Shaping microbial exposures and the immune system in childhood: Can sandboxes be probiotic?

By Prof. Seppo Salminen, University of Turku, Finland

Gut microbiota researchers have established that microbial exposures in early life can be influential on health later in life. Children who develop asthma in early childhood, for example, have an altered gut microbiota linked with exposure to less diverse microorganisms in their first year. The ‘biodiversity hypothesis’ has been advanced recently, suggesting that western lifestyles and low biodiversity in urban environments reduce contact with microbes both via food and via the natural environment, presenting fewer opportunities for children to be exposed to a diversity of microbes in their earliest years and increasing the risk of non-communicable diseases. If this is the case, the environments of daycare and kindergarten facilities come under scrutiny as a source of microbial exposures at a crucial time of life. So is it beneficial to intervene in children’s environments to ensure more diverse microbial exposures? Can we enhance gut microbial diversity and richness in children through environmental interventions?

A new study provided proof that shaping children’s microbial exposures may be possible. The study was the first of its kind – a placebo-controlled, double-blinded study on the effect of environmental exposures on gut microbiota diversity and immune parameters in young children. The study used playground sandboxes at daycare facilities as sources of environmental microbial diversity and explored whether these could have effects on the children.

Six day-care centers in southern Finland were enrolled in the study, with two randomly assigned to intervention and four to placebo. Identical-looking playground sandboxes were used. Intervention sandboxes were filled with sand of glacial origin enriched with a known biodiversity powder (including commercial soil, deciduous leaf litter, peat, and Sphagnum moss; described in detail by Hui et al., 2019 ; Grönroos et al, 2018). In control centers the sand was regular sandbox sand and placebo peat material. Altogether, 26 children ages 3-5 participated in supervised play for 20 minutes in the morning and afternoon for two weeks. Researchers measured the composition of gut and skin microbiota, as well as blood immune markers.

The results demonstrated that exposure to diverse environmental microbiota enhanced both the bacterial richness and diversity of the skin bacterial community. The microbiome of the skin changed only in those children who had played in a sandbox enriched with natural materials. The authors also found that the daily exposure to higher microbial biodiversity resulted in positive differences in immune response. For instance, the authors reported shifts in skin microbiota associated with IL-10 and T cell frequencies. This provides the first evidence from a placebo-controlled, double-blinded study in young children showing the differential effects on microbiota and immunity of daily exposure to defined microbial biodiversity.

An interesting follow-up could be using sandboxes to deliver probiotics with a proven health impact to children. Since the sandbox microbes were shown to influence children’s immune systems, could researchers go one step further and modulate children’s microbiota in a targeted manner? A probiotic must be defined, shown to have a health benefit and administered in an efficacious dose. In the case of sandboxes, the health benefit would need to be demonstrated for a certain level or duration of environmental exposure.

Playgrounds and sandboxes require materials that tolerate heavy wear and tear and are safe at the same time. Such materials need to be kept free of unnecessary contamination as sandboxes, for example, can also be good reservoirs of some detrimental bacteria. Therefore, it could be important to have defined natural materials for a positive impact on health. In the future, we may see many creative approaches to ensuring children receive appropriate health-supporting microbial exposures early in life. However, creating probiotic approaches requires identification of specific microbes in the biodiversity powder.

Why researchers need to understand more about the small intestinal microbiome

By Prof. Eamonn M. M. Quigley, MD, The Methodist Hospital and Weill Cornell School of Medicine, and Prof. Purna Kashyap, MD, Mayo Clinic

The phrase “gut microbiota” properly refers to the microorganisms living throughout the entire digestive tract, including the mouth and the upper digestive tract, through the length of the small intestine as well as the large intestine. Yet the vast majority of scientific studies on the gut microbiota make conclusions based only on stool samples, meaning that the contributions to health and disease of microorganisms from most of the digestive tract are largely unexplored.

Researchers have established that the microorganisms throughout different parts of the digestive tract vary greatly. In particular, the microorganisms living in the small intestine are fewer in number than those in the colon. They are less diverse, and they change more over time because of their dynamic environment (fluctuations in oxygen, digestive secretions, dietary substrates, among other influences).

The dynamic composition and biologic functions of the small intestinal microbiome in health and disease are mostly unknown. Research has been hampered by the difficulty in obtaining samples from this area of the digestive tract and, in particular, its more distal reaches. Participants in a 2022 ISAPP discussion group argued, however, there are some good reasons to dedicate more effort to investigation of the small intestinal microbiome:

  • The small intestine has critical homeostatic functions in relation to nutrient digestion and absorption, immune engagement and interactions with the enteric and central nervous systems, as well as the neuroendocrine system. Each of these could be influenced by microbiota-host interactions. Important locations for these interactions include the gut barrier and mucosa- or gut-associated lymphoid tissue. The nature of microbiota-host interactions in these particular areas needs to be better understood, as they could have implications for systemic host health.
  • Diet plays a critical role in symptom generation in many gastrointestinal disorders; it is important to better understand diet-microbe interactions in the gut lumen to determine how the small intestinal microbiome may be contributing to diet-triggered symptoms.
  • A disordered small intestinal microbiome is commonly implicated in the pathogenesis of various gastrointestinal and non-gastrointestinal symptoms, from irritable bowel syndrome to Alzheimer’s disease, through the much-disputed concept of small intestinal bacterial overgrowth (SIBO). A precise definition of the normal small intestinal microbiome is a prerequisite to the accurate diagnosis of SIBO and linking it with various disease states.

How can we gain more information on the small intestinal microbiome? Our group tackled the limitations of current definitions and diagnostic methods, noting that this field may be advanced in the near future by new technologies for real-time sampling of intestinal gases and contents. The group discussed optimal methods for the sampling of small intestinal microbes and their metabolic products—noting that a full range of ‘omics technologies applied in well-defined populations could lead to further insights. In the meantime, the gastroenterologists in our group advised restraint in the diagnosis of SIBO and the need to exert caution in identifying it as the cause of symptoms. Clinical progress in this area is best achieved through the application of modern molecular methods to the study of human small intestinal microorganisms.

Are probiotics effective in improving symptoms of constipation?

By Eirini Dimidi, PhD, Lecturer at King’s College London

Constipation is a common disorder that affects approximately 8% of the general population and is characterised by symptoms of infrequent or difficult bowel movements (1). People who suffer with constipation often report that it negatively affects their quality of life and the majority use some sort of treatment, such as fibre supplements and laxatives, to alleviate their symptoms (2). However, approximately half of those report they are not completely satisfied with the treatment options currently available to them, mainly due to lack of effectiveness in improving their symptoms (2).

Could probiotics offer an effective alternative way to treat constipation symptoms?

Our team at the Department of Nutritional Sciences at King’s College London has investigated the potential benefits of probiotic supplements in chronic constipation. We have extensively reviewed the available evidence on their mechanisms of action in affecting gut motility and their effectiveness in improving symptoms, and we have also conducted a randomised controlled trial of a novel probiotic in 75 people with chronic constipation (3-5).

USE OF PROBIOTICS

Before looking at the evidence on the effectiveness of probiotics in constipation, it is easy to see that some people with constipation already choose to try probiotics for their gut health. A national UK survey of over 2,500 members of the public, which included people with and without constipation, showed that people with constipation have a 5.2 higher chance of currently using probiotics for gut health, compared to people who don’t suffer from it (3).

However, the majority of doctors do not recommend probiotics for the relief of constipation symptoms, nor do they believe there is enough evidence to support their use in this condition (3).

So, what is the current evidence on probiotics and constipation?

MECHANISMS OF ACTION OF PROBIOTICS

Probiotics may impact gut motility and constipation through several mechanisms of action. Depending on the strain, they may affect the number and composition of gut microbes, as well as the compounds they release. The gut microbiota and their released compounds can then interact with our immune and nervous system, with the latter being the primary regulator of gut motility, ultimately improving constipation symptoms. Therefore, there is a rationale to support a potential improvement in constipation. But is this supported by evidence from clinical trials?

EFFECTIVENESS OF PROBIOTICS

A systematic review of the literature showed Bifidobacterium lactis strains appear to improve several symptoms of constipation, such as infrequent bowel movements and hard stools (4). At the same time, other probiotic species did not improve any symptoms. This is an important finding as it highlights that not all probiotics have the same effects in constipation, and that only certain probiotics may improve constipation. Therefore, people with constipation may only benefit from specific probiotic products – but which products would those be? Since the systematic review above showed that several B. lactis strains were effective, does this means that people with constipation may benefit from any B. lactis-containing product?

Unfortunately, it is a bit more complicated. Since the publication of the aforementioned review, new studies have been published showing that, while some probiotic products with B. lactis are effective, various other B. lactis probiotics do not impact constipation (5-6). This may be explained by strain-specific effects, but also other methodological differences among studies (e.g. probiotic dose).

TAKE HOME MESSAGE

Can we recommend probiotics for the management of constipation? At the moment, there is some low quality evidence to support the use of certain Bifidobacterium lactis strains to help manage symptoms of constipation. Further high-quality studies are needed to clarify which specific probiotic strains may be effective. However, given that there is some evidence in this area (albeit limited), along with the fact probiotics are safe for the general population to consume (unless clinically contraindicated), people with constipation could try a probiotic product of their choice for four weeks, should they wish to, bearing in mind the uncertainty in the evidence so far. But scientists continue to work to answer this question because the evidence is promising enough to warrant continued study of probiotics for constipation.

 

    1. Palsson, Gastroenterol 2020;158:1262-1273
    2. Johanson & Kralstein, Aliment Pharmacol Ther 2007;25(5):599-608
    3. Dimidi et al, Nutrition 2019;61:157-163
    4. Dimidi et al, Am J Clin Nutr 2014;100(4):1075-84
    5. Dimidi et al, Aliment Pharmacol Ther 2019;49:251-264
    6. Wang et al, Beneficial Microbes 2021;12:31-42

 

 

Can diet shape the effects of probiotics or prebiotics?

By Prof. Maria Marco PhD, University of California – Davis and Prof. Kevin Whelan PhD, King’s College London

If you take any probiotic or prebiotic product off the shelf and give it to several different people to consume, you might find that each person experiences a different effect. One person may notice a dramatic reduction in gastrointestinal symptoms, for example, while another person may experience no benefit. On one level this is not surprising, since every person is unique. But as scientists, we are interested in finding out exactly what makes a person respond to a given probiotic or prebiotic to help healthcare providers know which products to recommend to which people.

Among factors that might impact someone’s response to a probiotic or prebiotic – such as baseline microbiota, medications, and host genetics – diet emerges as a top candidate. Ample evidence has emerged over the past ten years that diet has direct and important effects on the structure and function of the gut microbiome. Overall the human gut microbiome is shaped by habitual diet (that is, the types of foods consumed habitually over time), but the microbes can also can fluctuate in response to short-term dietary shifts. Different dietary patterns are associated with distinct gut microbiome capabilities. Since probiotics and prebiotics may then interact with gut microbes when consumed, it is plausible that probiotic activity and prebiotic-mediated gut microbiome modulation may be impacted by host diet.

A discussion group convened at ISAPP’s 2022 annual meeting brought together experts from academia and industry to address whether there is evidence to support the impact of diet on the health effects of probiotics and prebiotics. To answer this question, we looked at how many probiotic or prebiotic studies included data on subjects’ diets.

  • Prebiotics: Our review of the literature showed that only a handful of prebiotic intervention studies actively measured background diet as a potential confounder of the effect of the prebiotic. One such study (Healey, et al., 2018) classified individuals based on habitual fiber intake, and in doing so found that the gut microbiome of individuals consuming high fiber diets exhibited more changes to microbiome composition than individuals with low fiber intake. While both groups consuming prebiotics showed enrichment of Bifidobacterium, those with high fiber intake uniquely were enriched in numerous other taxa, including butyrate-producing groups of microbes. Prebiotics also resulted in improved feelings of satiety, but only among the high fiber diet consumers.
  • Probiotics: We found no evidence of published human RCTs on probiotics that investigated diet as a possible confounding factor. This is a significant gap, since we know from other studies that host diet affects the metabolic and functional activity of probiotic lactobacilli in the digestive tract. Moreover, the food matrix for the probiotic may further shape its effects, via the way in which the probiotic is released in situ.

Our expert group agreed that diet should be included in the development of new human studies on probiotics and prebiotics, as well as other ‘-biotics’ and fermented foods. These data are urgently needed because although diet may be a main factor affecting outcomes of clinical trials for such products, it is currently a “hidden” factor.

We acknowledge there will be challenges in taking diet into account in future trials. For one, should researchers merely record subjects’ habitual dietary intake, or should they provide a prescribed diet for the duration of the trial? The dietary intervention (nutrient, food, or whole diet) must also be clearly defined, and researchers should carefully consider how to measure diet (e.g. using prospective or retrospective methods). In the nutrition field, it is well known that there are challenges and limitations in the ways dietary intake is recorded as well as the selection of dietary exclusion criteria. Hence, it is crucial that dietitians knowledgeable in dietary assessment and microbiome research contribute to the design of such trials.

If more probiotic and prebiotic trials begin to include measures of diet, perhaps we will get closer to understanding the precise factors that shape someone’s response to these products, ultimately allowing people to have more confidence that the product they consume will give them the benefits they expect.

Human milk oligosaccharides as prebiotics to be discussed in upcoming ISAPP webinar

Human milk oligosaccharides (HMOs), non-digestible carbohydrates found in breast milk, have beneficial effects on infant health by acting as substrates for immune-modulating bacteria in the intestinal tract. The past several years have brought an increase in our understanding of how HMOs confer health benefits, prompting the inclusion of synthetic HMOs in some infant formula products.

These topics will be covered in an upcoming webinar, “Human milk oligosaccharides: Prebiotics in a class of their own?”, with a presentation by Ardythe Morrow PhD, Professor of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine. The webinar will provide an overview of what HMOs are, how they are breaking new ground with the types of health benefits they can provide to infants and the recent technological innovations that will facilitate their translation into new infant formulas.

Dr. Karen Scott, Rowett Institute, University of Aberdeen, and Dr. Margriet Schoterman, FrieslandCampina, will host the webinar. All are welcome to join this webinar, scheduled for Wednesday, Oct 19th, 2022, from 10-11 AM Eastern Daylight Time.

Registration is now closed. Please watch the recording of this webinar below.

Can Probiotics Cause Harm? The example of pregnancy

By Prof. Dan Merenstein MD, Georgetown University School of Medicine, Washington DC, USA and Dr. Maria Carmen Collado, Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Valencia, Spain

Limiting excessive weight gain and controlling blood pressure during pregnancy are important to prevent pre-eclampsia and other complications of pregnancy. Researchers have examined if there is a role for probiotics in maintaining a healthy pregnancy. A recent Cochrane review, which evaluated evidence on probiotics for preventing gestational diabetes (GDM), concluded, “Low-certainty evidence from six trials has not clearly identified the effect of probiotics on the risk of GDM. However, high-certainty evidence suggests there is an increased risk of pre-eclampsia with probiotic administration.” This was an unexpected conclusion, which raised concerns about probiotic safety. A close look at the basis for this statement is warranted to determine if certain strains of probiotics are contraindicated for pregnant women.

