Am J Clin Nutr 2004 Adolfsson 245 56

Embed Size (px)

Citation preview

  • 8/12/2019 Am J Clin Nutr 2004 Adolfsson 245 56

    1/12

    Review Article

    Yogurt and gut function1,2

    Oskar Adolfsson, Simin Nikbin Meydani, and Robert M Russell

    ABSTRACT

    In recent years, numerous studies have been published on the health

    effects of yogurt and the bacterial cultures used in the production of

    yogurt. In the United States, these lactic acidproducing bacteria

    (LAB) includeLactobacillusandStreptococcusspecies. The bene-

    fits of yogurt and LAB on gastrointestinal health have been inves-

    tigatedin animalmodels and, occasionally,in human subjects.Some

    studies using yogurt, individual LAB species, or both showedprom-

    ising health benefits for certain gastrointestinal conditions, includ-

    ing lactose intolerance, constipation, diarrheal diseases, colon can-cer, inflammatory bowel disease,Helicobacter pylori infection, and

    allergies. Patients with any of theseconditionscould possibly benefit

    from theconsumptionof yogurt. Thebenefitsof yogurtconsumption

    to gastrointestinal function are most likely due to effects mediated

    through the gut microflora, bowel transit, and enhancement of gas-

    trointestinal innate and adaptive immune responses. Although sub-

    stantial evidence currently exists to support a beneficial effect of

    yogurt consumptionon gastrointestinalhealth, thereis inconsistency

    in reported results, which may be due to differences in the strains of

    LAB used, in routes of administration, or in investigational proce-

    dures or to the lack of objective definition of gut health. Further

    well-designed, controlled human studies of adequate duration are

    needed to confirm or extend these findings. Am J Clin Nutr2004;80:24556.

    KEY WORDS Yogurt, gut function, gut immunity, gastroin-

    testinal diseases, gut microflora

    INTRODUCTION

    Components of the human intestinal microflora and of the

    foodenteringthe intestine mayhave harmful or beneficial effects

    on human health. Abundant evidence implies that specific bac-

    terial species used for the fermentation of dairy products such as

    yogurt and selected from the healthy gut microflora have pow-erful antipathogenic and antiinflammatory properties. These mi-

    croorganisms are therefore involved with enhanced resistance to

    colonization of pathogenic bacteriain the intestine, which has led

    to the introduction of novel modes of therapeutic and prophy-

    lactic interventions based on the consumption of monocultures

    and mixed cultures of beneficial live microorganisms as probi-

    otics. Probiotics are defined as living microorganisms, which

    on ingestion in sufficient numbers, exert health benefits beyond

    inherent basic nutrition (1).

    Yogurt is one of the best-known of the foods that contain

    probiotics. Yogurt is defined by the Codex Alimentarius of 1992

    as a coagulated milk product that results from thefermentation of

    lactic acid in milk by Lactobacillus bulgaricusand Streptococ-

    cus thermophilus(2). Other lactic acid bacteria (LAB) species

    are now frequently used to give the final product unique charac-

    teristics. As starter cultures for yogurt production, LAB species

    display symbiotic relations during their growth in milk medium

    (3). Thus, a carefully selected mixture of LAB species is used to

    complement each other and to achieve a remarkable efficiency in

    acid production. Furthermore, to increase the number of LAB

    that survive the low pH and high acidity of the gastrointestinalenvironment, some LAB species that are indigenous to the hu-

    man intestine have been used in yogurt production. To meet the

    National Yogurt Associations criteria for live and active cul-

    ture yogurt, the finished yogurt product must contain live LAB

    in amounts108organisms/g at the time of manufacture (3),and

    the cultures must remain active at the end of the stated shelf life,

    as ascertained with the use of a specific activity test.

    In many modern societies, fermented dairy products make up

    a substantial proportion of the total daily food consumption.

    Furthermore, it has long been believed that consuming yogurt

    and other fermented milk products provides various health ben-

    efits (4). Studies from the1990son thepossiblehealth properties

    of yogurt added to this belief (1, 5).Probiotic therapy is based on the notion that there is such a

    thing as a normal healthy microflora, but normal healthy mi-

    croflora has not been defined except perhaps as microflora with-

    out a pathogenic bacterial overgrowth. The development of

    novel means of characterizing and modifying the gut microflora

    has opened up new perspectives on the role of the gut microflora

    in health and disease. Numerous studies suggested beneficial

    therapeutic effects of LAB on gut health. However, results have

    been inconsistent, which may be due to differences in the strains

    of LAB, routes of administration, and investigational procedures

    used in these studies.

    Several LAB species are currently used in the production of

    yogurt. This review focuses on the current evidence suggestingthat yogurt and specific LAB species that are used for the fer-

    mentation of milk may or may not have valuable health-

    promoting properties or therapeutic effects on various gastroin-

    testinal functions and diseases.

    1 From theJeanMayerUSDAHumanNutritionResearch Centeron Aging

    at Tufts University, Boston.2 Address reprint requests to SN Meydani, Nutritional Immunology Lab-

    oratory, JM USDA-HNRCA at Tufts University, 711 Washington Street,

    Boston, MA 02111. E-mail: [email protected].

    Received October 3, 2003.Accepted for publication February 12, 2004.

    245Am J Clin Nutr2004;80:24556. Printed in USA. 2004 American Society for Clinical Nutrition

    bygues

    tonMarch2,2014

    ajcn.nutrition.org

    Downloadedfrom

    http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/
  • 8/12/2019 Am J Clin Nutr 2004 Adolfsson 245 56

    2/12

  • 8/12/2019 Am J Clin Nutr 2004 Adolfsson 245 56

    3/12

    biohydrogenated derivative of linoleic acid, than does themilk from

    which the yogurt was processed (24). A fermented dairy product

    from India, referred to as dahi, has also been shown to have higher

    CLA content than does nonfermented dahi (25). The major sources

    of CLA in our diets are animal products from ruminants, in which

    CLA is synthesized by rumen bacteria. Increased consumption of

    dairy fat was shown to be associated with increased concentrations

    of CLA in both human adipose tissue (26) and human milk (27). It

    was hypothesizedthatbiohydrogenation alsooccursduring fermen-tationof milk andresultsinhigherconcentrationsof CLAin thefinal

    product (28).

    CLA was reported to have immunostimulatory and anticarci-

    nogenic properties (29). In a recent study of breast and colon

    cancer cells, Kemp et al (30) showed that the anticarcinogenic

    propertiesofCLAmaybeduetotheabilityofsomeCLAisomers

    to inhibit the expression of cyclins and thus halt the progression

    of thecellcycle from G1 to S phase.In addition,CLA induced the

    expression of the tumor suppressor p53.

    Minerals

    In addition to being a good source of protein, yogurt is an

    excellent source of calcium and phosphorus. In fact, dairy prod-ucts such as milk, yogurt, and cheese provide most of the highly

    bioavailable calcium in the typical Western diet. Because of the

    lower pH of yogurt compared with that of milk, calcium and

    magnesium are present in yogurt mostly in their ionic forms.

    Oneof the major functionsof calcium istherole itplaysin bone

    formation and mineralization. The calcium requirements during

    growth, pregnancy, and lactation are increased. However, the aver-

    age calcium intake of women of childbearing age is consistently

    less than is recommended (31). In addition, calcium intake of

    women tends to fall even lower during thepostmenopausalyears

    (32). This is especially important for postmenopausal women,

    who are at increased risk of bone loss and osteoporosis. Dietary

    fiber has an adverse effect on calcium absorption, whereas lac-tose may enhance the absorption of calcium (33). In the rat

    model, calcium retention was greater with consumption of a diet

    in which lactose made up half the total carbohydrates ingested

    than with consumption of the control diet (34). Schaafsma et al

    (35), investigating the effect of dairy products on mineral ab-

    sorption by using rat models, reported that lactose enhances the

    absorption of calcium,magnesium, and zinc. Because yogurt has

    a lactose content lower than that of milk, the bioavailability of

    these minerals may be negatively affected, although the effect is

    likely to be small.

    The acidic pH of yogurt ionizes calcium and thus facilitates

    intestinal calcium uptake (36). The low pH of yogurt also may

    reduce the inhibitory effect of dietary phytic acid on calcium

    bioavailability. Vitamin D plays a major regulatory role in in-

    testinal calcium absorption. The active, saturable, transcellular

    route of calcium absorption in the duodenum and proximal jeju-

    num requires calbindin-D, a vitamin D dependent calcium-

    binding protein (37). In the United States, milk and infant for-

    mula are fortified with vitamin D, and hence they serve as good

    dietary sources,with2.5 g (100IU) vitamin D/237-mLserving.

    However, other dairy products, such as yogurt, typically are not

    fortified with vitamin D.

    Few studies have investigated the effect of yogurt-derived

    calcium on bone mineralization in animals (34, 38). Kaup et al

    (34) reported that yogurt-fed rats showed greater bone mineral-

    ization than did rats fed a diet containing calcium carbonate.

    These studies may suggest that the bioavailability of calcium in

    yogurt is greater and yogurt may increase bone mineralization

    more than do nonfermented milk products. However, there are

    currently no published studies that show a superior effect of

    yogurt on bone mineralization in human subjects.

    MECHANISTIC RATIONALE FOR POTENTIALBENEFITS OF YOGURT ON GUT FUNCTION AND

    HEALTH

    It has been suggested that yogurt and LAB contribute to sev-

    eral facets of gastrointestinal health: the makeup of the gastro-

    intestinal flora, the immune response, and laxation.

    Gut microflora

    Lactobacilliare among the components of microbial flora in

    both the small and large intestines. The ability of nonpathogenic

    intestinal microflora, such as LAB, to associate with and bind to

    the intestinal brush border tissue is thought to be an important

    attribute that prevents harmful pathogens from accessing the

    gastrointestinal mucosa (39). For LAB to have an effect, theymust adapt to the host intestinal environment and be capable of

    prolonged survival in the intestinal tract (40 43). LAB survival

    is influenced by gastric pH as well as by exposure to digestive

    enzymes and bile salts (42), and LAB species differ in their

    ability to survive in the gastrointestinal environment (43).

