-course
analysis of hepcidin, serum iron, and plasma cytokine levels in humans injected with LPS.
Blood 106: 1864-1866.
Kern, P. A., Ranganathan, S., Li, C., Wood, L. and Ranganathan, G. (2001). Adipose tissue
tumor necrosis factor and interleukin-6 expression in human obesity and insulin resistance.
Am. J. Physiol. Endocrinol. Metab. 280: E745-E751.
Kießling, G., Schneider, J. and Jahreis, G. (2002). Long-term consumption of fermented dairy
products over 6 months increases HDL cholesterol. Eur. J. Clin. Nutr. 56: 843-849.
Kim, C. S., Park, H. S., Kawada, T., Kim, J. H., Lim, D., Hubbard, N. E., et al. (2006).
Circulating levels of MCP-1 and IL-8 are elevated in human obese subjects and associated
with obesity-related parameters. Int. J. Obes (Lond). 30: 1347-1355.
Kim, F., Pham, M., Luttrell, I., Bannerman, D. D., Tupper, J., Thaler, J., et al. (2007). Toll-like
receptor-4 mediates vascular inflammation and insulin resistance in diet-induced obesity.
Circ. Res. 100: 1589-1596.
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
45
Kim, Y. S. and Ho, S. B. (2010). Intestinal goblet cells and mucins in health and disease: Recent
insights and progress. Curr. Gastroenterol. Rep. 12: 319-330.
Kinugasa, T., Sakaguchi, T., Gu, X. and Reinecker, H. (2000). Claudins regulate the intestinal
barrier in response to immune mediators. Gastroenterology 118: 1001-1011.
Kolars, J. C., Levitt, M. D., Aouji, M. and Savaiano, D. A. (1984). Yogurt - an autodigesting
source of lactose. N. Engl. J. Med. 310: 1-3.
Labonté, M. E., Couture, P., Richard, C., Desroches, S. and Lamarche, B. (2013). Impact of
dairy products on biomarkers of inflammation: A systematic review of randomized
controlled nutritional intervention studies in overweight and obese adults. Am. J. Clin. Nutr.
97: 706-717.
Larsson, S. C., Andersson, S. O., Johansson, J. E. and Wolk, A. (2008). Cultured milk, yogurt,
and dairy intake in relation to bladder cancer risk in a prospective study of Swedish women
and men. Am. J. Clin. Nutr. 88: 1083-1087.
Larsson, S. C. and Wolk, A. (2007). Obesity and colon and rectal cancer risk: A meta-analysis of
prospective studies. Am. J. Clin. Nutr. 86: 556-565.
Laugerette, F., Vors, C., Géloën, A., Chauvin, M. A., Soulage, C., Lambert-Porcheron, S., et al.
(2011). Emulsified lipids increase endotoxemia: Possible role in early postprandial low-grade
inflammation. J. Nutr. Biochem. 22: 53-59.
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
46
Lavie, C. J., Milani, R. V. and Ventura, H. O. (2009). Obesity and Cardiovascular Disease. Risk
Factor, Paradox, and Impact of Weight Loss. J. Am. Coll. Cardiol. 53: 1925-1932.
Leal, E. C., Martins, J., Voabil, P., Liberal, J., Chiavaroli, C., Bauer, J., et al. (2010). Calcium
dobesilate inhibits the alterations in tight junction proteins and leukocyte adhesion to retinal
endothelial cells induced by diabetes. Diabetes 59: 2637-2645.
Lewington, S., Clarke, R., Qizilbash, N., Peto, R. and Collins, R. (2002). Age-specific relevance
of usual blood pressure to vascular mortality: A meta-analysis of individual data for one
million adults in 61 prospective studies. Lancet 360: 1903-1913.
Ley, R. E., Turnbaugh, P. J., Klein, S. and Gordon, J. I. (2006). Microbial ecology: Human gut
microbes associated with obesity. Nature 444: 1022-1023.
Li, Q., Zhang, Q., Wang, M., Zhao, S., Ma, J., Luo, N., et al. (2008). Interferon-γ and tumor
necrosis factor-α disrupt epithelial barrier function by altering lipid composition in
membrane microdomains of tight junction. Clin. Immunol. 126: 67-80.
Link-Amster, H., Rochat, F., Saudan, K. Y., Mignot, O. and Aeschlimann, J. M. (1994).
Modulation of a specific humoral immune response and changes in intestinal flora mediated
through fermented milk intake. FEMS Immunol. Med. Microbiol. 10: 55-63.
Lomer, M. C. E., Parkes, G. C. and Sanderson, J. D. (2008). Review article: Lactose intolerance
in clinical practice - Myths and realities. Aliment. Pharmacol. Ther. 27: 93-103.
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
47
Lorea Baroja, M., Kirjavainen, P. V., Hekmat, S. and Reid, G. (2007). Anti-inflammatory effects
of probiotic yogurt in inflammatory bowel disease patients. Clin. Exp. Immunol. 149: 470-
479.
Lozano, R., Naghavi, M., Foreman, K., Lim, S., Shibuya, K., Aboyans, V., et al. (2012). Global
and regional mortality from 235 causes of death for 20 age groups in 1990 and 2010: A
systematic analysis for the Global Burden of Disease Study 2010. Lancet 380: 2095-2128.
Mack, D. R., Michail, S., Wei, S., McDougall, L. and Hollingsworth, M. A. (1999). Probiotics
inhibit enteropathogenic E. coli adherence in vitro by inducing intestinal mucin gene
expression. Am. J. Physiol. Gastrointest. Liver Physiol. 276: G941-G950.
Macpherson, A. J. and Uhr, T. (2004). Induction of Protective IgA by Intestinal Dendritic Cells
Carrying Commensal Bacteria. Science 303: 1662-1665.
Madsen, K. L., Lewis, S. A., Tavernini, M. M., Hibbard, J. and Fedorak, R. N. (1997).
Interleukin 10 prevents cytokine-induced disruption of T84 monolayer barrier integrity and
limits chloride secretion. Gastroenterology 113: 151-159.
Manco, M., Putignani, L. and Bottazzo, G. F. (2010). Gut microbiota, lipopolysaccharides, and
innate immunity in the pathogenesis of obesity and cardiovascular risk. Endocr. Rev. 31:
817-844.
Mankertz, J., Tavalali, S., Schmitz, H., Mankertz, A., Riecken, E. O., Fromm, M., et al. (2000).
Expression from the human occludin promoter is affected by tumor necrosis factor α and
interferon γ. J. Cell Sci. 113: 2085-2090.
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
48
Marconi, E., Messia, M. C., Amine, A., Moscone, D., Vernazza, F., Stocchi, F., et al. (2004).
Heat-treated milk differentiation by a sensitive lactulose assay. Food Chem. 84: 447-450.
Margolis, K. L., Wei, F., de Boer, I. H., Howard, B. V., Liu, S., Manson, J. E., et al. (2011). A
diet high in low-fat dairy products lowers diabetes risk in postmenopausal women. J. Nutr.
141: 1969-1974.
Maroof, H., Hassan, Z. M., Mobarez, A. M. and Mohamadabadi, M. A. (2012). Lactobacillus
acidophilus could modulate the immune response against breast cancer in murine model. J.
Clin. Immunol. 32: 1353-1359.
Martínez-Maqueda, D., Miralles, B., De Pascual-Teresa, S., Reverón, I., Muñoz, R. and Recio, I.
(2012). Food-derived peptides stimulate mucin secretion and gene expression in intestinal
cells. J. Agric. Food. Chem. 60: 8600-8605.
