SEMDAR

Sustainable Enteric Microbiome Diversity Amelioration & Restitution

Microbiota Integrated Symbiotic Therapeutics (MIST)

http://bit.ly/semdar69

While there exists no current marker for assessing and monitoring the concept of a flourishing microbiome which is herein being called Sustainable Enteric Microbiome Diversity Amelioration and Restitution (SEMDAR) its existence can be inferred from the health outcome. By using the example of fermented non-dairy food item collectively called “Sauerkrautes” use and for ease of communication and comprehension the concept of SEMDAR would be used interchangeably and synonymously with “Sumptuous Sauerkraut Effect”. (sumptuous being synonymous with lavish, splendid, opulent, magnificent, elaborate, spectacular etc.). This connotes consumption of very generous servings and fairly regularly and not just a token half a bite of the item. Restitution is appended to the microbiome diversity amelioration to connote that resilience of the gut microbiome gets enhanced when like the healed strained and exercise torn muscle fibres and bone septa becoming stronger post-shock microbiome when restored and repaired becomes more resilient with resilient microbiome defined by exhibiting a higher tolerance to adverse environmental conditions like including pH, thermal shocks etc.. . Monotony or homogeneity of the enteric microbiome tends to produce adverse health impacts possibly through amplification of inflammaging, stress level, sleep disruption and worsening of the symptoms of the coexisting health conditions such that escaping microbiome homogeneity should be made a very high priority for wellness amelioration. Noteworthy is the connections of SEMDAR to the following important health marker influencers


1. Genetically determined propensities

2. Epigenomic structure (innate) and status (lifestyle dependent)

3. Enteric brain or enteric encephalon synchronizing with the cephalic encephalon.

4. Ambient baseline level of stress or chronic anxiety state being experiences.

5. Inflammaging intensity (CAIRO) and SEMDAR bidirectional relationship or dance.

Microbiome Diversity and health is extremely fluid and shifts within 48 hours and is in need of a continuous monitoring, maintenance and if damaged it needs rapid restitution or restoration. The positive nuance of this fluid nature of the microbiome health is that by adopting a probiotic lifestyle restoration of wellness can be accomplished speedily as well.


Limitations of returns

Limitations of returns from adoption of Lifespan Enhancing Enteric Eubiosis (LEEE) strategies or attempting Sustainable Enteric Microbiome Diversity Amelioration and Restitution (SEMDAR). In a majority of circumstances self initiated intervention to modify the microbiome would fail to exhibit any notable benefits because of imprecision in the specific aspects of the dysbiosis and foggy and vague nature of the intervention due to the following:

1. After the age of 5 the microbiome gets relatively stabilized and despite minor day to day variability tends not to undergone enduring transformation such that the best period for intervention is very early in life although elderly is also susceptible to enteric microbiome transformation.

2. Ineffective intervention due to actual absence or paucity of the micro-organisms being administered in the intervention or failure of the intervention micro-organisms to get colonized

3. Dominant impact of enteric biome refractoriness inducing impact of major lifestyle factors like sleep problems, social jetlag, stress, smoking, street substance etc.

4. Short lived or transient nature off the impact generated by the intervention such that without a permanent dietary shift the benefits might be lost after a few weeks.

5. Microbiome expert supervision is required for a successful admonition of the paired probiotic diet and the supplemental probiotics under specific indications. In the most impactful fecal transplant style interventions it might require antibiotic administration to shift the microbial flora for the implant to coniine successfully.


10 Easy Ways to Improve Gut Health

Microbiota Integrated Symbiotic Therapeutics (MIST)

Mind Microbiota Messaging Management Mastery

Sustainable Enteric Microbiome Diversity Amelioration and Restitution (SEMDAR) is proposed as a comprehensive lifestyle intervention that expands upon the Symbiotic therapy of a probiotic diet and supplemental use of probiotics or prebiotics as a Lifespan Enhancing Enteric Eubiosis (LEEE) strategy. It is to be ranked at par with and used complementarily to the epigenomic transformation for healthspan amelioration. The set of seven interventions included in MIST impact the epigenome as well as the microbiome at the same time and there likely exist interactions between these two systems.

Eubiotic effect is obtained by combining two process: Noxious Innocuous-looking Morbidogenic Microorganism Preponderance Attenuation (NIMMPA) and Inflammaging Snuffing & Extinguishing Microorganism Population Escalation (enhancement) (ISEMPE)

Cellulose type extremely complex non-digestible sturdy “super-starches” contained in whole grains seem to provide the optimal nutrient requirement for ISEMPE. and boost metabolic health.

Note of caution in interpretation of studies on microbiome especially in the context of healthspan promoting impact. Dietary and exercise results generated in prospective research trials tend not to be replicated in real life because of dismal compliance in real life and the presence or absence of associated factors in the healthspan promotion cluster or ensemble and omission of the weighting of the five major healthspan drivers namely sleep, sustained sports & sexercise, super supper, (Mediterranean diet), sociao-economic status, stress-slaying spirituality & Samadhi. In some cases, substances (smoking, spirits, street stuff) come into play in a major way. The impact of probiotics being a fraction of that of the other four components such that more attention needs to be paid to sleep, sports and stress slaying. The ensemble operates best as an orchestra when attention is paid to all these simultaneously.

Limitations of returns

Limitations of returns from adoption of Lifespan Enhancing Enteric Eubiosis (LEEE) strategies or attempting Sustainable Enteric Microbiome Diversity Amelioration and Restitution (SEMDAR). In a majority of circumstances self initiated intervention to modify the microbiome would fail to exhibit any notable benefits because of imprecision in the specific aspects of the dysbiosis and foggy and vague nature of the intervention due to the following:

1. After the age of 5 the microbiome gets relatively stabilized and despite minor day to day variability tends not to undergone enduring transformation such that the best period for intervention is very early in life although elderly is also susceptible to enteric microbiome transformation.

2. Ineffective intervention due to actual absence or paucity of the micro-organisms being administered in the intervention or failure of the intervention micro-organisms to get colonized

3. Dominant impact of enteric biome refractoriness inducing impact of major lifestyle factors like sleep problems, social jetlag, stress, smoking, street substance etc.

4. Short lived or transient nature off the impact generated by the intervention such that without a permanent dietary shift the benefits might be lost after a few weeks.

5. Microbiome expert supervision is required for a successful admonition of the paired probiotic diet and the supplemental probiotics under specific indications. In the most impactful fecal transplant style interventions it might require antibiotic administration to shift the microbial flora for the implant to coniine successfully.

While there exists no current marker for assessing and monitoring the concept of a flourishing microbiome which is herein being called Sustainable Enteric Microbiome Diversity Amelioration and Restitution (SEMDAR) its existence can be inferred from the health outcome. By using the example of fermented non-dairy food item collectively called “Sauerkrautes” use and for ease of communication and comprehension the concept of SEMDAR would be used interchangeably and synonymously with “Sumptuous Sauerkraut Effect”. (sumptuous being synonymous with lavish, splendid, opulent, magnificent, elaborate, spectacular etc.). This connotes consumption of very generous servings and fairly regularly and not just a token half a bite of the item. Restitution is appended to the microbiome diversity amelioration to connote that resilience of the gut microbiome gets enhanced when like the healed strained and exercise torn muscle fibres and bone septa becoming stronger post-shock microbiome when restored and repaired becomes more resilient with resilient microbiome defined by exhibiting a higher tolerance to adverse environmental conditions like including pH, thermal shocks etc.. . Monotony or homogeneity of the enteric microbiome tends to produce adverse health impacts possibly through amplification of inflammaging, stress level, sleep disruption and worsening of the symptoms of the coexisting health conditions such that escaping microbiome homogeneity should be made a very high priority for wellness amelioration. Noteworthy is the connections of SEMDAR to the following important health marker influencers

1. Genetically determined propensities

2. Epigenomic structure (innate) and status (lifestyle dependent)

3. Enteric brain or enteric encephalon synchronizing with the cephalic encephalon.

4. Ambient baseline level of stress or chronic anxiety state being experiences.

5. Inflammaging intensity (CAIRO) and SEMDAR bidirectional relationship or dance.

Microbiome Diversity and health is extremely fluid and shifts within 48 hours and is in need of a continuous monitoring, maintenance and if damaged it needs rapid restitution or restoration. The positive nuance of this fluid nature of the microbiome health is that by adopting a probiotic lifestyle restoration of wellness can be accomplished speedily as well.

Optimal composition and activity output of enteric microbiome can be termed entero-eubiosis

Term dysbiosis can be used to connoted malfunctioning or unhealthy gut microbiome that lacks diversity.

Ten dietary strategies for SEMDAR

1. Striving diligently for a splendidly high diversity of diet. Corinne Maurice, https://www.mcgill.ca/microimm/corinne-maurice an assistant professor at McGill University, who is also the Canada Research Chair in Gut Microbial Physiology and a CIFAR Azrieli Global Scholar for Humans and the Microbiome emphasizes the need for food type variability and ending of the food monotony. A good starting point is to divide the macronutrients into nearly equal thirds or starch: protein: fat ratio of 40%: 30%: 30% such that it is desirable to refrain from a diet that is high or contains in excess of 80% of any macronutrient or the microbiome wellness would suffer. Those with medically prescribed dietary restrictions due to celiac disease, Crohn’s ilieo-colitis, ulcerative colitis, irritable bowel syndrome and other gut-related disease, diabetes, food allergies, respiratory allergies aggravated by some food items, therapeutic ketogenic diet, tend to suffer from lack of food items diversity in their microbial populations. Food faddism, religious veganism, JUNK food addiction etc., tend to adversely impact enteric biodiversity. It’s clear that a healthy gut is a diversely-populated gut, and one of the best ways to build a diverse bacterial community is to eat a wide range of healthy foods. Focussing on ample food type diversity tends to include ample fibre as well which is another strongly prescribed directive for gut microbe wellness.

2. Striving diligently for very generous fibre ingestion. Next to sleep and exercise, a high fibre content diet seems to act as a fertilizer for the enteric microbiome such that it is essential to learn of ways to increase your dietary fibre. https://www.readersdigest.ca/food/healthy-food/increase-dietary-fibre/ High fibre content ensures a generous food type diversity and fibre makes a superb nutrient substrate for the health of the enteric microbiome. Benefits of high fibre diet likely derive from improved sleep quality and several other routes. Besides beans, lentils and legumes, even cooked whole grains like quinoa, barley and oats, have dietary fibre that can’t be broken down quickly which is a feature of food items that helps with SEMDAR. There is a very powerful bidirectional relationship between sleep-rest (sleep) cycle on one hand and high fibre intake on the other with this type of diet amplifying the quality of sleep and benefit of working out and in turn these two habits promote the inclination to eat fibre. There is no special benefit and even a possible drawback in ingestion of psyllium so the fibres best consumed in the format of natural food items not out of bottles.

3. Refraining from highly processed meats and extreme sugar overloading. The processed meats which tend to contain high sugar content and anti-bacterial and anti-fungal preservatives, tend to adversely impact enteric flora. While most of the sugar gets absorbed in the stomach the enteric flora tends not to exposed to the ingested simple sugars but the speedy absorption of sugars tends to impact the enteric pH adversely such that the diabetogenic effects of ingested sugars are enteric flora and inflammaging mediation to a greater extent than appreciated at this time. An extremely high energy density of processed meats defeats the overarching purpose of enteric wellness of food item diversity.

4. Correcting the gastric achlorhydria (neutralizing the gastric alkaline pH). Age related or genetically linked gastric achlorhydria likely adversely impacted enteric microbiome wellness. Partial correction of this can be achieved by steps like dieting the regurgitated jejunal alkaline contents due to bile by drinking ample water, ingestion of vinegar, ample citrus fruit intake etc. Excessive ingestion sodium bicarbonates of antacids can worsen the situation and raise the pH of gastric content which impedes digestion in the stomach which impacts digestion in the intestine adversely and tends to hurt the microbiome health.

5. Generous fermented dairy product consumption. Items like cheese, yogurt and kefir are known to promote enteric microbiome diversity. Fermented dairy products tend to be tolerated fairly well even by those with partial lactose deficiency therefore preferable to milk, ice cream etc., that tend to produce crampy diarrhea. Whether the probiotic enriched yogurt and other fermented dairy products are superior remains to be proved. Part of the benefit of the fermented diary products likely stems from their acidic nature and correction of the gastric achlorhydria. Another part of the benefit likely stems from promoting the diversity of the enteric bacteria even if the bacteria of the ingested food items do not actually colonize. Thirdly there is a possible stress lowering effect which helps the wellness of microbiome given that hormonal shifts linked to chronic anxiety state adversely impacts the gut flora. A 2011 study found that, when a strain of bacteria that’s common to yogurt was ingested by mice, it regulated their moods.

Cheese like tofu seems to have a unique role in gut health. A study conducted by the American Chemical Society found that people who ate cheese had higher levels of a certain by-product of gut bacteria that’s been associated with a reduction in cholesterol.

https://www.readersdigest.ca/food/healthy-food/5-heart-healthy-foods-lower-your-cholesterol/

The benefits have been shown to be linked only to unpasteurized cheeses that contain microbes conducive to SEMDAR but tend to be nearly eliminated by pasteurization.

Generous fermented vegetable products and fermented beans or tofu type product consumption. The benefits of tofu consumption on the enteric flora are likely even more powerful than those from fermented diary products. These products tend to bring their positive effects through acidity in the stomach, offering food and fertilizer to the enteric flora etc. Fermented beans tend to have a pewful inflammaging suppressive effect as well as sleep promoting effect which boosts enteric biome wellness. https://en.wikipedia.org/wiki/Natt%C5%8D

https://en.wikipedia.org/wiki/Natt%C5%8D

Nattō (納豆) is a legendary traditional smelly Japanese fermented soybean curd that uses Bacillus subtilis usually served as breakfast and is seen to have remarkable longevity effects.

6. Maintaining a rigorous oro-dental hygiene and health. Besides the fact of possibility that overgrowth of advice to the enteric microbiome oral bacterial and fungal flora the adverse impact on enteric biome of a “foul mouth” (dismal oro-dental hygiene,

The fact of being edentulous tends to so seriously impact the oral hygiene linked inflating amplification that having a decent set of own teeth during the third phase of life tends to be linked with longevity. Part of the benefits are derived from the masticatory pleasures derived in the process which are bypassed through use of puree diet that is consumed by the edentulous or those with oral health conditions that interfere with the chewing. Some pleasure comes from the chips crunching sounds by biting on crackling items reminiscent of biting the bone marrow during energy consume from scavenging on the bones left bending by the carnivores in the prehistoric era. The lowered chronic anxiety state having an ameliorative impact on the enteric microbiome. The inflammaging impact of the “foul mouth” is deemed to be the intrusion of the oral bacteria and fungi into bloodstream and act as foreign particles that amplify inflammaging effect.

7. Enrichment of the diet with food “Krim Kram” like phytochemicals, polyphenols, spicy and aromatic herbal products so called anti-oxidants and other enteric biome nutrients and toys.

In a 2016 study published in the journal Advances in Nutrition, researchers found that mice that were fed walnuts experienced changes in their gut microbes and developed fewer instances of colon cancer.

Cooking with spices like garlic, ginger and turmeric, doesn’t only make your meals delicious; it can also curb the growth of harmful bacteria in your gut. “These spices actually contain very powerful antibacterial chemicals,” says Maurice. “But they’re not bad for your good bacteria; they’re bad for your bad bacteria.”