    When 4 strains ofBifidobacterium(B. infantis, B. bifidum, B.

    adolescentis, and B. longum) were compared,B. longum was the

    most resistant to the effects of gastric acid (44).Bifidobacterium

    animaliswas reported to have a high survival rate during intes-

    tinal transit in human subjects (45).

    The effect of feeding yogurt fermented withS. thermophilus,

    L. bulgaricus, andLactobacillus casei on the fecal microflora of

    healthy infants aged 10 18 mo was investigated by Guerin-Danan et al (46). Whereas the number of infants with fecal

    Lactobacillus increased after the feeding, the total numbers of

    anaerobes,Bifidobacteria, bacteroides, and enterobacteria were

    not affected by yogurt intake. In a group of elderly patients with

    atrophic gastritis and hypochlorhydria, Lactobacillus gasseri

    survived passage through the gastrointestinal tract, butS. ther-

    mophilusandL. bulgaricuswere not recovered (43).Bifidobac-

    teriumsp has also been shown to survive passage through the

    gastrointestinal tract: fecal concentrations were detectable for

    8 d after the cessation of intake (47).

    Another important factor that limits the survival of lactobacilli

    within the upper gastrointestinaltract is the inherent ability of the

    organisms to adhere to intestinal epithelial cells (42). With the

    use of scanning electron microscopy, Plant and Conway (48)

    screened 16 strains ofLactobacillusfor their capacity to associ-

    ate with Peyers patches and the lymphoid villous intestinal tis-

    sues in mice. Two of the 16 strains investigated, Lactobacillus

    acidophilus andL. bulgaricus, are of interest because they relate

    to yogurt. It was found, in both in vitro and in vivo models using

    BALB/c mice, thatL. bulgaricusdid not associate with Peyers

    patches or with the lymphoid villous intestinal tissues. L. aci-

    dophilushad a low degree of association with Peyers patches

    and no association to the lymphoid villous intestinal tissue. Nev-

    ertheless, the authors stated that the strains ofLactobacillus

    tested showed high rates of survival when Lactobacillus was

    administered orally.

    YOGURT AND GUT FUNCTION 247

    bygues

    tonMarch2,2014

    ajcn.nutrition.org

    Downloadedfrom

    http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/
  • 8/12/2019 Am J Clin Nutr 2004 Adolfsson 245 56

    4/12

    Theability of LAB to decrease thegastrointestinal invasion of

    pathogenic bacteria has also been described (39, 49). Bernet et al

    (39) reported a dose-dependentL. acidophilusmediated inhibi-

    tion of the adherence of enteropathogenic Escherichia coliand

    Salmonella typhimurium to the enterocyte cell-line Caco-2. In

    addition,L. acidophilusinhibited the entry ofE. coli,S. typhi-

    murium, and Yersinia pseudotuberculosisinto Caco-2 cells. In

    another report (49), the sameauthorsdescribed similarinhibitory

    effects when 2 different strains ofBifidobacteria (B. breve andB.infantis) were used. Inaddition, long-termfeedingof yogurtdoes

    not result in a significant change in the results of breath-hydrogen

    tests, which indicates the absence of a significant change in the

    intestinal survival of the yogurt organisms (50). Furthermore, it

    is possible that the ability of LAB to compete with pathogens for

    adhesion to the intestinal wall is influenced by their membrane

    fluidity. This possibility was suggested by studies indicating that

    the type and quantities of polyunsaturated fatty acids in the ex-

    tracellular milieu influence the adhesive properties of LAB to the

    epithelium (51, 52).

    Gut-associated immune response

    The mucosal lymphoid tissue of the gastrointestinaltract playsan important role as a first line of defense against ingested patho-

    gens. The interactions of LAB with the mucosal epithelial lining

    of the gastrointestinal tract, as well as with the lymphoid cells

    residing in the gut, have been suggested as the most important

    mechanism by which LAB enhances gut immune function. Sev-

    eral factors have been identified as contributing to the immuno-

    modulating and antimicrobial activities of LAB, including the

    production of low pH, organic acids, carbon dioxide, hydrogen

    peroxide, bacteriocins, ethanol, and diacetyl; the depletion of

    nutrients; and competition for available living space (1, 5, 53).

    The gastrointestinal tract is a complex immune system tissue.

    Themainsiteof themucosalimmunesystemin thegut is referred

    to as gut-associated lymphoid tissue (GALT), which can be di-vided into inductive and effector sites. In the small intestine, the

    inductive sites are in the Peyers patches, which consist of large

    lymphoid follicles in the terminal small intestine. The best-

    defined effector component of the mucosal adaptive immune

    system is secretory immunoglobulin A (sIgA). sIgA is the main

    immunoglobulin of the humoral immune response, which to-

    gether with the innate mucosal defenses provides protection

    against microbial antigens at the intestinal mucosal surface (54).

    In a healthy person, sIgA inhibits the colonization of pathogenic

    bacteria in the gut, as well as the mucosal penetration of patho-

    genic antigens. At least 80% of all the bodys plasma cells, the

    source of sIgA, are located in the intestinal lamina propria

    throughout the length of the small intestine. IgA is the most

    abundantly produced immunoglobulin in the human body. The

    production of intestinal sIgArequiresthe presenceof commensal

    microflora (55), which indicates that the production of intestinal

    sIgA is induced in response to antigenic stimulation. It is not yet

    clear, however, how lamina propria B cells are activated to be-

    come IgA-secreting plasma cells or how the intestinal microflora

    influence this process. Most studies on the effect of fermented

    milk or specific LAB on gut immune function have centered on

    their immune adjuvant effects in the gut.

    The ability of LAB to modulate IgA concentrations in the gut

    has also been the subject of several studies. Orally administered

    L. acidophilus and L. caseiand the feeding of yogurt increased

    both IgA production and the number of cells secreting IgA in the

    small intestine of mice in a dose-dependent manner (5). Simi-

    larly, a report by Puri et al (56) indicated that S. typhimurium-

    induced serum IgA concentrations were significantly higher in

    mice fed yogurt over a period of 4 wk than in milk-fed control

    mice. Thisreport suggeststhat the IgAsecretedby thechallenged

    intestinal B cells enters the circulation and increases the concen-

    trations of IgA in the serum. Thus the IgA-enhancing effect of

    yogurt intake may have both an effect on the gut and a systemic

    effect. The same study also showed that intestinal lymphocytes

    from micefed yogurt had a higher mitogen-induced proliferative

    response after a challenge with S. typhimurium than did those

    from control-fed mice.

    In a study using human subjects, Link-Amster et al (57)

    showed that the specific anti-IgA titer to S. typhimuriumwas 4

    times greater in subjects fed fermented milk containing L. aci-

    dophilus than in control subjects fed diets without fermented

    milk. Total sIgA concentrations also increased in subjects fed

    fermented milk.

    Macrophages play an important role as a part of the innate

    immune response in the gut, and they represent one of the first

    lines of nonspecific defense against bacterial invasion. The ef-

    fects of feeding milk fermented with either L. casei or L. aci-

    dophilusor both on the specific and nonspecific host defense

    mechanisms in Swiss mice were investigated by Perdigon et al

    (58). They showed that feeding milk fermented withL. casei, L.

    acidophilus, or both for 8 d increased the in vitro and in vivo

    phagocytic activity of peritoneal macrophages and the produc-

    tion of antibodies to sheep red blood cells. The activation of the

    immune system began on day 3, peaked on day 5, and decreased

    somewhat on day 8 of feeding. Phagocytic activity was further

    boosted in mice given a single dose of fermented milk on day 11

    of feeding.

    Modulation of cytokine production by yogurt and LAB has

    also been the focus of several studies. In addition to interleukin

    (IL)-1 and tumor necrosis factor (TNF) , which are mainlyproduced by macrophages, T lymphocytes are thesource of most

    cytokines investigated in those reports. T cells are frequently

    classified into2 categoriestype 1 (Th1)and type 2 (Th2) helper

    T cells. On activation, these cells produce 2 diverse patterns of

    cytokines (59). Th1 cells are the main producers of interferon-

    (IFN-) and IL-2, and Th2 cells produce IL-4, IL-5, IL-6, and

    IL-10. The Th1 cytokines boost cell-mediated immunity, and the

    Th2 cytokines augment humoral immunity. IFN-plays a criti-

    cal role in the induction of other cytokines and in mediation of

    macrophage and natural killer cell activation.

    Several reports indicated that consumption of yogurt or intake of

    LAB by themselves modulates the production of several cytokines,

    such as IL-1, IL-6, IL-10, IL-12, IFN-, and TNF- (60 63).Moreover, the production of IFN- in an in vitro culture system

    using human lymphocytes was reported to be greater with cultures

    in the presence of LAB (L. bulgaricus and S. thermophilus) than

    with those without LAB (64). Yogurt containing liveL. bulgaricus

    andS. thermophiluswasalso reported to augment IFN-production

    by purified T cells from young adults after 4 mo feeding (62).

    Effects of yogurt consumption on the modulation of cytokine

    production in the human gastrointestinal tract, whether by cells

    of the GALT or by others, have not been investigated. These

    types of studies, althoughfeasiblewith theuse of biopsysamples

    from the intestines of healthy subjects (65), are difficult to carry

    out, and good animal models currently do not exist.