Most people familiar with probiotic science understand that giving anyone live bacteria carries some risk. The definition of probiotics is live microorganisms that, when administered in adequate amounts, confer a health benefit on the host. It is not live microorganisms that, when administered in adequate amounts, confer a health benefit on the host that outweighs potential adverse events. But clinicians understand that risk versus benefit must be considered for all interventions.

Many interventions associated with significant positive outcomes also are associated with some adverse events, some quite significant. For example, a recent United States Preventive Services Task Force report found that beta carotene, with or without vitamin A, was significantly associated with an increased risk of lung cancer and cardiovascular disease mortality. Aspirin kills thousands of people each year, with many more hospitalized with significant bleeds. While for an exercise doctors recommend all the time, biking, the CDC reports nearly 1,000 bicyclists die and over 130,000 are injured in crashes every year in the US.

Studies that led to the Cochrane conclusion

But let’s get back to trying to understand what made the Cochrane review come out with this warning about probiotics and pre-eclampsia. Turns out the conclusion was based on four randomized clinical trials which reported pre-eclampsia as an adverse event. All four studies were well done with low risk of bias per the Cochrane report.

Here is a summary of the four studies that collected preeclampsia data, included in the Cochrane review:

Callaway et al.(2019) studied  a mixture of Lactobacillus rhamnosus (LGG) and Bifidobacterium animalis subspecies lactis BB-12 for the prevention of gestational diabetes The reported pre-eclampsia in the probiotic group was 19 (9.2%) participants compared to 10 (4.9%) in the placebo group, p-value=0.09. This was in an obese cohort, with an average BMI of both groups near 32 (kg/m2).

Lindsay et al. (2014) evaluated the effect of Lactobacillus salivarius UCC118 on maternal fasting glucose. They reported preeclampsia in 3/62 in the probiotic group versus 2/74 in the placebo group (p-value >0.366).  Again, this was in an obese cohort with early pregnancy BMIs in the probiotic group, averaging 32.9 versus 34.1 in the placebo group.

Pellonpera et al. (2019) conducted a 4-arm study to determine if fish oil and or Lactobacillus rhamnosus HN001 and Bifidobacterium animalis ssp. lactis 420 could prevent gestational diabetes. In total there were 10 cases of pre-eclampsia among the four groups as shown below, (each group had about 95 total participants) and no significant differences between them, p value=0.80.

  • Fish oil + placebo, 1 of 95 participants (1.1%)
  • Probiotics + placebo, 4 of 96 participants (4.2%)
  • Fish oil + probiotics 3, of 96 participants (3.1%)
  • Placebo + placebo, 2 of 93 participants (2.2%)

Okesene-Gafa et al. (2019) published in the American Journal of Obstetrics and Gynecology in 2019 looking at culturally tailored dietary intervention and or daily probiotic capsules containing lactobacillus rhamnosus GG and Bifidobacterium lactis BB12 impact pregnancy weight-gain and birthweight. (This was also an obese cohort with an average BMI of 38.8.) They found pregnancy induced hypertension in the probiotic group in 4/96 (4.2%) of women versus 2/93 (2.2%) in the placebo group (p value=0.31).

Is there a rationale for the preeclampsia warning?

The increased rate of preeclampsia in probiotic groups was only with studies using obese subjects. Importantly, obesity has been associated with a higher risk of preeclampsia (see here and here). A recent meta-analysis, which included 86 studies representing 20,328,777 pregnant women, showed that higher BMI is associated with adverse pregnancy outcomes, among them, gestational diabetes and preeclampsia. Furthermore, the adjusted risk of preeclampsia is estimated to be double for overweight mothers and almost triple for obese mothers, compared to normal weight mothers.

It has been reported that pro-inflammatory signals (TNF-alpha, IL6) produced in adipose tissue of obese individuals induces a proinflammatory state characterized by insulin resistance and altered endothelial function. The gut microbiota is also disrupted in these individuals, consistent with observations that report an altered gut microbiota composition in obese versus lean individuals (see here, and the effects on offspring here and here). This suggests that obese mothers may have an increased risk of adverse events, but still the evidence supports that the addition of certain strains of probiotics may exacerbate this risk. Furthermore, it is relevant to mention the accumulating data showing that during gestation in parallel to the physiological, immune and metabolic adaptations, gut microbiota changes over the pregnancy (see here, here, here and here) although little is known on the impact of pre-gestational BMI on gut microbiota changes during pregnancy. However, specific microbial shifts have been reported to be predictive of GDM and also, gut microbial differences in women with and without GDM have been reported (here and here) . It has been also reported that the gut microbiota shifts (in composition and activity metabolites) in women with preeclampsia (see here). Thus, it is quite possible that the women in these studies, obese women, react to gut-microbiota-related interventions differently than non-obese women and that their pre-pregnancy weight puts them at an increased risk of complications.

It is worth noting that the total number of cases cited in the Cochrane review supporting their conclusion was 31 cases of preeclampsia in 472 women who took probiotics versus 17 in 483 women in the placebo groups. Thus, 14 more women who experienced preeclampsia, 9 of whom came from one of the studies [“probiotics increase the risk of pre-eclampsia compared to placebo (RR 1.85, 95% CI 1.04 to 3.29; p-value=0.04; 4 studies, 955 women; high-certainty evidence”] This is not a very large number of subjects for such a strong conclusion. The authors don’t mention if this high-certainty evidence is in all women or just obese women. By combining four studies, in which none found a significant increase in preeclampsia, the authors did find significance. Is this a convincing number of subjects? The Cochrane author, Dr. Marloes Dekker Nitert replied to an inquiry from us that she believes that this difference makes it unethical to conduct further studies in pregnant women, stating, “I think that there now is a lack of clinical equipoise to do an RCT on a combination of Lactobacillus/Bifidobacterium.

This is a strong statement but is consistent with their high-certainty of evidence statement. We acknowledge that something does appear to be going on. It is possible that certain populations react differentially to certain strains. Thus, maybe mild to morbidly obese women are a subgroup that needs closer monitoring during pregnancy and maybe even in non-pregnant settings, as they may react differently to probiotic interventions. Maybe it is just certain strains, as the Cochrane author was very clear in her email to state, “a combination of Lactobacillus/Bifidobacterium” and not generalize to all probiotics. We agree and in fact it is possible that different strains of Lactobacillus/Bifidobacterium will have different outcomes. Pregnancy is also a continuum and to think that giving an intervention during the first trimester is the same as during the third makes little scientific or clinical sense. Along these lines, one study showed the association of probiotic intake with different effects in early versus late pregnancy; an analysis that specifically focused on women in the third trimester of pregnancy found no association between probiotics and adverse fetal outcomes.

Conclusions

In summary, we must recognize that certain strains of probiotics may cause harm in certain populations. This reinforces the importance of diligent collection of adverse event data during all clinical trials. Although Cochrane is renowned to conduct analyses of the highest caliber, we wonder if four studies of 955 mostly obese women, in which 14 more in the probiotic group than the placebo group have a secondary outcome of harm, warrant the conclusion that there is “high-certainty evidence” that probiotics cause harm. This seems overstated based on our review of the literature. Should women and clinicians pay particular attention to this subgroup (obese pregnant women) and this outcome (preeclampsia, hypertension)? We think the answer is yes. But we do not conclude that all women at all stages of pregnancy need to refrain from probiotics. Fortunately, at the time of writing there appear to be 87 trials listed on clinicaltrials.gov looking at probiotics and pregnancy. As in many things the details still need to be further elucidated and we expect more clarification on this issue over the next 5-10 years.

A pediatrician’s perspective on c-section births and the gut microbiome

By Prof. Hania Szajewska, MD, Medical University of Warsaw, Poland and Kristina Campbell, MSc, ISAPP Consulting Communications Director

The decision to have a Cesarean section (C-section) should always depend on whether this is the best choice for the mother and baby, and it is never made by pediatricians. However, pediatricians are often asked about the consequences of C-section delivery for a child later in life and whether potential C-section-related harms may be reduced.

The data show that delivery by C-section is now more common than ever globally. The World Health Organization estimates the  C-section rate is around 21% of all births, and predicted to continue increasing. Although C-section rates are increasing both in developed and developing countries, Korea, Chile, Mexico, and Turkey have the highest rates in the world, with C-sections constituting 45% to 53% of all births. C-sections outnumber vaginal births in countries that include Dominican Republic, Brazil, Cyprus, Egypt, and Turkey.

Cesarean delivery is a medical procedure that can of course save an infant (or a mother) in a moment of danger, making birth less risky overall. But analyses have shown not all C-sections are initiated for safety reasons—some are driven by convenience and other non-medical factors. In areas with the highest C-section rates, only around half of the time are they required for life-saving reasons. Although the rate of medically necessary C-sections globally is difficult to establish, the WHO estimates it is between 10-15% of all births.

Non-essential C-sections would be perfectly reasonable if the health risks later in life were negligible. But are they? Scientific work in the past decade has shown that, in fact, there may be downsides to being born by C-section—and these health risks may manifest later in a child’s life.

By now, many observational studies have associated Cesarean births with an increased risk of various chronic health conditions that appear long after birth. C-section is associated with a higher risk of asthma and allergy, as well as obesity and type 2 diabetes. A systematic review and meta-analysis (incorporating 61 studies, which together included more than 20 million deliveries) also linked C-sections with autism spectrum disorders and attention deficit hyperactivity disorder (ADHD). Type 1 diabetes is also more prevalent in children born by c-section.

Since association is not the same as causation, scientists have looked at possible biological correlates of C-section and how they could be tied to future health problems. A leading hypothesis is that C-section deliveries cause health problems by disrupting the infant’s normal gut microbiota (i.e. the collection of microorganisms in specific ‘habitats’ on the infant’s body, such as the gut) within a critical time window for immune system development.

An altered microbiota in C-section births

One of the main clues about whether C-section births affect health via the microbiota is the consistent observation that infants born by C-section have a different collection of microorganisms in their digestive tracts and elsewhere on their bodies immediately after birth, compared with vaginally-born controls. Newborns delivered by C-section tend to harbor in their guts disease-causing microbes commonly found in hospitals (e.g. Enterococcus and Klebsiella), and lack strains of gut bacteria found in healthy children (e.g. Bacteroides species). Because it is known that gut microbiota are in close communication with the immune system, this difference in birth microbes may set the immune system up for later dysfunction.

However, an important confounding factor exists. Antibiotic administration is a recommended medical practice for C-section births in order to prevent infections. Antibiotics are potent disruptors of microbial communities – in this case the mother’s, or perhaps the infant’s if antibiotics are administered prior to umbilical cord clamping. It is not yet clear whether the timing of antibiotic administration can prevent such disruptions. (See conflicting evidence here and here; also see here.).

Gut microbiota disruption is associated with C-sections, but since C-section and antibiotics nearly always go together (with potential exposure of the infant to these drugs), it is not clear to what extent C-section and/or antibiotic treatments drive increased risk of chronic disease later in life. Antibiotic treatments within the first 2 years of life are independently associated with an increased risk of several conditions: childhood-onset asthma, allergic rhinitis, atopic dermatitis, celiac disease, overweight / obesity, and ADHD.

Options for microbiota ‘restoration’

If mechanistic studies continue to support the idea that the C-section-disrupted gut microbiota is the trigger for chronic diseases later in life, strategies could be proposed for ‘restoring’ or normalizing the infant gut microbiota after such births. Already some microbiota modifying interventions have been evaluated:

  • Probiotics: Undesired changes in microbiota composition and function caused by antibiotic treatments and/or caesarean birth may be addressed by probiotics—i.e. “live microorganisms that, when administered in adequate amounts, confer a health benefit on the host”. In one study, a mixture of several probiotic strains along with at least partial breastfeeding shifted the infant gut microbiota toward a more favorable profile. Intriguingly, probiotics (of various strains) prevent IgE-associated allergy until age 5 years, specifically in cesarean-delivered children but not in all children.
  • Synbiotics: A study showed supplementation with scGOS/lcFOS and B. breve M-16V appeared to compensate for delayed Bifidobacterium colonization in the guts of C-section delivered infants.
  • Maternal vaginal microbial transfer (otherwise known as ‘vaginal seeding’ or ‘microbial bath’: This is a procedure in which infants born by C-section, immediately after birth, are swabbed with gauze that contains microbes from the mother’s vaginal tract. After the media attention given to early studies, there is increased demand from parents for this procedure. Although some studies have found it effective for normalizing the infant gut microbiota, safety is not completely established and it is too early for routine use of this procedure. Parents wishing to try this approach are advised to participate in a study as part of an institutional review board-approved research protocol.
  • Maternal fecal microbiota transplantation: This procedure involves fecal microbes from the mother, orally administered to C-section infants after birth. A proof-of-concept study showed that after this intervention the gut microbiota of C-section-born infants looked more similar to that of vaginally born infants. But for this procedure, as above, the risk of transmitting harmful microbes is a concern, making it too early to recommend the procedure unless it is part of an institutional review board-approved research protocol.
  • Breastfeeding: Breastfeeding is the gold standard for infant nutrition, and breast milk contains live microorganisms as well as other components that interact with the gut microbiota. Exclusive breastfeeding for about 6 months of C-section infants helps the gut microbiota shift toward a profile seen in vaginally born infants.

So far, probiotics, synbiotics, and microbiota ‘restoration’ are not sufficiently reliable solutions for correcting the microbiota disruptions that accompany C-section births. Further studies are needed to develop these approaches.

A leading strategy

At present, breastfeeding is the main strategy for supporting the infant gut microbiota after C-section for the greatest chance of avoiding negative health consequences. Breastfeeding has multiple benefits, but may be of increased importance after C-section birth. Mothers should be supported after giving birth by C-section to breastfeed the infant during this critical period of early life and immune system development.

 

What is a strain in microbiology and why does it matter?

By Prof. Colin Hill, Microbiology Department and APC Microbiome Ireland, University College Cork, Ireland

At the recent ISAPP meeting in Sitges we had an excellent debate on the topic of ‘All probiotic effects must be considered strain-specific’. Notwithstanding which side of the debate prevailed, it does raise the question: what exactly is a strain? As a card-carrying microbiologist I should probably be able to simply define the term and give you a convincing answer, but I find that it is a surprisingly difficult concept to capture. It is unfortunately a little technical as a topic for a light-hearted blog, but here goes. Let me start by saying that the term ‘strain’ is important largely because we like to name things and then use those names when we share information, but that the concept of ‘strain’ may have no logical basis in nature where mutations and changes to a bacterial genome are constantly occurring events.