Sip some polyphenols A 2013 study from the journal Food Research International found that drinking black tea and red wine could improve the bacterial composition in the gut. That’s because, like chocolate, wine and tea contain microbe-feeding polyphenols.

Indulge in dark chocolate. Dark chocolate contains fibre and plant-based molecules called polyphenols. Since both of these compounds are difficult for the intestines to digest, they can travel deep into your gut where they’re fermented and metabolized by microbes. This process in turn releases health-promoting anti-inflammatory chemicals.

surprising health benefits of turmeric.

Consumption of pomegranate justice, grape juice, red wine etc., in modest amounts.

8. Refraining from using colonic washes, colon detoxification and other forms of laxative use.

9. Antibiotics Avoidance Attitude Adoption. Probiosis preservation is promoted via restriction of antibiotic use to diligently medically prescribed types alone. The recognition of the health impacts of enteric bacteria was identified when patients treated with broad spectrum antibiotics developed superinfection by Clostridium difficile producing hemorrhagic enterocolitis. While super-infection with fungi like Candida, C. difficile are rare moderate injury to the bacterial flora and result from even narrow spectrum antibiotics. While there are serious consuetudes of untreated infection there remains a clear role of ingestion of antibiotics for health status amelioration but their indiscriminate use must be refrained from and that they must not be self prescribed or consume without a good reason and that incudes not pressuring the physicians to prescribe antibiotics for viral conditions and as prophylactic agents unless specifies indications for their prophylactic use is indicated for dental procedures etc.

Find out what happens when you drink a glass of red wine every night.

10. foods that fight inflammation.

Canadian Digestive Health Foundation

10 Ways to Strengthen Your Microbiome

https://cdhf.ca/health-lifestyle/10-ways-to-strengthen-your-microbiome/

Medical News Today10 ways to improve gut health

https://www.medicalnewstoday.com/articles/325293

Gut health refers to the balance of microorganism’s residents symbiotically as commensals in the digestive tract.

Looking after the health of the gut and maintaining the right balance of these microorganisms is vital for physical and mental health, immunity, and more.

1. Take probiotics and eat fermented foods

Hemarajata and Versalovic published a review Effects of probiotics on gut microbiota: mechanisms of intestinal immunomodulation and neuromodulation in Therapeutic Advances in Gastroenterology 2013 Jan; 6(1): 39–51. doi: 10.1177/1756283X12459294 Describing how diet and intestinal luminal conversion by gut microbes play a role in shaping the structure and function of intestinal microbial communities. Proposed mechanisms of probiosis include alterations of composition and function of the human gut microbiome, and corresponding effects on immunity and neurobiology. Recent explorations of the human gut microbiota suggest that perturbations of microbial communities may increase predisposition to different disease phenotypes. Dietary nutrients may be converted into metabolites by intestinal microbes that serve as biologically active molecules affecting regulatory functions in the host. Probiotics may restore the composition of the gut microbiome and introduce beneficial functions to gut microbial communities, resulting in amelioration or prevention of gut inflammation and other intestinal or systemic disease phenotypes.

Popular fermented non-dairy products (collectively called “Sauerkrautes”), include home fermented or pickled vegetables, sauerkraut kefir, kimchi, Kombucha, miso, tempeh, dill pickle.

For grains or starch sources to effect SEMDAR they must be ingested with the nondigestible kernel which form the nutritional substrate for gut microbiome. This feature makes potatoes an inferior starch source and explains the reduced harm from fibre fastened fructose of whole fruits instead of drinking fruit juices.

Markowiak and Śliżewska published Effects of Probiotics, Prebiotics, and Synbiotics on Human Health in Nutrients. Sep 2017; 9(9): 1021. doi: 10.3390/nu9091021 attempting to summarize the current knowledge on the effects of probiotics, prebiotics, and synbiotics on human health. The mechanism of beneficial action of those substances is discussed, and verified study results proving their efficacy in human nutrition are presented. The human gastrointestinal tract is colonised by a complex ecosystem of microorganisms. Intestinal bacteria are not only commensal, but they also undergo a synbiotic co-evolution along with their host. Beneficial intestinal bacteria have numerous and important functions, e.g., they produce various nutrients for their host, prevent infections caused by intestinal pathogens, and modulate a normal immunological response. Therefore, modification of the intestinal microbiota in order to achieve, restore, and maintain favourable balance in the ecosystem, and the activity of microorganisms present in the gastrointestinal tract is necessary for the improved health condition of the host. The introduction of probiotics, prebiotics, or synbiotics into human diet is favourable for the intestinal microbiota. They may be consumed in the form of raw vegetables and fruit, fermented pickles, or dairy products. Another source may be pharmaceutical formulas and functional food.

People who want to enhance their gut health may wish to include more of the following prebiotic-rich foods in their diet: asparagus, Jerusalem artichoke, chicory, garlic, onions

3. Sugar and sweetener sparing strategy.

Laboratory animals fed standard Western diet tend to develop metabolic syndrome.

Magnusson et al published Relationships between diet-related changes in the gut microbiome and cognitive flexibility in Neuroscience Aug 2015 6;300:128-40. doi: 10.1016/j.neuroscience.2015.05.016. Western diets are high in fat and sucrose and can influence behavior and gut microbiota. There is growing evidence that altering the microbiome can influence the brain and behavior. This study was designed to determine whether diet-induced changes in the gut microbiota could contribute to alterations in anxiety, memory or cognitive flexibility. Two-month-old, male C57BL/6 mice were randomly assigned high-fat (42% fat, 43% carbohydrate (CHO), high-sucrose (12% fat, 70% CHO (primarily sucrose) or normal chow (13% kcal fat, 62% CHO) diets. Fecal microbiome analysis, step-down latency, novel object and novel location tasks were performed prior to and 2weeks after diet change. Water maze testing for long- and short-term memory and cognitive flexibility was conducted during weeks 5-6 post-diet change. Some similarities in alterations in the microbiome were seen in both the high-fat and high-sucrose diets (e.g., increased Clostridiales), as compared to the normal diet, but the percentage decreases in Bacteroidales were greater in the high-sucrose diet mice. Lactobacillales was only significantly increased in the high-sucrose diet group and Erysipelotrichales was only significantly affected by the high-fat diet. The high-sucrose diet group was significantly impaired in early development of a spatial bias for long-term memory, short-term memory and reversal training, compared to mice on normal diet. An increased focus on the former platform position was seen in both high-sucrose and high-fat groups during the reversal probe trials. There was no significant effect of diet on step-down, exploration or novel recognitions. Higher percentages of Clostridiales and lower expression of Bacteroidales in high-energy diets were related to the poorer cognitive flexibility in the reversal trials. These results suggest that changes in the microbiome may contribute to cognitive changes associated with eating a Western diet.

Dysbiotic effect of artificial sweetener aspartame that lowers microbiome diversity.

Palmnäs et al published Low-Dose Aspartame Consumption Differentially Affects Gut Microbiota-Host Metabolic Interactions in the Diet-Induced Obese Rat in PLoS One. Oct 2014; 9(10): e109841. doi: 10.1371/journal.pone.0109841

Aspartame consumption is implicated in the development of obesity and metabolic disease despite the intention of limiting caloric intake. The mechanisms responsible for this association remain unclear, but may involve circulating metabolites and the gut microbiota. Aims were to examine the impact of chronic low-dose aspartame consumption on anthropometric, metabolic and microbial parameters in a diet-induced obese model. Male Sprague-Dawley rats were randomized into a standard chow diet (CH, 12% kcal fat) or high fat (HF, 60% kcal fat) and further into ad libitum water control (W) or low-dose aspartame (A, 5–7 mg/kg/d in drinking water) treatments for 8 week (n = 10–12 animals/treatment). Animals on aspartame consumed fewer calories, gained less weight and had a more favorable body composition when challenged with HF compared to animals consuming water. Despite this, aspartame elevated fasting glucose levels and an insulin tolerance test showed aspartame to impair insulin-stimulated glucose disposal in both CH and HF, independently of body composition. Fecal analysis of gut bacterial composition showed aspartame to increase total bacteria, the abundance of Enterobacteriaceae and Clostridium leptum. An interaction between HF and aspartame was also observed for Roseburia ssp wherein HF-A was higher than HF-W (P<0.05). Within HF, aspartame attenuated the typical HF-induced increase in the Firmicutes:Bacteroidetes ratio. Serum metabolomics analysis revealed aspartame to be rapidly metabolized and to be associated with elevations in the short chain fatty acid propionate, a bacterial end product and highly gluconeogenic substrate, potentially explaining its negative affects on insulin tolerance. How aspartame influences gut microbial composition and the implications of these changes on the development of metabolic disease require further investigation.

Diabestogenic impact of diet sodas.

Suez et al published Artificial sweeteners induce glucose intolerance by altering the gut microbiota in Nature Oct 2014; 514(7521):181-86. doi: 10.1038/nature13793. reporting identifying NAS-altered (Non-caloric artificial sweeteners altered) microbial metabolic pathways that are linked to host susceptibility to metabolic disease, and demonstrate similar NAS-induced dysbiosis and glucose intolerance in healthy human subjects. Collectively, our results link NAS consumption, dysbiosis and metabolic abnormalities, thereby calling for a reassessment of massive NAS usage. Non-caloric artificial sweeteners (NAS) are among the most widely used food additives worldwide, regularly consumed by lean and obese individuals alike. NAS consumption is considered safe and beneficial owing to their low caloric content, yet supporting scientific data remain sparse and controversial. Here we demonstrate that consumption of commonly used NAS formulations drives the development of glucose intolerance through induction of compositional and functional alterations to the intestinal microbiota. These NAS-mediated deleterious metabolic effects are abrogated by antibiotic treatment, and are fully transferrable to germ-free mice upon faecal transplantation of microbiota configurations from NAS-consuming mice, or of microbiota anaerobically incubated in the presence of NAS.

Dysnoligosomnia linked dysbiosis or impaired gut microbiome diversity.

Managing stress is important for many aspects of health, including gut health.

Even short-term stress tends to inflict dysbiosis and in turn dysbiosis amplifies chronic anxiety state.

Galley et al published Exposure to a social stressor disrupts the community structure of the colonic mucosa-associated microbiota in BMC Microbiology Jul 2014; 14: 189. http://doi: 10.1186/1471-2180-14-189 pointing out that stress or anxiety-like behavior with elevated corticosterone outputs in mice tends to impact the colonic microbiota in inbred C57BL/6 mice and outbred CD-1 mice but different strains of mice tend to show different changes. The microbiota of the mammalian gastrointestinal (GI) tract consists of diverse populations of commensal bacteria that interact with host physiological function. Dysregulating these populations, through exogenous means such as antibiotics or dietary changes, can have adverse consequences on the health of the host. Studies from laboratories such as ours have demonstrated that exposure to psychological stressors disrupts the population profile of intestinal microbiota. To date, such studies have primarily focused on prolonged stressors (repeated across several days) and have assessed fecal bacterial populations. It is not known whether shorter stressors can also impact the microbiota, and whether colonic mucosa-associated populations can also be affected. The mucosa-associated microbiota exists in close proximity to elements of the host immune system and the two are tightly interrelated. Therefore, alterations in these populations should be emphasized.

In humans, a variety of stressors can negatively affect gut health, including:

  • psychological stress

  • environmental stress, such as extreme heat, cold, or noise

  • sleep deprivation

Social jet lag induced dysbiosis can explain the inflammaging intensification from circadian dysrhythmia.

Circadian Disorganization Alters Intestinal Microbiota

Voigt et al published Circadian Disorganization Alters Intestinal Microbiota

in PLoS One May 2014; 9(5): e97500. doi: 10.1371/journal.pone.0097500

Intestinal dysbiosis and circadian rhythm disruption are associated with similar diseases including obesity, metabolic syndrome, and inflammatory bowel disease. Despite the overlap, the potential relationship between circadian disorganization and dysbiosis is unknown; thus, in the present study, a model of chronic circadian disruption was used to determine the impact on the intestinal microbiome. Male C57BL/6J mice underwent once weekly phase reversals of the light:dark cycle (i.e., circadian rhythm disrupted mice) to determine the impact of circadian rhythm disruption on the intestinal microbiome and were fed either standard chow or a high-fat, high-sugar diet to determine how diet influences circadian disruption-induced effects on the microbiome. Weekly phase reversals of the light:dark (LD) cycle did not alter the microbiome in mice fed standard chow; however, mice fed a high-fat, high-sugar diet in conjunction with phase shifts in the light:dark cycle had significantly altered microbiota. While it is yet to be established if some of the adverse effects associated with circadian disorganization in humans (e.g., shift workers, travelers moving across time zones, and in individuals with social jet lag) are mediated by dysbiosis, the current study demonstrates that circadian disorganization can impact the intestinal microbiota which may have implications for inflammatory diseases.

Stress slashing interventions like yogic practices, meditation, deep breathing exercises, and progressive muscle relaxation would tend to promote symbiosis or effect SEMDAR,

Karl et al published Effects of Psychological, Environmental and Physical Stressors on the Gut Microbiota in Frontiers of Microbiology Sep 2018; 9: 2013. http://doi: 10.3389/fmicb.2018.02013 reporting that while gaps in data preclude concluding with high confidence that transient or cumulative exposures to psychological, environmental, and physical stressors has a consistent, meaningful impact on the human gut microbiota but that can be strongly speculated because provocative preclinical evidence highlights a need for translational research aiming to elucidate the impact of stressors on the human gut microbiota, and how the gut microbiota can be manipulated, for example by using nutrition, to mitigate adverse stress responses.

Stress, a ubiquitous part of daily human life, has varied biological effects which are increasingly recognized as including modulation of commensal microorganisms residing in the gastrointestinal tract, the gut microbiota. In turn, the gut microbiota influences the host stress response and associated sequelae, thereby implicating the gut microbiota as an important mediator of host health. This narrative review aims to summarize evidence concerning the impact of psychological, environmental, and physical stressors on gut microbiota composition and function. The stressors reviewed include psychological stress, circadian disruption, sleep deprivation, environmental extremes (high altitude, heat, and cold), environmental pathogens, toxicants, pollutants, and noise, physical activity, and diet (nutrient composition and food restriction). Stressors were selected for their direct relevance to military personnel, a population that is commonly exposed to these stressors, often at extremes, and in combination. However, the selected stressors are also common, alone or in combination, in some civilian populations. Evidence from preclinical studies collectively indicates that the reviewed stressors alter the composition, function and metabolic activity of the gut microbiota, but that effects vary across stressors, and can include effects that may be beneficial or detrimental to host health. Translation of these findings to humans is largely lacking at present.

5. Antibiotics Avoidance Attitude

Antibiotic abuse induced dysbiosis can be protracted and endure from 6 months to a year.