Martini, M. C., Lerebours, E. C., Lin, W. J., Harlander, S. K., Berrada, N. M., Antoine, J. M., et
al. (1991). Strains and species of lactic acid bacteria in fermented milks (yogurts): Effect on
in vivo lactose digestion. Am. J. Clin. Nutr. 54: 1041-1046.
Massiéra, F., Bloch-Faure, M., Ceiler, D., Murakami, K., Fukamizu, A., Gasc, J. M., et al.
(2001). Adipose angiotensinogen is involved in adipose tissue growth and blood pressure
regulation. FASEB J. 15: 2727-2729.
Mater, D. D. G., Bretigny, L., Firmesse, O., Flores, M. J., Mogenet, A., Bresson, J. L., et al.
(2005). Streptococcus thermophilus and Lactobacillus delbrueckii subsp. bulgaricus survive
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
49
gastrointestinal transit of healthy volunteers consuming yogurt. FEMS Microbiol. Lett. 250:
185-187.
Mathieu, P., Lemieux, I. and Després, J. P. (2010). Obesity, inflammation, and cardiovascular
risk. Clin. Pharmacol. Ther. 87: 407-416.
Matsumoto, M., Aranami, A., Ishige, A., Watanabe, K. and Benno, Y. (2007). LKM512 yogurt
consumption improves the intestinal environment and induces the T-helper type 1 cytokine in
adult patients with intractable atopic dermatitis. Clin. Exp. Allergy 37: 358-370.
Matsumoto, M., Ohishi, H. and Benno, Y. (2001). Impact of LKM512 yogurt on improvement of
intestinal environment of the elderly. FEMS Immunol. Med. Microbiol. 31: 181-186.
Mauras, N., DelGiorno, C., Kollman, C., Bird, K., Morgan, M., Sweeten, S., et al. (2010).
Obesity without established comorbidities of the metabolic syndrome is associated with a
proinflammatory and prothrombotic state, even before the onset of puberty in children. J.
Clin. Endocrinol. Metab. 95: 1060-1068.
McNulty, N. P., Yatsunenko, T., Hsiao, A., Faith, J. J., Muegge, B. D., Goodman, A. L., et al.
(2011). The impact of a consortium of fermented milk strains on the gut microbiome of
gnotobiotic mice and monozygotic twins. Sci Transl Med 3: 106ra106.
Miele, E., Pascarella, F., Giannetti, E., Quaglietta, L., Baldassano, R. N. and Staiano, A. (2009).
Effect of a probiotic preparation (VSL#3) on induction and maintenance of remission in
children with ulcerative colitis. Am. J. Gastroenterol. 104: 437-443.
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
50
Moghaddam, A. A., Woodward, M. and Huxley, R. (2007). Obesity and risk of colorectal
cancer: A meta-analysis of 31 studies with 70,000 events. Cancer Epidemiol. Biomarkers
Prev. 16: 2533-2547.
Morikawa, K., Kondo, I., Kanamaru, Y. and Nagaoka, S. (2007). A novel regulatory pathway for
cholesterol degradation via lactostatin. Biochem. Biophys. Res. Commun. 352: 697-702.
Muccioli, G. G., Naslain, D., Bäckhed, F., Reigstad, C. S., Lambert, D. M., Delzenne, N. M., et
al. (2010). The endocannabinoid system links gut microbiota to adipogenesis. Mol. Syst. Biol.
6.
Muoio, D. M. and Newgard, C. B. (2008). Mechanisms of disease: Molecular and metabolic
mechanisms of insulin resistance and β-cell failure in type 2 diabetes. Nat. Rev. Mol. Cell
Biol. 9: 193-205.
Nakamura, T., Mizutani, J., Sasaki, K., Yamamoto, N. and Takazawa, K. (2009). Beneficial
potential of casein hydrolysate containing Val-Pro-Pro and Ile-Pro-Pro on central blood
pressure and hemodynamic index: A preliminary study. J. Med. Food 12: 1221-1226.
National-Yogurt-Association (2008). Live and Active Culture Yogurt Seal Program. McLean,
VA.
Nicklas, T. A., Qu, H., Hughes, S. O., He, M., Wagner, S. E., Foushee, H. R., et al. (2011). Self-
perceived lactose intolerance results in lower intakes of calcium and dairy foods and is
associated with hypertension and diabetes in adults. Am. J. Clin. Nutr. 94: 191-198.
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
51
O'Hara, A. M. and Shanahan, F. (2006). The gut flora as a forgotten organ. EMBO Rep. 7: 688-
693.
O'Sullivan, M. G., Thornton, G., O'Sullivan, G. C. and Collins, J. K. (1992). Probiotic bacteria:
myth or reality? Trends Food Sci. Tech. 3: 309-314.
Ohmura, K., Ishimori, N., Ohmura, Y., Tokuhara, S., Nozawa, A., Horii, S., et al. (2010).
Natural killer T cells are involved in adipose tissues inflammation and glucose intolerance in
diet-induced obese mice. Arterioscler. Thromb. Vasc. Biol. 30: 193-199.
Okamura, Y., Watari, M., Jerud, E. S., Young, D. W., Ishizaka, S. T., Rose, J., et al. (2001). The
Extra Domain A of Fibronectin Activates Toll-like Receptor 4. J. Biol. Chem. 276: 10229-
10233.
Ortakci, F. and Sert, S. (2012). Stability of free and encapsulated Lactobacillus acidophilus
ATCC 4356 in yogurt and in an artificial human gastric digestion system. J. Dairy Sci. 95:
6918-6925.
Oshima, T., Laroux, F. S., Coe, L. L., Morise, Z., Kawachi, S., Bauer, P., et al. (2001).
Interferon-γ and interleukin-10 reciprocally regulate endothelial junction integrity and barrier
function. Microvasc. Res. 61: 130-143.
Pala, V., Sieri, S., Berrino, F., Vineis, P., Sacerdote, C., Palli, D., et al. (2011). Yogurt
consumption and risk of colorectal cancer in the Italian European prospective investigation
into cancer and nutrition cohort. Int. J. Cancer 129: 2712-2719.
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
52
Panagiotakos, D. B., Pitsavos, C., Yannakoulia, M., Chrysohoou, C. and Stefanadis, C. (2005).
The implication of obesity and central fat on markers of chronic inflammation: The ATTICA
study. Atherosclerosis 183: 308-315.
Park, B. S., Song, D. H., Kim, H. M., Choi, B. S., Lee, H. and Lee, J. O. (2009). The structural
basis of lipopolysaccharide recognition by the TLR4-MD-2 complex. Nature 458: 1191-
1195.
Park, Y., Albright, K. J., Liu, W., Storkson, J. M., Cook, M. E. and Pariza, M. W. (1997). Effect
of conjugated linoleic acid on body composition in mice. Lipids 32: 853-858.
Parvez, S., Malik, K. A., Ah Kang, S. and Kim, H. Y. (2006). Probiotics and their fermented
food products are beneficial for health. J. Appl. Microbiol. 100: 1171-1185.
Peeters, A., Barendregt, J. J., Willekens, F., Mackenbach, J. P., Al Mamun, A., Bonneux, L., et
al. (2003). Obesity in adulthood and its consequences for life expectancy: A life-table
analysis. Ann. Intern. Med. 138: 24-32.
Perdigon, G., Alvarez, S., Rachid, M., Agüero, G. and Gobbato, N. (1995). Immune system
stimulation by probiotics. J. Dairy Sci. 78: 1597-1606.