Let’s suppose I have a culture of Lactobacillus acidophilus growing in a test-tube, grown from a single colony. This clonal population is obviously a single strain that I will name strain Lb. acidophilus ISAPP2022. That was easy! I am aware of course that within this population there will almost certainly be a small number of individual cells with mutations (single nucleotide polymorphisms, or SNPs), cells that may have lost a plasmid, or cells that have undergone small genomic rearrangements. Nonetheless, because this genetic heterogeneity is unavoidable, I still consider this to be a pure strain. If I isolate an antibiotic resistant version of this strain by plating the strain on agar containing streptomycin and selecting a resistant colony I will now have an alternative clonal population all sharing a SNP (almost certainly in a gene encoding a ribosomal subunit). Even though there is a potentially very important genotypic and phenotypic difference I would not consider this to be a new strain, but rather it is a variant of Lb. acidophilus ISAPP2022. To help people in the lab or collaborators I might call this variant ISAPP2022SmR, or ISAPP2022-1. In my view, I could continue to make changes to ISAPP2022 and all of those individual clonal populations will still be variants of the original strain. So, the variant concept is that any change in the genome, no matter how small, creates a new variant. When I grow ISAPP2022 in my lab for many years, or share it with others around the word, it is my view that we are all working with the same strain, despite the fact that different variants will inevitably emerge over time and in different labs.

Where the strain concept becomes more difficult is when I isolate a bacterium from a novel source and I want to determine if it is the same strain as ISAPP2022. If the whole genome sequence (WGS) is a perfect match (100% average nucleotide identity or ANI) then both isolates are the same strain and both can be called ISAPP2022. If they have only a few SNPs then they are variants of the same strain. If the two isolates only share 95% ANI then they are obviously not the same strain and cannot even be considered as members of the same species (I am using a species ANI cut-off of 96% that I adopted from a recent paper in IJSEM.

Where it gets really tricky is when the ANI lies between 96% (so that we know that the isolates are both members of the same species) and 100% (where they are unequivocally the same strain). Where should we place the cut-off to define a strain? At what point is a threshold crossed and an isolate goes from being a variant to becoming a new strain? Should this be a mathematical decision based solely on ANI, or do we have to consider the functionality of the changes? If it is mathematical then we could simply choose a specific value, say 99.95% or 99.99% ANI, and declare anything below that value is a new strain. Remember that the 2Mb genome size of Lb. acidophilus would mean that two isolates sharing 99.99% ANI could differ by up to 200 SNPs. This could lead to a situation where an isolate with 199 SNPs compared to ISAPP2022 is considered a variant, but an isolate with 201 SNPs is a new strain (even though it only differs from the variant with 199 SNPs by two additional SNPs). This feels very unsatisfactory. But what about an isolate with only 50 SNPs, but one that has a very different phenotype to ISAPP2022 because the SNPs are located in important genes? Or what about an isolate with an additional plasmid, or missing a plasmid, or with a chromosomal deletion or insertion? I would argue we should not have a hard and fast cut-off based on SNPs alone, but we should continue to call all of these variants, and not define them as new strains.

So, by how much do two isolates have to differ before we no longer consider them as variants of one another, but as new strains? I will leave that question to taxonomists and philosophers since for me it falls into the territory of ‘how many angels can dance on the head of pin?’

All this may seem somewhat esoteric, but there are practical implications. Can we translate the findings from a clinical trial done with a specific variant of a strain to all other variants of the same strain? If Lactobacillus acidophilus ISAPP2022 has been shown to deliver a health benefit (and is therefore a probiotic), can we assume that Lb acidophilus ISAP2022-1 or any other variant will have the same effect? What if a variant has only one mutation, but that mutation eliminates an important phenotype required for the functionality of the original strain? I am afraid that at the end of all this verbiage I have simply rephrased the original debate topic from ‘All probiotic effects must be considered strain-specific’ to ‘All probiotic effects must be considered variant-specific’. Looks like we might be heading back to the debate stage in 2023!

Bifidobacteria in the infant gut use human milk oligosaccharides: how does this lead to health benefits?

By Martin Frederik Laursen, Technical University of Denmark, 2022 co-recipient of Glenn Gibson Early Career Research Prize

Breast milk is the ‘gold standard’ of infant nutrition, and recently scientists have zeroed in on human milk oligosaccharides (HMOs) as key components of human milk, which through specific interaction with bifidobacteria, may improve infant health. Clarifying mechanisms by which HMOs act in concert with bifidobacteria in the infant gut may lead to better nutritional products for infants.

Back in early 2016, I was in the middle of my PhD studies working on determinants of the infant gut microbiota composition in the Licht lab at the National Food Institute, Technical University of Denmark. I had been working with fecal samples from a Danish infant cohort study, called SKOT (Danish abbreviation for “Diet and well-being of young children”), investigating how the diet introduced in the complementary feeding period (as recorded by the researchers) influences the gut microbiota development 1,2. Around the same time, Henrik Munch Roager, PostDoc in the lab, was developing a liquid chromatography mass spectrometry (LC-MS)-based method for quantifying the aromatic amino acids (AAA) and their bacterially produced metabolites in fecal samples (the 3 AAAs and 16 derivatives thereof). These bacterially produced AAA metabolites were starting to receive attention because of their role in microbiota-host cross-talk and interaction with various receptors such as the Aryl Hydrocarbon Receptor (AhR) expressed in immune cells and important for controlling immune responses at mucosal surfaces 3,4. However, virtually nothing was known about bacterial metabolism of the AAAs in the gut in an early life context. Further, the fecal samples collected from the SKOT cohort were obtained in a period of life when infants are experiencing rapid dietary changes (e.g. cessation of breastfeeding and introduction of various new foods). Thus, we wondered whether the AAA metabolites would be affected by diet and whether these metabolites might contribute to the development of the infant’s immune system. Our initial results quickly guided us on the track of breastfeeding and bifidobacteria! Here is a summary of the story, published last year in Nature Microbiology5. (See the accompanying News & Views article here.)

We initially looked at the data from a subset of 59 infants, aged 9 months, from the SKOT cohort. Here we found that both the gut microbiome and the AAA metabolome were affected by breastfeeding status (breastfed versus weaned). It is well established that certain bifidobacteria dominate the bacterial gut community in breastfed infants due to their efficient utilization of HMOs – which are abundant components of human breastmilk 6. Our data showed the same, namely enrichment of Bifidobacterium in the breastfed infants, but also indicated that the abundance of specific AAA metabolites were dependent on breastfeeding.

Trying to connect the gut microbiome and AAA metabolome, we found striking correlations between the relative abundance of Bifidobacterium and specifically abundances of three aromatic amino acid catabolites – namely indolelactic acid (ILA), phenyllactic acid (PLA) and 4-hydroxyphenyllactic acid (4-OH-PLA), collectively aromatic lactic acids. These metabolites are formed in two enzymatic reactions (a transamination followed by a hydrogenation) of the aromatic amino acids tryptophan, phenylalanine and tyrosine. However, the genes involved in this pathway were not known for bifidobacteria. Digging deeper we discovered that not all Bifidobacterium species found in the infant’s gut correlated with these metabolites. This was only true for the Bifidobacterium species enriched in the breastfed infants (e.g. B. longum, B. bifidum and B. breve), but not post-weaning/adult type bifidobacteria such as B. adolescentis and B. catenulatum group.

We decided to go back to the lab and investigate these associations by culturing representative strains of the Bifidobacterium species found in the gut of these infants. Indeed, our results confirmed that Bifidobacterium species are able to produce aromatic lactic acids, and importantly that the ability to produce them was much stronger for the HMO-utilizing (e.g. B. longum, B. bifidum and B. breve) compared to the non-HMO utilizing bifidobacteria (e.g. B. adolescentis, B. animalis and B. catenulatum). Next, in a series of experiments we identified the genetic pathway in Bifidobacterium species responsible for production of the aromatic lactic acids and performed enzyme kinetic studies of the key enzyme, an aromatic lactate dehydrogenase (Aldh), catalyzing the last step of the conversion of aromatic amino acids into aromatic lactic acids. Thus, we were able to demonstrate the genetic and enzymatic basis for production of these metabolites in Bifidobacterium species.

To explore the temporal dynamics of Bifidobacteria and aromatic lactic acids and validate our findings in an early infancy context (a critical phase of immune system development), we recruited 25 infants (Copenhagen Infant Gut [CIG] cohort) from which we obtained feces from birth until six months of age. These data were instrumental for demonstrating the tight connection between specific Bifidobacterium species, HMO-utilization and production of aromatic lactic acids in the early infancy gut and further indicated that formula supplementation, pre-term delivery and antibiotics negatively influence the concentrations of these metabolites in early life.

Having established that HMO-utilizing Bifidobacterium species are key producers of aromatic lactic acids in the infant gut, we focused on the potential health implications of this. We were able to show that the capacity of early infancy feces to in vitro activate the AhR, depended on the abundance of aromatic lactic acid producing Bifidobacterium species and the concentrations of ILA (a known AhR agonist) in the fecal samples obtained from the CIG cohort. Further, using isolated human immune cells (ex vivo) we showed that ILA modulates cytokine responses in Th17 polarized cells – namely it increased IL-22 production in a dose and AhR-dependent manner. IL-22 is a cytokine important for protection of mucosal surfaces, e.g. it affects secretion of antimicrobial proteins, permeability and mucus production 7. Further, we tested ILA in LPS/INFγ induced monocytes (ex vivo), and found that ILA was able to decrease the production of the proinflammatory cytokine IL-12p70, in a manner dependent upon both AhR and the Hydroxycarboxylic Acid (HCA3) receptor, a receptor expressed in neutrophils, macrophages and monocytes and involved in mediation of anti-inflammatory processes 8,9. Overall, our data reveal potentially important ways in which bifidobacteria influence the infant’s developing immune system.

Figure 1 – HMO-utilizing Bifidobacterium species produce immuno-regulatory aromatic lactic acids in the infant gut.

Our study provided a novel link between HMO-utilizing Bifidobacterium species, production of aromatic lactic acids and immune-regulation in early life (Figure 1). This may explain previous observations that the relative abundance of bifidobacteria in the infant gut is inversely associated with development of asthma and allergic diseases 10–12 and our results, together with other recent findings13–15 are pointing towards aromatic lactic acids (especially ILA) as potentially important mediators of beneficial immune effects induced by HMO-utilizing Bifidobacterium species.

 

References

  1. Laursen, M. F. et al. Infant Gut Microbiota Development Is Driven by Transition to Family Foods Independent of Maternal Obesity. mSphere 1, e00069-15 (2016).
  2. Laursen, M. F., Bahl, M. I., Michaelsen, K. F. & Licht, T. R. First foods and gut microbes. Front. Microbiol. 8, (2017).
  3. Zelante, T. et al. Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22. Immunity 39, 372–385 (2013).
  4. Sridharan, G. V. et al. Prediction and quantification of bioactive microbiota metabolites in the mouse gut. Nat. Commun. 5, 1–13 (2014).
  5. Laursen, M. F. et al. Bifidobacterium species associated with breastfeeding produce aromatic lactic acids in the infant gut. Nat. Microbiol. 6, 1367–1382 (2021).
  6. Sakanaka, M. et al. Varied pathways of infant gut-associated Bifidobacterium to assimilate human milk oligosaccharides: Prevalence of the gene set and its correlation with bifidobacteria-rich microbiota formation. Nutrients 12, 71 (2020).
  7. Keir, M. E., Yi, T., Lu, T. T. & Ghilardi, N. The role of IL-22 in intestinal health and disease. J. Exp. Med. 217, (2020).
  8. Peters, A. et al. Metabolites of lactic acid bacteria present in fermented foods are highly potent agonists of human hydroxycarboxylic acid receptor 3. PLoS Genet. 15, e1008145 (2019).
  9. Peters, A. et al. Hydroxycarboxylic acid receptor 3 and GPR84 – Two metabolite-sensing G protein-coupled receptors with opposing functions in innate immune cells. Pharmacol. Res. 176, (2022).
  10. Fujimura, K. E. et al. Neonatal gut microbiota associates with childhood multisensitized atopy and T cell differentiation. Nat. Med. 22, 1187–1191 (2016).
  11. Stokholm, J. et al. Maturation of the gut microbiome and risk of asthma in childhood. Nat. Commun. 9, 141 (2018).
  12. Seppo, A. E. et al. Infant gut microbiome is enriched with Bifidobacterium longum ssp. infantis in Old Order Mennonites with traditional farming lifestyle. Allergy Eur. J. Allergy Clin. Immunol. 76, 3489–3503 (2021).
  13. Meng, D. et al. Indole-3-lactic acid, a metabolite of tryptophan, secreted by Bifidobacterium longum subspecies infantis is anti-inflammatory in the immature intestine. Pediatr. Res. 88, 209–217 (2020).
  14. Ehrlich, A. M. et al. Indole-3-lactic acid associated with Bifidobacterium-dominated microbiota significantly decreases inflammation in intestinal epithelial cells. BMC Microbiol. 20, 357 (2020).
  15. Henrick, B. M. et al. Bifidobacteria-mediated immune system imprinting early in life. Cell 184, 3884-3898.e11 (2021).

Probiotics vs. prebiotics: Which to choose? And when?

By Dr. Karen Scott, PhD, Rowett Institute, University of Aberdeen, Scotland

As consumers we are constantly bombarded with information on what we should eat to improve our health. Yet the information changes so fast that it sometimes seems that what was good for us last week should now be avoided at all costs!

Probiotics and prebiotics are not exempt from such confusing recommendations, and one area lacking clarity for many is which of them we should pick, and when. In this blog I will consider the relative merits of probiotics and prebiotics for the gut environment and health.

By definition, both probiotics and prebiotics should ‘confer a health benefit on the host’. Since an improvement in health can be either subjective (simply feeling better) or measurable (e.g. a lowering in blood pressure) it is clear that there is not a single way to define a ‘health benefit’. This was discussed nicely in a previous blog by Prof Colin Hill.

Although consumption of both probiotics and prebiotics should provide a health benefit, this does not mean that both need to act through the gut microbiota. Prebiotics definitively need to be selectively utilised by host microorganisms – they are food for our existing microbiota. However, depending on the site of action, this need not be the gut microbiota, and prebiotics targeting other microbial ecosystems in or on the body are being developed. Traditionally prebiotics have specifically been used to boost numbers of gut bacteria such as Bifidobacterium and the Lactobacilliaceae family, but new prebiotics targeting different members of the gut microbiota are also currently being researched.