Palleja et al published Recovery of gut microbiota of healthy adults following antibiotic exposure in Nature Microbiology Nov 2018;3(11):1255-1265. doi: 10.1038/s41564-018-0257-9 pointing out that despite a mild yet long-lasting imprint following antibiotics exposure, the gut microbiota of healthy young adults is resilient to a short-term broad-spectrum antibiotic’s intervention and their antibiotics resistance gene carriage modulates their recovery processes. Recovery of gut microbiota of healthy adults following antibiotic exposure to minimize the impact of antibiotics, gut microorganisms harbour and exchange antibiotics resistance genes, collectively called their resistome. Using shotgun sequencing-based metagenomics, we analysed the partial eradication and subsequent regrowth of the gut microbiota in 12 healthy men over a 6-month period following a 4-day intervention with a cocktail of 3 last-resort antibiotics: meropenem, gentamicin and vancomycin. Initial changes included blooms of enterobacteria and other pathobionts, such as Enterococcus faecalis and Fusobacterium nucleatum, and the depletion of Bifidobacterium species and butyrate producers. The gut microbiota of the subjects recovered to near-baseline composition within 1.5 months, although 9 common species, which were present in all subjects before the treatment, remained undetectable in most of the subjects after 180 days. Species that harbour β-lactam resistance genes were positively selected for during and after the intervention. Harbouring glycopeptide or aminoglycoside resistance genes increased the odds of de novo colonization, however, the former also decreased the odds of survival. Compositional changes under antibiotic intervention in vivo matched results from in vitro susceptibility tests.

It is a view held by the Centers for Disease Control and Prevention (CDC), that American doctors prescribe antibiotics unjustifiably in about 1 out of 4 times they are prescribed which adds up to a massive amount at national scale.

Dudek-Wicher et al published The influence of antibiotics and dietary components on gut microbiota in Przegląd Gastroenterologiczny. May 2018; 13(2): 85–92. http://doi: 10.5114/pg.2018.76005 The gut microbiota acts as a real organ. It exerts important metabolic functions, and regulates the inflammatory response by stimulating the immune system. Gut microbial imbalance (dysbiosis) has been linked to important human diseases and inflammation-related disorders. The symbiotic interactions between resident microorganisms and the gastrointestinal tract significantly contribute to maintaining gut homeostasis. The present review summarizes our knowledge regarding the impact of different antibiotics causing such long-term consequences as decreased microbial diversity, modulation of the Bacteroidetes/Firmicutes ratio, Clostridium difficile overgrowth, and increased expansion of the opportunistic pathogens Salmonella typhimurium, Escherichia spp., and Klebsiella spp. Also, food additives, such as emulsifiers and artificial sweeteners, which are meant to reduce the risk of obesity and diabetes, may actually increase the risk of diseases due to microbial alterations. On the other hand, dietary components such as polyphenols, omega-3 acids or curcumin may positively affect gut microbiota composition.

6. Aging reversal or inflammaging and symbiosis promotion

Health benefits of physical exertion linked thermogenesis (PELT) habit cannot be well explained by caloric expenditure alone and aerobics training fails to remedy diabesity. This observation could be explained why diet and exercise-based approach to diabesity is flawed because it overlooks the inflammaging -dysbiosis duo. For a regularly exercising contributes to good heart health and weight loss or weight maintenance it must soothe inflammaging, protect against oxidative assault and improve gut microbiome wellness via SEMDAR and as a result the improved symbiosis should promote the adherence to the exercise routine and shift the food preference in a strongly positive way away from sugary and processed food items that in turn set up a virtuous cycle of amplified microbiome resilience.

Petriz et al published Exercise induction of gut microbiota modifications in obese, non-obese and hypertensive rats in BMC Genomics. Jun 2014; 15(1): 511. doi: 10.1186/1471-2164-15-511 reporting that non-obese and hypertensive rats harbor a different gut microbiota from obese rats and that exercise training alters gut microbiota from an obese and hypertensive genotype background. Exercise altered the composition and diversity of gut bacteria at genus level in all rat lineages. Allobaculum (Hypertensive rats), Pseudomonas and Lactobacillus (Obese rats) were shown to be enriched after exercise, while Streptococcus (Wistar rats), Aggregatibacter and Sutturella (Hypertensive rats) were more enhanced before exercise. A significant correlation was seen in the Clostridiaceae and Bacteroidaceae families and Oscillospira and Ruminococcus genera with blood lactate accumulation. Moreover, Wistar and Hypertensive rats were shown to share a similar microbiota composition, as opposed to Obese rats. Finally, Streptococcus alactolyticus, Bifidobacterium animalis, Ruminococcus gnavus, Aggregatibacter pneumotropica and Bifidobacterium pseudolongum were enriched in Obese rats.

Obesity is a multifactor disease associated with cardiovascular disorders such as hypertension. Recently, gut microbiota was linked to obesity pathogenesis and shown to influence the host metabolism. Moreover, several factors such as host-genotype and life-style have been shown to modulate gut microbiota composition. Exercise is a well-known agent used for the treatment of numerous pathologies, such as obesity and hypertension; it has recently been demonstrated to shape gut microbiota consortia. Since exercise-altered microbiota could possibly improve the treatment of diseases related to dysfunctional microbiota, this study aimed to examine the effect of controlled exercise training on gut microbial composition in Obese rats (n = 3), non-obese Wistar rats (n = 3) and Spontaneously Hypertensive rats (n = 3). Pyrosequencing of 16S rRNA genes from fecal samples collected before and after exercise training was used for this purpose.

Athletic adults tend to had a more variegated gut flora than sedentary individuals and an inference from that is unless exercise brings out a shift in the microbiome it would fail to prevent diabesity.

https://www.ncbi.nlm.nih.gov/pubmed/25021423

Clarke et al published Exercise and associated dietary extremes impact on gut microbial diversity in Gut Dec 2014; 63(12):1913-20. doi: 10.1136/gutjnl-2013-306541 presenting evidence for a beneficial impact of exercise on gut microbiota diversity but also indicate that the relationship is complex and is related to accompanying dietary extremes. The commensal microbiota, host immunity and metabolism participate in a signalling network, with diet influencing each component of this triad. In addition to diet, many elements of a modern lifestyle influence the gut microbiota but the degree to which exercise affects this population is unclear. Therefore, we explored exercise and diet for their impact on the gut microbiota. Since extremes of exercise often accompany extremes of diet, we addressed the issue by studying professional athletes from an international rugby union squad. Two groups were included to control for physical size, age and gender. Compositional analysis of the microbiota was explored by 16S rRNA amplicon sequencing. Each participant completed a detailed food frequency questionnaire.

As expected, athletes and controls differed significantly with respect to plasma creatine kinase (a marker of extreme exercise), and inflammatory and metabolic markers. More importantly, athletes had a higher diversity of gut micro-organisms, representing 22 distinct phyla, which in turn positively correlated with protein consumption and creatine kinase.

Part of the reason for the difference is due to shifted diet preferences of athlete’s hat is in a virtuous cycle, and because sedentary people tend to prefer food items that promote dysbiosis which forces their bodies to stagnate in the vicious cycle.

The Physical Activity Guidelines for Americans recommend that adults engage in at least 150 minutes of moderate intensity exercise each week, along with muscle strengthening activities on 2 or more days each week.

7. Get enough sleep

Getting enough good-quality sleep can improve mood, cognition, and gut health.

A 2014 animal study indicated that irregular sleep habits and disturbed sleep can have negative outcomes for the gut flora, which may increase the risk of inflammatory conditions.

Establish healthful sleep habits by going to bed and getting up at the same time each day. Adults should get at least 7 hours of sleep per night.

MEDICAL NEWS TODAY NEWSLETTER

Stay in the know. Get our free daily newsletter

Expect in-depth, science-backed toplines of our best stories every day. Tap in and keep your curiosity satisfied.

Severely limiting usage of detergents and household cleaning products and switching to the organic home made variety.

Microbiome preponderance patterns shift with the use of laundry detergants and toilet/kitchen cleaning products as it happens with the antibiotics consumption. Whereas adults are resilient infants and toddlers tend to be significiantly impacted because they are in the phase of life when enteric microbiome is becoming established for the rest of the life making it imperative to limit their exposure to chemicals of all variety.

The 2018 research analyzed the gut flora of over 700 infants ages 3–4 months.

Tun et al published Postnatal exposure to household disinfectants, infant gut microbiota and subsequent risk of overweight in children in CMAJ. 2018 Sep 17; 190(37): E1097–E1107. doi: 10.1503/cmaj.170809 reporting that exposure to household disinfectants was associated with higher BMI at age 3, mediated by gut microbial composition at age 3–4 months. Although child overweight was less common in households that cleaned with eco-friendly products, the lack of mediation by infant gut microbiota suggests another pathway for this association.

Emerging links between household cleaning products and childhood overweight may involve the gut microbiome. We determined mediating effects of infant gut microbiota on associations between home use of cleaning products and future overweight. From the Canadian Healthy Infant Longitudinal Development (CHILD) birth cohort, we tested associations between maternal report of cleaning product use and overweight at age 3, and whether associations were mediated by microbial profiles of fecal samples in 3- to 4-month-old infants. Among 757 infants, the abundance of specific gut microbiota was associated with household cleaning with disinfectants and eco-friendly products in a dose-dependent manner. With more frequent use of disinfectants, Lachnospiraceae increasingly became more abundant (highest v. lowest quintile of use: adjusted odds ratio [AOR] 1.93, 95% confidence interval [CI] 1.08 to 3.45) while genus Haemophilus declined in abundance (highest v. lowest quintile of use: AOR 0.36, 95% CI 0.20 to 0.65). Enterobacteriaceae were successively depleted with greater use of eco-friendly products (AOR 0.45, 95% CI 0.27 to 0.74). Lachnospiraceae abundance significantly mediated associations of the top 30th centile of household disinfectant use with higher body mass index (BMI) z score (p = 0.02) and with increased odds of overweight or obesity (p = 0.04) at age 3. Use of eco-friendly products was associated with decreased odds of overweight or obesity independently of Enterobacteriaceae abundance (AOR 0.44, 95% CI 0.22 to 0.86), with no significant mediation (p = 0.2).

Infants growing up in homes where disinfectant cleaning products are used more than twice a week appear to have a higher levels of Lachnospiraceae gut microbes, type associated with type 2 diabetes and obesity. At age 3, these infants had a higher body mass index (BMI) than children without exposure to such high levels of disinfectants.

https://www.nature.com/articles/s41598-018-29687-x

de la Cuesta-Zuluaga et al published Gut microbiota is associated with obesity and cardiometabolic disease in a population in the midst of Westernization in Scientific Reports Jul 2018;8(1):11356. doi: 10.1038/s41598-018-29687-x.

Westernization and its accompanying epidemiological transitions are associated with changes in gut microbiota. While the extremes of this lifestyle spectrum have been compared (hunter-gatherers, industrialized countries), populations undergoing such shifts have received little attention. To fill the gap of knowledge about the microbiome evolution following broad lifestyle changes and the emergence of disease-associated dysbiosis, we performed a cross-sectional study in which we characterized the microbiota of 441 Colombian adults through 16S rRNA gene sequencing and determined its relationship with demographic, health-related and dietary parameters. We showed that in the gut microbiota of this cohort thrive taxa proper of both hunter-gatherers (Prevotella, Treponema) and citizens of industrialized countries (Bacteroides, Bifidobacterium, Barnesiella); the relative abundances of these taxa differed from those in Western and non-Western populations. We also showed that the Colombian gut microbiota is composed of five consortia of co-abundant microorganisms that are differentially associated with lifestyle, obesity and cardiometabolic disease, and highlighted metabolic pathways that might explain associations between microbiota and host health. Our results give insights into the evolution of the gut microbiota, and underscore the importance of this community to human health. Promoting the growth of specific microbial consortia could help ameliorating physiological conditions associated with Western lifestyles.

Smoking Succession

A part of harm to heart and lungs from smoking likely get mediated through the enteric biome and gut-brain axis. Smoking associated dysbiosis that tilts the microbiome towards noxious enteric morbidogenic microorganism species preponderance. One route of injury to microbiome could be the swallowing of smoked tobacco products.

Savin et al published a review Smoking and the intestinal microbiome in Archives of Microbiology 2018 Jul;200(5):677-684. doi: 10.1007/s00203-018-1506-2 concluding that smoking has an effect on intestinal microbiome and is suggested to alter its composition. This interaction may contribute to the development of intestinal and systemic diseases, particularly inflammatory bowel diseases. Studies are emerging alluding to the role of intestinal microbiome in the pathogenesis of diseases. Intestinal microbiome is susceptible to the influence of environmental factors such as smoking, and recent studies have indicated microbiome alterations in smokers. The aim of the study was to review the literature regarding the impact of smoking on the intestinal microbiome. A literature review of publications in PUBMED was performed using combinations of the terms "Intestinal/Gut/Gastrointestinal/Colonic" with "Microbiome/Microbiota/Microbial/Flora" and "Smoking/Smoker/Tobacco". We selected studies that were published between the years 2000 and 2016 as our inclusion criteria. Observational and interventional studies suggest that the composition of intestinal microbiome is altered due to smoking. In these studies, Proteobacteria and Bacteroidetes phyla were increased, as well as the genera of Clostridium, Bacteroides and Prevotella. On the other hand, Actinobacteria and Firmicutes phyla as well as the genera Bifidobacteria and Lactococcus were decreased. Smoking also decreased the diversity of the intestinal microbiome. Mechanisms that have been suggested to explain the effect of smoking on intestinal microbiome include: oxidative stress enhancement, alterations of intestinal tight junctions and intestinal mucin composition, and changes in acid-base balance. Interestingly, some smoking-induced alterations of intestinal microbiome resemble those demonstrated in conditions such as inflammatory bowel disease and obesity. Further studies should be performed to investigate this connection.

Summary

By making appropriate lifestyle and dietary changes, people can alter the diversity and number of microbes in their gut for the better. Positive changes a person can make include taking probiotics, following a fiber-rich vegetarian diet, and avoiding the unnecessary use of antibiotics and disinfectants.

lifestyle changes a person can make include getting enough sleep and exercising regularly.

Shunning Super Sanitary Stance. Lifespan Enhancing Enteric Eubiosis (LEEE) is fostered through shunning the mindset of germaphobia and engaged in excessively disinfection super-clean habits and pursuing that objective through a super-robust use of detergents and antiseptic chemicals.

Drastic dietary deviance to foster LEEE can prove hazardous if done without physician supervision. Commitment to pursue a LEEE lifestyle is a very serious undraping and should be undertaken with ample planning and consultation with dietician, physician or microbiome expert especially if the individual has a resisting Non-communicable Disease (NCD) or metabolic syndrome or gastrointestinal condition like enteral allergies, Crohn’s disease , ulcerative colitis, irritable bowel syndrome, asthma, hay fever or any other major allergic medical conditions, probiotics and fiber-rich or vegetarian diets may not be helpful.

10 Ways to Strengthen Your Microbiome

https://cdhf.ca/health-lifestyle/10-ways-to-strengthen-your-microbiome/

1. Make sure to eat your vegetables!

Especially the leafy green ones! Vegetables are loaded with fibres, which cannot be digested by people but are consumed by the good bacteria in your gut. It has been observed that people who follow a diet rich with fruits and vegetables are less likely to grow disease-causing bacteria. Some great examples of vegetables that feed your microbes are:

Broccoli, Asparagus, Artichokes, Leeks, garlic, Onions, Spinach

2. Cut out sugar and avoid processed foods

You’re sweet enough already! Fast digesting sugars, otherwise known as Monnosaccarides, are digested so quickly that your little microbes don’t get a chance to take a bite out of them! If you eat too many simple sugars too regularly, you run the risk of literally starving your microbiome to death. Additionally, hungry microbes will resort to munching away at the lining in your intestine, which can lead to inflammation. Try to alter your diet to include more foods with complex sugars, to ensure a happy and healthy microbiome. Here’s a list of some sweet foods that will keep both you and your gut happy!