Pineiro, M. and Embarek, B. (2002). Guidelines for the Evaluation of Probiotics in Food.
London Ontario, Canada, FAO/WHO.
Porter, E. M., Bevins, C. L., Ghosh, D. and Ganz, T. (2002). The multifaceted Paneth cell. Cell
Mol. Life Sci. 59: 156-170.
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
53
Pugin, J., Heumann, D., Tomaszi, A., Kravchenko, V. V., Akamatsu, Y., Nishijima, M., et al.
(1994). CD14 is a pattern recognition receptor. Immunity 1: 509-516.
Puri, P., Rattan, A., Bijlani, R. L., Mahapatra, S. C. and Nath, I. (1996). Splenic and intestinal
lymphocyte proliferation response in mice fed milk or yogurt and challenged with
Salmonella typhimurium. Int. J. Food Sci. Nutr. 47: 391-398.
Raetz, C. R. H. and Whitfield, C. (2002). Lipopolysaccharide endotoxins. Annu. Rev. Biochem.
71: 635-700.
Rahmouni, K., Correia, M. L. G., Haynes, W. G. and Mark, A. L. (2005). Obesity-associated
hypertension: New insights into mechanisms. Hypertension 45: 9-14.
Rallabhandi, P., Bell, J., Boukhvalova, M. S., Medvedev, A., Lorenz, E., Arditi, M., et al. (2006).
Analysis of TLR4 polymorphic variants: New insights into TLR4/MD-2/CD14
stoichiometry, structure, and signaling. J. Immunol. 177: 322-332.
Ralston, R. A., Lee, J. H., Truby, H., Palermo, C. E. and Walker, K. Z. (2012). A systematic
review and meta-analysis of elevated blood pressure and consumption of dairy foods. J.
Hum. Hypertens. 26: 3-13.
Recker, R. R., Bammi, A., Barger-Lux, M. and Heaney, R. P. (1988). Calcium absorbability
from milk products, an imitation milk and calcium carbonate. Am. J. Clin. Nutr. 47: 93-95.
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
54
Reeves, G. K., Pirie, K., Beral, V., Green, J., Spencer, E. and Bull, D. (2007). Cancer incidence
and mortality in relation to body mass index in the Million Women Study: Cohort study.
BMJ. 335: 1134-1139.
Rombaut, R., Camp, J. V. and Dewettinck, K. (2005). Analysis of phospho- and sphingolipids in
dairy products by a new HPLC method. J. Dairy Sci. 88: 482-488.
Saavedra, J. M., Bauman, N. A., Oung, I., Perman, J. A. and Yolken, R. H. (1994). Feeding of
Bifidobacterium bifidum and Streptococcus thermophilus to infants in hospital for prevention
of diarrhoea and shedding of rotavirus. Lancet 344: 1046-1049.
Sadrzadeh-Yeganeh, H., Elmadfa, I., Djazayery, A., Jalali, M., Heshmat, R. and Chamary, M.
(2010). The effects of probiotic and conventional yoghurt on lipid profile in women. Br. J.
Nutr. 103: 1778-1783.
Sakamoto, I., Igarashi, M., Kimura, K., Takagi, A., Miwa, T. and Koga, Y. (2001). Suppressive
effect of Lactobacillus gasseri OLL 2716 (LG21) on Helicobacter pylori infection in humans.
J. Antimicrob. Chemother. 47: 709-710.
Salzman, N. H., Underwood, M. A. and Bevins, C. L. (2007). Paneth cells, defensins, and the
commensal microbiota: A hypothesis on intimate interplay at the intestinal mucosa. Semin.
Immunol. 19: 70-83.
Salzman, N. H., Underwood, M. A. and Bevins, C. L. (2007). Paneth cells, defensins, and the
commensal microbiota: A hypothesis on intimate interplay at the intestinal mucosa. Seminars
in Immunology 19: 70-83.
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
55
Scaldaferri, F., Pizzoferrato, M., Gerardi, V., Lopetuso, L. and Gasbarrini, A. (2012). The gut
barrier: New acquisitions and therapeutic approaches. J. Clin. Gastroenterol. 46: S12-S17.
Schaafsma, G., Meuling, W. J. A., Van Dokkum, W. and Bouley, C. (1998). Effects of a milk
product, fermented by Lactobacillus acidophilus and with fructo-oligosaccharides added, on
blood lipids in male volunteers. Eur. J. Clin. Nutr. 52: 436-440.
Scharl, M., Geisel, S., Vavricka, S. R. and Rogler, G. (2010). Dying in yoghurt: The number of
living bacteria in probiotic yoghurt decreases under exposure to room temperature. Digestion
83: 13-17.
Schiffrin, E. J., Brassart, D., Servin, A. L., Rochat, F. and Donnet-Hughes, A. (1997). Immune
modulation of blood leukocytes in humans by lactic acid bacteria: Criteria for strain
selection. Am. J. Clin. Nutr. 66: 515S-520S.
Schiffrin, E. J., Parlesak, A., Bode, C., Bode, J. C., van't Hof, M. A., Grathwohl, D., et al.
(2009). Probiotic yogurt in the elderly with intestinal bacterial overgrowth: Endotoxaemia
and innate immune functions. Br. J. Nutr. 101: 961-966.
Schillinger, U. (1999). Isolation and identification of lactobacilli from novel-type probiotic and
mild yoghurts and their stability during refrigerated storage. Int. J. Food Microbiol. 47: 79-
87.
Schlee, M., Harder, J., Köten, B., Stange, E. F., Wehkamp, J. and Fellermann, K. (2008).
Probiotic lactobacilli and VSL#3 induce enterocyte β-defensin 2. Clin. Exp. Immunol. 151:
528-535.
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
56
Schlee, M., Wehkamp, J., Altenhoefer, A., Oelschlaeger, T. A., Stange, E. F. and Fellermann, K.
(2007). Induction of human β-defensin 2 by the probiotic Escherichia coli Nissle 1917 is
mediated through flagellin. Infect. Immun. 75: 2399-2407.
Schmitz, H., Fromm, M., Bentzel, C. J., Scholz, P., Detjen, K., Mankertz, J., et al. (1999). Tumor
necrosis factor-alpha (TNFα) regulates the epithelial barrier in the human intestinal cell line
HT-29/B6. J. Cell Sci. 112: 137-146.
Shah, N. P. (2007). Functional cultures and health benefits. Int. Dairy J. 17: 1262-1277.
Shahani, K. M. and Chandan, R. C. (1979). Nutritional and healthful aspects of cultured and
culture-containing dairy foods. J. Dairy Sci. 62: 1685-1694.
Shaw, J. E., Sicree, R. A. and Zimmet, P. Z. (2010). Global estimates of the prevalence of
diabetes for 2010 and 2030. Diabetes Res. Clin. Pract. 87: 4-14.
Sheikh, M. S., Santa Ana, C. A., Nicar, M. J., Schiller, L. R. and Fordtran, J. S. (1987).
Gastrointestinal absorption of calcium from milk and calcium salts. N. Engl. J. Med. 317:
532-536.
Shoelson, S. E., Lee, J. and Goldfine, A. B. (2006). Inflammation and insulin resistance. J. Clin.
Invest. 116: 1793-1801.
Shulman, G. I. (2000). Cellular mechanisms of insulin resistance. J. Clin. Invest. 106: 171-176.