    248 ADOLFSSON ET AL

    bygues

    tonMarch2,2014

    ajcn.nutrition.org

    Downloadedfrom

    http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/
  • 8/12/2019 Am J Clin Nutr 2004 Adolfsson 245 56

    5/12

    Even though cytokines play diverse roles in regulating

    immune functions, some cytokines, eg, IL-1, IL-6, and TNF-,

    have been given more attention than others because they have

    traditionally been classified as proinflammatory and as such are

    known to be associated with inflammatory conditions such as

    Crohn disease and ulcerative colitis (66). Another diverse family

    of immune modulators that play important roles in the health of

    the gastrointestinal tract consists of chemokines and their recep-

    tors (67). Currently, only limited data have been published on theeffect of yogurt or its components on chemokine modulation in

    the gastrointestinal tract. The effects of different strains ofLac-

    tobacilluson chemokine production by the intestinal epithelial

    cell-line, HT-29, were investigated by Wallace et al (68). All 3

    LAB species investigatedL. acidophilus, Lactobacillus rham-

    nosus,and Lactobacillus delbrueckii had suppressive effects

    on the production of 2 chemokines, RANTES (a member of the

    IL-8 superfamily of cytokines) and IL-8, by activated HT-29

    cells. As is the case with proinflammatory cytokines, these che-

    mokines are necessary for normal immune function. However, a

    high production of these chemokines during an inflammatory

    condition is believed to exacerbate the inflammatory response.

    Laxation

    Few reports have discussed the effects of yogurt and LAB on

    laxation. In the studies published, however, both significant ef-

    fects (G Wilhelm, unpublished observations, 1993; 69) and no

    effects (70) of yogurt or LAB on laxation and gastrointestinal

    transit time were described.

    Strandhagen et al (69) reported that the transit time for 50%

    (t50) of gastric content was significantly greater for ropymilk, an

    L. bulgaricus and S. thermophilusfermented milk product

    indigenous to Sweden, than for unfermented milk. Another study

    showed thatmilk fermented withL. bulgaricusand S. thermophi-

    lus reduced intestinal transit time in human subjectswith habitual

    constipation (G Wilhelm, unpublished observations, 1993). Inthe same study, subjects consuming fermented milk also had

    improved bowel function. The number of defecations increased

    from 3/wk during a control period to 7/wk when fermented milk

    was consumed. When milk fermented with L. acidophiluswas

    consumed,the number of defecations increasedfurther to 15/wk.

    Studies were conductedof theeffects of a commerciallyavail-

    able yogurt fermented with B. animalis on orofecal gut transit

    time (71, 72). In a double-blind, randomized, crossover design,

    B. animalis reduced the colonic transit time in a group of healthy

    women aged 18 45 y (72). Likewise, in a group of elderly

    subjects experiencing lengthy orofecal gut transit time but oth-

    erwise free of any gastrointestinal pathology,B. animalisintake

    provided led to a significant reduction in transit time (71). Thus,the effect of LAB ingestion on orofecal gut transit time appears

    to be dependent on the bacterial strain used and the population

    being studied.

    YOGURT AND DISEASES OF THE

    GASTROINTESTINAL TRACT

    Lactase deficiency and lactose maldigestion

    Lactase deficiency among adults is the most common of all

    known enzyme deficiencies. More than half of the worlds adult

    population is lactose intolerant.In developmentalterms,this may

    not necessarily be considered abnormal, because humans are the

    only known mammal in whom lactase activity in the small

    intestine is sustained after weaning. In the case of lactose mal-

    digestion, undigested lactose remains in the intestinal lumen,

    and, as it reaches the colon, it is fermented by colonic bacteria.

    Byproducts of thisprocess include short-chain fatty acids such as

    lactate, butyrate, acetate, and propionate. These fatty acids as-

    sociate with electrolytes and lead to an osmotic load that can

    induce diarrhea. Furthermore, fermentation of lactose by colonic

    bacteriaproducesmethane, hydrogen,and carbon dioxide.Thesegases may stay in the lumen and eventually will both be excreted

    as flatus, diffusing into the circulation, and be exhaled via the

    lungs. Exhaled hydrogen after a lactose load has been used as an

    indirect but measurable indicator of lactose maldigestion. In

    addition to lactose, some sources of dietary fiber and other un-

    absorbed carbohydrates can serve as substrates for colonic fer-

    mentation that results in increased hydrogen production.

    Inability to digest lactose varies widely among ethnic and

    geographic populations (73, 74). In the United States, the prev-

    alence of primary lactose intolerance in adults is 53% among

    Mexican Americans, 75% among African Americans, and 15%

    among whites. The prevalence among adults in South America

    and Africa is 50% and that in some Asian countries is close to100%. Lactose intolerance varies greatly between European

    countries, from 2% prevalence in Scandinavian adults to

    70% among Southern Italian adults (74).

    Lactose maldigestionmay develop secondary to inflammation

    oras a resultof functional loss ofthe smallintestinalmucosa(14),

    which can result from conditions such as Crohn disease, celiac

    sprue, short bowel syndrome, or bacterial and parasitic infec-

    tions. In addition, lactose maldigestion may develop as a conse-

    quence of severe protein calorie malnutrition. The disorder is

    clinically expressed by symptoms of abdominal cramps, diar-

    rhea, and flatulence after milk ingestion. However, most persons

    who have symptoms of lactose intolerance can endure small

    amounts (210 g) of lactose in a meal without becoming symp-tomatic (14).

    It is well known that, for many lactose-intolerant people, fer-

    mented milk products are better accepted than are unfermented

    milk products. There may be more than one reason for this.

    During fermentation of milk, lactose is partially hydrolyzed,

    which results in a lower lactose content in yogurtthanin milk (2).

    However, this reduction in lactose may not be significant, be-

    cause milk solids are usually added during processing. The

    greater tolerance of lactose from yogurt than of that from milk

    among lactose-intolerant subjects may be due to the endogenous

    lactase activity of yogurtorganisms (13, 15, 75). Kolarset al (15)

    used a series of breath hydrogen tests as well as a subjective

    assessment to ascertain whether subjects who were identified as

    lactose-intolerant digested and absorbed lactose in yogurt better

    than they digested and absorbed lactose in milk. The area under

    thecurvefor breathhydrogenwas smaller after yogurtconsump-

    tion than after consumption of milk or lactose in water, which

    indicates better digestion and absorption of lactose from yogurt

    than of that from either milk or lactose in water. Subjective

    assessment by the subjects in the study of Kolars et al also

    indicatedthat lactose in yogurtwas bettertolerated than thesame

    amount of lactose from milk or in water. Using breath hydrogen

    measurement, Savaiano et al (75) investigated the effects of 3

    varieties of cultured milk products on the digestion of lactose by

    9 lactase-deficient human subjects. When yogurt, cultured milk

    (buttermilk), and sweet acidophilus milk were compared, yogurt

    YOGURT AND GUT FUNCTION 249

    bygues

    tonMarch2,2014

    ajcn.nutrition.org

    Downloadedfrom

    http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/
  • 8/12/2019 Am J Clin Nutr 2004 Adolfsson 245 56

    6/12

    had the most beneficial effect on lactose digestion in these sub-

    jects. Lactase activity and the number of surviving LAB were

    significantly reduced when the yogurt was pasteurized.

    The enzyme activity of lactase is generally stable in response

    to environmental factors.Forexample,it wasshownthat thelactase

    activity ofyogurtwas preservedand even increasedwhen theyogurt

    was subjected to an environment that simulated the temperature

    and low pH values of the gut(15). As suggested by the authors, this

    study supports the notion that lactose in yogurt is autohydrolyzedonce it is in the jejunal environment. Other studies reported that

    lactase activity is less stable in response to acidic environment.

    Pochartet al(76)reported that lactase activityin yogurtdecreased by

    80% at a pH of 5.0 in an in vitro model.

    However, heating yogurt does significantly decrease lactase

    activity, which indicates that yogurt that has been heat treated is

    not as beneficial for lactose-intolerant persons is yogurt contain-

    ing live and active cultures. Thus, there is a growing body of

    evidence that yogurt containing live and active cultures is better

    tolerated by lactose malabsorbers than are heat-treated fer-

    mented milks (50). During the fermentation process, the amount

    of lactose present in yogurt is reduced. The lactose content also

    varies withthe durationof storage after fermentation. In addition,the bacterial lactaseactivity corresponds withthe survivaltime of

    lactobacilli after ingestion. The enhanced digestion of lactose is

    explained partly by the improved lactase activity after yogurt

    ingestion and partly by other enzymatic functions, such as the

    activity of the lactose transport system (permease) that allows

    lactose to enter the probiotic cell (77, 78). Furthermore, animal

    studies have suggested that LAB may induce lactase activity of

    the gut intestinal endothelial cells (79).

    A study by Martini et al (80) supports the microbial mediation

    of lactase activity in the gastrointestinal tract. Those authors

    showed that lactase activity in yogurt was stable at pH 4.0, but

    that microbial cell disruption resulted in 80% loss of lactase

    activity and a twofold increase in lactose malabsorption in agroup of lactose maldigesters.

    Although the organisms that make up the live cultures in

    yogurt are recognized as having functional lactase activityand as

    contributing to the digestion of lactose, their survival in the

    gastrointestinal tract is short. On average, significant numbers

    survive for 1 h after ingestion (15, 50). Regardless of this

    somewhat limited survival time, the beneficial effect of LAB on

    lactose digestion in those suffering from lactose intolerance is

    now widely accepted.

    Diarrheal diseases

    Diarrheais a common problemamong childrenworldwide and

    has been reported to contribute substantially to pediatric physi-

    cian visits and hospitalizations in the United States (81). Since

    the early 20thcentury,it has beenhypothesized thatlive bacterial

    cultures, such as those used for the fermentation of dairy prod-

    ucts, may offer benefits in preventing and treating diarrhea (4).

    A recent meta-analysis of randomized, controlled studies by

    Van Neil et al (82) found that therapy usingLactobacillus strains

    offered a safe and effective means of treating acute infectious

    diarrheain children. Boththe durationand frequencyof diarrheal

    episodes were reduced when compared with those in control

    subjects. The benefit ofLactobacillus therapywas seenin diarrheal

    diseases caused by various pathogens. The effect of supplementing

    formula withB. bifidum andS. thermophiluson preventingthe onset

    of acute viral diarrhea in infants was examined in a double-blind,

    placebo-controlledtrial(83).The infants receiving bacterial therapy

    developed diarrhea and shed rotavirus less than did the infants fed

    the control formula. Evidence of the beneficial effect of LABon the

    occurrence of diarrhea of bacterial origin is more contradictory be-

    cause both benefits (84, 85) and no effects (86, 87) of feeding LAB

    were reported.