· Honey

· Dark Chocolate

· Coconut Flour

· Apples

· Berries

· Bananas

· Mango

· Sweet Potatoes

Also make sure you keep out an eye for dreaded hidden sources of monosaccharides. Sugar can sneak into foods you would never expect them to be. Keep an eye on sugar levels in things like smoothies, nut butters, protein bars, salad dressings and even in a gut-favourite… yogurt!

3. Probiotics are your gut’s best friend

Probiotics are chock-full of live bacteria that will help ensure your gut is populated by mostly the good types of microbes. You can get a good probiotic supplement at your local health food store, however, make sure you ask your doctor what strains of cultures are best for you, as each person’s microbiome is different. There are many probiotic products out there that claim to have live cultures but do not, so it is important to do your research beforehand. For example, Align is a great brand of probiotic for people who suffer from IBS.

4. Avoid Antibiotics

If probiotics are your gut’s best friend, then Antibiotics are your gut’s worst enemy!

Antibiotics work buy wiping out any and all bacteria, which makes them very effective for treating illnesses, but very bad for your microbiome. The antibiotic cannot recognize the difference between good gut bacteria and bad bacteria. They work on a ‘kill now ask questions later’ model. Try to buy meat products that were raised without antibiotics, and if you do have to take an antibiotic to treat a virus, make sure to take a probiotic daily for the duration of your prescription to help replenish your gut bacteria.

5. Stock up on dietary sources of prebiotics

Prebiotics are food for your microbiome! It’s important to feed these little guys to give them the energy they need to complete their very important task of managing your enteric nervous system. Here is a list of dietary prebiotics that should be staples in your home kitchen:

· Whole Grains

· Apples

· Leeks

· Onions

· Garlic

· Cocoa Extracts

· Garlic

· Bananas

· Asparagus

· Nuts

· Seeds

· Red Wine Extracts

· Root Vegetables

· Beans

· Lentils

· Chickpeas

· Green Tea Extracts

6. Fermented Foods are gut-friendly

Fermented foods are another great source of probiotics. The crowd favourite is yogurt, however, if you’re going to be eating a lot of yogurt, make sure that it is sugar-free! There are several other options that are a great source of good bacteria. Kombucha is becoming a very popular source of probiotics. You can also eat things like pickles, kimchee, and kefir to ensure that you’re getting enough live cultures to keep your gut healthy and happy.

7. Try to cut back on the red meat

Aside from the fact that these days, many meat brands are known for raising their livestock with antibiotics, which is detrimental to your gut, there have been several studies that show healthier microbiomes in vegetarians. A vegetarian’s gut for example, will have a significantly smaller number of disease-causing bacteria that an omnivore’s gut. However, it is still unclear if this is due to the lack of meat being consumed, or the fact that vegetarians and plant-based individuals tend to consume a great deal more fibre than the average person.

8. It’s past your bedtime!

Getting enough rest is so important! Studies have shown that people with erratic sleeping patterns run the risk of disrupting their microbiome and running the risk of developing inflammatory diseases. Try to make sure that you get at least 8 hours of sleep a night.

9. Hit the Gym

Your microbes feel that if they’re working hard to keep you healthy, then you should be working hard too! The microbiomes of physically active people are more healthy and diverse. It also has to be said that one of the best ways to de-stress after a long day is by working out. Even just walking for 30 minutes a day could really impact your gut health, and help these little microbes continue to make sure that your stress levels are managed and your mental health stays intact.

10. Make time for you!

Say ‘no’ more often, explore meditation, mindfulness, yoga, or tai. Establishing balance in your life will support your mental and emotional health and optimize your gut and overall health. Stress can negatively affect your microbiome and you need a healthy microbiome to manage help you manage your stressors. If you’re not careful, and you may get caught in an unhealthy cycle if you do not give yourself time to re-energize.

******8

Your gut microbiome is a vast community of trillions of bacteria and fungi that inhabit every nook and cranny of your gastrointestinal tract, and have a major influence on your metabolism, body weight, propensity to illness, immune system, appetite and mood. These microbes mostly live in your lower intestine (the colon) and outnumber all the other cells in your body put together.

Conceptually, we should view these microbes as a newly discovered organ, weighing slightly more than our brains and nearly as vital. There are some organs we can live without, including our spleen, gall bladder, tonsils and appendix, but we wouldn’t survive long without our gut microbes. Intriguingly, no two microbiomes are the same – we are all unique. And more than ever, we’re finding out just how important these microbes are.

Read more about the microbiome:

· The British microbiome: how our guts can tell us more than our genes

· Are you a super pooper?

According to research, the richer and more diverse the community of gut microbes are, the lower your risk of disease and allergies. This has been shown in animal tests and also in human studies comparing the microbes of people with and without particular diseases. Examples from recent work at King’s College London include studies of diabetes, obesity, allergy and inflammatory diseases like colitis and arthritis.

Meanwhile, there is mounting evidence that babies born via caesarean section miss out on some of the microbes they would obtain through a vaginal birth, which may make them more vulnerable to allergies and asthma.

So how can you restore healthy gut flora, increase the good bacteria in your body, and give your microbiome a healthy boost? Here are some tips to get your gut going:

1

Increase your fibre intake

Aim for more than 40g per day, which is about double the current averages. Fibre intake has been shown to reduce heart disease and some cancers, as well as reduce weight gain.

2

Eat as many types of fruit and veg as possible, and try to eat seasonally

The variety may be as important as the quantities, as the chemicals and types of fibre will vary, and each support different microbial species.

3

Pick high-fibre vegetables

Good examples are artichokes, leeks, onions and garlic, which all contain high levels of inulin (a prebiotic fibre). Some vegetables like lettuce have little fibre or nutrient value.

Read more about microbes:

· Miracle microbes: bacterial buddies fuelling our future

· Eating insects is good for your gut

4

Choose food and drinks with high levels of polyphenols

Polyphenols are antioxidants that act as fuel for microbes. Examples are nuts, seeds, berries, olive oil, brassicas, coffee and tea – especially green tea.

5

Avoid snacking

Also, try to increase intervals between meals to give your microbes a rest. Occasionally skip meals or have an extended fast – this seems to reduce weight gain.

6

Eat plenty of fermented foods containing live microbes

Good choices are unsweetened yoghurt; kefir, which is a sour milk drink with five times as many microbes as yoghurt; raw milk cheeses; sauerkraut; kimchi, a Korean dish made from garlic, cabbage and chilli; and soybean-based products such as soy sauce, tempeh and natto.

Organic probiotic kefir drink and kefir Tibetan mushrooms © Getty

7

Drink a bit of alcohol

In small quantities, alcohol has been shown to increase your gut diversity, but large amounts are harmful to your microbes and your health.

8

Steer clear of artificial sweeteners like aspartame, sucralose and saccharine

These disrupt the metabolism of microbes and reduce gut diversity – in animal studies this has led to obesity and diabetes. Ditch the processed foods too, as these also upset microbes’ metabolism.

Read more about processed food:

· Ultra-processed food and the risk of death: will fish fingers and fizzy drinks kill you?

· Michael Mosley: Why do you think they call it junk food?

9

Spend more time in the countryside

People living in rural areas have better microbes than city-dwellers. While you’re at it, dust off your trowel: gardening and other outdoor activities are good for your microbiome.

10

Stroke animals

Studies have shown that people living with dogs have more microbial diversity.

11

Avoid antibiotics and non-essential medicines

Antibiotics destroy good and bad microbes, and it can take weeks to recover, so don’t take them unless you need them. Their use is also associated with obesity and allergies in animals. Even common medications like paracetamol and antacids can interfere with microbes.

12

Don’t be hygiene obsessed

Fastidious washing and overuse of antibacterial sprays may not be good for your gut.

Read more about hygiene:

· Personal hygiene: Is there such thing as ‘too clean’?

· Many cleaning products say they kill 99.9 per cent of germs. Should I worry about the remaining 0.1 per cent?

13

Spend time close to a lean person

Studies in mice have shown that leanness may be contagious. Microbes from a lean animal can reverse obesity in a fat one, but strangely, obesity microbes are harder to transmit than lean ones.

14

Avoid food and vitamin supplements

Only a tiny proportion of supplements have been shown to be beneficial. Instead, focus on eating a diverse range of real food to get all your nutrients.

15

Eat like the Hadza

Hadza hunt for food using traditional bows and arrows. Here, they’ve killed several vervet monkeys © Getty

The Hadza people of Tanzania have a gut microbiome diversity that is one of the richest on the planet and about 40 per cent higher than the average American and about 30 per cent higher than the average Brit.

The average Hadza person eats around 600 species of plants and animals in a year and has huge seasonal variation. They have virtually none of the common Western diseases such as obesity, allergies, heart disease and cancer. In contrast, most Westerners have fewer than 50 species in their diet and are facing an epidemic of illness and obesity.

--------------------é

BBC Science Focus Magazine

The microbes in your gut can help you to get thinner, be happier and live longer. Here’s how you can give them a helping hand…

Prof Tim Spector 15 tips to boost your gut microbiome

https://www.sciencefocus.com/the-human-body/how-to-boost-your-microbiome/

Your gut microbiome is a vast community of trillions of bacteria and fungi that inhabit every nook and cranny of your gastrointestinal tract, and have a major influence on your metabolism, body weight, propensity to illness, immune system, appetite and mood. These microbes mostly live in your lower intestine (the colon) and outnumber all the other cells in your body put together.

Conceptually, we should view these microbes as a newly discovered organ, weighing slightly more than our brains and nearly as vital. There are some organs we can live without, including our spleen, gall bladder, tonsils and appendix, but we wouldn’t survive long without our gut microbes. Intriguingly, no two microbiomes are the same – we are all unique. And more than ever, we’re finding out just how important these microbes are.

Read more about the microbiome:

· The British microbiome: how our guts can tell us more than our genes

· Are you a super pooper?

According to research, the richer and more diverse the community of gut microbes are, the lower your risk of disease and allergies. This has been shown in animal tests and also in human studies comparing the microbes of people with and without particular diseases. Examples from recent work at King’s College London include studies of diabetes, obesity, allergy and inflammatory diseases like colitis and arthritis.

Meanwhile, there is mounting evidence that babies born via caesarean section miss out on some of the microbes they would obtain through a vaginal birth, which may make them more vulnerable to allergies and asthma.

So how can you restore healthy gut flora, increase the good bacteria in your body, and give your microbiome a healthy boost? Here are some tips to get your gut going:

1

Increase your fibre intake

Aim for more than 40g per day, which is about double the current averages. Fibre intake has been shown to reduce heart disease and some cancers, as well as reduce weight gain.

2

Eat as many types of fruit and veg as possible, and try to eat seasonally

The variety may be as important as the quantities, as the chemicals and types of fibre will vary, and each support different microbial species.

3

Pick high-fibre vegetables

Good examples are artichokes, leeks, onions and garlic, which all contain high levels of inulin (a prebiotic fibre). Some vegetables like lettuce have little fibre or nutrient value.

Read more about microbes:

· Miracle microbes: bacterial buddies fuelling our future

· Eating insects is good for your gut

4

Choose food and drinks with high levels of polyphenols

Polyphenols are antioxidants that act as fuel for microbes. Examples are nuts, seeds, berries, olive oil, brassicas, coffee and tea – especially green tea.

5

Avoid snacking

Also, try to increase intervals between meals to give your microbes a rest. Occasionally skip meals or have an extended fast – this seems to reduce weight gain.

6

Eat plenty of fermented foods containing live microbes

Good choices are unsweetened yoghurt; kefir, which is a sour milk drink with five times as many microbes as yoghurt; raw milk cheeses; sauerkraut; kimchi, a Korean dish made from garlic, cabbage and chilli; and soybean-based products such as soy sauce, tempeh and natto.

Organic probiotic kefir drink and kefir Tibetan mushrooms © Getty

7

Drink a bit of alcohol

In small quantities, alcohol has been shown to increase your gut diversity, but large amounts are harmful to your microbes and your health.

8

Steer clear of artificial sweeteners like aspartame, sucralose and saccharine

These disrupt the metabolism of microbes and reduce gut diversity – in animal studies this has led to obesity and diabetes. Ditch the processed foods too, as these also upset microbes’ metabolism.

Read more about processed food:

· Ultra-processed food and the risk of death: will fish fingers and fizzy drinks kill you?

· Michael Mosley: Why do you think they call it junk food?

9

Spend more time in the countryside

People living in rural areas have better microbes than city-dwellers. While you’re at it, dust off your trowel: gardening and other outdoor activities are good for your microbiome.

10

Stroke animals

Studies have shown that people living with dogs have more microbial diversity.

11

Avoid antibiotics and non-essential medicines

Antibiotics destroy good and bad microbes, and it can take weeks to recover, so don’t take them unless you need them. Their use is also associated with obesity and allergies in animals. Even common medications like paracetamol and antacids can interfere with microbes.

12

Don’t be hygiene obsessed

Fastidious washing and overuse of antibacterial sprays may not be good for your gut.

Read more about hygiene:

· Personal hygiene: Is there such thing as ‘too clean’?

· Many cleaning products say they kill 99.9 per cent of germs. Should I worry about the remaining 0.1 per cent?

13

Spend time close to a lean person

Studies in mice have shown that leanness may be contagious. Microbes from a lean animal can reverse obesity in a fat one, but strangely, obesity microbes are harder to transmit than lean ones.

14

Avoid food and vitamin supplements

Only a tiny proportion of supplements have been shown to be beneficial. Instead, focus on eating a diverse range of real food to get all your nutrients.

15

Eat like the Hadza

Hadza hunt for food using traditional bows and arrows. Here, they’ve killed several vervet monkeys © Getty

The Hadza people of Tanzania have a gut microbiome diversity that is one of the richest on the planet and about 40 per cent higher than the average American and about 30 per cent higher than the average Brit.

The average Hadza person eats around 600 species of plants and animals in a year and has huge seasonal variation. They have virtually none of the common Western diseases such as obesity, allergies, heart disease and cancer. In contrast, most Westerners have fewer than 50 species in their diet and are facing an epidemic of illness and obesity.

----------------------------------é

Science Alert

https://www.sciencealert.com/here-s-how-to-make-your-microbiome-healthier

5 Science-Backed Ways to Make Your Microbiome Healthier in 2020

It's common for people to focus on their health at the start of the year.

But few consider the well being of the microbes that live inside the human gut – the microbiome – which are vital to an individual's good health.

How important are these bacteria? There are as many bacterial cells in us as there are human cells, and they help control everything from inflammation and the development and treatment of cancer to how much energy we get from our foods and perhaps even what foods we crave and our moods.

When our microbiome becomes unbalanced, often indicated when certain species or groups of bacteria become overly abundant, these functions can be disrupted, contributing to the development of a wide range of diseases such as obesity, cancer, inflammatory bowel disease and many others.

Our gut microbes are also responsible for gas production when we eat new foods as those microbes adapt to this new nutrient source in their environment.

So it is clear we want to have a healthy microbiome, but what is that?

There is a lot of debate regarding what exactly constitutes a healthy community of gut microbes, but one thing has become clear.