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
57
Sluijs, I., Forouhi, N. G., Beulens, J. W. J., Van Der Schouw, Y. T., Agnoli, C., Arriola, L., et al.
(2012). The amount and type of dairy product intake and incident type 2 diabetes: Results
from the EPIC-InterAct Study. Am. J. Clin. Nutr. 96: 382-390.
Smith, T. M., Kolars, J. C., Savaiano, D. A. and Levitt, M. D. (1985). Absorption of calcium
from milk and yogurt. Am. J. Clin. Nutr. 42: 1197-1200.
Soedamah-Muthu, S. S., Verberne, L. D. M., Ding, E. L., Engberink, M. F. and Geleijnse, J. M.
(2012). Dairy consumption and incidence of hypertension: A dose-response meta-analysis of
prospective cohort studies. Hypertension 60: 1131-1137.
Solinas, G. and Karin, M. (2010). JNK1 and IKKβ: Molecular links between obesity and
metabolic dysfunction. FASEB J. 24: 2596-2611.
Sommansson, A., Nylander, O. and Sjöblom, M. (2013). Melatonin decreases duodenal epithelial
paracellular permeability via a nicotinic receptor-dependent pathway in rats in vivo. J. Pineal
Res. 54: 282-291.
Sood, A., Midha, V., Makharia, G. K., Ahuja, V., Singal, D., Goswami, P., et al. (2009). The
Probiotic Preparation, VSL#3 Induces Remission in Patients With Mild-to-Moderately
Active Ulcerative Colitis. Clin. Gastroenterol. Hepatol. 7: 1202-1209.e1201.
Stoll, L. L., Denning, G. M., Li, W. G., Rice, J. B., Harrelson, A. L., Romig, S. A., et al. (2004).
Regulation of endotoxin-induced proinflammatory activation in human coronary artery cells:
Expression of functional membrane-bound CD14 by human coronary artery smooth muscle
cells. J. Immunol. 173: 1336-1343.
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
58
Stuart, R. O., Sun, A., Panichas, M., Hebert, S. C., Brenner, B. M. and Nigam, S. K. (1994).
Critical role for intracellular calcium in tight junction biogenesis. J. Cell Physiol. 159: 423-
433.
Sun, L., Yu, Z., Ye, X., Zou, S., Li, H., Yu, D., et al. (2010). A marker of endotoxemia is
associated with obesity and related metabolic disorders in apparently healthy Chinese.
Diabetes Care 33: 1925-1932.
Teixeira, T. F. S., Souza, N. C. S., Chiarello, P. G., Franceschini, S. C. C., Bressan, J., Ferreira,
C. L. L. F., et al. (2012). Intestinal permeability parameters in obese patients are correlated
with metabolic syndrome risk factors. Clin. Nutr. 31: 735-740.
Tenenbaum, A. and Fisman, E. Z. (2012). Fibrates are an essential part of modern anti-
dyslipidemic arsenal: spotlight on atherogenic dyslipidemia and residual risk reduction.
Cardiovasc. Diabetol. 11.
Thomas, A. P., Dunn, T. N., Drayton, J. B., Oort, P. J. and Adams, S. H. (2012). A high calcium
diet containing nonfat dry milk reduces weight gain and associated adipose tissue
inflammation in diet-induced obese mice when compared to high calcium alone. Nutr.
Metab. (Lond). 9.
Tilg, H. and Kaser, A. (2011). Gut microbiome, obesity, and metabolic dysfunction. J. Clin.
Invest. 121: 2126-2132.
Tong, X., Dong, J. Y., Wu, Z. W., Li, W. and Qin, L. Q. (2011). Dairy consumption and risk of
type 2 diabetes mellitus: A meta-analysis of cohort studies. Eur. J. Clin. Nutr. 65: 1027-1031.
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
59
Tsuchiya, A., Almiron-Roig, E., Lluch, A., Guyonnet, D. and Drewnowski, A. (2006). Higher
satiety ratings following yogurt consumption relative to fruit drink or dairy fruit drink. J. Am.
Diet. Assoc. 106: 550-557.
Tsukita, S., Furuse, M. and Itoh, M. (2001). Multifunctional strands in tight junctions. Nat. Rev.
Mol. Cell Biol. 2: 285-293.
Turnbaugh, P. J., Ley, R. E., Mahowald, M. A., Magrini, V., Mardis, E. R. and Gordon, J. I.
(2006). An obesity-associated gut microbiome with increased capacity for energy harvest.
Nature 444: 1027-1031.
Turner, J. R. (2009). Intestinal mucosal barrier function in health and disease. Nat. Rev.
Immunol. 9: 799-809.
Van Bossuyt, H., De Zanger, R. B. and Wisse, E. (1988). Cellular and subcellular distribution of
injected lipopolysaccharide in rat liver and its inactivation by bile salts. J. Hepatol. 7: 325-
337.
Van Kruijsdijk, R. C. M., Van Der Wall, E. and Visseren, F. L. J. (2009). Obesity and cancer:
The role of dysfunctional adipose tissue. Cancer Epidemiol. Biomarkers Prev. 18: 2569-
2578.
Van Meijl, L. E. C., Vrolix, R. and Mensink, R. P. (2008). Dairy product consumption and the
metabolic syndrome. Nutr. Res. Rev. 21: 148-157.
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
60
Van Niel, C. W., Feudtner, C., Garrison, M. M. and Christakis, D. A. (2002). Lactobacillus
therapy for acute infectious diarrhea in children: A meta-analysis. Pediatrics 109: 678-684.
Vélez, M. P., Hermans, K., Verhoeven, T. L. A., Lebeer, S. E., Vanderleyden, J. and De
Keersmaecker, S. C. J. (2007). Identification and characterization of starter lactic acid
bacteria and probiotics from Columbian dairy products. J. Appl. Microbiol. 103: 666-674.
Wadden, T. A., Webb, V. L., Moran, C. H. and Bailer, B. A. (2012). Lifestyle modification for
obesity: New developments in diet, physical activity, and behavior therapy. Circulation 125:
1157-1170.
Warensjö, E., Jansson, J. H., Cederholm, T., Boman, K., Eliasson, M., Hallmans, G., et al.
(2010). Biomarkers of milk fat and the risk of myocardial infarction in men and women: A
prospective, matched case-control study. Am. J. Clin. Nutr. 92: 194-202.
Weisberg, S. P., McCann, D., Desai, M., Rosenbaum, M., Leibel, R. L. and Ferrante Jr, A. W.
(2003). Obesity is associated with macrophage accumulation in adipose tissue. J. Clin.
Invest. 112: 1796-1808.
Whigham, L. D., Cook, M. E. and Atkinson, R. L. (2000). Conjugated linoleic acid: Implications
for human health. Pharmacol. Res. 42: 503-510.
Woof, J. M. and Ken, M. A. (2006). The function of immunoglobulin A in immunity. J. Pathol.
208: 270-282.
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
61
Wu, H., Ghosh, S., Perrard, X. D., Feng, L., Garcia, G. E., Perrard, J. L., et al. (2007). T-cell
accumulation and regulated on activation, normal T cell expressed and secreted upregulation
in adipose tissue in obesity. Circulation 115: 1029-1038.
Xu, H., Barnes, G. T., Yang, Q., Tan, G., Yang, D., Chou, C. J., et al. (2003). Chronic
inflammation in fat plays a crucial role in the development of obesity-related insulin
resistance. J. Clin. Invest. 112: 1821-1830.
Yang, H., Kiristioglu, I., Fan, Y., Forbush, B., Bishop, D. K., Antony, P. A., et al. (2002).