    Several studies investigatedthe effectsof probiotic bacteriaon

    diarrhea associated with the use of antibiotics. The most likely

    cause of diarrhea associated with antibiotic use is the negativeinfluence of antibiotics on the bacterial steady state of the intes-

    tines (88). Most cases of antibiotic-associated diarrhea are mild,

    and they end shortly after antibiotic therapy is discontinued. A

    less common but more serious type of antibiotic-associated di-

    arrhea is due to antibiotic-mediated overgrowth of pathogenic

    bacterial species such as Clostridium difficilethat is associated

    with pseudomembranous colitis (89).

    A recent meta-analysis evaluated the ability of several differ-

    ent probiotic LAB species to prevent antibiotic-associated diar-

    rhea (90). Of the 9 studies that were included in the analysis, 4

    used Lactobacilli strains or a combination ofLactobacilli and

    Bifidobacteria(9194). Of those 4 studies, 2 showed a signifi-

    cant benefit of probioticuse in comparison with placebo (93, 94).The authors concluded that probiotic bacteria supplied in cap-

    sules or as yogurt-based products may be useful in preventing

    antibiotic-associated diarrhea. However, none of these studies

    provide evidence for a role of probiotic bacteria in the treatment

    of such diarrhea.

    The mechanisms by which LAB may provide a beneficial

    effect against some forms of diarrheal disease are unknown. It

    has been suggested that the beneficial effect may stem from the

    ability of LAB to reestablish the intestinal microflora,to increase

    the intestinal barrier by competing with pathogenic bacteria for

    adhesion to the enterocytes, or to increase mucosal IgA response

    to pathogens.

    Colon cancer

    According to the National Cancer Institute, cancer of the colon

    is the second leading cancer diagnosis among both women and

    men in the United States (95). Colon cancer is also the second

    most common cause of cancer death. Risk factors for colorectal

    cancer include both genetic and environmental factors, and sev-

    eral reports have suggested that interactions between dietary

    factors, colonic epithelium, and intestinal flora are central to the

    development of colon cancer.

    Theroleof diet in theetiology of cancerhas been given greater

    attention in recent years. Although the relation between colon

    cancer and certain food constituents, such as fiber and fat, gen-

    erated the mostinterest, the possibility thatfermented dairy prod-

    ucts may protect against tumor formation in the colon was also

    investigated. Epidemiologic evidence suggests a negative corre-

    lation between the incidence of certain cancers, including colon

    cancer, and the intake of fermented dairy products (96). More-

    over, fermented dairy products or the bacteria used for milk

    fermentation were shown to have an effect on colon cancer and

    certain other tumors in murine models of carcinogenesis (97

    100). However, a number of animal studies investigating the

    effectof various strains of LAB on colon carcinogenesis showed

    inconsistent results.

    Wollowski et al (100) investigatedtheprotective effectof several

    strains of LAB, traditionally used for milk fermentation, against

    1,2-dimethylhydrazine (DMH)induced colon carcinogenesis in

    250 ADOLFSSON ET AL

    bygues

    tonMarch2,2014

    ajcn.nutrition.org

    Downloadedfrom

    http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/
  • 8/12/2019 Am J Clin Nutr 2004 Adolfsson 245 56

    7/12

    rats. Oral treatment with L. bulgaricus for 4 d protected against

    DMH-induced DNAdamage in the colon. In contrast, there was no

    protective effect whenS. thermophiluswas administered. The au-

    thors did not ascertain the mechanisms of protection byL. bulgari-

    cus, buttheyspeculatedthatthiol-containingbreakdownproducts of

    proteins that resultfromtheproteolyticactivityofL. bulgaricusmay

    have produced the effect.

    In a previousstudy using a similar DMH-induced colon cancer

    model in rats, Shackelford et al (99) showed that milk fermentedwithL. bulgaricus resulted in greater survival than did nonfer-

    mented milk. However, in contrast to the findings of Wollowski

    et al (100), L. bulgaricus-fermented milk did not reduce the

    number of rats that developed colon tumors, whereas S.

    thermophilus-fermented milk did do so (99). In a study using

    azoxymethane to induce aberrant crypt foci in the colon of rats,

    no significant effects were seen with eitherB. longum orL. casei

    (101). Those authors did, however, observe a protective effect of

    L. acidophilusand inulin, but only when the total fat content of

    the diet was increased.

    Using a colon carcinoma cell culture system, Ganjam et al

    (102) isolated a yogurt fraction that decreased cell proliferation,

    as ascertained with the use of thymidine incorporation. Cell pro-liferation was not inhibited in response to a similarly isolated

    milk fraction or to lactic acid.

    Elevated activity of several bacterial fecal enzymes, some of

    which are involved in the metabolism of genotoxic nitrates, was

    associated with an increasedrisk of colon cancer (103, 104). The

    activity of these enzymes can be altered by diet or antibiotic

    intake (10, 105). L. acidophilus(106) andL. gasseri(43) were

    shown to reduce the fecal enzyme activity of nitroreductase,

    azoreductase, and-glucuronidase in humans, with a reduction

    by 50% or 75% in the activities of these enzymes during a period

    ofLactobacilli feeding. Likewise, Guerin-Danan et al (46) re-

    ported that 10 18-mo-old infants fed yogurt fermented withS.

    thermophilus, L. bulgaricus, and L. casei had lower fecal-glucuronidase activity than did a similar group of infants fed

    milk or yogurt not fermented with L. casei.

    The mechanism by which LAB may have an effect on colon

    carcinogenesis is currently unknown. Some of the mechanisms

    thatmay be involved includeenhancement ofthehosts gutimmune

    response, suppression of harmful intestinal bacteria, sequestration

    of potential mutagens, production of antimutagenic compounds,

    reduction of pH concentrations in the colon, and alteration of other

    physiologicconditions(107). Furthermore,it wasshownby Pedrosa

    et al (43) that the feeding of yogurt or Lactobacillusreduced fecal

    enzymes, which convert procarcinogens to carcinogens, such as

    azoreductase and nitroreductase.

    Inflammatory bowel disease

    Inflammatory bowel disease (IBD) is a term used for certain

    chronic immunemediated conditions of the intestinal tract.

    These chronic diseases include Crohn disease and ulcerative

    colitis,conditions thathave comparable symptomsbut thataffect

    the digestive tract in very different ways (66). Ulcerative colitis

    involves inflammationof thecolon andrectum andnot that of the

    uppergastrointestinal tract, whereas Crohn disease can affect the

    upperintestinaldigestive tract and thuscan leadto malabsorption

    of both macronutrients and micronutrients. The etiologies of

    these diseasesare unknown,but studies suggest thatthe intestinal

    microflora play a crucial pathogenic role (108). This notion is

    supported by animal models of Crohn disease, in which the

    presence of intestinal microflora is absolutely required for the

    development of disease.

    Proinflammatory cytokines, particularly TNF-, have also

    been recognized as playing a central role in the pathogenesis of

    Crohn disease. However, despite earlier hopes, the results from

    studies using TNF-antagonists were disappointing, and there

    were some reports of severe complications (109). Nevertheless,

    reducing the production or effect of TNF-(or both) in Crohndisease patients is belived to be beneficial. Bourrel et al (63)

    reported that, when inflamed intestinal mucosa from a group of

    Crohns disease patients was cocultured in the presence of L.

    casei or L. bulgaricus, expression and release of TNF- by

    intraepithelial lymphocytes were reduced.

    Normally, a healthy mucosal barrier provides a first defense

    mechanism against both the intestinal microflora and invading

    pathogens. It hasbeen suggestedthat theproportions of different

    intestinal microflora are altered in patients with IBD. For exam-

    ple, colonic biopsy specimens have shown lower concentrations

    ofLactobacillus and lower fecal concentrations of both Lacto-

    bacillus andBifidobacterium species in patients with Crohn dis-

    ease than in healthy subjects (110). This disturbance in intestinalflora may increase the opportunity for colonization of pathogens

    and bring about a subsequent proinflammatory response.

    In the case of IBD, a defective mucosal barrier allows for in-

    creased uptake of antigens and proinflammatory mediators origi-

    nating from luminal bacteria. It has been reported that patients with

    IBDhavediminished mucosal protectionas a resultof changesin the

    composition andthicknessof themucosallayerandalterationsin the

    glycosylationstatusof mucosal glycoproteins(111). Thesechanges

    in theintestinalmucosa arealso associatedwith decreasedintestinal

    IgA activity and increased IgG activity, which coincides with re-

    duced state of protection and a proinflammatory condition. With

    weakened mucosalbarrier and thereby increased adherence of bac-

    terial pathogens to the mucosa, sustained inflammation results, andthat leads to further damage to the gut mucosa. In recent years,

    immunosuppressive and immunomodulating therapies, such as the

    steroids usedsincethe1960s,havebecome more andmore frequent

    in the treatment of these conditions. Although efficacious, these

    types of drugs can increase the prevalence of opportunistic infec-

    tions as well as the severity of any underlying infection that may be

    present (112). Other side effects of these treatments may include

    hepatotoxicity, fibrosis, lymphoma, and pathologic suppression of

    bone marrow function.

    The roleof beneficial intestinal microflora in the prevention of

    intestinal inflammation was investigated by using gene-targeted

    IL-10 knockout (IL-10/) mice (113, 114). These IL-10 defi-

    cient mice spontaneously develop ileocolitis with many similar-

    ities to Crohn disease in humans. Furthermore, affected mice

    respond favorably to immunosuppression or immunomodula-

    tory drugs that are similar to those used to treat human IBD. The

    immunoregulatory activity of IL-10 has been studied exten-

    sively. It is now well established that IL-10 plays a role in down-

    regulating both the synthesis of inflammatory cytokines and

    the presentation of antigens. Thus, IL-10 has been suggested for

    use as an immunomodulator for the treatment of Crohn disease.

    Targeted in vivo delivery of IL-10 to the affected intestinal ep-

    ithelium by using genetically engineeredLactococcus lactishas

    shown great promise in 2 mouse models of IBD (114).