Probiotics are live bacteria and despite a wealth of scientific evidence that specific probiotic bacterial strains confer specific health benefits, we often still do not know the exact mechanisms of action. This can make it difficult both to explain how or why they work, and to select new strains conferring similar health benefits. Many probiotics exert their effects within the gut environment, but they may or may not do this by interacting with the resident gut microbiota. For instance probiotics that reduce inflammation do so by interacting directly with cells in the mucosal immune system. Yet strains of lactobacilli (see here for what’s included in this group of bacteria) may do this by modulating cytokine production while Bifidobacterium strains induce tolerance acquisition. These very different mechanisms are one reason why mixtures containing several probiotic species or strains may in the end prove the most effective way to improve health. On the other hand, some probiotics do interact with the resident gut microbes: probiotics that act by inhibiting the growth of pathogenic bacteria clearly interact with other bacteria. Sometimes these may be potential disease-causing members of the resident microbiota, normally kept in check by other commensal microbes that themselves have become depleted due to some external impact, and some may be incoming pathogens. Such interactions can occur in the gut or elsewhere in the body.

This brings me back to the original question, and one I am frequently asked – should I take a probiotic or a prebiotic? The true and quick answer to this question is ‘it depends’! It depends why you are asking the question, and what you want to achieve. Let’s think about a few possible reasons for asking the question.

I want to improve the diversity of my microbiota. Should I take a prebiotic or a probiotic?

My first reaction was that there is an easy answer to this question – a prebiotic. Prebiotics are ‘food’ for your resident bacteria, so it follows that if you want to improve the diversity of your existing microbiota you should take a prebiotic. However, in reality this is too simplistic. Since prebiotics are selectively utilised by a few specific bacteria within the commensal microbiota to provide a health benefit, taking a prebiotic will boost the numbers of those specific bacteria. If the overall bacterial diversity is low, this may indeed improve the diversity. However, if the person asking the question already has a diverse microbiota, although taking one specific prebiotic may boost numbers of a specific bacterium, it may not change the overall diversity in a measurable way. In fact the best way to increase the overall diversity of your microbiota is to consume a diverse fibre-rich diet – in that way you are providing all sorts of different foods for the many different species of bacteria living in the gut, and this will increase the diversity of your microbiota.  Of course, if you already consume a diverse fibre-rich diet your microbiota may already be very diverse, and any increased diversity may not be measurable.

I want to increase numbers of bifidobacteria in my microbiota. Should I take a prebiotic or a probiotic?

Again, I initially thought this was easy to answer – a prebiotic. There is a considerable amount of evidence that prebiotics based on fructo-oligosaccharides (FOS or inulin) boost numbers of bifidobacteria in the human gut. But this is only true as long as there are bifidobacteria present that can be targeted by consuming suitable prebiotics. Some scientific studies have shown that there are people who respond to prebiotic consumption and people who do not (categorised as responders and non-responders). This can be for two very different reasons. If an individual is devoid of all Bifidobacterium species completely, no amount of prebiotic will increase bifidobacteria numbers, so they would be a non-responder. In contrast if someone already has a large, diverse bifidobacteria population, a prebiotic may not make a meaningful impact on numbers – so they may also be a non-responder.

However, for those people who do not have any resident Bifidobacterium species, the only possible way to increase them would indeed be to consume a probiotic- specifically a probiotic containing one or several specific Bifidobacterium species. Consuming a suitable diet, or a prebiotic alongside the probiotic, may help retention of the consumed bifidobacteria, but this also depends on interactions with the host and resident microbiota.

I want to increase numbers of ‘specific bacterium x’ in my microbiota. Should I take a prebiotic or a probiotic?

The answer here overlaps with answer 2, and depends on the specific bacterium, and what products are available commercially, but the answer could be to take either, or a combination of both – i.e. a synbiotic.

If bacterium x is available as a probiotic, consuming that particular product could help. If bacterium x has been widely researched, and the specific compounds it uses for growth have been established, identifying and consuming products containing those compounds could boost numbers of bacterium x within the resident microbiota. Such research may already have identified combination products – synbiotics – that could also be available.

One caveat for the answers to questions 2 and 3 is that probiotics do not need to establish or alter the gut microbiota to have a beneficial effect on health. In fact, a healthy large intestine has a microbial population of around 1011-1012 bacterial cells per ml, or up to 1014 cells in total, while a standard pot of yogurt contains 1010 bacterial cells (108 cells/ml). Assuming every probiotic bacterial cell reaches the large intestine alive, they would be present in a ratio of 1: 10,000. This makes it difficult for them to find a specific niche to colonise, so consuming a probiotic may not “increase numbers of ‘specific bacterium x’ in my microbiota”, but this does not mean that the function of the probiotic within the gut ecosystem would not provide a health benefit. Many probiotics act without establishing in the microbiota.

I’ve been prescribed antibiotics. Should I take a prebiotic or a probiotic?

In this case the answer is clear cut – a probiotic.

There is a lot of evidence that consumption of probiotics can alleviate symptoms of, or reduce the duration of, antibiotic associated diarrhoea. From what we know about mechanisms of action, consumption of antibiotics kills many resident gut bacteria, reducing the overall bacterial population and providing an opportunity for harmful bacteria to become more dominant. Consuming certain probiotics can either help boost bacterial numbers in the large intestine, preventing the increased growth in pathogenic bacteria until the resident population recovers, or can increase production of short chain fatty acids, decreasing the colonic pH, preventing growth of harmful bacteria. Ideally probiotics would be taken alongside antibiotics, from day 1, to avoid the increase in numbers of the potentially harmful bacteria in the first place. This has been shown to be more effective. Consuming the probiotic alongside prebiotics that could help the resident microbiota recover more quickly may be even more effective. Even if you’ve already started the course of antibiotics, it’s not too late to start taking probiotics to reduce any side-effects. Always remember to complete taking the course of antibiotics as prescribed.

 

 

Putting all of this together to answer the initial question of whether it’s better to take probiotics or prebiotics, a better answer may in fact be take both to cover the different effects each has, maximising the benefit to health. There are specific times when probiotics are better, and other times when prebiotics are better, and consuming both together may make each more effective. In any case care has to be taken to consume a product that has been confirmed through robust studies to have the specific benefit that is required.

 

Do polyphenols qualify as prebiotics? The latest scientific perspectives

Kristina Campbell, Consulting Communications Director, ISAPP

When the ISAPP scientific consensus definition of ‘prebiotic’ was published in 2017, the co-authors on the paper included polyphenols as potential prebiotic substances. At the time, the available data on the effect of polyphenols on the gut microbiota were insufficient to show a true prebiotic effect.

An ISAPP webinar held in April 2022, aimed to give an update on the health effects of polyphenols and their mechanisms of action, along with how well polyphenols fit the prebiotic definition. Prof. Daniele Del Rio from University of Parma, Italy, and Prof. Yves Desjardins from Université Laval, Canada, presented the latest perspectives in the field.

What are polyphenols?

Polyphenols are a group of compounds found in plants, with over 6000 types identified to date. They can be divided into two main categories, flavonoids and non-flavonoids.

Polyphenols are absorbed in two different ways in the body. A very small fraction is absorbed in the small intestine, but 95% of them reach the lower gut and interact with gut microbiota. Although polyphenols have a special capacity to influence the activities of microorganisms, some resident microorganisms, in turn, can change the chemical structure of polyphenols through enzymatic action. These interactions produce a unique array of metabolites, which may be responsible for some of polyphenols’ prebiotic effects.

What are the health effects of polyphenols?

Epidemiological studies show that polyphenols in the diet are associated with many health benefits, including prevention of cardiovascular disease, certain cancers, and metabolic disease. These effects occur through various mechanisms. However, association is not proof of causation. So how good is the evidence that polyphenols can lead to health benefits?

Numerous human studies exist, but the most robust study to date for the health benefits of polyphenols is a randomized, controlled trial of over 20,000 adults, published in 2022, which showed supplementation with cocoa extract reduced death from cardiovascular events (although it did not reduce the number of cardiovascular events).

What are the mechanisms of action for polyphenols?

Polyphenols have multiple mechanisms of action. Del Rio focuses on the metabolites produced from dietary polyphenols called flavan-3-ols, which are found in red wine, grapes, tea, berries, chocolate and other foods. Along with colleagues, he showed that the metabolites produced in response to a polyphenol-rich food occur two ‘waves’: a small wave in the first 2 hours after ingestion, and a larger wave 5-35 hours after ingestion. The second wave is produced when flavan-3-ols reach the colon and interact with gut microbiota.

Work is ongoing to link these metabolites to specific health effects. Along these lines, Del Rio described a study showing how cranberry flavan-3-ol metabolites help defend against infectious Escherichia coli in a model system of bladder epithelial cells. These polyphenols are transformed by the gut microbiota into smaller compounds that are absorbed—so the health benefit comes not from the activity of polyphenols directly, but from the molecule(s) that the gut microbiota has produced from the polyphenols.

How else do polyphenols work? Ample evidence suggests polyphenols interact in different ways with gut microbes: they have direct antimicrobial effects, they affect quorum sensing, they compete with bacteria for some minerals, and/or they modify ecology, thereby affecting biofilm formation. Desjardins explained that these interactions may occur in parallel: for example, polyphenols may exert antimicrobial effects when they reach the colon, and at the same time, microorganisms in the gut begin to degrade them.

The mode of action of polyphenols Desjardins studies is the prebiotic mode of action—or as he describes it, “prebiotic with a twist”. A landmark paper from 2015 showed how cranberry polyphenols had protective effects on metabolism and obesity through the creation of mucin in the intestine, which formed a good niche for Akkermansia muciniphila, a keystone bacterial species for good metabolic health. Other polyphenols have since been shown to work the same way: by stimulating production of mucin, thereby providing ideal conditions for beneficial bacteria to grow. In this way, polyphenols appear to show small-scale effects comparable to the effects of probiotics, by inducing a host response that alters the bacterial niche.

Are the effects of polyphenols individual?

Del Rio offered some evidence that the health effects of polyphenols, via metabolites, is personalized: a study showed the existence of three distinct patterns of metabolite production in response to dietary polyphenols (ellagitannins). These may depend on the particular microbes of the gut and their ability to produce the relevant metabolites—so in essence, in each case the gut microbiota is equipped to produce a certain set of metabolites in response to polyphenols. More work is needed, however, to be able to personalize polyphenol intake.

Do polyphenols qualify as prebiotic substances?

Polyphenols clearly interact with gut microbiota to influence human health. The definition of a prebiotic is “a substrate that is selectively utilized by host microorganisms conferring a health benefit”. Given the available evidence that polyphenols are not metabolized or utilized by bacteria in all cases in the same direct way as carbohydrate prebiotics, Desjardins sees them as having a “prebiotic-like effect”. Rather, polyphenols are transformed into other biologically active molecules that ultimately provide health benefits to the host. These prebiotic-like properties of polyphenols are nicely summarized in a 2021 review paper and include decreasing inflammation, increasing bacteriocins and defensins, increasing gut barrier function (thereby reducing low-grade inflammation), modulating bile acids, and increasing gut immuno-globulins.

Overall, the speakers showed that polyphenols exert their health effects in several ways—and while the gut microbiota are important for their health effects, polyphenols, as a heterogenous group, may not strictly meet the criteria for prebiotics. Clearly, more research on polyphenols may reveal other mechanisms by which these important nutrients influence the gut microbiome and contribute to host health, and they may someday be regarded as prebiotics.

Watch the replay of the ISAPP webinar here.

The gut mycobiome and misinformation about Candida

By Prof. Eamonn Quigley, MD, The Methodist Hospital and Weill Cornell School of Medicine, Houston

As a gastroenterologist, I frequently meet with patients who are adamant that a Candida infection is the cause of their ailments. Patients experiencing a range of symptoms, including digestive problems, sometimes believe they have an overgrowth of Candida in their gastrointestinal (GI) tract and want to know what to do about it. Their insistence is perhaps not surprising, given how many many websites and social media ‘gurus’ share lists of symptoms supposedly tied to Candida infections. Even cookbooks exist with recipes specifically tailored to “cure” someone of Candida infection through dietary changes. Some articles aim to counter the hype – for example, an article titled “Is gut Candida overgrowth actually real, and do Candida diets work?” Yet patients are too often confused about the evidence on Candida and other fungi in the GI tract. In a 2021 ISAPP presentation on the gut mycobiome, I provided a clinical perspective on fungal infections and the related evidence base.

Fungal infections do occur

Much of the misinformation I encounter on Candida infections focuses on selling a story that encourages people to blame Candida overgrowth as the cause of their symptoms and undertake expensive or complicated dietary and supplement regimens to “cure” the infection. This is not to say that fungal infections do not take place in the body. Fungal infections, from Candida or other fungi, frequently occur on the nails or skin. Patients taking oral or inhaled steroids may develop Candida infections in the oropharynx and esophagus. Immunocompromised patients also face a greater risk of Candidiasis and Candidemia—these include HIV patients; patients undergoing chemotherapy; transplant patients; and patients suffering from malnutrition.

Fungal infections are rare in the GI tract

Regardless, instances of documented Candida infection in the GI tract remain few in number. One study published in the 90s reported 10 patients hospitalized with severe diarrhea1. These patients suffered from chronic illness, underwent intense antimicrobial treatment or chemotherapy, and faced severe outcomes such as dehydration—and clinicians consistently identified the growth of Candida albicans in the patient fecal samples. Other studies on the matter lack the clinical evidence to conclude that fungal infections drive GI disease. A study examining small intestinal fungal overgrowth identified instances of fungal overgrowth among 150 patients with unexplained symptoms2. However, the lack of documentation of response to an antifungal treatment protocol makes it difficult to attribute the observed symptoms to the presence of fungal organisms.

 The gut mycobiome in IBS

The gut microbiome has taken centre stage in common discourse about gut health. In line with this movement, my colleagues at Cork investigated the fungal members of the gut microbiome – that is, the gut mycobiome – in the guts of patients diagnosed with irritable bowel syndrome (IBS)3 to ascertain whether there was any correlation with symptoms. This effort revealed DNA sequences belonging to many fungal species. However, no significant differences in the number of fungal species were observed between IBS patients and volunteers. A smaller study done on a Dutch cohort, on the other hand, detected significantly reduced total fungal diversity among IBS patients4. So, it’s not yet clear whether mycobiome differences exist across populations with IBS.

Studying the gut mycobiome for further insights

The few studies that have examined the human gut mycobiome expose the need to answer basic questions about the fungal components of the gut microbiome. For instance, what is the gut mycobiome composition among people not suffering from GI-related symptoms? Efforts to answer these questions would require longitudinal sample collection to account for the high turnover of microbes in the GI tract. We would also need to perform stool measures not typically performed in the clinic to better correlate fungal overgrowth with GI-related symptoms. Overall, any gut mycobiome study requires careful and detailed experimental design.

We also have to consider where the gut mycobiome originates. A recent study in mSphere showed that the increased amount of DNA belonging to S. cerevisiae in stool samples coincided with the number of times subjects consumed bread and other fungi-rich foods5. S. cerevisiae also failed to grow in lab conditions mimicking the gut environment after 7 days of incubation. These findings suggest that the fungi identified in gut mycobiome profiles are not persistent gut colonizers, but transient members of the gut microbiome that come from the food we digest or our saliva.