Interferon-gamma expression by intraepithelial lymphocytes results in a loss of epithelial
barrier function in a mouse model of total parenteral nutrition. Ann. Surg. 236: 226-234.
Yang, R., Han, X., Uchiyama, T., Watkins, S. K., Yaguchi, A., Delude, R. L., et al. (2003). IL-6
is essential for development of gut barrier dysfunction after hemorrhagic shock and
resuscitation in mice. Am. J. Physiol. Gastrointest. Liver Physiol. 285: G621-G629.
Yang, Y. J. and Sheu, B. S. (2012). Probiotics-Containing Yogurts Suppress Helicobacter pylori
Load and Modify Immune Response and Intestinal Microbiota in the Helicobacter pylori-
Infected Children. Helicobacter 17: 297-304.
Youakim, A. and Ahdieh, M. (1999). Interferon-γ decreases barrier function in T84 cells by
reducing ZO-1 levels and disrupting apical actin. Am. J. Physiol. Gastrointest. Liver Physiol.
276: G1279-G1288.
Zasloff, M. (2002). Antimicrobial peptides of multicellular organisms. Nature 415: 389-395.
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
62
Zemel, M. B., Shi, H., Greer, B., Dirienzo, D. and Zemel, P. C. (2000). Regulation of adiposity
by dietary calcium. FASEB J. 14: 1132-1138.
Zemel, M. B., Thompson, W., Milstead, A., Morris, K. and Campbell, P. (2004). Calcium and
dairy acceleration of weight and fat loss during energy restriction in obese adults. Obes. Res.
12: 582-590.
Zhao, Y., Martin, B. R. and Weaver, C. M. (2005). Calcium bioavailability of calcium carbonate
fortified soymilk is equivalent to cow's milk in young women. J. Nutr. 135: 2379-2382.
Zsivkovits, M., Fekadu, K., Sontag, G., Nabinger, U., Huber, W. W., Kundi, M., et al. (2003).
Prevention of heterocyclic amine-induced DNA damage in colon and liver of rats by
different lactobacillus strains. Carcinogenesis 24: 1913-1918.
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
63
TABLES
Table 1. Representative nutrient data bank values for plain yogurts in the U.S.A
Nutrient
(per 6 oz) Whole Low-fat Fat-free Fat-free (Greek)
calories 104 107 95 100
total fat (g) 5.9 2.64 0.31 0.66
saturated fat (g) 3.56 1.7 0.12 0.20
MUFA (g) 1.52 0.72 0.05 0.09
PUFA (g) 0.16 0.08 0.01 0.02
cholesterol (mg) 22 10 2 8
carbohydrates (g) 7.92 11.97 13.06 6.12
sugar (g)
B
7.92 11.97 13.06 5.51
dietary fiber (g) 0.0 0.0 0.0 0.0
protein (g) 5.9 6.77 5.73 17.32
thiamin (mg) 0.05 0.08 0.08 0.04
riboflavin (mg) 0.24 0.36 0.40 0.47
niacin (mg) 0.13 0.19 0.21 0.35
vitamin B6 (mg) 0.05 0.08 0.09 0.11
folate (mcg) 12 19 20 12
vitamin B12 (mcg) 0.63 0.95 1.04 1.28
vitamin A (RAE) 46 24 3 2
vitamin C (mg) 0.8 1.4 1.50 0.0
vitamin D (mcg) 3.0 2.0 0.0 0.0
vitamin E (mg) 0.1 0.05 0.0 0.02
vitamin K (mcg) 0.3 0.3 0.30 0.0
calcium (mg) 206 311 338 187
phosphorus (mg) 162 245 267 230
magnesium (mg) 20 29 32 19
sodium (mg) 78 119 131 61
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
64
potassium (mg) 264 398 434 240
iron (mg) 0.08 0.14 0.15 0.12
zinc (mg) 1 1.51 1.65 0.88
A Derived from USDA National Nutrient Database for Standard Reference, Release 26.
BSweetened or fruit yogurts typically have an additional 20 g sugars per 6 oz.
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
65
Table 2. Clinical studies of yogurt on biomarkers relevant to obesity and chronic disease risk.
Category Reference Population Treatment Outcome
Lipid profiles
(Schaafsma et al.,
1998)
n = 30 healthy men
375 mL/d for 3 wk, 0.5%
fat, + L. acidophilus
serum total cholesterol, LDL, and
LDL/HLD-ratio.
(Anderson and
Gilliland, 1999)
n = 40
hypercholesterolemic
individuals
200 mL/d for 4 wk, + L.
acidophilus, unspecified
fat content
serum cholesterol by 3.2%
(Kießling et al., 2002)
n = 29 women (14
hypercholesterolemic)
300 g/day for 6 mo, 3.5%
fat, + L. acidophilus and
B. longum
HDL
(Fabian and Elmadfa,
2006)
n = 33 lean women
100 g/d for 2 wk and then
200 g/d for another 2 wk,
3.6% fat
LDL/HLD ratio
(Sadrzadeh-Yeganeh
et al., 2010)
n = 90 lean women
300 g/d for 6 wk, 2.5%
fat
total cholesterol and total:HDL
cholesterol ratio
Inflammation (Matsumoto et al.,
2001)
n = 6 elderly (3 M, 3 F) 100 g/day for 2 wk, + L.
acidophilus and B. lactis,
unspecified fat content
haptoglobin in feces
(Sakamoto et al.,
2001)
n = 31 elderly (29 M, 2
F)
180 g/day for 8 wk, + L.
gasseri, unspecified fat
content
H. pylori-induced gastric mucosal
inflammation
(Schiffrin et al., 2009) n = 36 elderly (9 M, 27 300 g/day for 4 wk, + L. plasma LBP, sCD14 and surrogate
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
66
F) johnsonii, unspecified fat
content
markers of LPS permeability
(Yang and Sheu,
2012)
n = 38 children 400 mL/day for 4 wk, +
L. acidophilus and B.
lactis, unspecified fat
content
serum IL-6
Appetite (Tsuchiya et al., 2006) n = 32 healthy men and
women
acute yogurt intake (200
Kcal)
hunger, fullness, subsequent
food intake
(Chapelot and Payen,
2010)
n = 18 lean men acute yogurt intake (287
Kcal)
satiety, subsequent food intake
(Dougkas et al., 2012) n = 40 overweight men acute yogurt intake (201
Kcal)
appetite, subsequent energy intake
(Douglas et al., 2013) n = 15 women acute yogurt intake (160
Kcal)
hunger, fullness, and delayed
subsequent eating
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
67
Table 3. Obesity-related changes in biomarkers of inflammation.