    Madsen et al (113) found that IL-10/ mice had increased

    adherence of luminal bacteria to the mucosal layer in the colon

    YOGURT AND GUT FUNCTION 251

    bygues

    tonMarch2,2014

    ajcn.nutrition.org

    Downloadedfrom

    http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/
  • 8/12/2019 Am J Clin Nutr 2004 Adolfsson 245 56

    8/12

    that preceded the development of colitis. This occurred in par-

    allel to decreased numbers of luminal Lactobacillus. When the

    concentrations ofLactobacillus in the gastrointestinal lumen

    were restored by rectal delivery ofLactobacillus reuteri or by

    oral lactulose therapy, colitis was attenuated. The concentrations

    of adherent and translocated bacteria in the mucosal wall also

    were reduced.

    Another benefit of LAB in Crohn disease may be due to the

    stimulation of the IgA response. A report by Malin et al (115)suggests that oral bacteriotherapy using L. casei can restore

    antigen-specific IgA immune response in persons with Crohn

    disease. In a previous study from the same laboratory (116), oral

    administration ofL. casei to patients with viral gastroenteritis

    promoted antigen-specific IgA responses and shortened the pa-

    tient diarrhea.

    Although experimental evidence exists indicating beneficial

    effects of LAB on Crohn disease and ulcerative colitis, the exact

    mechanism through which LAB species antagonize the progres-

    sion of these diseases is poorlyunderstood. The exact etiology of

    IBD is also unknown, but it is likely that, in susceptible persons,

    IBD results from an ongoing inflammatoryresponse, which may

    be due to a defect in both the regulation of the mucosal proin-flammatory response and the function of the intestinal epithe-

    lium. Currently, evidence suggests that yogurt and LAB have

    modestclinical benefits andare safe foruse in patients with these

    conditions. Further studies are required to ascertain whether yo-

    gurt is beneficial as a prophylactic or a therapeutic regimen for

    IBD (or both) and to establish exactly which mechanisms are

    involved.

    Helicobacter pylori

    It has only been 20 y since Helicobacter pylori, a gram-

    negative, spiral-shaped bacterium that is found in the gastric

    mucous layer or adherent to the epithelial lining of the stomach,

    was discovered (117). H. pylori relies on the ammonia-producing surface protein urease for adherence and colonization

    to the gastric epithelium. Urease allows H. pylorito survive by

    neutralizing the acidic gastric environment (118).H. pyloripro-

    duces catalase, which may play a role in protecting the bacteria

    from free radicals that are released by activated leukocytes. H.

    pyloriinfection is associated with a massive infiltration of neu-

    trophils into the gastric wall and local production of IFN-,

    proinflammatory cytokines eg,TNF-, IL-1, and IL-6and

    the chemokine IL-8.

    Infection withH. pyloriis now known to play a role in peptic

    ulcer disease, chronic gastritis, gastric adenocarcinoma, and

    mucosa-associated lymphoid tissue lymphoma. The association

    between duodenal ulcer disease and H. pyloriis also well doc-

    umented:H. pyloriinfection is reported in 90% of duodenal

    ulcer patients (119). Treatment of this infection involves the use

    of proton pump inhibitors, often in combination with antibiotics.

    However, the use of antibiotics to treat H. pyloriinfection has

    been associated with adverse effects and frequently leads to

    resistance to antibiotic therapy.

    Several in vitro and animal studies have shown reduced via-

    bility ofH. pylori and less adhesion of the bacteria to human

    intestinal mucosal cells after treatment with variousLactobacil-

    lusstrains (120). In series of in vitro assays, Midolo et al (121)

    showed that the growth ofH. pyloriwas inhibited by lactic acid

    in a pH-independent manner. They also found that 6 strains ofL.

    acidophilus and L. casei inhibited the growth of H. pylori,

    whereas B. bifidus and L. bulgaricus did not. The inhibitory

    effect correlated with the concentrations of lactic acid produced

    by the LAB examined. In another study, Coconnier et al (122)

    reported that conditioned media fromL. acidophilus reduced the

    viability ofH. pyloriin vitro, independent of lactic acid concen-

    trations. In addition, the adhesion ofH. pylori to human mucose-

    creting HT-29 cells decreased. Several in vitro studies were con-

    ducted to ascertain whether the effects of LAB on H. pylori

    survival and function are due to lactic acid or to other antibac-terial products generated by LAB, such as bacteriocins. Of the

    several bacteriocins tested, lacticins produced byLactoc. lactis

    were shown to have the greatest anti-Helicobacteractivity when

    used against several strains ofH. pylori(123).

    Studies that indicate promising inhibitory effects of LAB onH.

    pylori survival andfunction in vitro were extended to in vivo studies

    using human patients. Armuzzi et al (124) reported that, when 120

    asymptomatic subjects who were positive for H. pyloriinfection

    received anL. caseistrain GG supplement over a 14-d period in

    addition to a standard 1-wk antibiotic therapy regimen, the eradica-

    tion ofH. pyloriwas faster than that in control subjects.

    Although promising results have been reported, the effects of

    LAB on H. pyloriinfection in humans remain ambiguous. Forexample, L. acidophilus and L. gasseri were both shown to

    decreaseH. pyloriinfection, as indicated by reduced [13C] urea

    breath testvalues(125, 126), and therapy withL. acidophiluswas

    shown to reduce gastric mucosal inflammation (125). However,

    gastric biopsies did not show eradication ofH. pylori. Similarly,

    Cats et al (127) reported that viable L. casei was required to

    inhibit the growth ofH. pyloriin vitro, but only a slight nonsig-

    nificant trend was observed toward an in vivo suppressive effect

    of anL. casei-supplemented milk drink.

    Allergic reactions

    The effects of yogurt and LAB on allergic reactions in thegastrointestinal tract have received some interest (128, 129). It

    was reported that a delay in the development ofBifidobacterium

    andLactobacillusin the gastrointestinal microflora is a general

    finding in children with allergic reactions (128). Isolauri (130)

    reported data suggesting that Lactobacillus GG can be used to

    prevent food allergies.

    Heat treatment was suggested as a way of reducing the ability

    of milk proteins to cause allergic reactions, which would make

    milk a more suitable source of protein for persons with an im-

    munologic sensitization to cow milk protein (131). However,

    Kirjavainen et al (129) used a randomized double-blind design to

    investigate in a recent study the effects of heat-inactivated and

    viableL. rhamnosusGG on infants with atopic eczema and cow

    milk allergy. Milk formula supplemented with viable but not

    heat-inactivatedL. rhamnosusGG significantlyimproved atopic

    eczema andsubjectivesymptoms of cowmilk allergy in subjects

    in comparison with the control group. These results suggest that,

    in persons with cow milk allergy, the presence of viable LAB

    may provide benefits that outweigh the possible detrimental

    effects that undenatured milk proteins may have on milk al-

    lergy. Furthermore, the immunologic response to native milk

    proteins may differ from that to heat-denatured milk proteins. A

    recent study using a rat model showed that heat-denaturated

    -lactoglobulin induced a local mucosal inflammatory response,

    whereas native -lactoglobulin induced an IgE-mediated

    systemic response (132). Heat denaturation is likely to result in

    252 ADOLFSSON ET AL

    bygues

    tonMarch2,2014

    ajcn.nutrition.org

    Downloadedfrom

    http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/
  • 8/12/2019 Am J Clin Nutr 2004 Adolfsson 245 56

    9/12

    conformational changes that expose or hide (or both) epitopes

    and lead to the activation of different subpopulations of immune

    cells and thus to different end results.

    The mechanisms of the protective effects of LAB on allergic

    reactions are not known. A proinflammatory response in the gut

    mucosathat is induced by food allergensmay impairthe function

    of the intestinal barrier. It is possible that LAB may prevent

    allergic reactions by having a protective effect on the function of

    theintestinal barrier, although themechanism of such an effectispoorly understood. A more direct link between the function of

    GALT andallergic responsesis also possible. Oneof theprimary

    mechanisms of active cellular suppression of proinflammatory

    events in the gut after antigen-specific triggering is the secretion

    of suppressive cytokines, such as transforming growth factor

    and IL-10. Transforming growth factor is produced by both

    CD4 and CD8 GALT-derived T cells and is an important

    mediator of the active suppression component of oral tolerance.

    Furthermore, IL-4 mediated isotype switching of immunoglob-

    ulin from IgM to IgE and IgE-dependent degranulation of mast

    cells has been shown to be involved in the pathogenesis of food

    allergyrelated enteropathy (133).

    Yogurts LAB are known to enhance the production of IFN-(62, 134), which acts to inhibit isotype switching to IgE. IgE-

    mediated hypersensitivity reaction, also known as type 1 allergy,

    is triggered by the cross-linking of antigens with IgE antibodies

    that are bound to Fc receptors on mast cells. It was reported that

    L. casei inhibited antigen-induced IgE production by mouse

    splenocytes (135). In addition, production of the immunosup-

    pressive cytokine IL-10 is induced by LAB (60).

    A combination of enhancing and suppressive effects is the

    most likely mechanism by which LAB may have their effects.

    However, theways in which LAB or other components of yogurt

    influence the production of these immunoregulatorycytokines in

    thegut remainto be elucidated, as do thepossiblemechanisms of

    LAB-mediated protection.

    SAFETY

    Although the safe use of nonsporing anaerobic LAB in fer-

    mented foods is widespread and has a long history, there have

    been occasional reports associating LAB with clinical infections

    (53, 136) because benignmicroorganisms have been shown to be

    infective when a patient is severely debilitated or immunosup-

    pressed (137, 138). Some of the diseases that have been associ-

    ated with LAB infection include septicemia, infective endocar-

    ditis, and dental caries.

    Very rarely, cases of lactobacillemia have been reported in

    patients with severe underlying illness, many of whom received

    a prior antibiotic therapy that may have selected-out for the

    organism (139, 140). Moreover, Husni et al (141) reviewed the

    cases of 45 patients with clinically significant lactobacillemia

    and reported that 11 of the patients were receiving immunosup-

    pressive therapy and23 hadreceived antibiotics.In none of these

    reports was a definitive link made between the consumption of

    fermented milk products and infection.