A survey of the literature on the gut mycobiome and fungal infections in the GI tract highlights the need to conduct more studies on the role fungi play in gut and overall health. My clinical approach when I encounter someone claiming to have GI symptoms caused by Candida infection is a skeptical, yet empathetic response. Through proper communication of the evidence, we can investigate the origin of symptoms together and identify the best treatment methods for any GI-related disease, whether caused by fungal infections or not.

ISAPP held a mini-symposium featuring six short lectures that explore different aspects of the human mycobiome, including research, clinical and industry perspectives. See here for the replay, with Dr. Quigley’s talk at 1:12:30.

References

(1)        Gupta, T. P.; Ehrinpreis, M. N. Candida-Associated Diarrhea in Hospitalized Patients. Gastroenterology 1990, 98 (3), 780–785. https://doi.org/10.1016/0016-5085(90)90303-i.

(2)        Jacobs, C.; Coss Adame, E.; Attaluri, A.; Valestin, J.; Rao, S. S. C. Dysmotility and Proton Pump Inhibitor Use Are Independent Risk Factors for Small Intestinal Bacterial and/or Fungal Overgrowth. Aliment Pharmacol Ther 2013, 37 (11), 1103–1111. https://doi.org/10.1111/apt.12304.

(3)        Das, A.; O’Herlihy, E.; Shanahan, F.; O’Toole, P. W.; Jeffery, I. B. The Fecal Mycobiome in Patients with Irritable Bowel Syndrome. Sci Rep 2021, 11 (1), 124. https://doi.org/10.1038/s41598-020-79478-6.

(4)        Botschuijver, S.; Roeselers, G.; Levin, E.; Jonkers, D. M.; Welting, O.; Heinsbroek, S. E. M.; de Weerd, H. H.; Boekhout, T.; Fornai, M.; Masclee, A. A.; Schuren, F. H. J.; de Jonge, W. J.; Seppen, J.; van den Wijngaard, R. M. Intestinal Fungal Dysbiosis Is Associated With Visceral Hypersensitivity in Patients With Irritable Bowel Syndrome and Rats. Gastroenterology 2017, 153 (4), 1026–1039. https://doi.org/10.1053/j.gastro.2017.06.004.

(5)        Auchtung, T. A.; Fofanova, T. Y.; Stewart, C. J.; Nash, A. K.; Wong, M. C.; Gesell, J. R.; Auchtung, J. M.; Ajami, N. J.; Petrosino, J. F. Investigating Colonization of the Healthy Adult Gastrointestinal Tract by Fungi. mSphere 2018, 3 (2), e00092-18. https://doi.org/10.1128/mSphere.00092-18.

 

 

Improving the quality of microbiome studies – STORMS

By Mary Ellen Sanders, PhD, ISAPP Executive Science Officer

In mid-March I attended the Gut Microbiota for Health annual meeting. I was fortunate to participate in a short workshop chaired by Dr. Geoff Preidis MD, PhD, a pediatric gastroenterologist from Baylor College of Medicine and Dr. Brendan Kelly MD, MSCE, an infectious disease physician and clinical epidemiologist from University of Pennsylvania. The topic of this workshop was “Designing microbiome trials – unique considerations.”

Dr. Preidis introduced the topic by recounting his effort (Preidis et al. 2020) to review evidence for probiotics for GI endpoints, including for his special interest, necrotizing enterocolitis (NEC). After a thorough review of available studies testing the ability of probiotics to prevent morbidity and mortality outcomes for premature neonates, he and the team found 63 randomized controlled trials that assessed close to 16,000 premature babies. Although the effect size for the different clinical endpoints was impressive and clinically meaningful, AGA was only able to give a conditional recommendation for probiotic use in this population.

Why? In part, because inadequate conduct or reporting of these studies led to reduced confidence in their conclusions. For example, proper approaches to mitigate selection bias must be reported. Some examples of selection bias include survival bias (where part of the target study population is more likely to die before they can be studied), convenience sampling (where members of the target study population are not selected at random), and loss to follow-up (when probability of dropping out is related to one of the factors being studied). These are important considerations that might influence microbiome results. If the publication on the trial does not clearly indicate how these potential biases were addressed, then the study cannot be judged as low risk of bias. It’s possible in such a study that bias is addressed correctly but reported incompletely. But the reader cannot ascertain this.

With an eye toward improving the quality and transparency of future studies that include microbiome endpoints, Dr. Preidis shared a paper by a multidisciplinary team of bioinformaticians, epidemiologists, biostatisticians, and microbiologists titled Strengthening The Organization and Reporting of Microbiome Studies (STORMS): A Reporting Checklist for Human Microbiome Research.

Dr. Preidis kindly agree to help the ISAPP community by answering a few questions about STORMS:

Dr. Preidis, why is the STORMS approach so important?

Before STORMS, we lacked consistent recommendations for how methods and results of human microbiome research should be reported. Part of the problem was the complex, multi-disciplinary nature of these studies (e.g., epidemiology, microbiology, genomics, bioinformatics). Inconsistent reporting negatively impacts the field because it renders studies difficult to replicate or compare to similar studies. STORMS is an important step toward gaining more useful information from human microbiome research.

One very practical outcome of this paper is a STORMS checklist, which is intended to help authors provide a complete and concise description of their study. How can we get journal editors and reviewers to request this checklist be submitted along with manuscripts for publication?

We can reach out to colleagues who serve on editorial boards to initiate discussions among the editors regarding how the STORMS checklist might benefit reviewers and readers of a specific journal.

How does this checklist differ from or augment the well-known CONSORT checklist?

Whereas the CONSORT checklist presents an evidence-based, minimum set of recommendations for reporting randomized trials, the STORMS checklist facilitates the reporting of a comprehensive array of observational and experimental study designs including cross-sectional, case-control, cohort studies, and randomized controlled trials. In addition to standard elements of study design, the STORMS checklist also addresses critical components that are unique to microbiome studies. These include details on the collection, handling, and preservation of specimens; laboratory efforts to mitigate batch effects; bioinformatics processing; handling of sparse, unusually distributed multi-dimensional data; and reporting of results containing very large numbers of microbial features.

How will papers reported using STORMS facilitate subsequent meta-analyses?

When included as a supplemental table to a manuscript, the STORMS checklist will facilitate comparative analysis of published results by ensuring that all key elements are reported completely and organized in a way that makes the work of systematic reviewers more efficient and more accurate.

I have been struck through the years of reading microbiome research that primary and secondary outcomes seem to be rarely stated up front. Or if such trials are registered, for example on clinicaltrials.gov, the paper does not necessarily focus on the pre-stated primary objectives. This approach runs the risk of researchers finding the one positive story to tell out of the plethora of data generated in microbiome studies. Will STORMS help researchers design more hypothesis driven studies?

Not necessarily. The STORMS checklist was not created to assess study or methodological rigor; rather, it aims to aid authors’ organization and ease the process of reviewer and reader assessment of how studies are conducted and analyzed.  However, if investigators use this checklist in the planning phases of a study in conjunction with sound principles of study design, I believe it can help improve the quality of human microbiome studies – not just the writing and reporting of results.

Do you have any additional comments?

One of the strengths of the STORMS checklist is that it was developed by a multi-disciplinary team representing a consensus across a broad cross-section of the microbiome research community. Importantly, it remains a work in progress, with planned updates that will address evolving standards and technological processes.  Anyone interested in joining the STORMS Consortium should visit the consortium website (www.stormsmicrobiome.org).

See related blog:   ISAPP take-home points from American Gastroenterological Association guidelines on probiotic use for gastrointestinal disorders

 

ISAPP’s Guiding Principles for the Definitions of ‘Biotics’

By Mary Ellen Sanders, PhD, ISAPP Executive Science Officer

Articulating a definition for a scientific concept is a significant challenge. Inevitably, scientists have different perspectives on what falls inside and outside the bounds of a term. Prof. Glenn Gibson, ISAPP co-founder and longtime board member, recently published a paper that describes his path to coining the word ‘prebiotic’, with this observation: “One thing I have learned about definitions is that if you propose one, then be ready for it to be changed, dismissed or ignored!”

Mary Ellen Sanders with Glenn Gibson

Members of the ISAPP board, however, have remained steadfast in their belief that such definitions are worth creating. They are the basis for shared understanding and coordinated progress across a scientific field.

Developing the consensus definition papers on probiotics, prebiotics, synbiotics, postbiotics and fermented foods was demanding on the part of all involved. The objective of the panels that met to discuss these definitions was clear – to provide common ground for consistent use of this growing body of terms for all stakeholders. Although some disagreement among the broader scientific community exists about some of the definitions, ISAPP’s approach relied on important, underlying principles:

  • Don’t unnecessarily limit future innovation
  • Don’t unnecessarily limit mechanisms of action
  • Don’t unnecessarily limit scope (host, regulatory category, mechanism, site of action, etc.)
  • Require a health benefit on a target host to be demonstrated – otherwise, what is the value of these biotic substances? (Of course, fermented foods were the exception in this criterion, because the value of consuming fermented foods even in the absence of an established health benefit is evident.)
  • Limit to preparations that are administered, not substances produced by in situ activities

In my opinion, many published definitions, including previous ones for postbiotics (see supplementary table here), are untenable because they don’t recognize these principles. There may also be a tendency to rely on historical use of terms, rather than to describe what is justified by current scientific knowledge. A good example of this is provided by the first definition of probiotics, published in 1965. It was “substances secreted by one microorganism that stimulate another microorganism” (Lily and Stillwell, 1965), which is far from the current definition of “live microorganisms that, when administered in adequate amounts, confer a health benefit on the host (Hill et al. 2014).

If you’re looking for a concise summary of the five published ISAPP definitions, see here for our definitions infographic.

Additional reflections: I noted with a smile Glenn’s views on ISAPP, specifically on the appropriate pronunciation of the abbreviation ‘ISAPP’. “My only negative is that everyone involved in the organisation aside from 2 or 3 of us pronounce its acronym wrongly.” Most board members, including myself, have always pronounced this as ‘eye-sap’. Glenn opines, “The abbreviation is not eye-SAPP, it is ISAPP (with the ‘I’ – remarkably enough – being spoken as it is in the word ‘International’).” I wonder how he pronounces IBM?

 

 

 

 

Domestic horses from different geographical locations harbor antibiotic resistant gut bacteria, unlike their wild counterparts

By Dr. Gabriel Vinderola, PhD,  Associate Professor of Microbiology at the Faculty of Chemical Engineering from the National University of Litoral and Principal Researcher from CONICET at Dairy Products Institute (CONICET-UNL), Santa Fe, Argentina

It all started on the 12-hour ferry trip that links Turku with Stockholm during one of the last still warm- summer days of September 2016, when a group of scientists met: Seppo Salminen, Miguel Gueimonde, Carlos Gómez- Galllego and Akihito Endo (joining us virtually from Japan well before the pandemic made these virtual meetings so popular). One of the topics was the possibility of conducting a study comparing the gut microbiome of feral and domestic horses. We had no specific funding for the project but we agreed it would be worthwhile and all agreed to participate.

Misaki wild horses from Cape Toi’s Reserve, Japan. Photo courtesy of Seppo Salminen.

Domesticated horses live under different conditions compared with their wild ancestors. We hypothesized that the animals’ housing, regular veterinary care and feeds would lead to an altered microbiota compared to wild horses. The project was ambitious and challenging in several ways: we aimed at sequencing all microbes, not just bacteria, by using whole genome sequencing; sampling droppings from feral horses needed special permission from the parks or reserves where these horses were held; the project required shipping samples from different parts of the world to the same place where they would be processed; and this all had to be managed without specific financial support to cover the expenses. Curiosity, personal dedication and funding from each end fueled this project.

Little by little, samples of feces of feral and domestic horses were collected in Argentina, Finland, Spain, Russia and Japan. Fecal DNA was extracted in every sampling location and sent to Prof. Li Ang in China for whole genome sequencing and data analysis.  A remarkable contribution was made by Prof. Ang and his team from the Zhengzhou University in China. In his words:

The biggest challenge was that very few sequences (less than 5%) were from known species hosted in the gut of horses. This number is usually 50-60% or 80-90% in human adults or infants gut microbiota, respectively. Thus we had to use ‘old school methods’ to get microbiota profiles, by constructing a custom reference database with whole genomes and then choosing specific alignments, a process that required thousands of computing hours. Interestingly, we found some specific species in horses from different locations. For example, we found shiitake mushroom in Japanese horses, a common food in East Asia families, but not in horses from other locations.

Cimarron wild horses from the State Park Ernesto Tornquist, Argentina. Photo courtesy of Seppo Salminen.

The fecal microbiome of 57 domestic and feral horses from five different locations on three continents were analyzed, observing geographical differences. A higher abundance of eukaryota (p < 0.05) and viruses (p < 0.05) and lower abundance of archaea (p < 0.05) were found in feral animals when compared with domestic ones. The abundance of genes coding for microbe-produced enzymes involved in the metabolism of carbohydrates was significantly higher (p < 0.05) in feral animals regardless of the geographic origin, which may reflect the fact that feral horses are exposed to a much more diverse natural vegetal diet than their domesticated counterparts. Differences in the fecal resistomes between both groups of animals were also observed. The domestic/captive horse microbiomes were enriched in genes conferring resistance to tetracycline, likely reflecting the use of this antibiotic in the management of these animals. Our data also showed an impoverishment of the fecal microbiome in domestic horses with diet, antibiotic exposure and hygiene being likely drivers, a fact that has been also reported for us, humans.

Almost 6 years passed since the results of those ideas discussed on board a ferry slowly galloped into the cover of the February edition of Nature Communications Biology. We hope this will be a starting point for more work that can help uncover the best ways to support equine health.

Mini-tutorial on statistical analysis: Correcting a common misinterpretation of p-values

Daniel Tancredi PhD, Professor of Pediatrics, UC Davis School of Medicine and Center for Healthcare Policy and Research, Sacramento, CA.

Decision makers frequently rely on p-values to decide whether and how to use a study to inform their decisions. Many misinterpret what a low p-value actually means, however. I will attempt to correct this common misinterpretation and explain how to use small p-values to evaluate whether a null hypothesis is plausible. I will show that a low p-value should be used in the same way that a clinician should use a valuable but imperfect clinical diagnostic test result; as one factor, but not the only one, on which to base a decision.

Typically, readers assume that if a p value is low, such as less than 0.05, and thus the test statistic is statistically significant at the conventional level, that there is a good chance that the study results are not “due to chance”. But it is not as simple as that. Let’s suppose that one has a p-value that was generated in a well-designed placebo-controlled randomized clinical trial. We will assume that the trial had a sample size that would provide 80% power to detect what the investigators considered to be the minimum clinical significant difference. The null hypothesis in a trial like this would predict that there is no difference between the placebo and intervention arms for the primary outcome. Once the data were analyzed and reported properly, the p-value was estimated to be just under 0.05. Does this p value less than 0.05 mean that the null hypothesis has no more than a 5% probability of being true? Does it even ensure that the null hypothesis is unlikely? If the calculated p-value was greater than 0.05 (let’s say p=0.10) would that be enough to ensure that the probability of the null hypothesis being true is definitely greater than 5%?