Reference Samples Population Markers
(Hotamisligil et al.,
1995)
adipose tissue premenopausal
women, n = 18
lean/n = 19 obese
TNF-α mRNA;
body weight reduction TNF-α
mRNA
(Kern et al., 2001) adipose tissue n = 50 lean/n = 50
obese
TNF-α secretion
plasma IL-6
(Panagiotakos et al.,
2005)
serum 3042 adults CRP, TNF-α, amyloid A,
IL-6 in subjects with central
adiposity
(Kim et al., 2006) serum 50 obese and 50
lean adults
MCP-1, IL-8 and CRP
(Herder et al., 2007)
serum 519 adolescents IL-6, IL-18 and interferon-γ-
inducible protein-10 positively
associated with BMI and waist
circumference
(Mauras et al.,
2010)
plasma 203 children hsCRP, fibrinogen, IL-6 and
plasminogen activator inhibitor-1
(Brake et al., 2006) adipose tissue High-fat-fed male
mice
ICAM-I, IL-6 and MCP-1
mRNA
(Ehses et al., 2007) pancreatic
islets
High-fat-fed mice IL-6, IL-8 and macrophage
inflammatory protein 1α
(De Souza et al.,
2005)
hypothalamus High-fat-fed rats TNF-α, IL-1β, and IL-6
Downloaded by [University of Wisconsin - Madison] at 06:45 20 April 2015
... The ingestion of fermented milk has been shown to transitorily modify the intestinal microbiota because of the survival of external species. Thus, yogurt ingestion may increase the amount of lactic acid bacteria at different levels of the human intestine [6,7] and modify the functions of the intestinal microbiota. The presence of lactobacilli in the diet was associated with a reduction in human fecal enzymatic activity of beta-glucuronidase, nitroreductase, and glycocholic acid reductase and the urinary excretion of p-cresol, among other changes in bacterial enzymes. ...
... Pasteurized yogurts are available in the market, and it seems reasonable to expect that the benefits associated to the presence of viable bacteria in yogurt would not be present if the product was pasteurized, as it has been reported in some clinical trials [8,9,12]. According to the published literature, with the exception of some small studies [6,13], the studies that have compared the benefits associated with the routine intake of fresh versus pasteurized yogurt have been limited to the assimilation of nutrients, oral cecal transit time, survival of probiotics or changes in fecal microbiota. There is preliminary evidence that immunostimulation by fresh yogurt is associated with a decreased incidence of certain tumors, gastrointestinal disorders, and allergic conditions [11]. ...
Article
Full-text available
The immune system is affected by the dietary products humans intake. Immune system regulation by nutrition has uses in the clinical context, but it can also benefit healthy populations by delaying or preventing the emergence of immune-mediated chronic illnesses. In this study, the purpose was to describe and compare the modulator effects on the immune system of the routine ingestion of fresh vs. pasteurized yogurt. A unicentral, prospective, randomized, double-blind, parallel group 8-week nutritional study was carried out comparing the ingestion of 125 g of the products in healthy adults three times a day. A complete battery of in vitro tests on the activity of the immune system, processes and phenomena was performed. Exclusive immune-modulatory effects of fresh yogurt with respect to base line were found in terms of increased systemic IgM (primary immune responses), increased synthesis of IFN-gamma upon stimulation (Th1) and increased peripheral T cells (mainly “naive” CD4s). In the three interventions, we observed an increased phagocytic activity and burst test in granulocytes, together with increased secretion of IL-6, IL-1 β and IL-8 (pro-inflammatory) and increased CD16 expression (FcR favoring phagocytosis) in granulocytes. Overall, it is concluded that regardless of bacteria being alive or thermally inactivated, yogurt has common effects on the innate system, but the presence of live bacteria is necessary to achieve a potentiating effect on the specific immune response.
... Several studies have confirmed that yogurt has positive effects on human health, reducing the risk of disease [3,4]. In fact, it has been observed that yogurt consumption helps treat diseases including obesity, allergies, intestinal tract inflammation, colon cancer, cardiovascular disease, and Helicobacter pylori infection [5][6][7][8][9][10]. ...
... Although many studies have explored the positive factors registered by the consumption of yogurt in relation to the health and well-being of children and adolescents [5][6][7][8][9][10], little is known about their tastes in relation to these products and the factors influence their choices in purchasing functional foods, particularly yogurt. ...
... The straining process that creates Greek yoghurt also results in a reduction of lactose, the natural sugar found in milk. This reduction in lactose content can make Greek yoghurt easier to digest for individuals who are sensitive to lactose (Pei et al., 2017). ...
... The administration of fermented products may have effect on the intestinal microflora, restoring immune cells correlated to the intestine and it is beneficial in case of intestinal infection and colon cancer [13]. Additionally, Lactobacillus acidophilus isolated from yogurt reduced tumor growth rate and increased lymphocyte proliferation in a mouse model of breast cancer [38].Fermented dairy products brings about apoptosis, cell cycle arrest and turned down proliferation of tumor in breast cancer cells; as a result, possibly acceptable in the prohibition or cure of breast cancer [43]. ...
Chapter
Agriculture and Food Science Book series aims to bring together leading academic scientists, researchers and research scholars to publish their experiences and research results on all aspects of Agriculture and Food Science. It also provides a premier interdisciplinary platform for researchers, practitioners and educators to present and discuss the most recent innovations, trends, and concerns as well as practical challenges encountered and solutions adopted in the fields of Agriculture and Food Science. High quality research contributions describing original and unpublished results of conceptual, constructive, empirical, experimental, or theoretical work in all areas of Agriculture and Food Science are cordially invited for publication.
... Obesity is an increased risk of chronic diseases and affects both industrialized and developing nations (30). According to the latest review and meta-analysis of 39 randomized controlled trials, probiotic fermented milk products might be used as adjuvant therapy to lower total cholesterol, LDL cholesterol, and triglycerides in the blood, Frontiers in Nutrition 03 frontiersin.org ...
Article
Full-text available
In the current arena of time, the transformation of society has improved the standard of living in terms of lifestyle and their nutritional demands and requirements. The microorganisms under controlled conditions and the enzymatic transformation of dietary components are the processes that resulted in fermented foods and beverages. Fermented dairy products with high nutritional value are “the pearls of the dairy industry.” During fermentation, fermented dairy products produce bioactive compounds and metabolites derived from bacteria. Research indicates the beneficial effects of probiotics found in dairy products on human health is making lightning-fast headway these days. The utilization of lactic acid bacteria as probiotics for the prevention or treatment of disease has been a driving force behind the discovery of novel potential probiotics found in naturally fermented milk. Probiotics such as lactic acid bacteria and bifidobacteria found in fermented dairy products have a variety of health benefits, including innate immune enhancement, diarrhea treatment, inflammatory bowel disease, diabetes, Tuberculosis, and obesity, relieving irritable bowel disease symptoms, preventing cancer, improving lactose tolerance, lowering cholesterol, enhancing antioxidant activity, and antimicrobial activity against pathogens. This review aims to evaluate the therapeutic efficacy and nutritional and microbiological properties of popular fermented dairy products and their health benefits.
... It is a particular probiotic microorganism that provides health benefits when administered in appropriate amounts (WHO 2006). This bacterium exerts probiotic effects through its activities, such as modulation of the intestinal microbiota, inhibition of specific pathogens, antioxidant activities, and prevention of obesity (Cui et al. 2016;Pei et al. 2017). The other essential activity of S. thermophilus is to produce exopolysaccharide (EPS), which has a significant role in forming the texture, viscosity, and mouthfeel of dairy products (Vaningelgem et al. 2004;De Vuyst et al. 2011). ...
Article
Streptococcus thermophilus, the only Streptococcus species considered “Generally Recognized Safe”, has been used widely in the food industry. This bacterium is one of the most valuable industrial lactic acid bacterial species. Due to the importance of this bacterium in industrial applications, it should be stored for a long time without losing its metabolic properties. The present study aimed to investigate the cryoprotectant effect of three compatible solutes (ectoine, trehalose, and sucrose) on bacterial cells stored at different temperatures (frozen at -80 °C or freeze-dried and subsequently stored at +4, -20, and -80 °C) for three months. The bacterial cells were tested for cell viability, bile salt tolerance, and lactic acid production before and after processing. The highest cell viability, bile salt tolerance, and lactic acid production were obtained with ectoine and under frozen (storage at -80 °C) conditions. In freeze-dried and subsequently stored at various temperatures, the best preservation was obtained at -80 °C, followed by -20 °C and +4 °C. Moreover, when ectoine’s preservation potential was compared to other cryoprotectants, ectoine showed the highest preservation, followed by trehalose and sucrose. Although ectoine has a variety of qualities that have been proven, in the current work, we have shown for the first time that ectoine has cryoprotectant potential in yogurt starter cultures (S. thermophilus).