    In addition, rare cases of endocarditis have been associated

    withL. rhamnosus, a LAB indigenous to the human gastrointes-

    tinal tract (142144). However, as with lactobacillemia, no re-

    ports to date have been able to identify a connection between

    LAB from fermented milk and infection in humans. In most of

    these cases,the originof theLactobacillus is most likely thehost.

    There is also a hypothetical risk of the transfer of antimicrobial

    resistance from LAB to other microorganisms with which LAB

    might come in contact, but this has not yet been described in the

    literature.

    In the past,Lactobacilliisolated from infections were habit-

    ually dismissed as contaminants or secondary invaders. However,

    recent evidence suggests that they might function as opportunistic

    pathogens in a small number of severely immunosuppressed

    persons. Even in these patients,this is a very rare event,and it hasnot yet been reported in a large group of immunosuppressed

    persons, such as the elderly or persons with AIDS. LAB have a

    long history of safe use in foods and also in products that have

    been tested in clinical trials. However, as with any new food

    ingredient, the safety of a new strain of LAB must be clearly

    established before it is introduced into fermented dairy products.

    CONCLUDING REMARKS AND RECOMMENDATIONS

    FOR FUTURE STUDIES

    It has long been believed that the consumption of yogurt and

    other fermented milk products provides various health benefits.

    Recent studies of the possible health benefits of yogurt in gut-associated diseases substantiate some of these beliefs. Of partic-

    ular interest are the reduction by yogurt, yogurt bacteria, or

    bothin the duration of diarrheal diseases in children, the pre-

    ventive or therapeutic (or both) effects on IBD and colon cancer

    as suggested by epidemiologic evidence and animal studies, and

    the possiblebeneficial effectsin increasing the eradication rateof

    H. pylori as indicated by in vitro and preliminary human studies.

    In addition, there is ever-increasing evidence of the beneficial

    effect of yogurt containing live and active cultures on the diges-

    tion of lactose in persons with lactose intolerance.

    These findings are interesting and should encourage future

    studies to 1) substantiate or extend these findingsby using animal

    models and clinical trials;2) ascertain whether these effects areage-specific or can be observed across all age groups: eg, ascer-

    tain whether yogurt would have effects similar to those observed

    in children on attenuation of the incidence or duration of diar-

    rheal diseases in elderly people, a group that has high morbidity

    and mortality from these infections; and 3) investigate the mech-

    anisms through which yogurt exerts its effects and ascertain the

    critical components of yogurt involved in its mechanisms of

    action. Finally, in recent years, yogurt has been touted as im-

    provinggut health. In the absence of a universally accepted

    definition or any definition ofgut health,it is difficult to sub-

    stantiate these claims. Studies focused on determining the char-

    acteristics of a healthy gut would be extremely helpful in eval-

    uating the effect of yogurt on gut health.

    All 3 authors participated in the literature review and the development of

    the manuscript outline, and SNM and RMR determined the areas to be

    discussed. OA conducted the literature search and organized and wrote the

    manuscript. SNM provided corrections. RMR revised the manuscript.

    This review was prepared in response to a request from the National

    Yogurt Association for a critical and objective review, for which the authors

    received an honorarium.

    REFERENCES1. Guarner F, Schaafsma GJ. Probiotics. Int J Food Microbiol 1998;39:

    237 8.

    2. Bourlioux P, Pochart P. Nutritional and health properties of yogurt.World Rev Nutr Diet 1988;56:21758.

    YOGURT AND GUT FUNCTION 253

    bygues

    tonMarch2,2014

    ajcn.nutrition.org

    Downloadedfrom

    http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/
  • 8/12/2019 Am J Clin Nutr 2004 Adolfsson 245 56

    10/12

    3. Chandan RC, Shahani KM. Yogurt. In: Hui YH, ed. Dairy science and

    technology handbook. New York: VCH Publishers, Inc, 1993:157.

    4. Metchnikoff E. Surla floredu corpshumain. (Onthe flora ofthe human

    body. ) Manch Lit Philos Soc 1901;45:138 (in French).

    5. Perdigon G, Alvarez S, Rachid M, Aguero G, Gobbato NJ. Immune

    system stimulation by probiotics. J Dairy Sci 1995;78:1597 606.

    6. Buttriss J. Nutritionalproperties of fermented milkproducts.Int J Dairy

    Tech 1997;50:217.

    7. Reddy KP,Shahani KM,Kulkarni SM.B-complex vitamins in cultured

    and acidified yogurt. J Dairy Sci 1976;59:1915.

    8. Shahani KM, Chandan RC. Nutritional and healthful aspects of cul-

    turedand culture-containingdairy foods. J DairySci 1979;62:168594.

    9. Kneifel W, Mayer HK. Vitamin profiles of kefirs made from milks of

    different species. Int J Food Sci Technol 1991;26:423 8.

    10. Kneifel W, Kaufmann M, Fleischer A, Ulberth F. Screening of com-

    mercially available mesophilic dairy starter cultures: biochemical, sen-sory and morphological properties. J Dairy Sci 1992;75:3158 66.

    11. Crittenden RG, Martinez NR, Playne MJ. Synthesis and utilisation offolate by yoghurt starter cultures and probiotic bacteria. Int J Food

    Microbiol 2003;80:21722.

    12. Wigertz K, Svensson UK, Jagerstad M. Folate and folate binding pro-

    tein content in dairy products. J Dairy Res 1996;64:239 54.

    13. Rosado JL, Solomons NW, Allen LH. Lactose digestion from unmod-

    ified,low-fat and lactose-hydrolyzedyogurt in adult lactosemaldigest-ers. Eur J Clin Nutr 1992;46:617.

    14. Vesa TH, Marteau P, Korpela R. Lactose intolerance. J Am Coll Nutr2000;19:165S75S.

    15. KolarsJC, LevittMD, Aouji M, Savaiano DA.Yogurtan autodigest-ing source of lactose. N Engl J Med 1984;310:13.

    16. Goodenough ER, Kleyn DH. Influence of viable yogurt microflora ondigestion of lactose by the rat. J Dairy Sci 1976;59:601 6.

    17. Rasic JL, Kurmann JA. Yoghurt: scientific grounds, technology, man-ufacture and preparations. Vol 1 of Rasic JL, Kurmann JA, eds. Fer-

    mented fresh milk products and their cultures. Copenhagen: TechnicalDairy Publishing House, 1978.

    18. Loones A. Transformation of milk components during yogurt fermen-tation. In: Chandan RC, ed. Yogurt: nutritional and health properties.

    McLean, VA: National Yogurt Association, 1989:95114.

    19. Beshkova DM, Simova ED, Frengova GI, Simov ZI, Adilov EF. Pro-

    duction of amino acids by yogurt bacteria. Biotechnol Prog 1998;14:9635.

    20. Hewitt D, Bancroft HJ. Nutritional value of yogurt. J Dairy Res 1985;

    52:197207.21. Bissonnette DJ, Jeejeebhoy KN, eds. Meeting dietary nutrient require-

    ments with cows milk and milk products. Rotterdam: Balkema, 1994.

    22. Gaudichon C, Roos N, Mah S, Sick H, Bouley C, Tom D. Gastricemptying regulates the kinetics of nitrogen absorption from 15N-

    labeled milk and 15N-labeled yogurt in miniature pigs. J Nutr 1994;124:1970 7.

    23. Gaudichon C, Mah S, Roos N, et al. Exogenous and endogenousnitrogen flow rates and level of protein hydrolysis in the human jeju-

    num after [15N] milk and [15N] yogurt ingestion. Br J Nutr 1995;74:251 60.

    24. Shantha NC, Ram LN, OLeary J, Hicks CL, Decker EA. Conjugatedlinoleic acidconcentrations in dairy products as affected by processingand storage. J Food Sci 1995;60:695 8.

    25. Aneja RP, MurthiTN. Conjugatedlinoleic acid contents of Indiancurd

    and ghee. Indian J Dairy Sci 1990;43:231 8.

    26. JiangJ, Wolk A,VessbyB. Relation betweenthe intake ofmilkfat andthe occurrence of conjugated linoleic acid in human adipose tissue.Am J Clin Nutr 1999;70:217.

    27. Park Y, McGuire MK, Behr R, McGuire MA, Evans MA, Schultz TD.High-fat dairy product consumption increases 9c; 11t18:2 (rumenicacid) and total lipid concentrations of human milk. Lipids 1999;34:

    5439.

    28. BoccignoneM, Brigidi R, Sarra C. Studi effettuatisullacompo-sizionein trigliceridi ed acidi grassi liberi nello yogurt preparato dalatte vac-cino, pecorinoe, caprino. (Studies on triglyceride and free fatty acid

    composition of yogurt prepared from cow, goat, and sheep milk.) AnnFac Med Vet (Torino) 1984;28:22333 (in Italian).

    29. Whigham LD, Cook ME, Atkinson RL. Conjugated linoleic acid: im-plications for human health. Pharmacol Res 2000;42:50310.

    30. Kemp MQ,Jeffy BD,Romagnolo DF.Conjugated linoleic acidinhibitscell proliferation through a p53-dependent mechanism: effects on the

    expression of G1-restriction points in breast and colon cancer cells. J

    Nutr 2003;133:3670 7.

    31. Block G, Abrams B. Vitamin and mineral status of women of child-bearing potential. Ann N Y Acad Sci 1993;678:244 54.

    32. Ervin RB, Kennedy-Stephenson J. Mineral intakes of elderly adultsupplement and non-supplement users in the third National Health andNutrition Examination Survey. J Nutr 2002;132:34227.

    33. Allen LH.Calcium bioavailability andabsorption: a review. Am J ClinNutr 1982;35:783 808.

    34. Kaup SM,ShahaniKM, Amer MA,Peo ER.(Bioavailabilityof calcium

    in yogurt.) Milchwissenschaft 1987;42:513 6 (in German).