Humans need a diverse microbiome with a variety of bacterial species that can quickly adapt to the wide range of foods that we might want to consume while still performing all those important functions like preventing inflammation.

So what are some things that you can do to support a healthy, diverse microbiome?

Eat your fruits and veggies

While all the different foods that make up your diet can influence the gut microbiome, it is the fiber – the carbohydrates in our diet that we cannot break down ourselves but the bacteria in our gut can use readily – that drives the formation of a healthy microbiome.

Eating a diverse and abundant selection of fruits and veggies is a great way to feed some of the most health-promoting bacteria in our gut.

Add resistant starch

Most of the starch in our diet - like white bread and pasta - is quickly broken down and absorbed. But a fraction of that starch is resistant to digestion and acts more like a fiber, feeding the bacteria in our gut.

Resistant starch has been identified as particularly beneficial for supporting all of those healthy functions of the gut microbiome.

Some sources of resistant starch include potatoes and legumes.

All sources of starch can also become more resistant after cooking and then cooling in the fridge. So those leftover potatoes and pasta, cold or reheated, may have some added microbiome-promoting punch.

Experiment with different fibers

Not all gut microbiomes are the same and not all fibers are the same. Certain fibers and microbiomes will mix better than others, depending on what functions are present.

This means that you need to do some experimentation to see what fibers will make you and your gut feel the best. You can do this with fiber supplements or with different categories of fiber sources such as whole grains, legumes or cruciferous vegetables like broccoli.

Give your microbiome a couple of weeks to adjust to each fiber source to see how it responds.

Exercise for both you and your microbes

Regular physical activity is not only good for your heart, it is good for your gut, too. Studies recently showed that some of the lactate produced during exercise can impact certain gut microbes – although we don't yet know how and why.

Start slow if you haven't had regular physical activity as part of your daily life. If you start on New Year's, by Valentine's Day you could be walking daily, or doing some time of activity that you like, to help your heart, mind and gut.

Add probiotic foods into your diet

What are probiotic foods? These are foods that contain microorganisms that have a health benefit.

There are several different kinds of helpful microorganisms that are added to foods like yogurt, or are naturally found in other fermented foods – like sauerkraut or kimchi – that give them a health-promoting effect. Give one of these foods a try in the new year.

You might be wondering if probiotic supplements are as beneficial as probiotic food. So far there isn't enough evidence to say that – so stick with food.

Connie Rogers, Associate Professor of Nutritional Sciences, Pennsylvania State University and Darrell Cockburn, Assistant Professor of Food Science, Pennsylvania State University.

This article is republished from The Conversation under a Creative Commons license.

----------------------------------------é

oxidative assault

Healthline 10 Ways to Improve Your Gut Bacteria, Based on Science

There are around 40 trillion bacteria in your body, most of which are in your intestines.

https://www.healthline.com/nutrition/improve-gut-bacteria

here are around 40 trillion bacteria in your body, most of which are in your intestines.

Collectively, they are known as your gut microbiota, and they are hugely important for your health.

It is intuitively assumed that SEMDAR bring out positive health impacts via suppression of the content of the viruses, fungi, monocellular parasites like morbidogenic and pro-inflammaging bacteria by choking their nutrient supply by competitively consuming those nutrients by their preponderance.

1. Eat a Diverse Range of Foods

The rationale bending diversification of food composition is most readily grasped by viewing the enteric microbes akin to zoo animal population, whereby the management of the zoo has to purchase a wide variety of food for the zoo residents from fish, birds, reptiles, to wide variety of mammals who have very different dietary preferences. The wide diversity of the consents of gut biome mandates that for their collective good a wide variety of food must be consumed because each microbe category has somewhat distinctive nutritional preference. With a monotonous monoculture type diet several of the species of gut biome would undergo attrition which harms the SEMDAR objective. It is also possible that there happens a shift in their dietary preferences with changes in the gut environment. The precise mechanism how SEMDAR brings about positive health impacts is not yet well investigated but there are widely recognized association with benefits to SEMDAR.

Healthspan SHEAL benefits of enteric eubiosis

Claesson et al published Gut microbiota composition correlates with diet and health in the elderly in Nature 2012 Aug 9;488(7410):178-84. doi: 10.1038/nature11319.

Alterations in intestinal microbiota composition are associated with several chronic conditions, including obesity and inflammatory diseases. The microbiota of older people displays greater inter-individual variation than that of younger adults. Here we show that the faecal microbiota composition from 178 elderly subjects formed groups, correlating with residence location in the community, day-hospital, rehabilitation or in long-term residential care. However, clustering of subjects by diet separated them by the same residence location and microbiota groupings. The separation of microbiota composition significantly correlated with measures of frailty, co-morbidity, nutritional status, markers of inflammation and with metabolites in faecal water. The individual microbiota of people in long-stay care was significantly less diverse than that of community dwellers. Loss of community-associated microbiota correlated with increased frailty. Collectively, the data support a relationship between diet, microbiota and health status, and indicate a role for diet-driven microbiota alterations in varying rates of health decline upon ageing.

Lozupone et al published Diversity, stability and resilience of the human gut microbiota in Nature 2012 Sep 13;489(7415):220-30. doi: 10.1038/nature11550.

DOI: 10.1038/nature11550

Trillions of microbes inhabit the human intestine, forming a complex ecological community that influences normal physiology and susceptibility to disease through its collective metabolic activities and host interactions. Understanding the factors that underlie changes in the composition and function of the gut microbiota will aid in the design of therapies that target it. This goal is formidable. The gut microbiota is immensely diverse, varies between individuals and can fluctuate over time - especially during disease and early development. Viewing the microbiota from an ecological perspective could provide insight into how to promote health by targeting this microbial community in clinical treatments.

3Trusted Source,

Human Microbiome Project Consortium published Structure, function and diversity of the healthy human microbiomein Nature . 2012 Jun 13;486(7402):207-14. doi: 10.1038/nature11234.

Studies of the human microbiome have revealed that even healthy individuals differ remarkably in the microbes that occupy habitats such as the gut, skin and vagina. Much of this diversity remains unexplained, although diet, environment, host genetics and early microbial exposure have all been implicated. Accordingly, to characterize the ecology of human-associated microbial communities, the Human Microbiome Project has analysed the largest cohort and set of distinct, clinically relevant body habitats so far. We found the diversity and abundance of each habitat's signature microbes to vary widely even among healthy subjects, with strong niche specialization both within and among individuals. The project encountered an estimated 81-99% of the genera, enzyme families and community configurations occupied by the healthy Western microbiome. Metagenomic carriage of metabolic pathways was stable among individuals despite variation in community structure, and ethnic/racial background proved to be one of the strongest associations of both pathways and microbes with clinical metadata. These results thus delineate the range of structural and functional configurations normal in the microbial communities of a healthy population, enabling future characterization of the epidemiology, ecology and translational applications of the human microbiome.

Wu et al published Linking long-term dietary patterns with gut microbial enterotypes in Science. 2011 Oct 7;334(6052):105-8.

DOI: 10.1126/science.1208344

Diet strongly affects human health, partly by modulating gut microbiome composition. We used diet inventories and 16S rDNA sequencing to characterize fecal samples from 98 individuals. Fecal communities clustered into enterotypes distinguished primarily by levels of Bacteroides and Prevotella. Enterotypes were strongly associated with long-term diets, particularly protein and animal fat (Bacteroides) versus carbohydrates (Prevotella). A controlled-feeding study of 10 subjects showed that microbiome composition changed detectably within 24 hours of initiating a high-fat/low-fiber or low-fat/high-fiber diet, but that enterotype identity remained stable during the 10-day study. Thus, alternative enterotype states are associated with long-term diet.

Diversity dedicated diet which may derived from over a hundred or at a minimum fifty different plant and animal species fosters enteric eubiosis or SEMDAR.\

Heiman & Greenway published A healthy gastrointestinal microbiome is dependent on dietary diversity in Molecular Metabolism 2016 Mar 5;5(5):317-320. doi: 10.1016/j.molmet.2016.02.005.

PMID: 27110483 PMCID: PMC4837298 DOI: 10.1016/j.molmet.2016.02.005

Like all healthy ecosystems, richness of microbiota species characterizes the GI microbiome in healthy individuals. Conversely, a loss in species diversity is a common finding in several disease states. This biome is flooded with energy in the form of undigested and partially digested foods, and in some cases drugs and dietary supplements. Each microbiotic species in the biome transforms that energy into new molecules, which may signal messages to physiological systems of the host.

Dietary choices select substrates for species, providing a competitive advantage over other GI microbiota. The more diverse the diet, the more diverse the microbiome and the more adaptable it will be to perturbations. Unfortunately, dietary diversity has been lost during the past 50 years and dietary choices that exclude food products from animals or plants will narrow the GI microbiome further.

Additional research into expanding gut microbial richness by dietary diversity is likely to expand concepts in healthy nutrition, stimulate discovery of new diagnostics, and open up novel therapeutic possibilities.

David et al published Diet rapidly and reproducibly alters the human gut microbiome in Nature Jan 2014;505(7484):559-63. doi: 10.1038/nature12820.

PMID: 24336217 PMCID: PMC3957428 DOI: 10.1038/nature12820

Long-term dietary intake influences the structure and activity of the trillions of microorganisms residing in the human gut, but it remains unclear how rapidly and reproducibly the human gut microbiome responds to short-term macronutrient change. Here we show that the short-term consumption of diets composed entirely of animal or plant products alters microbial community structure and overwhelms inter-individual differences in microbial gene expression. The animal-based diet increased the abundance of bile-tolerant microorganisms (Alistipes, Bilophila and Bacteroides) and decreased the levels of Firmicutes that metabolize dietary plant polysaccharides (Roseburia, Eubacterium rectale and Ruminococcus bromii). Microbial activity mirrored differences between herbivorous and carnivorous mammals, reflecting trade-offs between carbohydrate and protein fermentation. Foodborne microbes from both diets transiently colonized the gut, including bacteria, fungi and even viruses. Finally, increases in the abundance and activity of Bilophila wadsworthia on the animal-based diet support a link between dietary fat, bile acids and the outgrowth of microorganisms capable of triggering inflammatory bowel disease. In concert, these results demonstrate that the gut microbiome can rapidly respond to altered diet, potentially facilitating the diversity of human dietary lifestyles.

Sonnenburg et al published Diet-induced extinctions in the gut microbiota compound over generations in Nature. Jan 2016;529(7585):212-5. doi: 10.1038/nature16504.

The gut is home to trillions of microorganisms that have fundamental roles in many aspects of human biology, including immune function and metabolism. The reduced diversity of the gut microbiota in Western populations compared to that in populations living traditional lifestyles presents the question of which factors have driven microbiota change during modernization. Microbiota-accessible carbohydrates (MACs) found in dietary fibre have a crucial involvement in shaping this microbial ecosystem, and are notably reduced in the Western diet (high in fat and simple carbohydrates, low in fibre) compared with a more traditional diet. Here we show that changes in the microbiota of mice consuming a low-MAC diet and harbouring a human microbiota are largely reversible within a single generation. However, over several generations, a low-MAC diet results in a progressive loss of diversity, which is not recoverable after the reintroduction of dietary MACs. To restore the microbiota to its original state requires the administration of missing taxa in combination with dietary MAC consumption. Our data illustrate that taxa driven to low abundance when dietary MACs are scarce are inefficiently transferred to the next generation, and are at increased risk of becoming extinct within an isolated population. As more diseases are linked to the Western microbiota and the microbiota is targeted therapeutically, microbiota reprogramming may need to involve strategies that incorporate dietary MACs as well as taxa not currently present in the Western gut.

The sugar, salt and stereo-fat studded monotonous, mundane, metabulopathic, morbidogenic, metropolitan maintained, diversity devoid non-variegated; “meat & potato” based Standard Western Diet is promotes enteric dysbiosis IAcODcIM mediated aging accelerating

Food surveyors estimated that 75% of the world’s food is derived from mere a dozen plant species and a half dozen animal species.

Heiman and Greenway published A healthy gastrointestinal microbiome is dependent on dietary diversity in Molecular Metabolism May 2016 5;5(5):317-320.

doi: 10.1016/j.molmet.2016.02.005. Lamenting over their observation that, dietary diversity has been lost during the past the past 5 decades and dietary choices that exclude food products from animals or plants will further narrow the GI microbiome in the future five decades, adding that diets of several rural region residents tend to be more diverse and richer in plant sourced foods. Like all healthy ecosystems, richness of microbiota species characterizes the GI microbiome in healthy individuals. Conversely, a loss in species diversity is a common finding in several disease states. This biome is flooded with energy in the form of undigested and partially digested foods, and in some cases drugs and dietary supplements. Each microbiotic species in the biome transforms that energy into new molecules, which may signal messages to physiological systems of the host.

Dietary choices select substrates for species, providing a competitive advantage over other GI microbiota. The more diverse the diet, the more diverse the microbiome and the more adaptable it will be to perturbations.

Highest enteric microbiota diversity is noted among residents of rural southern hemisphere (Africa, Asia and South America) relative to the Northern hemisphere (North American and Europe) with the lowest diversity. Greater ease to access of food among the wealthy tends to narrow their diversity leading to favourite dishes and food faddism. Having to forage and rummage for own food practiced by the poorer rural denizens tends to insert great diversity.

De Filippo et al published Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa in Proceedings of the National Academy of Sciences of U. S. A. Aug 2010;107(33):14691-6. doi: 10.1073/pnas.1005963107. Epub 2010 Aug 2.

Gut microbial composition depends on different dietary habits just as health depends on microbial metabolism, but the association of microbiota with different diets in human populations has not yet been shown. In this work, we compared the fecal microbiota of European children (EU) and that of children from a rural African village of Burkina Faso (BF), where the diet, high in fiber content, is similar to that of early human settlements at the time of the birth of agriculture. By using high-throughput 16S rDNA sequencing and biochemical analyses, we found significant differences in gut microbiota between the two groups. BF children showed a significant enrichment in Bacteroidetes and depletion in Firmicutes (P < 0.001), with a unique abundance of bacteria from the genus Prevotella and Xylanibacter, known to contain a set of bacterial genes for cellulose and xylan hydrolysis, completely lacking in the EU children. In addition, we found significantly more short-chain fatty acids (P < 0.001) in BF than in EU children. Also, Enterobacteriaceae (Shigella and Escherichia) were significantly underrepresented in BF than in EU children (P < 0.05). We hypothesize that gut microbiota coevolved with the polysaccharide-rich diet of BF individuals, allowing them to maximize energy intake from fibers while also protecting them from inflammations and noninfectious colonic diseases. This study investigates and compares human intestinal microbiota from children characterized by a modern western diet and a rural diet, indicating the importance of preserving this treasure of microbial diversity from ancient rural communities worldwide.

Clemente et al published The microbiome of uncontacted Amerindians in Scientific Advances . Apr 2015;1(3):e1500183. doi: 10.1126/sciadv.1500183.