... Commonly used microorganisms are lactic acid bacteria, bacillus, yeast and mold, which can produce a variety of highly active proteases and peptidases [32] . Playing a powerful role in cow's milk are lactic acid bacteria, which secrete cell wall proteases and many different intracellular peptidases to convert proteins into small molecules and free amino acid forms during the fermentation of milk [33] . Bacillus is often used in industrial fermentation, yeast can give dairy products unique flavor characteristics, mold can hydrolyze a variety of proteins, and probiotics can enhance the functional activity of commercially available milk health products. ...
... Dairy products prepared from CM could have the possibility for progress in the dairy market because of the potential therapeutic attributes of CM, such as tuberculosis, asthma, diabetes, jaundice, dropsy, and visceral leishmaniasis treatments (Sulieman and Alayan, 2022). Yogurt is one of the most common fermented foods worldwide, with established beneficial health effects (Pei et al., 2017). The structural and rheological properties of BM, goat milk, or sheep milk yogurts have been widely investigated (Nguyen et al., 2018). ...
Article
The manufacture of camel milk (CM) yogurt has been associated with several challenges, such as the weak structure and watery texture, thereby decreasing its acceptability. Therefore, this study aimed to investigate the effect of whey protein isolate (WPI) addition on the health-promoting benefits, texture profile, and rheological properties of CM yogurt after 1 and 15 d of storage. Yogurt was prepared from CM was supplemented with 0, 3, and 5% of WPI and compared with bovine milk yogurt. The results show that the water holding capacity was affected by WPI addition representing 31.3%, 56.8%, 64.7%, and 45.1% for yogurt from CM containing 0, 3 or 5% WPI, and bovine milk yogurt, respectively, after 15 d. The addition of WPI increased yogurt hardness, adhesiveness, and decreased the resilience. CM yogurt without WPI showed lower apparent viscosity, storage modulus, and loss modulus values compared with other samples. The supplementation of CM with WPI improved the rheological properties of the obtained yogurt. Furthermore, the antioxidant activities of yogurt before and after in vitro digestion varied among yogurt treatments, which significantly increased after digestion except the superoxide anion scavenging and lipid oxidation inhibition. After in vitro digestion at d 1, the superoxide anion scavenging of the 4 yogurt treatments respectively decreased from 83.7%, 83.0%, 79.1%, and 87.4% to 36.7%, 38.3%, 44.6%, and 41.3%. The inhibition of ⍺-amylase and ⍺-glucosidase, angiotensin-converting enzyme inhibition, cholesterol removal, and degree of hydrolysis exhibited different values before and after in vitro digestion.
Article
Scope Yogurt consumption is related to a decreased risk of colorectal cancer (CRC), but whether such association is causal remains unclear. Patients with familial adenomatous polyposis (FAP) are at increased risk of CRC development. Here, the study investigates the efficacy of yogurt for intestinal polyposis chemoprevention in Apc Min/+ mice, a preclinical model for human FAP. Methods and results A 10‐week yogurt supplementation (15 g kg ⁻¹ ) in Apc Min/+ mice significantly reduces the intestinal polyp number (6.50 ± 0.97 versus 1.80 ± 0.49; p < 0.001) compared to controls. 16S rRNA gene‐based microbiota analysis suggests that yogurt supplementation may greatly modulate the gut microbiome composition, especially in the relative abundance of Lactobacillus and Bifidobacterium . Importantly, the fecal concentration of d ‐lactate ( d ‐Lac, 0.39 ± 0.04 µmol g ⁻¹ versus 8.14 ± 0.62 µmol g ⁻¹ ; p < 0.001) is boosted by yogurt, while oral administration with d ‐Lac (125 or 250 mg kg ⁻¹ ) reduces the polyp number by 71.43% or 77.14% ( p < 0.001), respectively. The study also observes that d ‐Lac does not affect cell viability and anchorage‐independence in CRC cells, but it greatly suppresses epidermal growth factor (EGF) or 12‐O‐tetradecanoylphorbol‐13‐acetate (TPA)‐induced cell transformation in preneoplastic cells. Mechanistically, it demonstrates that d ‐Lac may attenuate epithelial cell transformation by targeting PI3K/AKT/β‐catenin axis. Conclusion Yogurt protects against intestinal polyposis in Apc Min/+ mice, and d ‐Lac may partially account for the chemopreventive effects above.
Article
Scope Epidemiological studies indicate an inverse correlation between yogurt consumption and colorectal cancer (CRC), but whether there is a cause‐and‐effect relationship has not yet been validated. This study aims to investigate the effects and possible mechanisms of yogurt on colitis‐associated colorectal cancer (CAC) in mice. Methods and results Experimental CAC is induced by azoxymethane (AOM, 10 mg kg ⁻¹ , ip) followed by three cycles of dextran sulfate sodium (DSS, 3%) treatment. Colitis is induced by adding DSS (3%) in drinking water for 5 days. Primary mouse macrophages are isolated for mechanistic studies. Data clearly show that yogurt (15 g kg ⁻¹ body weight) significantly reduces the multiplicity of colonic neoplasms by 38.83% in mice. Yogurt protects mice from colitis dependent on lactate receptor GPR81. The deficiency of Gpr81 exacerbates colitis and CAC in mice. Further investigation reveals that GPR81 may be dispensable for gut barrier function but essential for colonic mucosal repair. d ‐lactate in yogurt can activate GPR81 to suppress proinflammatory macrophage polarization, thereby facilitating inflammatory resolution after colonic injury and consequently suppressing CAC progression. Conclusion Yogurt effectively protects against colitis‐associated colorectal tumorigenesis in mice, and this study provides a rationale for introducing yogurt supplementation to patients with chronic inflammatory bowel diseases.
Article
Full-text available
Consumption of lactic acid bacteria (LAB) has been suggested to confer a range of health benefits including stimulation of the immune system and increased resistance to malignancy and infectious illness. In the present study, the effects of feeding Lactobacillus rhamnosus(HN001, DR20(TM)),Lactobacillus acidophilus(HN017)and Bifidobacterium lactis(HN019, DR10(TM))on in vivo and in vitro indices of natural and acquired immunity in healthy mice were examined. Mice were fed daily with L. rhamnosus, L. acidophilusor B. lactis(10(9) colony forming units) and their immune function was assessed on day 10 or day 28. Supplementation with L. rhamnosus, L. acidophilusor B. lactis resulted in a significant increase in the phagocytic activity of peripheral blood leucocytes and peritoneal macrophages compared with the control mice. The proliferative responses of spleen cells to concanavalin A (a T-cell mitogen) and lipopolysaccharide (a B-cell mitogen) were also significantly enhanced in mice given different LAB. Spleen cells from mice given L. rhamnosus, L. acidophilusor B. lactis also produced significantly higher amounts of interferon-gamma in response to stimulation with concanavalin A than cells from the control mice. LAB feeding had no significant effect on interleukin-4 production by spleen cells or on the percentages of CD4(+), CD8(+) and CD40(+) cells in the blood. The serum antibody responses to orally and systemically administered antigens were also significantly enhanced by supplementation with L. rhamnosus, L. acidophilusor B. lactis. Together, these results suggest that supplementation of the diet with L. rhamnosus(HN001),L. acidophilus(HN017)or B. lactis(HN019)is able to enhance several indices of natural and acquired immunity in healthy mice.