    35. Schaafsma GJ,Dekker PR,de Ward H. Nutritional aspectsof yogurt. 2.

    Bioavailability of essential minerals and trace elements. Neth MilkDairy J 1988;42:135 46.

    36. Bronner F, Pansu D. Nutritional aspects of calcium absorption. J Nutr1999;129:9 12.

    37. Norman AW. Intestinal calcium absorption: a vitamin D-hormonemediated adaptive response. Am J Clin Nutr 1990;51:290 300.

    38. Pointillart A, Cayron B, Gueguen L. Calcium and phosphorus utiliza-

    tion and bone mineralization in yogurt-fed pigs. Sci Alim 1986;6:1530.

    39. Bernet MF,Brassart D, NeeserJR, Servin AL.Lactobacillus acidophi-

    lusLA 1 binds to cultured human intestinal cell lines and inhibits cell

    attachment and cell invasion by enterovirulent bacteria. Gut 1994;35:4839.

    40. Alm L, Pettersson L. Survival rate of lactobacilli during digestion: anin vitro study Am J Clin Nutr 1980;33(suppl):S2543 (abstr).

    41. Robins-Browne RM, Path FF, Levine MM. The fate of ingested lacto-bacilli in the proximal small intestine.Am J Clin Nutr 1981;34:5149.

    42. Conway PL,Gorbach SL,Goldin BR.Survival oflacticacid bacteria inthe human stomach and adhesion to intestinal cells. J Dairy Sci 1987;

    70:112.

    43. Pedrosa MC, Golner BB, Goldin BR, Barakat S, Dallal GE, RussellRM. Survival of yogurt-containing organisms andLactobacillus gas-

    seri(ADH) and their effect on bacterial enzyme activity in the gastro-

    intestinal tract of healthy and hypochlorhydric elderly subjects. Am JClin Nutr 1995;61:3539.

    44. Clark PA, Martin JH. Selection of bifidobacteria for use as dietaryadjuvants in cultured dairy foods: III. Tolerance to stimulated bile

    concentrations of human small intestines. Cult Dairy Prod J 1994;29:18 21.

    45. Duez H, Pelletier H, Cools S, et al. A colony immunoblotting method

    forquantitativedetectionof aBifidobacterium animalis probiotic strainin human faeces. J Appl Microbiol 2000;88:1019 27.

    46. Guerin-Danan C, Chabanet C, Pedone C, et al. Milk fermented with

    yogurt cultures and Lactobacillus casei compared with yogurt andgelled milk: influence on intestinal microflora in healthy infants. Am J

    Clin Nutr 1998;67:1117.

    47. Bouhnik Y, Pochart P, Marteau P, Arlet G, Goderel I, Rambaud JC.

    Fecal recovery in humans of viable Bifidobacterium sp ingested infermented milk. Gastroenterology 1992;102:875 8.

    48. Plant L, Conway P. Association of Lactobacillus spp. with Peyerspatches in mice. Clin Diagn Lab Immunol 2001;8:320 4.

    49. Bernet MF, Brassart D, Neeser JR, Servin AL. Adhesion of humanbifidobacterial strains to cultured human intestinal epithelial cells and

    inhibition of enteropathogen-cell interactions. Appl Env Microbiol1993;59:4121 8.

    50. LereboursE, N'Djitoyap Ndam C, Lavoine A, Hellot MF, Antoine JM,

    Colin R. Yogurt and fermented-then-pasteurized milk: effects of short-term and long-term ingestion on lactoseabsorption and mucosal lactaseactivity in lactase-deficient subjects. Am J Clin Nutr 1989;49:8237.

    51. Kankaanpaa P, Salminen SJ, Isolauri E, Lee YK. The influence ofpolyunsaturated fatty acids on probiotic growth and adhesion. FEMS

    Microbiol Lett 2001;194:149 53.

    52. KankaanpaaP, Yang B, Kallio H, Isolauri E, Salminen S. Effects ofpolyunsaturated fattyacids in growth mediumon lipidcomposition andon physicochemical surface properties ofLactobacilli. Appl Env Mi-

    crobiol 2004;70:129 36.

    53. Aguirre M, Collins MD. Lactic acid bacteria and human clinicalinfec-

    tion. J Appl Bacteriol 1993;75:95107.

    54. Brandtzaeg P, Baekkevold ES, Farstad IN, et al. Regional specializa-tion in the mucosal immune system: what happens in the microcom-partments? Immunol Today 1999;20:14151.

    55. Macpherson AJ, Gatto D, Sainsbury E, Harriman GR, Hengartner H,

    254 ADOLFSSON ET AL

    bygues

    tonMarch2,2014

    ajcn.nutrition.org

    Downloadedfrom

    http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/
  • 8/12/2019 Am J Clin Nutr 2004 Adolfsson 245 56

    11/12

    Zinkernagel RM. A primitive T cell-independent mechanism of intes-

    tinal mucosal IgA responses to commensal bacteria. Science2000;288:

    2222 6.

    56. Puri P, Rattan A, Bijlani RL, Mahapatra SC, Nath I. Splenic and intes-tinal lymphocyte proliferation response in mice fed milk or yogurt and

    challenged with Salmonella typhimurium. Int J Food Sci Nutr 1996;

    47:391 8.

    57. Link-Amster H, Rochat F, Saudan KY, Mignot O, Aeschlimann JM.

    Modulation of a specific humoral immune response and changes in

    intestinal flora mediated through fermented milk intakes. FEMS Im-munol Med Microbiol 1994;10:55 64.

    58. Perdigon G,de Macias ME, Alvarez S,OliverG, de RuizHolgadoAA.Systemic augmentation of the immune response in mice by feeding

    fermented milks withLactobacillus caseiandLactobacillus acidophi-

    lus. Immunology 1988;63:1723.

    59. Mosmann TR, Cherwinski H, Bond MW, Giedlin MA, Coffman RL.

    Two types of murine helper T cell clone. I. Definition according toprofiles of lymphokine activities and secreted proteins. J Immunol

    1986;136:2348 57.

    60. Miettinen M, Vuopio-Varkila J, VarkilaK. Productionof human tumor

    necrosis factor-alpha, interleukin-6 and interleukin-10 is induced by

    lactic acid bacteria. Infect Immun 1996;64:54035.

    61. Solis-Pereyra B, Aattouri N, Lemonnier D. Role of food in the stim-

    ulation of cytokine production. Am J Clin Nutr 1997;66(suppl):

    521S5S.

    62. Halpern GM, Vruwink KG, van de Water J, Keen CL, Gershwin ME.Influence of long-term yogurt consumption in young adults. Int J Im-munother 1991;7:20510.

    63. Borruel N, Carol M, Casellas F, et al. Increased mucosal tumour ne-

    crosisfactor alpha productionin Crohns diseasecan be downregulatedex vivo by probiotic bacteria. Gut 2002;51:659 64.

    64. De Simone C, Bianchi Salvadori B, Negri M, Ferrazzi M, BaldinelliL,Vesely R. The adjuvant effect of yogurt on production of gamma-

    interferon by Con A stimulated human peripheral blood lymphocytes.

    Nutr Rep Int 1986;33:419 33.

    65. Beharka AA, Paiva S, Leka LS, Ribaya-Mercado JD, Russell RM,

    Nibkin Meydani S. Effect of age on the gastrointestinal-associatedmucosal immune response of humans. J Gerontol A Biol Sci Med Sci

    2001;56:B218 23.

    66. Podolsky DK. Inflammatory bowel disease. N Engl J Med 2002;347:41729.

    67. Ajuebor MN,SwainMG.Roleof chemokines andchemokine receptorsin the gastrointestinal tract. Immunology 2002;105:137 43.

    68. Wallace TD, BradleyS, BuckleyND, Green-Johnson JM. Interactions

    of lactic acid bacteria with human intestinal epithelial cells: effects oncytokine production. Food Prot 2003;66:466 72.

    69. Strandhagen E, Lia A, Lindstrand S, et al. Fermented milk (ropy milk)replacing regular milkreduces glycemic responce and gastric emptying

    in healthy subjects. Scand J Nutr 1994;38:11721.

    70. Nakamura T, Nishida S, Mizutani M, Iino H. Effects of yogurt supple-

    mented with brewers yeast cell wall on constipation and intestinal

    microflora in rats. J Nutr Sci Vitaminol (Tokyo) 2001;47:36772.

    71. Meance S, Cayuela C, Turchet P, Raimondi A, Lucas C, Antoine JM.

    A fermented milk with aBifidobacteriumprobiotic strain DN-173 010

    shortened oro-fecal gut transit time in elderly. Microb Ecol Health Dis2001;13:21722.

    72. Marteau P, Cuillerier E, Meance S, et al. Bifidobacterium animalis

    strain DN-173 010 shortens the colonic transit time in healthy women:a double-blind, randomized,controlledstudy. Aliment Pharmacol Ther

    2002;16:58793.

    73. Rorick MH, Scrimshaw NS. Comparative tolerance of elderly from

    differing ethnic backgrounds to lactose-containing and lactose-freedairy drinks: a double-blind study. J Gerontol 1979;34:191 6.

    74. SahiT. Genetics andepidemiologyof adult-type hypolactasia. Scand J

    Gastroenterol 1994;202:720.

    75. Savaiano DA, AbouElAnouar A, Smith DE, Levitt MD. Lactose mal-

    absorption from yogurt, pasteurized yogurt, sweet acidophilus milk,and cultured milk in lactase-deficient individuals. Am J Clin Nutr

    1984;40:1219 23.

    76. Pochart P, Dewit O, Desjeux JF, Bourlioux P. Viable starter culture,

    -galactosidase activity, and lactose in duodenum after yogurt inges-

    tion in lactase-deficient humans. Am J Clin Nutr 1989;49:828 31.

    77. HickeyMW, Hillier AJ,JagoGR. Transportand metabolism oflactose,

    glucose, and galactose in homofermentative lactobacilli. Appl Environ

    Microbiol 1986;51:82531.