The answer to each of these questions is “no”! This is where, unfortunately, it begins to get (a little) complicated for many users of statistics. The p-value is calculated under the assumption that the null hypothesis is true, and so it does not and cannot measure the probability of that assumption being correct.1 Even though it is common to interpret a p-value as though it is an objective and sufficient statement about the probability that the null hypothesis is true, that is not the case. Statisticians have been trying to communicate this nuance for decades, including the issuance of a statement in 2016 on p-values by the American Statistical Association, a rare statement on statistical practice in that prominent organization’s long history1.

Using a p-value to calculate the probability that the null hypothesis is true

If one wants to use a p-value as one factor in a procedure that can produce a statement about the probability that the null hypothesis is true, one needs to supply an additional input that can be very difficult to obtain. This is a prior (or pre-study) probability, a quantitative estimate of the probability that the null hypothesis is true. This is based on a considered judgment of the state of existing knowledge, what is already known (outside the study results) about how and by how much the intervention may affect the outcomes being assessed.2 Of course, such a judgment can vary a great deal from one individual to the next, according to his/her ability to gather and appraise that knowledge. These judgments can also be influenced by other interests, including financial and ideological, how much of a stake one has in each of the various competing scientific explanations.  Depending on the prior (or pre-experiment) probability for the null hypothesis, a p-value of 0.01 may not be enough to ensure that the null hypothesis has less than a 50% posterior (or after-experiment) probability of being true, whereas a p-value of 0.10 may be enough to make the posterior probability of the null hypothesis be comfortably under 5% (for those interested in taking this discussion further, the final section of this post illustrates this in more detail and with examples).

Determining whether an intervention works

Fundamentally, p-values cannot be used by themselves as though they are objective and reliable ways to make prudent decisions about whether an intervention works. Statisticians emphasize the necessity for the results of individual studies to be interpreted in a broader context, one that involves both statistical judgment and judgment on the underlying scientific plausibility of the hypothesized effects. It is well known that when sample sizes are very large, such as in many observational studies involving tens or even hundreds of thousands of observations, p-values can be very low, even for effect sizes whose confidence intervals are relatively narrow yet do not include any effects that would be of practical importance. In evidence-based medicine we typically face the opposite challenge, where small sample sizes and/or relatively infrequent outcome events result in p-values greater than 0.05 and 95% confidence intervals that are ambiguous because they include the null value (as is implied by p>0.05), but with outcomes that would be very important clinically. Thankfully, in my own career, it does seem to me to have become better appreciated that simply describing studies as positive or negative depending on which side of 0.05 the p-value falls is an unreliable method for evaluating evidence.

p-values and meta-analysis

Another thing to keep in mind is that even when a majority of individual studies that address the same research question may have p-values above 0.05, the meta-analysis of those study results can still indicate a statistically and clinically significant effect. As an example I will use a 2017 Cochrane review of the use of probiotics for the prevention of Clostridioides difficile‐associated diarrhea (CDAD) in adults and children.3 The overwhelming majority of studies, 17 of 21, were supposedly “negative” in that they have confidence intervals that include the null value, but the overall pooled estimate reports a statistically significant and clinically important range of effects. Also note that the overwhelming majority of the studies report confidence intervals that are consistent with the confidence interval for the overall pooled estimate, when one considers the degree of overlap. See Figure 1 below.

Figure 1. Forest plot summarizing complete-case analyses from systematically reviewed clinical trials of probiotics for the prevention of Clostridium difficile‐associated diarrhea (CDAD) in adults and children. Although only 4 of the 31 individual trials had statistically significant results, the pooled estimate shows a statistically and clinically signficant reduction in risk of CDAD for the studied probiotics, without statistically significant heterogeneity among the individual trials’ relative risk estimates. Note that the confidence inferval for the pooled estimate is entirely contained by all but two of the confidence intervals from the individual trials and that even the confidence intervals from these two exceptions largely contain the pooled estimate.

Reprint of Figure 3 from Joshua Z Goldenberg, Christina Yap, Lyubov Lytvyn, et al’s “ Probiotics for the prevention of Clostridium difficile‐associated diarrhea in adults and children”, published December 12, 2017 in “Cochrane Database of Systematic Reviews” by John Wiley and Sons. Copyright by John Wiley and Sons. Reprinted under one-time use license from John Wiley.

Summary

In conclusion, p values are an important component of determining whether an outcome can be deemed to be statistically significant, but this depends on the question under investigation, and is only one part of a more complete analysis. When appraising evidence for whether an intervention works, it is important to keep in mind that if one relies only on statistical inferences from individual studies, one is vulnerable to making unreliable assessments that substantially misstate the plausibility that an intervention does (or does not) have an effect. Statistical analysis cannot replace context-specific scientific judgment; both are needed to make reliable evidence appraisals.

A deeper dive into how to use p-values to assess the probability that the null hypothesis is true

A common misinterpretation of p-values is that they measure the probability that the null hypothesis is true, given the sample data. As stated above, the p-value, by itself, cannot speak to this probability, but if one is willing to supply a judgment on the prior probability that the null hypothesis is true, one can use that and the p-value to get a lower bound on the probability of interest. The compelling figure that accompanies Regina Nuzzo’s terrific Nature article on p-values and their shortcomings nicely illustrates such results for six combinations involving three example prior probabilities and p-values of 0.05 and 0.01.4 Table 1 shows posterior probabilities for those and other input combinations.

 

Table 1. Plausible lower bound for the posterior (post-study) probability of the null hypothesis being true for a given prior (pre-study) probability and study p-value. Note that low p-values do not necessarily imply that the null hypothesis is unlikely to be true!
P-value
Prior Probability for Null 0.1000 0.0500 0.0100 0.0050 0.0010 0.0005 0.0001
5% 3.2% 2.1% 0.7% 0.4% 0.1% 0.1% 0.0%
10% 6.5% 4.3% 1.4% 0.8% 0.2% 0.1% 0.0%
25% 17.3% 12.0% 4.0% 2.3% 0.6% 0.3% 0.1%
50% 38.5% 28.9% 11.1% 6.7% 1.8% 1.0% 0.2%
75% 65.3% 55.0% 27.3% 17.8% 5.3% 3.0% 0.7%
90% 84.9% 78.6% 53.0% 39.3% 14.5% 8.5% 2.2%
95% 92.2% 88.6% 70.4% 57.8% 26.3% 16.4% 4.5%
Note: Calculations use the Bayes Factor – e p ln(p), which is shown to be a lower bound for the Bayes Factor among an appealing set of candidates, thus resulting in a plausible “lower bound” for the posterior probability that the null hypothesis is true. For example, when the prior probability is 50%, a p-value of 0.05 implies that the null hypothesis retains at least a 28.9% probability of being true.

 

The calculations used in that figure and in Table 1 for converting the two inputs, a prior probability for the null hypothesis and a p-value, into a posterior probability for the null hypothesis is simply an application of a much more general formula, one that has been known for over 200 years. This formula is simple to state and remember when expressed as odds. According to Bayes Theorem, Posteriors Odds equals Prior Odds multiplied by a term we call the Likelihood Ratio. The likelihood ratio is a ratio of two conditional probabilities for the observed data, with each computed under differing hypotheses.5 [Another very widely-used application of this general formula is when physicians use the results and the operational characteristics (e.g. the sensitivity and specificity) of clinical tests to inform medical diagnoses.6] The formula uses odds not in the way that they are defined in horse racing where long-shots have high odds, but in the way that statisticians define it, as the ratio of the probability of an event to the probability of the absence of that event. To a statistician, high odds mean high probability for the event. When the probability of an event P is greater than 0, the odds are P / (1 – P). For example, if the probability of an event is 0.75 (or 75%), then the odds would be 0.75 / ( 1 – 0.75 ) = 3. If one knows the odds O, then one can find the probability P, using the equation P = O / ( 1 + O ). For example, if the odds are 4:1, or 4, then the probability is 4/5 = 0.80, and if the odds are 1:4, or 0.25, then the probability is 0.25 / 1.25 = 0.20.

In order to use this long-known formula, one has to have a way to convert the p-value into a value to use for the “Likelihood Ratio” term, which in this context is called a Bayes Factor. For the Nature article, Nuzzo used a conversion proposed in the 1990s by Thomas Sellke, M. J. Bayarri, and James O. Berger and that they eventually published in the widely read American Statistician. That conversion has an appealing statistical motivation as the minimum possible value for the Bayesian Factor among a realistic set of candidates and thus it provides a useful plausible lower bound on the Bayesian Factor for p < 1 /e ≈ 0.368, where e is the Euler number, exp(1) ≈ 2.718,7 BayesFactor = – e * p * ln(p), where ln(p) is the natural logarithm of p. (For p ≥1/e, one can use BayesFactor=1.)  For example, p=0.04 would result in a BayesFactor of -exp(1) * 0.04 * ln( 0.04 ), approximately 0.35. So, if one specified that the prior probability for the null hypothesis is 50%, a toss-up, that corresponds to a prior odds of 1, then the BayesFactor for a p-value of 0.04 converts that prior odds of 1 into a posterior odds of 0.35, which corresponds to a posterior probability of 26% for the null hypothesis, substantially higher than 4%. In the analogous setting of diagnostic medicine, consider a test result that moves a physician’s suspicion for whether the patient has a disease from a pre-test value of 50% up to a post-test value of 74%. Such a result would be considered useful, but it would not be considered definitive, something for clinicians to keep in mind when they see that a study’s p-value was just under 0.05!

Another notable conversion of the p-value into a Bayes Factor was, as far as I can tell, first reported in a pioneering 1963 article in the social sciences literature that was authored by illustrious Bayesian statisticians. 8 That same Bayes Factor formula can be found clearly presented in the second5 of Steven N. Goodman’s excellent two-part set of Annals of Internal Medicine articles concerning fallacious use of p-values in evidence-based medicine. That conversion involves statistics that have an approximately normal distribution and is thus applicable to most statistics in the medical literature. That conversion reports the minimum theoretically possible value for the Bayes Factor, BayesFactormin = exp( – Z2 / 2 ), where Z is the number of standard errors the test statistic is from the null value.  (Z can be estimated in Microsoft Excel by using the formula Z = NORMSINV( p ) or Z = NORMSINV( p / 2 ). For example, a two-sided p-value of 0.04 corresponds to Z ≈ -2.054 and a BayesFactormin of exp( – (-2.054 * -2.054) / 2 ) ≈ 0.121. So, if the prior probability for the null hypothesis is 50%, a p-value of 0.04 would mean that, at the minimum, the null hypothesis has a posterior probability of 0.121 / 1.21 = 10.8% of being true, substantively higher than the 4% probability that the popular misinterpretation of p-values would yield. When that factor was introduced in the 1963 article, it was noted by the authors as not being one that would be realistically attained by any study, as it would involve an impossibly lucky guess for the best possible prior probability to use, but it is still useful mathematically because it results in a theoretical minimum for the posterior probability that the null hypothesis is true. In mathematics, we routinely use well-chose impractical scenarios to define the limits for what is practically possible. Given that decisionmakers want to know how probable the null hypothesis remains in light of the study data, it is helpful to know the minimum possible theoretical value for it. Table 2 shows these posterior probabilities for the same inputs used above in Table 1. Notably, a p-value of 0.05 may not even be enough to make the null hypothesis less likely than not!

P-value
Prior Probability for Null 0.1000 0.0500 0.0100 0.0050 0.0010 0.0005 0.0001
5% 1.3% 0.8% 0.2% 0.1% 0.0% 0.0% 0.0%
10% 2.8% 1.6% 0.4% 0.2% 0.0% 0.0% 0.0%
25% 7.9% 4.7% 1.2% 0.6% 0.1% 0.1% 0.0%
50% 20.5% 12.8% 3.5% 1.9% 0.4% 0.2% 0.1%
75% 43.7% 30.5% 9.8% 5.5% 1.3% 0.7% 0.2%
90% 69.9% 56.9% 24.6% 14.9% 3.9% 2.1% 0.5%
95% 83.1% 73.6% 40.8% 27.0% 7.8% 4.3% 1.0%
Note: For 2-sided p-values based on approximately normally distributed test statistics, using the mathematically lowest theoretically possible Bayesian Factor,5,8 thus ensuring the lowest possible value for the posterior probability for the null hypothesis. Although these lower bonds would never be attained in any realistic application, this table is useful in showing the smallest null hypothesis probability that is even theoretically possible. Note that even with a p-value of 0.05, the posterior probability for the null hypothesis may still be high.

References

  1. Wasserstein RL, Lazar NA. The ASA Statement on p-Values: Context, Process, and Purpose. The American Statistician. 2016;70(2):129-133.
  2. Goodman SN. Toward evidence-based medical statistics. 1: The P value fallacy. Annals of Internal Medicine. 1999;130(12):995-1004.
  3. Goldenberg JZ, Yap C, Lytvyn L, et al. Probiotics for the prevention of Clostridium difficile-associated diarrhea in adults and children. Cochrane Database Syst Rev. 2017;12:CD006095.
  4. Nuzzo R. Statistical Errors. Nature. 2014;506(7487):150-152.
  5. Goodman SN. Toward evidence-based medical statistics. 2: The Bayes factor. Annals of Internal Medicine. 1999;130(12):1005-1013.
  6. Deeks JJ, Altman DG. Diagnostic tests 4: likelihood ratios. Bmj. 2004;329(7458):168-169.
  7. Sellke T, Bayarri MJ, Berger JO. Calibration of p values for testing precise null hypotheses. Am Stat. 2001;55(1):62-71.
  8. Edwards W, Lindman H, Savage LJ. Bayesian Statistical-Inference for Psychological-Research. Psychol Rev. 1963;70(3):193-242.

Bacterial genes lead researchers to discover a new way that lactic acid bacteria can make energy and thrive in their environments

Lactic acid bacteria are an important group of bacteria associated with the human microbiome. Notably, they are also responsible for creating fermented foods such as sauerkraut, yogurt, and kefir. In the past two decades, culture-independent techniques have allowed scientists to sequence the genomes of these bacteria and discover more about their capabilities.

Researchers studying a type of lactic acid bacteria called Lactiplantibacillus plantarum found something unexpected: they contained genes for making energy in a way that had not been previously documented. Generally, living organisms obtain energy from their surroundings either by fermentation or respiration. L. plantarum have long been understood to obtain energy using fermentation, but the new genetic analysis found they had additional genes that were suited to respiration. Could they be using both fermentation and respiration?