Article
The 65 kDa protein occludin is a membrane-spanning part of the epithelial tight junction, which is the main barrier of the paracellular pathway. The function of occludin as part of tight junctions is still poorly understood and even less is known about the regulatory mechanisms that influence occludin gene expression. This study aimed to identify the sequences essential in cis for genomic regulation of tight junction formation and to investigate their funcional role in cytokine-dependent tight junction regulation. Using genome walking cloning of occludin-specific human genomic DNA sequences, a 1853 bp DNA fragment containing the transcription start point of occludin cDNA sequences was amplified and sequenced. Subcloning of this fragment in front of the luciferase reporter gene revealed strong expression of enzymatic activity after transfection of the human intestinal cell line HT-29/B6. With subsequent deletions of parts of the promoter fragment, its size was reduced to 280 bp that are necessary and sufficient to mediate promoter activity. Tumor necrosis factor α and another cytokine involved in inflammation, interferon γ, reduced transepithelial resistance in HT-29/B6 cells, which was preceded by a decrease in occludin mRNA expression as revealed by northern blot analysis. Tumor necrosis factor α and interferon γ diminished occludin promoter activity alone and even synergistically, suggesting a genomic regulation of alterations of the paracellular barrier. In conclusion, proinflammatory cytokines such as tumor necrosis factor α and interferon γ can downregulate the expression of the transmembrane tight junction strand protein occludin, paralleling the barrier disturbance detected electrophysiologically. This could be an important mechanism in gastrointestinal diseases accompanied by barrier defects, for example inflammatory bowel diseases.
Article
OBJECTIVE: The major objective of this study is to provide estimates of diarrhoea mortality at country, regional and global level by employing the Child Health Epidemiology Reference Group (CHERG) standard. METHODS: A systematic and comprehensive literature review was undertaken of all studies published since 1980 reporting under-5 diarrhoea mortality. Information was collected on characteristics of each study and its population. A regression model was used to relate these characteristics to proportional mortality from diarrhoea and to predict its distribution in national populations. FINDINGS: Global deaths from diarrhoea of children aged less than 5 years were estimated at 1.87 million (95% confidence interval, CI: 1.56-2.19), approximately 19% of total child deaths. WHO African and South-East Asia Regions combined contain 78% (1.46 million) of all diarrhoea deaths occurring among children in the developing world; 73% of these deaths are concentrated in just 15 developing countries. CONCLUSION: Planning and evaluation of interventions to control diarrhoea deaths and to reduce under-5 mortality is obstructed by the lack of a system that regularly generates cause-of-death information. The methods used here provide country-level estimates that constitute alternative information for planning in settings without adequate data.
Article
The age-specific relevance of blood pressure to cause-specific mortality is best assessed by collaborative meta-analysis of individual participant data from the separate prospective studies. Methods Information was obtained on each of one million adults with no previous vascular disease recorded at baseline in 61 prospective observational studies of blood pressure and mortality. During 12.7 million person-years at risk, there were about 56 000 vascular deaths (12 000 stroke, 34000 ischaemic heart disease [IHD], 10000 other vascular) and 66 000 other deaths at ages 40-89 years. Meta-analyses, involving "time-dependent" correction for regression dilution, related mortality during each decade of age at death to the estimated usual blood pressure at the start of that decade. Findings Within each decade of age at death, the proportional difference in the risk of vascular death associated with a given absolute difference in usual blood pressure is about the same down to at least 115 mm Hg usual systolic blood pressure (SBP) and 75 mm Hg usual diastolic blood pressure (DBP), below which there is little evidence. At ages 40-69 years, each difference of 20 mm Hg usual SBP (or, approximately equivalently, 10 mm Hg usual DBP) is associated with more than a twofold difference in the stroke death rate, and with twofold differences in the death rates from IHD and from other vascular causes. All of these proportional differences in vascular mortality are about half as extreme at ages 80-89 years as at,ages 40-49 years, but the annual absolute differences in risk are greater in old age. The age-specific associations are similar for men and women, and for cerebral haemorrhage and cerebral ischaemia. For predicting vascular mortality from a single blood pressure measurement, the average of SBP and DBP is slightly more informative than either alone, and pulse pressure is much less informative. Interpretation Throughout middle and old age, usual blood pressure is strongly and directly related to vascular (and overall) mortality, without any evidence of a threshold down to at least 115/75 mm Hg.
Article
Dietary regulation of appetite may contribute to the prevention and management of excess body weight. The present study examined the effect of consumption of individual dairy products as snacks on appetite and subsequent ad libitum lunch energy intake. In a randomised cross-over trial, forty overweight men (age 32 (sd 9) years; BMI 27 (sd 2) kg/m2) attended four sessions 1 week apart and received three isoenergetic (841 kJ) and isovolumetric (410 ml) servings of dairy snacks or water (control) 120 min after breakfast. Appetite profile was determined throughout the morning and ad libitum energy intake was assessed 90 min after the intake of snacks. Concentrations of amino acids, glucose, insulin, ghrelin and peptide tyrosine tyrosine were measured at baseline (0 min) and 80 min after the intake of snacks. Although the results showed that yogurt had the greatest suppressive effect on appetite, this could be confounded by the poor sensory ratings of yogurt. Hunger rating was 8, 10 and 24 % (P < 0·001) lower after the intake of yogurt than cheese, milk and water, respectively. Energy intake was 11, 9 and 12 % (P < 0·02) lower after the intake of yogurt, cheese and milk, respectively, compared with water (4312 (se 226) kJ). Although there was no difference in the postprandial responses of hormones, alanine and isoleucine concentrations were higher after the intake of yogurt than cheese and milk (P < 0·05). In conclusion, all dairy snacks reduced appetite and lunch intake compared with water. Yogurt had the greatest effect on suppressing subjective appetite ratings, but did not affect subsequent food intake compared with milk or cheese.
Article
The intestinal barrier serves 2 critical functions for the survival of the individual: first, it allows nutrient absorption and second, it defends the body from dangerous macromolecule penetration. It is a complex multilayer system, consisting of an external "anatomic" barrier and an inner "functional" immunological barrier. The interaction of these 2 barriers enables equilibrated permeability to be maintained. Many factors can alter this balance: gut microflora modifications, mucus layer alterations, and epithelial damage can increase intestinal permeability, allowing the translocation of luminal content to the inner layer of intestinal wall. Several techniques are now available that enable us to study gut permeability: "in vitro" models (Caco-2 and HT29-MTX cells) and "in vivo" not invasive tests (sugar tests and radioisotope scanning tests) are used to estimate permeability and to suggest molecular pathophysiological mechanisms of intestinal permeability in health and diseases. Many medicinal products used in the treatment of gastrointestinal diseases have also found to play an active role in modulate intestinal permeability: corticosteroids, 5-aminosalicylic acid, anti tumor necrosis factor, probiotics, and mucosal protectors, like gelatin tannate. This review will particularly address the role of the gut barrier in maintaining intestinal permeability (microbiota, mucus, and epithelial cells), the techniques used for estimating intestinal permeability and the therapeutic approaches able to modify it.