    78. Foucaud C, Poolman B. Lactose transport system of Streptococcusthermophilus. J Biol Chem 1992;267:2208794.

    79. Thoreux K, Balas D, Bouley C, Senegas-Balas F. Diet supplemented

    with yoghurt or milk fermented by Lactobacillus casei DN-114 001

    stimulates growth and brush-border enzyme activities in mouse smallintestine. Digestion 1998;59:349 59.

    80. Martini MC,Bollweg GL,LevittMD, Savaiano DA.Lactose digestion

    by yogurt-galactosidase: influence of pH and microbial cell integrity.Am J Clin Nutr 1987;45:432 6.

    81. Glass RI, Lew JF, Gangarosa RE, LeBaron CW, Ho MS. Estimates of

    morbidity and mortality rates for diarrheal diseases in American chil-dren. J Pediatr 1991;118:S2733.

    82. VanNeilCW, FeudtnerC, GarrisonMM, ChristakisDA.Lactobacillustherapy for acute infectious diarrhea in children: a meta-analysis. Pe-

    diatrics 2002;109:678 84.

    83. Saavedra JM,Bauman NA,OungI, PermanJA, YolkenRH. Feedingof

    Bifidobacterium bifidum andStreptococcus thermophilusto infants ina hospital forprevention of diarrhoeaand shedding of rotavirus. Lancet

    1994;344:1046 9.

    84. Gorbach SL, Chang TW, Goldin B. Successful treatment of relapsing

    Clostridium difficile colitis with Lactobacillus GG. Lancet 1987;2:1519(letter).

    85. Biller JA, Katz AJ, Flores AF, Buie TM, Gorbach SL. Treatment of

    recurrentClostridium difficilecolitis withLactobacillusGG. J PediatrGastroenterol Nutr 1995;21:224 6.

    86. Shornikova AV, Isolauri E, Burkanova L, Lukovnikova S, Vesikari T.

    A trial in the Karelian Republic of oral rehydration and LactobacillusGG for treatment of acute diarrhoea. Acta Paediatr 1997;86:460 5.

    87. Clements ML, Levine MM, Ristaino PA, Daya VE, Huges TP. Exog-enous lactobacilli fedto mantheirfate andability toprevent diarrheal

    disease. Prog Food Nutr Sci 1983;7:29 37.

    88. Bartlett JG. Antibiotic-associated diarrhea. Clin Infect Dis 1992;15:

    573 81.

    89. Van der Waaij D. The ecology of the human intestine and its conse-

    quences for overgrowth by pathogens such as Clostridium difficile.Annu Rev Microbiol 1989;43:69 87.

    90. DSouzaAL, Rajkumar C,CookeJ, BulpittCJ.Probioticsin preventionof antibiotic associated diarrhoea: meta-analysis. BMJ 2002;324:1 6.

    91. Gotz V, Romankiewicz JA, Moss J, Murray HW. Prophylaxis against

    ampicillin associated diarrhoea with Lactobacilluspreparation. Am JHosp Pharm 1979;36:754 7.

    92. Tankanow RM, Ross MB, Ertel IJ, Dickinson DG, McCormick LS,

    Garfinkel JF. A double blind, placebo-controlled study of the efficacyof Lactinex in the prophylaxis of amoxicillin-induced diarrhea. DICP

    1990;24:382 4.

    93. Orrhage K, Brismar B, Nord CE. Effects of supplements ofBifidobac-terium longumandLactobacillus acidophiluson intestinal microbiotaduring administration of clindamycin. Microb Ecol Health Dis 1994;

    7:1725.

    94. Vanderhoof JA, Whitney DB, Antonson DL, Hanner TL, Lupo JV,

    Young RJ.Lactobacillus GG in the prevention of antibiotic-associated

    diarrhoea in children. J Pediatr 1999;135:356 68.

    95. National Cancer Institute. SEER Cancer Incidence Public-Use Data-

    base, 1973-1996, August 1998 Submission. Bethesda, MD: US De-partment of Health and Human Services, Public Health Service, 1999.

    96. Peters RK, Pike MC, Garabrant D, Mack TM. Diet and colon cancerin Los Angeles County, California. Cancer Causes Control 1992;3:

    45773.

    97. Reddy BS,RivensonA. Inhibitory effectofBifidobacterium longum on

    colon, mammary, and liver carcinogenesis induced by 2-amino-3-methylimidazo[4,5-f]quinoline, a food mutagen. Cancer Res 1993;53:3914 8.

    98. Ayebo AD, Shahani KM, Dam R. Antitumor component(s) of yogurt:fractionation. J Dairy Sci 1981;64:2318 23.

    99. Shackelford LA, Rao DR, Chawan CB, Pulusani SR. Effect of feedingfermentedmilk onthe incidence of chemicallyinduced colon tumorsin

    rats. Nutr Cancer 1983;5:159 64.

    100. Wollowski I, Ji S, Bakalinsky AT, Neudecker C, Pool-Zobel BL. Bac-teria used for the production of yogurt inactivate carcinogens and pre-

    vent DNA damage in the colon of rats. J Nutr 1999;129:77 82.

    101. Bolognani F, Rumney CJ, Pool-Zobel BL, Rowland IR. Effect of

    YOGURT AND GUT FUNCTION 255

    bygues

    tonMarch2,2014

    ajcn.nutrition.org

    Downloadedfrom

    http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/http://ajcn.nutrition.org/
  • 8/12/2019 Am J Clin Nutr 2004 Adolfsson 245 56

    12/12

    lactobacilli,bifidobacteria and inulinon the formationof aberrant crypt

    foci in rats. Eur J Nutr 2001;40:293300.102. Ganjam LS, Thornton WH, Marshall RT, MacDonald RS. Antiprolif-

    erative effects of yogurt fractions obtained by membrane dialysis oncultured mammalian intestinal cells. J Dairy Sci 1997;80:2325 9.

    103. Kim DH, Jin YH. Intestinal bacterial beta-glucuronidase activity of

    patients with colon cancer. Arch Pharm Res 2001;24:564 7.104. Reddy BS, Engle A, Simi B, Goldman M. Effect of dietary fiber on

    colonic bacterial enzymes and bile acids in relation to colon cancer.Gastroenterology 1992;102:1475 82.

    105. GoldinBR, Gorbach SL. Alterationsof the intestinal microfloraby diet,

    oralantibiotics,andLactobacillus: decreased productionof freeaminesfrom aromatic nitro compounds, azo dyes, and glucuronides. J NatlCancer Inst 1984;73:689 95.

    106. GoldinBR,Gorbach SL.The effectof milk andlactobacillus feeding onhuman intestinal bacterial enzyme activity. Am J Clin Nutr 1984;39:

    756 61.107. Rafter JJ. The role of lactic acid bacteria in colon cancer prevention.

    Scand J Gastroenterol 1995;30:497502.

    108. Sartor RB. Pathogenesis and immune mechanisms of chronic inflam-matory bowel diseases. Am J Gastroenterol 1997;92:5S11S.

    109. Kwon HJ,Cote TR,CuffeMS, KramerJM, Braun MM.Case reports ofheart failure after therapy with a tumor necrosis factor antagonist. AnnIntern Med 2003;138:80711.

    110. FabiaR, ArRajabA, JohanssonML,et al.Impairmentof bacterialflorain human ulcerativecolitis andexperimentalcolitis in therat. Digestion

    1993;54:248 55.111. McCormick DA, Horton LW, Mee AS. Mucin depletion in inflamma-

    tory bowel disease. J Clin Pathol 1990;43:143 6.

    112. van Wijngaarden P, Meijssen MA. Tuberculous pleurisy: an unusualcomplication during treatment of Crohn disease with azathioprine.Scand J Gastroenterol 2001;37:1004 7.

    113. Madsen KL, Doyle JS, Jewell LD, Tavernini MM, Fedorak RN.Lac-tobacillus species prevents colitis in interleukin 10 gene-deficient

    mice. Gastroenterology 1999;116:110714.114. Steidler L, Hans W, Schotte L, et al. Treatment of murine colitis by

    Lactococcus lactissecreting interleukin-10. Science 2000;289:13525.

    115. Malin M, Suomalainen H, Saxelin M, Isolauri E. Promotion of IgAimmune response in patients with Crohns disease by oral bacterio-therapy withLactobacillusGG. Ann Nutr Metab 1996;40:137 45.

    116. Kaila M, Isolauri E, Soppi E, Virtanen E, Laine S, Arvilommi H.Enhancement of the circulating antibody secreting cell response in

    human diarrhea by a humanLactobacillus strain. PediatrRes 1992;32:141 4.

    117. Marshall BJ. Unidentified curved bacillus on gastric epithelium in

    active chronic gastritis. Lancet 1983;1:12735.118. Labigne A, de Reuse H. Determinants of Helicobacter pylori patho-

    genicity. Infect Agents Dis 1996;5:191202.119. Duggan A.Helicobacter pylori: when is treatment now indicated? Int

    Med J 2002;32:4659.

    120. Aiba Y, Suzuki N, Kabir AMA, Takagi A, Koga Y. Lactic acid-mediated supression ofHelicobacter pyloriby the oral administrationofLactobacillus salivariusas a probiotic in a gnobiotic murine model.

    Am J Gastroenterol 1998;93:2097101.121. MidoloPD, Lambert JR,HullR, LuoF, Grayson ML.In vitro inhibition

    ofHelicobacter pylori NCTC 11637 by organic acids and lactic acidbacteria. J Appl Bacteriol 1995;79:4759.

    122. Coconnier M, Lievin V, Hemery E, Servin AL. Antagonistic activity

    against Helicobacter infection in vitro and in vivo by the humanLact obaci llus acido philu sstrain LB. Appl Env Microbiol 1998;64:

    4573 80.123. Kim TS, Hur JW, YuMA, et al. Antagonism ofHelicobacter pylori by

    bacteriocins of lactic acid bacteria. J Food Prot 2003;66:3 12.

    124. Armuzzi A, Cremonini F, Ojetti V, et al. Effect ofLactobacillusGG

    su