ISAPP board member Prof. Maria Marco is a leading expert on lactic acid bacteria and their role in fermented foods and in human health. In her lab at University of California Davis, she decided to investigate why L. plantarum had genes equipping it for respiration. Her group recently published findings that show a new type of “hybrid” metabolism used by these lactobacilli.

Here is a Q&A with Prof. Marco about these exciting new findings.

What indicated to you that some of the genes in L. plantarum didn’t ‘belong’?

Organisms that use respiration normally require an external molecule that can accept electrons, such as oxygen. Interestingly, some microorganisms can also use solid electron acceptors located outside the cell, such as iron. This ability, called extracellular electron transfer, has been linked to proteins encoded by specific genes. L. plantarum had these genes, even though this species is known to use fermentation. We first learned about their potential function from Dr. Sam Light, now at the University of Chicago. Sam discovered a related pathway in the foodborne pathogen Listeria monocytogenes. Sam came across our research on L. plantarum because we previously published a paper showing that a couple of genes in this pathway are switched on in the mammalian digestive tract. We wondered what the proteins encoded by these genes were doing.

How did you set out to investigate the metabolism of these bacteria?

We investigated this hybrid metabolism in a variety of ways. Using genetic and biochemical approaches we studied the extent to which L. plantarum and other lactic acid bacteria are able to use terminal electron acceptors like iron. Our collaborators at Lawrence Berkeley National Lab and Rice University contributed vital expertise with their electrochemistry experiments, including making fermented kale juice in a bioelectrochemical reactor.

What did you find out?

We discovered a previously unknown method of energy metabolism in Lactiplantibacillus plantarum. This hybrid strategy blends features of respiration (a high NAD+/NADH ratio and use of a respiratory protein) with features of fermentation (use of endogenous electron acceptors and substrate-level phosphorylation).

We verified that this hybrid metabolism happens in different laboratory media and in kale juice fermentations. We also found that, in the complex nutritive environment of a kale juice fermentation, this hybrid metabolism increases the rate and extent of fermentation and increases acidification. Within the ecological context of the fermented food, this could give L. plantarum a fitness advantage in outcompeting other microorganisms. This could potentially be used to change the flavor and texture of fermented foods.

This discovery gives us a new understanding of the physiology and ecology of lactic acid bacteria.

Are there any indications about whether this energy-making strategy is shared by other lactic acid bacteria?

Some other fermentative lactic acid bacteria also contain the same genetic pathway. It is likely that we are just at the tip of the iceberg learning about the extent of this hybrid metabolism in lactobacilli and related bacteria.

Your finding means there is electron transfer during lactic acid bacteria metabolism. What does this add to previous knowledge about bacterially-produced ‘electricity’?

Certain soil and aquatic microbes have been the focus of research on bacterially-produced electricity. We found that by giving L. plantarum the right nutritive environment, it can produce current to the same level as some of those microbes. We believe there is potential to apply the findings from our studies to better inform food fermentation processes and to guide fermentations to generate new or improved products. Because strains of L. plantarum and related bacteria are also used as probiotics, this information may also be useful for understanding their molecular mechanisms of action in the human digestive tract.

How might this knowledge be applied in practice?

Our findings can lead to new technologies which use lactic acid bacteria to produce healthier and tastier fermented foods and beverages. Because this hybrid metabolism leads to efficient fermentation and a larger yield, it could also help minimize food waste. We plan to continue studying the diversity, expression, and regulation of this hybrid metabolism in the environments in which these bacteria are found.

ISAPP awards the Glenn Gibson Early Career Research Prize to two diet and gut health researchers

The ISAPP board of directors is pleased to announce that the 2022 Glenn Gibson Early Career Research Prize has been awarded to two promising researchers in the field of probiotics, prebiotics and related substances.

Dr. Martin Laursen, Senior Researcher at the National Food Institute, Technical University of Denmark, has demonstrated excellence in his work on the impact of probiotics and human milk oligosaccharides on infant gut microbiota and health. Dr. Eirini Dimidi, Lecturer at King’s College in London, UK, has carried out meaningful work on probiotics, prebiotics, and fermented foods and their impact on constipation.

The award criteria stipulated that the researchers must be fewer than five years from their terminal degree, and their scope of research must be basic or clinical research disciplines in the fields of probiotics, prebiotics, synbiotics, postbiotics or fermented foods. In addition, the researchers were required to show evidence of a significant research finding and its publication(s), new ideas that advance the field, and / or evidence of impact through citizenship, general outreach, social media or other means.

The prize committee chose the two recipients from among dozens of applicants and identified each of them as having made important contributions to the field at this early stage in their scientific careers. Each winner will receive a cash prize and an opportunity to speak at the ISAPP annual meeting, to be held in Spain in June, 2022.

Stay tuned to learn more about these rising star researchers!

See here for details about the 2022 Glenn Gibson Early Career Research Prize

Do fermented foods contain probiotics?

By Prof. Maria Marco, PhD, Department of Food Science & Technology, University of California, Davis

We frequently hear that “fermented foods are rich in beneficial probiotics.” But is this actually true? Do fermented foods contain probiotics?

The quick answer to this question is no – fermented foods are generally not sources of probiotics. Despite the popular assertion to the contrary, very few fermented foods contain microbes that fit the criteria to be called probiotic. But this fact does not mean that fermented foods are bad for you. To uphold the intent of the word probiotic and to explain how fermented foods actually are healthy, we need to find better ways to describe the benefits of fermented foods.

Probiotics are living microorganisms, that when administered in adequate amounts, confer a health benefit on the host (Hill et al 2014 Nat Rev Gastroenterol Hepatol). This current definition reflects minor updates to a definition offered by an expert consultation of scientists in 2001 convened by the Food and Agriculture Organization of the United Nations (FAO) and the World Health Organization. Evident from the definition, a microbial strain is not a probiotic unless a health benefit has been found with its use. At a minimum, the strain should be proven to be beneficial in at least one randomized controlled trial (RCT). Probiotics must also be defined at the strain level through genome sequencing (a strain is a single genotype of a species).

Fermented foods, on the other hand, have no requirement to improve health. Fermented foods are foods and beverages made through desired microbial growth and enzymatic conversion of food components. This definition was recently formulated by an ISAPP consensus panel of scientific experts to affirm the common properties of all foods of this type and to differentiate foods that may look or taste similar but are not made using microbes (Marco et al 2021 Nat Rev Gastroenterol Hepatol). Fermented foods encompass an expansive variety of foods made from animal and plant sourced ingredients and produced from all types of microbial metabolism. The desired characteristics of these foods are frequently how they look, smell, and taste. There no expectation in this definition that fermented foods alter health in any way.

There is also no requirement for fermented foods contain living microbes at the time they are ingested. Foods such as bread, chocolate, and beer are fermented but then are baked, roasted, and/or filtered. This means those fermented foods cannot be probiotic.

Some fermented foods, such as kimchi and kombucha, are typically eaten with living microbes present. However, the microbes in those foods usually do not meet the criteria to be called probiotic. Whether the fermented food was made at home or purchased from the supermarket, studies investigating whether the microbes in those fermented foods are specifically responsible for a health benefit remain to be done. Those foods also do not contain microbes defined to the strain level, nor is the number of living microbes typically known. An exception to this is if specific strains previously shown to provide a health benefit in one or more RCT are intentionally used in the production of the food and remain viable at expected numbers over the shelf-life of that fermented food product. An example of this would be a commercial fermented yogurt that has an added probiotic strain remaining viable at the time of consumption, beyond the strains that carried out the fermentation.

Despite these distinctions between probiotics an fermented foods, the probiotics term has pervaded common lexicon to mean “beneficial microbes”. In contrast to pathogenic or harmful microbes, beneficial microbes are those that are understood to help rather than hurt bodily functions. However, just as we do not assume that all pathogens cause the same disease or result in the same severity of symptoms, we should also not expect that beneficial microbes all serve the same purpose. By analogy, automobiles are useful vehicles which help us to get from place to place. We do not expect that all automobiles perform like those used for Formula 1 racing. Microbes are needed to make fermented foods and may be beneficial for us, but we should not assume that those drive health benefits like established probiotic strains.

What are the consequences of calling fermented foods probiotic when they include undefined numbers of living microbes for which strain identities are not known? One can suppose that there is no harm in labeling or describing those products as “probiotic” or “containing probiotics”. However, by doing so, confusion and misunderstanding is created and too often, spread by journalists, nutritionists, scientists, and medical professionals. For example, news articles in reputable sources have written that foods like kefir, kimchi, sauerkraut made from beets or cabbage, pickles, cottage cheese, olives, bread and chocolate are rich in probiotics. As misuse perpetuates, what becomes of bona fide probiotics shown with rigorous study to benefit health, such as reducing the incidence and duration of diarrhea or respiratory infections? It becomes difficult to know which strains have scientific proof of benefit. Just as there are laws for standards of food identity, we should strive to do the same when describing microbes in fermented foods.

Avoiding the term probiotic when describing fermented foods should not stop us from espousing the myriad of positive attributes of those foods. Besides their favorable sensory qualities, fermented foods are frequently safer and better tolerated in the digestive tract than the foods they are made from. During the production of fermented foods, microbes remove or reduce toxins in the ingredients and produce bioactive compounds that persist long after the microbes that make them are gone.

Even though the living microbes in fermented foods may not rise to the standard of a probiotic, they may provide health benefits. We just don’t have the studies to prove that they do. With more study, we may find that viable microbes in fermented foods work similarly to probiotics in the digestive tract through shared mechanisms. This is already known for yogurts. Yogurt cultures share the ability to deliver lactase to the intestine, thereby improving tolerance of lactose by intolerant individuals. Clinical and epidemiological studies performed on fermented foods already suggest an association between them and different health benefits but as we recently explained (Marco et al 2021 J Nutrition), more work is needed in order to understand if and what benefits these microbes provide.

For now, we should simply continue enjoying the making and eating of fermented foods and reserve the term probiotics for those specific microbial strains which have been shown to improve our health. Marketers should resist labeling products as containing probiotics if their products do not meet the criteria for a probiotic. Indeed, the descriptor “live and active cultures” more accurately reflects the microbial composition of many fermented foods, and should be used until controlled human trials demonstrating health benefits are conducted.

 

Additional resources:

How are probiotic foods and fermented foods different? ISAPP infographic.

Fermented foods. ISAPP infographic.

What are fermented foods? ISAPP video.

Are fermented foods probiotics? Webinar by Mary Ellen Sanders, PhD.

 

Pasteurized Akkermansia muciniphila as a postbiotic: EFSA approval and beyond

By Prof. Seppo Salminen, University of Turku, Finland

Earlier this year, the European Food Safety Authority (EFSA) delivered an opinion that heat-treated Akkermansia muciniphila is safe for use as a novel food in the European Union. EFSA described A. muciniphila as a “well‐characterised non‐toxin producing, avirulent microorganism that has been reported as part of normal gut microbiota” and determined based on a literature review that its safety is adequate for use as a food supplement or in foods for special medical purposes, at a specified maximum dose.

ISAPP connected with three individuals from A-Mansia Biotech, the company that initiated the EFSA request: Prof. Willem M. de Vos and Prof. Patrice D. Cani, as well as the company CEO Michael Oredsson. They jointly answered some questions on their EFSA success and plans for the future.

Originally, what led you to test whether the pasteurized form of the live microbe might be able to confer a health benefit?

We first noticed that killing Akkermansia by using autoclaving (121°C 20°C) completely abolished the beneficial effects of Akkermansia. However, we wanted to test whether a milder procedure (i.e. pasteurization) could keep some structures of the outer membrane of Akkermansia intact and therefore still able to interact with the host. We knew that several other classical probiotics (types of lactobacilli) partly retained their effects after pasteurization. Our surprise was to see that pasteurization successfully maintained the effects of Akkermansia compared to the live form, but even increased its efficacy.

Pasteurised Akkermansia has now been extensively studied for safety and health effects. Does this make it the first real postbiotic, as defined by ISAPP?

If we are accepting the ISAPP definition proposed in 2021, we can answer yes to this question. Prof. Cani in his scientific capacity believes indeed that the product (pasteurized Akkermansia) is unique and can fall under this definition. Whether A-Mansia will be positioning the pasteurized Akkermansia as a postbiotic according to that definition is still to be discussed.

Pasteurised Akkermansia has been demonstrated to control gut barrier and reduce inflammation associated with fat storage and obesity – will we see a product that helps in weight loss/control?

Akkermansia is clearly playing a major role by tackling the gut barrier dysfunction which is the root cause of the different metabolic problems mentioned here (i.e., inflammation, fat storage, liver/fat tissue inflammation) and they are all connected to better energy expenditure/oxidation when a lower inflammation/insulin resistance is observed. Therefore, pasteurized Akkermansia should help to maintain a healthy weight and abdominal fat. A product focusing on a better weight management is currently under development at A-Mansia.

Is the next step to apply for an EU health claim?

All the current human investigations and studies at our company are aiming at fulfilling future EU health claims.

It took two years to get the acceptance for the safety of inanimate pasteurised Akkermansia – what do you think of this timeframe for safety assessment?

This is perfectly in line with what the EFSA was expecting, although it was a few months delayed with the COVID-19 crisis. The assessment was very clear, smooth and well managed by the EFSA.

In general, what do you think the future holds for postbiotics as food ingredients?

We are entering into a new era, first with next-generation beneficial bacteria, and Akkermansia as one of the most studied (if not the most studied). The pasteurized form is so active, stable, and easy to use that the postbiotic era, as led by this example, is a novel and innovative manner of targeting the microbiome for improving/maintaining health.

 

As the science on health benefits for similar postbiotic substances continues to advance, we may see more ingredients qualifying as true postbiotics. More products are likely to follow a similar path, considering the practical advantages of delivering non-living substances to consumers.

 

ISAPP’s 2021 year in review

By Mary Ellen Sanders, PhD, ISAPP Executive Science Officer

The upcoming year-end naturally leads us to reflect about what has transpired over the past 12 months. From my perspective working with ISAPP, I witnessed ISAPP board members and the broader ISAPP community working creatively and diligently to find solutions to scientific challenges in probiotics, prebiotics and related fields. Let’s look back together at some of the key developments of 2021.

ISAPP published outcomes from two consensus panels this year, one on fermented foods and one on postbiotics. The popularity of these articles astounds me, with 49K and 29K accesses respectively, as of this writing. I think this reflects recognition on the part of the scientific community of the value – for all stakeholders – of concise, well-considered scientific definitions of terms that we deal with on a daily basis. If we can all agree on what we mean when we use a term, confusion is abated and progress is facilitated. The postbiotics definition was greeted with some resistance, however, and it will remain to be seen how this is resolved. But I think ISAPP’s response about this objection makes it clear that productive definitions are difficult to generate. Even if the field ultimately embraces another definition, it is heartening to engage in scientific debate about ideas and try to find alignment.