Most studies of the human microbiome have focused on westernized people with life-style practices that decrease microbial survival and transmission, or on traditional societies that are currently in transition to westernization. We characterize the fecal, oral, and skin bacterial microbiome and resistome of members of an isolated Yanomami Amerindian village with no documented previous contact with Western people. These Yanomami harbor a microbiome with the highest diversity of bacteria and genetic functions ever reported in a human group. Despite their isolation, presumably for >11,000 years since their ancestors arrived in South America, and no known exposure to antibiotics, they harbor bacteria that carry functional antibiotic resistance (AR) genes, including those that confer resistance to synthetic antibiotics and are syntenic with mobilization elements. These results suggest that westernization significantly affects human microbiome diversity and that functional AR genes appear to be a feature of the human microbiome even in the absence of exposure to commercial antibiotics. AR genes are likely poised for mobilization and enrichment upon exposure to pharmacological levels of antibiotics. Our findings emphasize the need for extensive characterization of the function of the microbiome and resistome in remote nonwesternized populations before globalization of modern practices affects potentially beneficial bacteria harbored in the human body.

Consumption of diverse diet rich in unprocessed whole foods consisting of the edible portions of the plant or the animal source of food promotes enteric eubiosis because the objectives of the food marketing from commercial food outlets tends to introduce these modifications that harm the food quality and produces dysbiosis.

1. Elimination of the non-calorie-dense and taste dulling fibre rich kernels etc. that reduces the volume and weight of the food item for reducing transpiration cost and ease of storage.

2. Addition of antibacterial and antifungal preservatives that boost shelf life but inhibit enteric eubiosis among the consumers.

3. Addition of sugar, salt, trans fats, high concentration fructose syrup (HCFS) in processing the convenience food that eliminates the need of meal preparation injures enteric eubiosis.

4. Addition of colorants and secret taste enhancing and addictive element elevating components likely injure enteric eubiosis but this cannot be tested.

2. Eat Lots of Vegetables, Legumes, Beans and Fruit

Mood elevation impact of a generous produce consumption of 8 to ten servings daily is proposed to bring that impact via enteric eubisosis.

They are high in fiber, which can’t be digested by your body. However, fiber can be digested by certain bacteria in your gut, which stimulates their growth.

Beans and legumes also contain very high amounts of fiber.

Some high-fiber foods that are good for your gut bacteria include:

  • Raspberries

  • Artichokes

  • Broccoli

  • Chickpeas, Green peas, Lentils and other beans (kidney, pinto and white)

Diets rich in produce tend to bring out SHEAL effects via suppressing preponderance of morbidogenic enteric microbes via promoting the preponderance of the healthspan promoting organisms

Kinder et al published Impact of increasing fruit and vegetables and flavonoid intake on the human gut microbiota in Food & Function 2016 Apr;7(4):1788-96. doi: 10.1039/c5fo01096a.

Epidemiological studies have shown protective effects of fruits and vegetables (F&V) in lowering the risk of developing cardiovascular diseases (CVD) and cancers. Plant-derived dietary fibre (non-digestible polysaccharides) and/or flavonoids may mediate the observed protective effects particularly through their interaction with the gut microbiota. The aim of this study was to assess the impact of fruit and vegetable (F&V) intake on gut microbiota, with an emphasis on the role of flavonoids, and further to explore relationships between microbiota and factors associated with CVD risk. In the study, a parallel design with 3 study groups, participants in the two intervention groups representing high-flavonoid (HF) and low flavonoid (LF) intakes were asked to increase their daily F&V intake by 2, 4 and 6 portions for a duration of 6 weeks each, while a third (control) group continued with their habitual diet. Faecal samples were collected at baseline and after each dose from 122 subjects. Faecal bacteria enumeration was performed by fluorescence in situ hybridisation (FISH). Correlations of dietary components, flavonoid intake and markers of CVD with bacterial numbers were also performed. A significant dose X treatment interaction was only found for Clostidium leptum-Ruminococcus bromii/flavefaciens with a significant increase after intake of 6 additional portions in the LF group. Correlation analysis of the data from all 122 subjects independent from dietary intervention indicated an inhibitory role of F&V intake, flavonoid content and sugars against the growth of potentially pathogenic clostridia. Additionally, we observed associations between certain bacterial populations and CVD risk factors including plasma TNF-α, plasma lipids and BMI/waist circumference.

Preponderance of the content of Bifidobacteria in human enteric biome which is considered a beneficial microorganism, as they tend to retard inflammaging has been linked with apples, artichokes, blueberries, almonds and pistachios etc.

(11Trusted Source, 12Trusted Source, 13Trusted Source, 14Trusted Source).

(15Trusted Source).

Fiber rich food items like whole grains, fruits, vegetables promote preponderance of Bifidobacteria. And other healthspan enhancing microorganisms also termed friendly or beneficial gut bacteria

Furrie et al published Synbiotic therapy (Bifidobacterium longum/Synergy 1) initiates resolution of inflammation in patients with active ulcerative colitis: a randomised controlled pilot trial in Gut Feb 2005;54(2):242-9. doi: 10.1136/gut.2004.044834 reporting that Short term synbiotic treatment of active Ulcerative colitis resulted in improvement of the full clinical appearance of chronic inflammation in patients receiving this therapy.

Ulcerative colitis (UC) is an acute and chronic inflammatory disease of the large bowel with unknown aetiology. The immune response against normal commensal microorganisms is believed to drive inflammatory processes associated with UC. Therefore, modulation of bacterial communities on the gut mucosa, through the use of probiotics and prebiotics, may be used to modify the disease state.

A synbiotic was developed for use in UC patients combining a probiotic, Bifidobacterium longum, isolated from healthy rectal epithelium, and a prebiotic (Synergy 1), a preferential inulin-oligofructose growth substrate for the probiotic strain. Treatment was employed in a double blinded randomised controlled trial using 18 patients with active UC for a period of one month. Clinical status was scored and rectal biopsies were collected before and after treatment, and transcription levels of epithelium related immune markers were measured.

Sigmoidoscopy scores (scale 0-6) were reduced in the test group (start 4.5 (1.4), end 3.1 (2.5)) compared with placebo (start 2.6 (2.1), end 3.2 (2.2)) (p=0.06). mRNA levels for human beta defensins 2, 3, and 4, which are strongly upregulated in active UC, were significantly reduced in the test group after treatment (p=0.016, 0.038, and 0.008, respectively). Tumour necrosis factor alpha and interleukin 1alpha, which are inflammatory cytokines that drive inflammation and induce defensin expression, were also significantly reduced after treatment (p=0.018 and 0.023, respectively). Biopsies in the test group had reduced inflammation and regeneration of epithelial tissue.

3. Eat Fermented Foods

The process

Fermentation tends to promote growth of wellness friendly bacteria or yeasts converting the sugars in food to organic acids or alcohol. Examples of fermented foods include:

Many of these foods are rich in lactobacilli, a type of bacteria that can benefit your health.

People who eat a lot of yogurt appear to have more lactobacilli in their intestines. These people also have fewer Enterobacteriaceae, a bacteria associated with inflammation and a number of chronic diseases (16Trusted Source).

Similarly, a number of studies have shown that yogurt consumption can beneficially modify intestinal bacteria and improve symptoms of lactose intolerance in both infants and adults (17Trusted Source, 18Trusted Source, 19Trusted Source).

Certain yogurt products may also reduce the abundance of certain disease-causing bacteria in people with irritable bowel syndrome.

Two studies showed that yogurt also enhanced the function and composition of the microbiota (20Trusted Source).

However, it is important to note that many yogurts, especially flavored yogurts, contain high levels of sugar.

Therefore, the best yogurt to consume is plain, natural yogurt. This kind of yogurt is made only of milk and bacteria mixtures, which are sometimes referred to as “starter cultures.”

Furthermore, fermented soybean milk may promote the growth of beneficial bacteria, such as Bifidobacteria and lactobacilli, while decreasing quantities of some other disease-causing bacteria.

Kimchi may also benefit the gut flora (21Trusted Source, 22Trusted Source).

BOTTOM LINE:

Fermented foods, particularly plain, natural yogurt, can benefit the microbiota by enhancing its function and reducing the abundance of disease-causing bacteria in the intestines.

4. Mobidogenic impact of Aspertame and its analogues (artificial sweetener) in sodas and beverages is largely dysbiosis mediate. It is reasonably presumed that artificial sweeteners induce diabesity and promote inflammaging through harming the enteric microbiome. The modest weight decline of reduced sugar caloric intake tends to be concealed by the insulin resistance enhancing morbidogenic impact of dysbiosis. Enteric preponderance of morbidogenic micro-organisms namely Clostridium and Enterobacteriaceae tends to be associated with inflammaging incrementing.

Palmnas et al published Low-dose aspartame consumption differentially affects gut microbiota-host metabolic interactions in the diet-induced obese rat in PLoS One 2014 Oct 14;9(10):e109841. https://doi.org/10.1371/journal.pone.0109841

Aspartame consumption is implicated in the development of obesity and metabolic disease despite the intention of limiting caloric intake. The mechanisms responsible for this association remain unclear, but may involve circulating metabolites and the gut microbiota. Aims were to examine the impact of chronic low-dose aspartame consumption on anthropometric, metabolic and microbial parameters in a diet-induced obese model. Male Sprague-Dawley rats were randomized into a standard chow diet (CH, 12% kcal fat) or high fat (HF, 60% kcal fat) and further into ad libitum water control (W) or low-dose aspartame (A, 5-7 mg/kg/d in drinking water) treatments for 8 week (n = 10-12 animals/treatment). Animals on aspartame consumed fewer calories, gained less weight and had a more favorable body composition when challenged with HF compared to animals consuming water. Despite this, aspartame elevated fasting glucose levels and an insulin tolerance test showed aspartame to impair insulin-stimulated glucose disposal in both CH and HF, independently of body composition. Fecal analysis of gut bacterial composition showed aspartame to increase total bacteria, the abundance of Enterobacteriaceae and Clostridium leptum. An interaction between HF and aspartame was also observed for Roseburia ssp wherein HF-A was higher than HF-W (P<0.05). Within HF, aspartame attenuated the typical HF-induced increase in the Firmicutes:Bacteroidetes ratio. Serum metabolomics analysis revealed aspartame to be rapidly metabolized and to be associated with elevations in the short chain fatty acid propionate, a bacterial end product and highly gluconeogenic substrate, potentially explaining its negative affects on insulin tolerance. How aspartame influences gut microbial composition and the implications of these changes on the development of metabolic disease require further investigation.

Inulins

Prebiotics are dietary items that promote the growth of beneficial microbes in the gut particularly Bifidobacteria. Indigestible highly complex digestion resistant starches found in what is included under the dietary definition of fibre tend to act as powerful SEMDAR agents and are categorizes under hard probiotics while fermented diary products that contain beneficial bacteria can be termed soft probiotics.

Eubiotics bring about healthspan promoting changes recognized by the biomarkers like lowered insulin, triglycerides and cholesterol levels that are particularly relevant for those afflicted with metabolic syndrome.

Dewulf et al published Insight into the prebiotic concept: lessons from an exploratory, double blind intervention study with inulin-type fructans in obese women in Gut Aug 2013;62(8):1112-21. doi: 10.1136/gutjnl-2012-303304 reporting that ITF prebiotics selectively changed the gut microbiota composition in obese women, leading to modest changes in host metabolism, as suggested by the correlation between some bacterial species and metabolic endotoxaemia or metabolomic signatures.

Objective: To highlight the contribution of the gut microbiota to the modulation of host metabolism by dietary inulin-type fructans (ITF prebiotics) in obese women.

Methods: A double blind, placebo controlled, intervention study was performed with 30 obese women treated with ITF prebiotics (inulin/oligofructose 50/50 mix; n=15) or placebo (maltodextrin; n=15) for 3 months (16 g/day). Blood, faeces and urine sampling, oral glucose tolerance test, homeostasis model assessment and impedancemetry were performed before and after treatment. The gut microbial composition in faeces was analysed by phylogenetic microarray and qPCR analysis of 16S rDNA. Plasma and urine metabolic profiles were analysed by 1H-NMR spectroscopy.

Treatment with ITF prebiotics, but not the placebo, led to an increase in Bifidobacterium and Faecalibacterium prausnitzii; both bacteria negatively correlated with serum lipopolysaccharide levels. ITF prebiotics also decreased Bacteroides intestinalis, Bacteroides vulgatus and Propionibacterium, an effect associated with a slight decrease in fat mass and with plasma lactate and phosphatidylcholine levels. No clear treatment clustering could be detected for gut microbial analysis or plasma and urine metabolomic profile analyses. However, ITF prebiotics led to subtle changes in the gut microbiota that may importantly impact on several key metabolites implicated in obesity and/or diabetes.

Vulevic et al published A mixture of trans-galactooligosaccharides reduces markers of metabolic syndrome and modulates the fecal microbiota and immune function of overweight adults in Journal of Nutrition 2013 Mar;143(3):324-31. doi: 10.3945/jn.112.166132.

Metabolic syndrome is a set of disorders that increases the risk of developing cardiovascular disease. The gut microbiota is altered toward a less beneficial composition in overweight adults and this change can be accompanied by inflammation. Prebiotics such as galactooligosaccharides can positively modify the gut microbiota and immune system; some may also reduce blood lipids. We assessed the effect of a galactooligosaccharide mixture [Bi2muno (B-GOS)] on markers of metabolic syndrome, gut microbiota, and immune function in 45 overweight adults with ≥3 risk factors associated with metabolic syndrome in a double-blind, randomized, placebo (maltodextrin)-controlled, crossover study (with a 4-wk wash-out period between interventions). Whole blood, saliva, feces, and anthropometric measurements were taken at the beginning, wk 6, and end of each 12-wk intervention period. Predominant groups of fecal bacteria were quantified and full blood count, markers of inflammation and lipid metabolism, insulin, and glucose were measured. B-GOS increased the number of fecal bifidobacteria at the expense of less desirable groups of bacteria. Increases in fecal secretory IgA and decreases in fecal calprotectin, plasma C-reactive protein, insulin, total cholesterol (TC), TG, and the TC:HDL cholesterol ratio were also observed. Administration of B-GOS to overweight adults resulted in positive effects on the composition of the gut microbiota, the immune response, and insulin, TC, and TG concentrations. B-GOS may be a useful candidate for the enhancement of gastrointestinal health, immune function, and the reduction of metabolic syndrome risk factors in overweight adults.

Parnell & Reimer published Prebiotic fiber modulation of the gut microbiota improves risk factors for obesity and the metabolic syndrome in Gut Microbes Jan-Feb 2012;3(1):29-34. doi: 10.4161/gmic.19246.

Prebiotic fibers are non-digestible carbohydrates that promote the growth of beneficial bacteria in the gut. Prebiotic consumption may benefit obesity and associated co-morbidities by improving or normalizing the dysbiosis of the gut microbiota. We evaluated the dose response to a prebiotic diet on the gut microbiota, body composition and obesity associated risk factors in lean and genetically obese rats. Prebiotic fibers increased Firmicutes and decreased Bacteroidetes, a profile often associated with a leaner phenotype. Bifidobacteria and Lactobacillus numbers also increased. Changes in the gut microbiota correlated with energy intake, glucose, insulin, satiety hormones, and hepatic cholesterol and triglyceride accumulation. Here we provide a comprehensive analysis evaluating the results through the lens of the gut microbiota. Salient, new developments impacting the interpretation and significance of our data are discussed. We propose that prebiotic fibers have promise as a safe and cost-effective means of modulating the gut microbiota to promote improved host:bacterial interactions in obesity and insulin resistance. Human clinical trials should be undertaken to confirm these effects.