Keeping with the idea of postbiotics, a noteworthy development this year was the opinion from the European Food Safety Authority that the postbiotic made from heat-treated Akkermansia muciniphila is safe for use as a novel food in the EU. Undoubtedly, this development is a bellwether for likely future developments in this emerging area as some technological advantages to postbiotics will make these substances an attractive alternative to probiotics IF the scientific evidence for health benefits becomes available.

Recognizing the existing need for translational information for clinicians, ISAPP developed a continuing education course for dietitians. Published in March, it has currently reached close to 6000 dietitians. This course focused on probiotics, prebiotics and fermented foods: what they and how they might be applied in dietetic practice. It is a freely available, self-study course and completion provides two continuing education credits for dietitians.

On a sad note, in March of this year, ISAPP suffered the loss of Prof. Todd Klaenhammer. Todd was a founding ISAPP board member, and directed many of our activities over the course of his 18-year term on the board. He was also my dear friend and major advisor for my graduate degrees at NC State many years ago.  As one former collaborator put it, “I was not prepared to finish enjoying his friendship and mentorship.” See here for a tribute to Prof. Klaenhammer on the ISAPP blog: In Memoriam: Todd Klaenhammer.

So where will 2022 lead ISAPP? The organization has now published five consensus definitions: probiotics, prebiotics, synbiotics, postbiotics and fermented foods – extending its purview beyond where it started, with probiotics and prebiotics. Through the year ahead, ISAPP is committed to providing science-based information on the whole ‘biotics’ family of substances as well as fermented foods. Our Students and Fellows Association is growing, supported by the opportunity for young scientists to compete for the Glenn Gibson Early Career Researcher Prize. We continue to see our industry membership expand. Through our new Instagram account and other online platforms, our overall community is increasing. The ISAPP board of directors continues to evolve as well, with several long-term members leaving the board to make room for younger leaders in the field who will direct the future of the organization. This applies to me as well, as I have made the difficult decision to depart ISAPP in June of 2023. Thus, hiring a new executive director/executive science officer is an important priority for ISAPP in 2022. My 20 years with ISAPP have seen the organization evolve tremendously, through the hard work of incredible board members as well as many external contributors. We will strive to make 2022 – our 20th anniversary – ISAPP’s best year yet.

ISAPP board members give a scientific overview of synbiotics in webinar

Many kinds of products are labeled as synbiotics – but how do they differ from each other? And do they all meet the scientific criteria for synbiotic ingredients?

To demystify the science of synbiotics – including ISAPP’s definition published in 2020 – ISAPP is holding a free webinar: Synbiotics: Definitions, Characterization, and Assessment. Two ISAPP board members, Profs. Bob Hutkins and Kelly Swanson, present on the implications of the synbiotic definition for science and industry. They clarify the difference between ‘complementary’ and ‘synergistic’ synbiotics and cover the basics of meeting the criteria for synbiotic efficacy and safety. One challenge is learning when a synbiotic is required to have demonstrated both selective utilization of the microbiota in the same study that measures the health outcome. A Q&A is scheduled for the last 20 minutes of the webinar.

This webinar is for scientists, members of the public, and media who want a scientific overview on synbiotics as they appear in more and more consumer products.

The live webinar was broadcast on Friday, January 28th, 2022, from 10:00 am – 11:10 New York (Eastern) time.

Find the webinar recording here.

Research on the microbiome and health benefits of fermented foods – a 40 year perspective

By Prof. Bob Hutkins, PhD, University of Nebraska Lincoln, USA

Many ISAPPers remember when fermented foods attracted hardly any serious attention from scientists outside the field. Certainly, most clinicians and health professionals gave little notice to fermented foods. In the decades before there were artisan bakeries and microbreweries proliferating on Main Street USA, even consumers did not seem very interested in fermented foods.

When I began my graduate program at the University of Minnesota in 1980, I was very interested in microbiology, but I did not know a lot about fermented foods. Accordingly, I was offered two possible research projects. One involved growing flasks of Staphylococcus aureus, concentrating the enterotoxins, feeding that material to lab animals, and then waiting for the emetic response.

My other option was to study how the yogurt bacterium, Streptococcus thermophilus, metabolized lactose in milk. This was the easiest career choice ever, and the rest, as they say, is history.

Indeed, that lab at Minnesota was one of only a handful in North America that conducted research on the physiology, ecology, and genetics of microbes important in fermented foods. Of the few labs in North America delving into fermented foods, most emphasized dairy fermentations, although some studied vegetable, meat, beer, wine, and bread fermentations. Globally, labs in Europe, Japan, Korea, Australia, and New Zealand were more engaged in fermented foods research than we were in North America, but overall, the field did not draw high numbers of interested researchers or students.

That’s not to say there weren’t exciting and important research discoveries occurring. Most research at that time was focused on the relevant functional properties of the microbes. This included carbohydrate and protein metabolism, flavor and texture development, tolerance to acid and salt, bacteriocin production, and bacteriophage resistance. Despite their importance, even fewer labs studied yeasts and molds, and the focus was on lactic acid bacteria.

Other researchers were more interested in the health benefits of fermented foods. Again, yogurt and other cultured dairy foods attracted the most interest. According to PubMed, there were about 70 randomized clinical trials (RCTs) with yogurt as the intervention between 1981 and 2001. Over the next 20 years, there were more than 400 yogurt RCTs.

Fast forward a generation or two to 2021, and now fermented foods and beverages are all the rage. Certainly, having the molecular tools to sequence genomes and interrogate entire microbiomes of these foods has contributed to this new-found interest. Scanning the recent literature, there are dozens of published papers on microbiomes (and metabolomes) of dozens of fermented foods, including kombucha (and their associated symbiotic cultures of bacteria and yeast, known as SCOBYs), kefir, kimchi, beer (and barrels), cheese (and cheese rinds), wine, vinegar, miso and soy sauce, and dry fermented sausage.

It’s not just fermentation researchers who are interested in fermented foods. For ecologists and systems biologists, fermented foods serve as model systems to understand succession and community dynamics and how different groups of bacteria, yeast, and mold compete for resources.

Moreover, consumers can benefit when companies that manufacture fermented foods take advantage of these tools. The data obtained from fermented food microbiota analyses can help to correlate microbiome composition to quality attributes or identify potential sources of contamination.

Importantly, it is also now possible to screen microbiomes of fermented foods for gene clusters that encode potential health traits. Indeed, in addition to microbiome analyses of fermented foods, assessing their health benefits is now driving much of the research wave.

As mentioned above, more than 400 yogurt RCTs were published in the past two decades, but alas, there were far fewer RCTs reported for other fermented foods. This situation, however, is already changing. The widely reported fiber and fermented foods clinical trial led by Stanford researchers was published in Cell earlier this year and showed both microbiome and immune effects. Other RCTs are now in various stages, according to clinicaltrials.gov.

Twenty years ago, when ISAPP was formed, I suspect few of us would have imagined that the science of fermented foods would be an ISAPP priority. If you need proof that it is, look no further than the 2021 consensus paper on fermented foods. It remains one of the most highly viewed papers published by Nature Reviews Gastroenterology and Hepatology.

Further evidence of the broad interest in fermented foods was the recently held inaugural meeting of The Fermentation Association. Participants included members of the fermented foods industry, culture suppliers, nutritionists, chefs, food writers, journalists, retailers, scientists and researchers.

Several ISAPP board members also presented seminars, including this one who remains very happy to have made a career of studying fermented foods rather than the emetic response of microbial toxins.

Lactobacilli dominate the vagina in Belgian women

By Prof. Sarah Lebeer, Research Professor in Microbiology and Molecular Biology, Department of Bioscience Engineering, University of Antwerp, Belgium

A little over a year ago, I wrote an ISAPP blog post about the setup of our Isala citizen science project on women’s health. Now, I can proudly say that we have the first results. Last year, more than 3300 women sent vaginal samples back to our lab, not only from the big cities but also from the smallest villages all over Flanders, Belgium (Figure 1). While Prof. Jack Ravel and many other colleagues have already done pioneering work in the US (e.g., Ravel et al. PNAS & Valencia study), Estonia and Africa, the vaginal microbiome of healthy women was less well mapped in the region where we live in Western Europe (Flanders, Belgium).

Figure 1: Map of Flanders (Belgium) showing regions from which the Isala participants sent their samples, with a gradient for the number of participants.

Last year, we managed to inspire women from a wide age range to donate two vaginal self-sampled swabs: the youngest participants were 18 years old, while a woman of 98 years old even participated. Each participant of Isala showed a unique vaginal microbiome (Figure 2).

Figure 2. Bar chart showing that each Isala participant had a unique vaginal microbiome composition, but also that lots of parallels could be drawn based on the most dominant bacterium.

Through various analyses, we were able to find parallels between the vaginal profiles of the Isala participants. We decided to divide the women in eight groups based on their most dominant microbe. Lactobacillus crispatus was found in 43% of all Isala women as most dominant bacterium, Lactobacillus iners in 28%, Lactobacillus jensenii in 4%, Lactobacillus gasseri in 3%, Gardnerella vaginalis in 12%, Prevotella in 6%, Bifidobacterium in 2% and Streptococcus in 2% of all Isala participants (Figure 3). Last June 2021, all women received this information, with a nice drawing for each bacterium and some interesting facts about these bacteria, as well as the relative abundance of this top 8. (See here.)

Figure 3. Chart showing the proportion of women participating in the Isala project that have a vaginal microbiota dominated by different bacterial genera or species.

Our work has only just begun. My team (see photo below) is now analyzing all the metadata collected via the detailed questionnaires and associating them with these microbiome profiles. The impact of the menstrual cycle, hormonal fluctuations, diet, smoking, sexual activity and other relevant factors is currently being explored. Hopefully, this will allow us to better understand for the vaginal tract what a ‘healthy microbiome’ really is and what action women can take to obtain or preserve  a ‘healthy’ or resilient microbiome. This is challenging to define with our current state of knowledge, but one characteristic of health of the microbiome may be its resilience. At the next annual ISAPP meeting, Karen Scott and I will co-chair a discussion group on ‘What do we really know about the microbiome and health?’. Now, I think it is fair to say that, compared to the gut, associations between specific microbiome members, such as lactobacilli, and health are quite strong for the vaginal tract. These lactobacilli form a protective barrier, are able to keep pathogens out, and prevent overt inflammation, so we could define lactobacilli-dominated vaginal communities as being resilient to many infections and disorders and thus probably ‘healthy’.

However, there is still much we do not know. Can women make certain changes in their lifestyle, diet, anticonception, underwear material etc. to promote lactobacilli such as L. crispatus in their vagina? What are the consequences of normal events in live such as pregnancy and menopause on these lactobacilli? Is a vaginal community with less lactobacilli always less healthy or resilient? On this page, you can get an overview of the different aspects we want to investigate. We hope to submit the first big Isala manuscript by the end of this year and will inform you as soon as possible about the results.

Lebeer lab, University of Antwerp

Scientists looking at a bottle of probiotic supplements.

Current issues in probiotic quality: An update for industry

By Dr. Mary Ellen Sanders, ISAPP, Dr. Kit Goldman, USP, Dr. Amy Roe, P&G, Dr. Christina Vegge, Dr. Jean Schoeni, Eurofins

With probiotic dietary supplement use growing globally and an increasing array of products on the market, probiotic quality is an issue of perpetual relevance to industry. Best practices for producing high-quality probiotics change frequently, making it important for companies to stay informed.

ISAPP convened a webinar on this topic, available to ISAPP members only. The webinar took place November 16, 2021, and was hosted by Executive Science Officer, Dr. Mary Ellen Sanders. Speakers focused on the activities of the United States Pharmacopoeia (USP), a non-profit organization based in the US and operating globally, which for the past 200 years has worked to improve public health through development of quality standards for medicines, dietary supplements and foods. In 2017 USP formed an Expert Panel on probiotics.

Dr. Kit Goldman, Sr Director, Dietary Supplements and Herbal Medicines, USP, spoke about the origin of USP and the USP activities related to probiotic quality. USPs expert volunteers have determined the necessary parameters for probiotic quality standards, which include tests for identification, assay/enumeration and contaminants, and have created standards for a number of probiotic species/strains. In the course of doing so, the Probiotics Expert Panel identified specific areas where more information was needed to fully understand issues related to probiotic quality. This led to the formation of sub-teams to consider aspects of probiotic identification, enumeration and safety.

Dr. Amy Roe, Principal Scientist at P&G, spoke on appropriate regulatory requirements for probiotic safety. Currently, there is no global harmonization on the requirements for establishing probiotic safety for use in foods and supplements. Although ‘history of safe use’ has been central to safety assessments for many current probiotic species, probiotic manufacturers are increasingly seeking to use new strains, species, and next-generation probiotics; justification of safety based on a significant history of use may be challenged. USP and other stakeholders are looking to develop best practices guidelines for assessing the quality and safety of probiotics. A current initiative of the USP seeks to provide expert advice specific to safety considerations for probiotics through reviewing global regulatory guidelines, evaluating appropriateness of traditional animal toxicology studies for studying the safety of probiotics, highlighting the importance of proper manufacturing practices with regard to final product safety, and outlining of essential parameters of a comprehensive safety assessment for a probiotic.

Dr. Jean Schoeni, Fellow at Eurofins, spoke on comparing probiotic enumeration methods. One challenge faced by the USP Probiotics Expert Panel is how to compare the increasing number of probiotic enumeration methods appearing in monograph submissions. A sub-team of the panel developed a solution that combines APLM (Analytical Procedures Lifecycle Management – a streamlined approach for determining the method’s fitness for intended use) with TI (tolerance interval) calculations. Schoeni encouraged companies to adopt this solution, highlighting tools that have been provided to the probiotics industry through publication of the sub-team’s work.

Dr. Christina Vegge spoke on quantification of multi-strain blends. For probiotic products comprising multiple strains, the viable numbers of each strain in these products would ideally be quantified. However, reliance on plate count methods creates analytical challenges regardless of whether the quantification of viable numbers of each strain in the blend is conducted prior to or after blending. Further challenges arise when addressing the reductions in potency over shelf life of the product. For multi-strain products, plate count procedures are insufficient—and currently no official guideline or general best practice exists to resolve this situation. Therefore, the USP Probiotics Expert Panel wants to conduct an explorative study to examine non-culture based technologies to quantify the viable composition of multi-strain blends.

A recording of this webinar is available for ISAPP industry members only. Please see here and email info@nullisappscience.org for the password to access this page.

Publications (open access) from USP Probiotics Expert Panel:

Jackson et al. Improving End-User Trust in the Quality of Commercial Probiotic Products. Front Microbiol. 2019 Apr 17;10:739.  doi: 10.3389/fmicb.2019.00739.

Weitzel MLJ, et al. Improving and Comparing Probiotic Plate Count Methods by Analytical Procedure Lifecycle Management. Front Microbiol. 2021 Jul 12;12: 693066. doi: 10.3389/fmicb.2021.693066.