Davis et al published A dose dependent impact of prebiotic galactooligosaccharides on the intestinal microbiota of healthy adults in International Journal of Food Microbiology Dec 2010 15;144(2):285-92. doi: 10.1016/j.ijfoodmicro.2010.10.007.

The goal of this research was to determine the effect of different doses of galactooligosaccharide (GOS) on the fecal microbiota of healthy adults, with a focus on bifidobacteria. The study was designed as a single-blinded study, with eighteen subjects consuming GOS-containing chocolate chews at four increasing dosage levels; 0, 2.5, 5.0, and 10.0g. Subjects consumed each dose for 3 weeks, with a two-week baseline period preceding the study and a two-week washout period at the end. Fecal samples were collected weekly and analyzed by cultural and molecular methods. Cultural methods were used for bifidobacteria, Bacteroides, enterobacteria, enterococci, lactobacilli, and total anaerobes; culture-independent methods included denaturing gradient gel electrophoresis (DGGE) and quantitative real-time PCR (qRT-PCR) using Bifidobacterium-specific primers. All three methods revealed an increase in bifidobacteria populations, as the GOS dosage increased to 5 or 10g. Enumeration of bifidobacteria by qRT-PCR showed a high inter-subject variation in bifidogenic effect and indicated a subset of 9 GOS responders among the eighteen subjects. There were no differences, however, in the initial levels of bifidobacteria between the responding individuals and the non-responding individuals. Collectively, this study showed that a high purity GOS, administered in a confection product at doses of 5g or higher, was bifidogenic, while a dose of 2.5g showed no significant effect. However, the results also showed that even when GOS was administered for many weeks and at high doses, there were still some individuals for which a bifidogenic response did not occur.

Ramirez-Farias et al published Effect of inulin on the human gut microbiota: stimulation of Bifidobacterium adolescentis and Faecalibacterium prausnitzii in British Journal of Nutrition 2009 Feb;101(4):541-50. doi: 10.1017/S0007114508019880.

Prebiotics are food ingredients that improve health by modulating the colonic microbiota. The bifidogenic effect of the prebiotic inulin is well established; however, it remains unclear which species of Bifidobacterium are stimulated in vivo and whether bacterial groups other than lactic acid bacteria are affected by inulin consumption. Changes in the faecal microbiota composition were examined by real-time PCR in twelve human volunteers after ingestion of inulin (10 g/d) for a 16-d period in comparison with a control period without any supplement intake. The prevalence of most bacterial groups examined did not change after inulin intake, although the low G+C % Gram-positive species Faecalibacterium prausnitzii exhibited a significant increase (10.3% for control period v. 14.5% during inulin intake, P=0.019). The composition of the genus Bifidobacterium was studied in four of the volunteers by clone library analysis. Between three and five Bifidobacterium spp. were found in each volunteer. Bifidobacterium adolescentis and Bifidobacterium longum were present in all volunteers, and Bifidobacterium pseudocatenulatum, Bifidobacterium animalis, Bifidobacterium bifidum and Bifidobacterium dentium were also detected. Real-time PCR was employed to quantify the four most prevalent Bifidobacterium spp., B. adolescentis, B. longum, B. pseudocatenulatum and B. bifidum, in ten volunteers carrying detectable levels of bifidobacteria. B. adolescentis showed the strongest response to inulin consumption, increasing from 0.89 to 3.9% of the total microbiota (P=0.001). B. bifidum was increased from 0.22 to 0.63% (P<0.001) for the five volunteers for whom this species was present.

Holscher et al published Agave Inulin Supplementation Affects the Fecal Microbiota of Healthy Adults Participating in a Randomized, Double-Blind, Placebo-Controlled, Crossover Trial in Journal of Nutrition 2015 Sep;145(9):2025-32. doi: 10.3945/jn.115.217331.

reporting that agave inulin supplementation shifted the gastrointestinal microbiota composition and activity in healthy adults. Further investigation is warranted to determine whether the observed changes translate into health benefits in human populations.

Prebiotics resist digestion, providing fermentable substrates for select gastrointestinal bacteria associated with health and well-being. Agave inulin differs from other inulin type fibers in chemical structure and botanical origin. Preclinical animal research suggests these differences affect bacterial utilization and physiologic outcomes. Thus, research is needed to determine whether these effects translate to healthy adults.

Objective: We evaluated agave inulin utilization by the gastrointestinal microbiota by measuring fecal fermentative end products and bacterial taxa.

Methods: A randomized, double-blind, placebo-controlled, 3-period, crossover trial was undertaken in healthy adults (n = 29). Participants consumed 0, 5.0, or 7.5 g agave inulin/d for 21 d with 7-d washouts between periods. Participants recorded daily dietary intake; fecal samples were collected during days 16-20 of each period and were subjected to fermentative end product analysis and 16S Illumina sequencing.

Results: Fecal Actinobacteria and Bifidobacterium were enriched (P < 0.001) 3- and 4-fold after 5.0 and 7.5 g agave inulin/d, respectively, compared with control. Desulfovibrio were depleted 40% with agave inulin compared with control. Agave inulin tended (P < 0.07) to reduce fecal 4-methyphenol and pH. Bivariate correlations revealed a positive association between intakes of agave inulin (g/kcal) and Bifidobacterium (r = 0.41, P < 0.001). Total dietary fiber intake (total fiber plus 0, 5.0, or 7.5 g agave inulin/d) per kilocalorie was positively associated with fecal butyrate (r = 0.30, P = 0.005), tended to be positively associated with Bifidobacterium (r = 0.19, P = 0.08), and was negatively correlated with Desulfovibrio abundance (r = -0.31, P = 0.004).

Holscher et al published Agave Inulin Supplementation Affects the Fecal Microbiota of Healthy Adults Participating in a Randomized, Double-Blind, Placebo-Controlled, Crossover Trial in Journal of Nutrition 2015 Sep;145(9):2025-32. doi: 10.3945/jn.115.217331. http://doi: 10.3945/jn.115.217331

Reporting that agave inulin supplementation shifted the gastrointestinal microbiota composition and activity in healthy adults. Further investigation is warranted to determine whether the observed changes translate into health benefits in human populations. This trial was registered at clinicaltrials.gov as NCT01925560.

Background: Prebiotics resist digestion, providing fermentable substrates for select gastrointestinal bacteria associated with health and well-being. Agave inulin differs from other inulin type fibers in chemical structure and botanical origin. Preclinical animal research suggests these differences affect bacterial utilization and physiologic outcomes. Thus, research is needed to determine whether these effects translate to healthy adults.

Objective: We evaluated agave inulin utilization by the gastrointestinal microbiota by measuring fecal fermentative end products and bacterial taxa.

Methods: A randomized, double-blind, placebo-controlled, 3-period, crossover trial was undertaken in healthy adults (n = 29). Participants consumed 0, 5.0, or 7.5 g agave inulin/d for 21 d with 7-d washouts between periods. Participants recorded daily dietary intake; fecal samples were collected during days 16-20 of each period and were subjected to fermentative end product analysis and 16S Illumina sequencing.

Results: Fecal Actinobacteria and Bifidobacterium were enriched (P < 0.001) 3- and 4-fold after 5.0 and 7.5 g agave inulin/d, respectively, compared with control. Desulfovibrio were depleted 40% with agave inulin compared with control. Agave inulin tended (P < 0.07) to reduce fecal 4-methyphenol and pH. Bivariate correlations revealed a positive association between intakes of agave inulin (g/kcal) and Bifidobacterium (r = 0.41, P < 0.001). Total dietary fiber intake (total fiber plus 0, 5.0, or 7.5 g agave inulin/d) per kilocalorie was positively associated with fecal butyrate (r = 0.30, P = 0.005), tended to be positively associated with Bifidobacterium (r = 0.19, P = 0.08), and was negatively correlated with Desulfovibrio abundance (r = -0.31, P = 0.004).

Gibson et al published Selective stimulation of bifidobacteria in the human colon by oligofructose and inulin in Gastroenterology 1995 Apr;108(4):975-82. doi: 10.1016/0016-5085(95)90192-2. reporting that A 15-g.day-1 dietary addition of oligofructose or inulin led to Bifidobacterium becoming the numerically predominant genus in feces. Thus, small changes in diet can alter the balance of colonic bacteria towards a potentially healthier microflora.

Oligofructose and inulin are naturally occurring indigestible carbohydrates. In vitro they selectively stimulate the growth of species of Bifidobacterium, a genus of bacteria considered beneficial to health. This study was designed to determine their effects on the large bowel microflora and colonic function in vivo.

Methods: Eight subjects participated in a 45-day study during which they ate controlled diets. For the middle 15 days, 15 g.day-1 oligofructose was substituted for 15 g.day-1 sucrose. Four of these subjects went on to a further period with 15 g.day-1 inulin. Bowel habit, transit time, stool composition, breath H2 and CH4, and the predominant genera of colonic bacteria were measured.

Results: Both oligofructose and inulin significantly increased bifidobacteria from 8.8 to 9.5 log10 g stool-1 and 9.2 to 10.1 log10 g stool-1, respectively, whereas bacteroides, clostridia, and fusobacteria decreased when subjects were fed oligofructose, and gram-positive cocci decreased when subjects were fed inulin. Total bacterial counts were unchanged. Fecal wet and dry matter, nitrogen, and energy excretion increased with both substrates, as did breath H2. Little change in fecal short-chain fatty acids and breath CH4 was observed.

Holscher et al published Agave Inulin Supplementation Affects the Fecal Microbiota of Healthy Adults Participating in a Randomized, Double-Blind, Placebo-Controlled, Crossover Trial in Journal of Nutrition 2015 Sep;145(9):2025-32. doi: 10.3945/jn.115.217331 reporting that agave inulin supplementation shifted the gastrointestinal microbiota composition and activity in healthy adults. Further investigation is warranted to determine whether the observed changes translate into health benefits in human populations

Prebiotics resist digestion, providing fermentable substrates for select gastrointestinal bacteria associated with health and well-being. Agave inulin differs from other inulin type fibers in chemical structure and botanical origin. Preclinical animal research suggests these differences affect bacterial utilization and physiologic outcomes. Thus, research is needed to determine whether these effects translate to healthy adults. We evaluated agave inulin utilization by the gastrointestinal microbiota by measuring fecal fermentative end products and bacterial taxa. A randomized, double-blind, placebo-controlled, 3-period, crossover trial was undertaken in healthy adults (n = 29). Participants consumed 0, 5.0, or 7.5 g agave inulin/d for 21 d with 7-d washouts between periods. Participants recorded daily dietary intake; fecal samples were collected during days 16-20 of each period and were subjected to fermentative end product analysis and 16S Illumina sequencing.

Fecal Actinobacteria and Bifidobacterium were enriched (P < 0.001) 3- and 4-fold after 5.0 and 7.5 g agave inulin/d, respectively, compared with control. Desulfovibrio were depleted 40% with agave inulin compared with control. Agave inulin tended (P < 0.07) to reduce fecal 4-methyphenol and pH. Bivariate correlations revealed a positive association between intakes of agave inulin (g/kcal) and Bifidobacterium (r = 0.41, P < 0.001). Total dietary fiber intake (total fiber plus 0, 5.0, or 7.5 g agave inulin/d) per kilocalorie was positively associated with fecal butyrate (r = 0.30, P = 0.005), tended to be positively associated with Bifidobacterium (r = 0.19, P = 0.08), and was negatively correlated with Desulfovibrio abundance (r = -0.31, P = 0.004).

Gibson et al published Selective stimulation of bifidobacteria in the human colon by oligofructose and inulin in Gastroenterology. 1995 Apr;108(4):975-82. doi: 10.1016/0016-5085(95)90192-2.

A 15-g.day-1 dietary addition of oligofructose or inulin led to Bifidobacterium becoming the numerically predominant genus in feces. Thus, small changes in diet can alter the balance of colonic bacteria towards a potentially healthier microflora.

Oligofructose and inulin are naturally occurring indigestible carbohydrates. In vitro they selectively stimulate the growth of species of Bifidobacterium, a genus of bacteria considered beneficial to health. This study was designed to determine their effects on the large bowel microflora and colonic function in vivo.

Methods: Eight subjects participated in a 45-day study during which they ate controlled diets. For the middle 15 days, 15 g.day-1 oligofructose was substituted for 15 g.day-1 sucrose. Four of these subjects went on to a further period with 15 g.day-1 inulin. Bowel habit, transit time, stool composition, breath H2 and CH4, and the predominant genera of colonic bacteria were measured.

Results: Both oligofructose and inulin significantly increased bifidobacteria from 8.8 to 9.5 log10 g stool-1 and 9.2 to 10.1 log10 g stool-1, respectively, whereas bacteroides, clostridia, and fusobacteria decreased when subjects were fed oligofructose, and gram-positive cocci decreased when subjects were fed inulin. Total bacterial counts were unchanged. Fecal wet and dry matter, nitrogen, and energy excretion increased with both substrates, as did breath H2. Little change in fecal short-chain fatty acids and breath CH4 was observed.

These results suggest that prebiotics may reduce the risk factors for many diseases associated with obesity, including heart disease and diabetes.

BOTTOM LINE:

Prebiotics promote the growth of beneficial bacteria, especially Bifidobacteria. This may help reduce symptoms of metabolic syndrome in obese people.

Boosting consumption of Whole Grains (preferably germinated or eat as whole without milling)

as beta-glucan which behaves like cellulose found in grass tends not to be digested and absorbed in jejunum and to some extent not even in colon where the glucan digestion is engaged in by the microbiome but human enzyme and there is no energy derived from these. Glucans are the nutritional substrate for the microbiota and promote the growth of the healthspan promoting micro organisms.

Whole grains tend to boost of Bifidobacteria, lactobacilli and Bacteroidetes preponderance in humans

Costabile et al published Whole-grain wheat breakfast cereal has a prebiotic effect on the human gut microbiota: a double-blind, placebo-controlled, crossover study in British Journal of Nutrition Jan 2008;99(1):110-20. doi: 10.1017/S0007114507793923. Epidemiological studies have shown an inverse association between dietary intake of whole grains and the risk of chronic disease. This may be related to the ability to mediate a prebiotic modulation of gut microbiota. However, no studies have been conducted on the microbiota modulatory capability of whole-grain (WG) cereals. In the present study, the impact of WG wheat on the human intestinal microbiota compared to wheat bran (WB) was determined. A double-blind, randomised, crossover study was carried out in thirty-one volunteers who were randomised into two groups and consumed daily 48 g breakfast cereals, either WG or WB, in two 3-week study periods, separated by a 2-week washout period. Numbers of faecal bifidobacteria and lactobacilli (the target genera for prebiotic intake), were significantly higher upon WG ingestion compared with WB. Ingestion of both breakfast cereals resulted in a significant increase in ferulic acid concentrations in blood but no discernible difference in faeces or urine. No significant differences in faecal SCFA, fasting blood glucose, insulin, total cholesterol (TC), TAG or HDL-cholesterol were observed upon ingestion of WG compared with WB. However, a significant reduction in TC was observed in volunteers in the top quartile of TC concentrations upon ingestion of either cereal. No adverse intestinal symptoms were reported and WB ingestion increased stool frequency. Daily consumption of WG wheat exerted a pronounced prebiotic effect on the human gut microbiota composition. This prebiotic activity may contribute towards the beneficial physiological effects of WG wheat.

Martínez et al published Gut microbiome composition is linked to whole grain-induced immunological improvements in ISME 2013 Feb;7(2):269-80. doi: 10.1038/ismej.2012.104. Epub 2012 Oct 4.

The involvement of the gut microbiota in metabolic disorders, and the ability of whole grains to affect both host metabolism and gut microbial ecology, suggest that some benefits of whole grains are mediated through their effects on the gut microbiome. Nutritional studies that assess the effect of whole grains on both the gut microbiome and human physiology are needed. We conducted a randomized cross-over trial with four-week treatments in which 28 healthy humans consumed a daily dose of 60 g of whole-grain barley (WGB), brown rice (BR), or an equal mixture of the two (BR+WGB), and characterized their impact on fecal microbial ecology and blood markers of inflammation, glucose and lipid metabolism. All treatments increased microbial diversity, the Firmicutes/Bacteroidetes ratio, and the abundance of the genus Blautia in fecal samples. The inclusion of WGB enriched the genera Roseburia, Bifidobacterium and Dialister, and the species Eubacterium rectale, Roseburia faecis and Roseburia intestinalis. Whole grains, and especially the BR+WGB treatment, reduced plasma interleukin-6 (IL-6) and peak postprandial glucose. Shifts in the abundance of Eubacterium rectale were associated with changes in the glucose and insulin postprandial response. Interestingly, subjects with greater improvements in IL-6 levels harbored significantly higher proportions of Dialister and lower abundance of Coriobacteriaceae. In conclusion, this study revealed that a short-term intake of whole grains induced compositional alterations of the gut microbiota that coincided with improvements in host physiological measures related to metabolic dysfunctions in humans.

Wang et al published High Molecular Weight Barley β-Glucan Alters Gut Microbiota Toward Reduced Cardiovascular Disease Risk in Frontiers of Microbiology Feb 2016 10;7:129. doi: 10.3389/fmicb.2016.00129 The physiological cholesterol-lowering benefits of β-glucan have been well documented, however, whether modulation of gut microbiota by β-glucan is associated with these physiological effects remains unknown. The objectives of this study were therefore to determine the impact of β-glucan on the composition of gut microbiota in mildly hypercholesterolemic individuals and to identify if the altered microbiota are associated with bioactivity of β-glucan in improving risk factors of cardiovascular disease (CVD). Using a randomized, controlled crossover study design, individuals received for 5-week either a treatment breakfast containing 3 g high molecular weight (HMW), 3 g low molecular weight (LMW), 5 g LMW barley β-glucan, or wheat and rice. The American Heart Association (AHA) diet served as the background diet for all treatment groups. Phases were separated by 4-week washout periods. Fecal samples were collected at the end of each intervention phase and subjected to Illumina sequencing of 16S rRNA genes. Results revealed that at the phylum level, supplementation of 3 g/d HMW β-glucan increased Bacteroidetes and decreased Firmicutes abundances compared to control (P < 0.001). At the genus level, consumption of 3 g/d HMW β-glucan increased Bacteroides (P < 0.003), tended to increase Prevotella (P < 0.1) but decreased Dorea (P < 0.1), whereas diets containing 5 g LMW β-glucan and 3 g LMW β-glucan failed to alter the gut microbiota composition. Bacteroides, Prevotella, and Dorea composition correlated (P < 0.05) with shifts of CVD risk factors, including body mass index, waist circumference, blood pressure, as well as triglyceride levels. Our data suggest that consumption of HMW β-glucan favorably alters the composition of gut microbiota and this altered microbiota profile associates with a reduction of CVD risk markers. Together, our study suggests that β-glucan induced shifts in gut microbiota in a MW-dependent manner and that might be one of the underlying mechanisms responsible for the physiological benefits of β-glucan.

Carvalho-Wells et al published Determination of the in vivo prebiotic potential of a maize-based whole grain breakfast cereal: a human feeding study British Journal of Nutrition Nov 2010;104(9):1353-6. doi: 10.1017/S0007114510002084.

Epidemiological studies have shown an inverse relationship between risk of CVD and intake of whole grain (WG)-rich food. Regular consumption of breakfast cereals can provide not only an increase in dietary WG but also improvements to cardiovascular health. Various mechanisms have been proposed, including prebiotic modulation of the colonic microbiota. In the present study, the prebiotic activity of a maize-derived WG cereal (WGM) was evaluated in a double-blind, placebo-controlled human feeding study (n 32). For a period of 21 d, healthy men and women, mean age 32 (sd 8) years and BMI 23·3 (sd 0·58) kg/m2, consumed either 48 g/d WG cereal (WGM) or 48 g placebo cereal (non-whole grain (NWG)) in a crossover fashion. Faecal samples were collected at five points during the study on days 0, 21, 42, 63 and 84 (representing at baseline, after both treatments and both wash-out periods). Faecal bacteriology was assessed using fluorescence in situ hybridisation with 16S rRNA oligonucleotide probes specific for Bacteroides spp., Bifidobacterium spp., Clostridium histolyticum/perfringens subgroup, Lactobacillus-Enterococcus subgroup and total bacteria. After 21 d consumption of WGM, mean group levels of faecal bifidobacteria increased significantly compared with the control cereal (P = 0·001). After a 3-week wash-out period, bifidobacterial levels returned to pre-intervention levels. No statistically significant changes were observed in serum lipids, glucose or measures of faecal output. In conclusion, this WG maize-enriched breakfast cereal mediated a bifidogenic modulation of the gut microbiota, indicating a possible prebiotic mode of action.

Cooper et al published Does Whole Grain Consumption Alter Gut Microbiota and Satiety? in Healthcare (Basel). Jun 2015; 3(2): 364–392. doi: 10.3390/healthcare3020364

This review summarizes recent studies examining whole grain consumption and its effect on gut microbiota and satiety in healthy humans. Studies comparing whole grains to their refined grain counterparts were considered, as were studies comparing different grain types. Possible mechanisms linking microbial metabolism and satiety are described. Clinical trials show that whole grain wheat, maize, and barley alter the human gut microbiota, but these findings are based on a few studies that do not include satiety components, so no functional claims between microbiota and satiety can be made. Ten satiety trials were evaluated and provide evidence that whole oats, barley, and rye can increase satiety, whereas the evidence for whole wheat and maize is not compelling. There are many gaps in the literature; no one clinical trial has examined the effects of whole grains on satiety and gut microbiota together. Once understanding the impact of whole grains on satiety and microbiota is more developed, then particular grains might be used for better appetite control. With this information at hand, healthcare professionals could make individual dietary recommendations that promote satiety and contribute to weight control.

https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4939539/

In these studies, whole grains also increased feelings of fullness and reduced inflammation and heart disease risk factors.

Probiotic Preponderance Production via Picking Plant-Predominant Portions

While there is a reason to speculate the elimination or reduction of the highly processed meat products is the real reason vegetarian diets may benefit the gut microbiota. This may be due to their higher fiber contents. It is not necessary to become strict vegetarian or vegan for probiotic propensity production.

David et al published Diet rapidly and reproducibly alters the human gut microbiome in Nature 2014 Jan 23;505(7484):559-63. doi: 10.1038/nature12820. doi: 10.1038/nature12820 Demonstrating that the gut microbiome can rapidly respond to altered diet, potentially facilitating the diversity of human dietary lifestyles.

Long-term dietary intake influences the structure and activity of the trillions of microorganisms residing in the human gut, but it remains unclear how rapidly and reproducibly the human gut microbiome responds to short-term macronutrient change. Here we show that the short-term consumption of diets composed entirely of animal or plant products alters microbial community structure and overwhelms inter-individual differences in microbial gene expression. The animal-based diet increased the abundance of bile-tolerant microorganisms (Alistipes, Bilophila and Bacteroides) and decreased the levels of Firmicutes that metabolize dietary plant polysaccharides (Roseburia, Eubacterium rectale and Ruminococcus bromii). Microbial activity mirrored differences between herbivorous and carnivorous mammals, reflecting trade-offs between carbohydrate and protein fermentation. Foodborne microbes from both diets transiently colonized the gut, including bacteria, fungi and even viruses. Finally, increases in the abundance and activity of Bilophila wadsworthia on the animal-based diet support a link between dietary fat, bile acids and the outgrowth of microorganisms capable of triggering inflammatory bowel disease.

Comparative metabolomics

Wu et al published Comparative metabolomics in vegans and omnivores reveal constraints on diet-dependent gut microbiota metabolite production in Gut 2016 Jan;65(1):63-72. doi: 10.1136/gutjnl-2014-308209 reporting that residence in globally distinct societies helps determine the composition of the gut microbiota that, in turn, influences the production of diet-dependent gut microbial metabolites.The consumption of an agrarian diet is associated with a reduced risk for many diseases associated with a 'Westernised' lifestyle. Studies suggest that diet affects the gut microbiota, which subsequently influences the metabolome, thereby connecting diet, microbiota and health. However, the degree to which diet influences the composition of the gut microbiota is controversial. Murine models and studies comparing the gut microbiota in humans residing in agrarian versus Western societies suggest that the influence is large. To separate global environmental influences from dietary influences, we characterised the gut microbiota and the host metabolome of individuals consuming an agrarian diet in Western society. Design and results: Using 16S rRNA-tagged sequencing as well as plasma and urinary metabolomic platforms, we compared measures of dietary intake, gut microbiota composition and the plasma metabolome between healthy human vegans and omnivores, sampled in an urban USA environment. Plasma metabolome of vegans differed markedly from omnivores but the gut microbiota was surprisingly similar. Unlike prior studies of individuals living in agrarian societies, higher consumption of fermentable substrate in vegans was not associated with higher levels of faecal short chain fatty acids, a finding confirmed in a 10-day controlled feeding experiment. Similarly, the proportion of vegans capable of producing equol, a soy-based gut microbiota metabolite, was less than that was reported in Asian societies despite the high consumption of soy-based products.

bacteria in obese people, as well as reduced weight,

Inflammation promoting foods

https://www.healthline.com/nutrition/13-anti-inflammatory-foods

Studies have demonstrated a significant difference between the gut microbiomes of vegetarians and those of people who eat meat.

For example, in one small study, people with obesity followed a strict vegetarian diet that eliminated all animal products, including meat, dairy, and eggs, for a month.

At the end of the study, the participants had lower levels of inflammation due to the altered types of gut microbes. They also lost weight.

Kim et al published Strict vegetarian diet improves the risk factors associated with metabolic diseases by modulating gut microbiota and reducing intestinal inflammation in Environmental microbiology reports. 2013 Oct;5(5):765-75. doi: 10.1111/1758-2229.12079. Low-grade inflammation of the intestine results in metabolic dysfunction, in which dysbiosis of the gut microbiota is intimately involved. Dietary fibre induces prebiotic effects that may restore imbalances in the gut microbiota; however, no clinical trials have been reported in patients with metabolic diseases. Here, six obese subjects with type 2 diabetes and/or hypertension were assigned to a strict vegetarian diet (SVD) for 1 month, and blood biomarkers of glucose and lipid metabolisms, faecal microbiota using 454-pyrosequencing of 16S ribosomal RNA genes, faecal lipocalin-2 and short-chain fatty acids were monitored. An SVD reduced body weight and the concentrations of triglycerides, total cholesterol, low-density lipoprotein cholesterol and haemoglobin A1c, and improved fasting glucose and postprandial glucose levels. An SVD reduced the Firmicutes-to-Bacteroidetes ratio in the gut microbiota, but did not alter enterotypes. An SVD led to a decrease in the pathobionts such as the Enterobacteriaceae and an increase in commensal microbes such as Bacteroides fragilis and Clostridium species belonging to clusters XIVa and IV, resulting in reduced intestinal lipocalin-2 and short-chain fatty acids levels. This study underscores the benefits of dietary fibre for improving the risk factors of metabolic diseases and shows that increased fibre intake reduces gut inflammation by changing the gut microbiota.

Dozen Most Anti Inflammaging food items

https://www.healthline.com/nutrition/13-anti-inflammatory-foods#1

A vegetarian diet was observed to adversely impact the preponderance of morbidogenic E. coli

Zimmer et al published A vegan or vegetarian diet substantially alters the human colonic faecal microbiota in European Journal of Clinical Nutrition - Jan 2012;66(1):53-60. doi: 10.1038/ejcn.2011.141 concluding that in selected cases maintaining a strict vegan or vegetarian diet results in a significant shift in the microbiota while total cell numbers remain unaltered.

Background/objectives: Consisting of ≈10(14) microbial cells, the intestinal microbiota represents the largest and the most complex microbial community inhabiting the human body. However, the influence of regular diets on the microbiota is widely unknown.

Subjects/methods: We examined faecal samples of vegetarians (n=144), vegans (n=105) and an equal number of control subjects consuming ordinary omnivorous diet who were matched for age and gender. We used classical bacteriological isolation, identification and enumeration of the main anaerobic and aerobic bacterial genera and computed absolute and relative numbers that were compared between groups.

Results: Total counts of Bacteroides spp., Bifidobacterium spp., Escherichia coli and Enterobacteriaceae spp. were significantly lower (P=0.001, P=0.002, P=0.006 and P=0.008, respectively) in vegan samples than in controls, whereas others (E. coli biovars, Klebsiella spp., Enterobacter spp., other Enterobacteriaceae, Enterococcus spp., Lactobacillus spp., Citrobacter spp. and Clostridium spp.) were not. Subjects on a vegetarian diet ranked between vegans and controls. The total microbial count did not differ between the groups. In addition, subjects on a vegan or vegetarian diet showed significantly (P=0.0001) lower stool pH than did controls, and stool pH and counts of E. coli and Enterobacteriaceae were significantly correlated across all subgroups.

Another factor in need of correction or accounting for is that vegetarians tend to be more fitness conscious and often religious and lead healthier and better controlled lifestyles than omnivores or heavy carnivores. Whether the benefits of a vegetarian diet on the gut microbiota are simply due to refraining from meat consumption is unproven nor does the research target the level of processing of the meat consumed.

Vegetarian or predominantly plant based diets tend to boost SEMDAR but there is no conclusive evidence that their positive effects associated can be directly attributed to a low or missing meat intake.

Polyphenols are plant compounds that have many health benefits, including reductions in blood pressure, inflammation, cholesterol levels

Polyphenols from cocoa tend to reduce the gut content of injurious to wellness bacteria of Clostridia species and tend to promote wellness promoting bacteria of Bifidobacteria, lactobacilli etc., in human gut. Popular polyphenol sources : Cocoa and dark chocolate, Red wine, Grape skins. Green tea. Almonds, Onions, Blueberries, Broccoli etc. Human digestive enzymes are ineffective indigestion of polyphenols so they get transported to colon where they assist with SEMSAR by nourishing the gut bacteria.

Pandey and Rizvi published Plant polyphenols as dietary antioxidants in human health and disease in Oxidative Medicine and Cellular Longevity. 2009 Nov-Dec; 2(5): 270–278. doi: 10.4161/oxim.2.5.9498 pointing out that Polyphenols are secondary metabolites of plants and are generally involved in defense against ultraviolet radiation or aggression by pathogens. In the last decade, there has been much interest in the potential health benefits of dietary plant polyphenols as antioxidant. Epidemiological studies and associated meta-analyses strongly suggest that long term consumption of diets rich in plant polyphenols offer protection against development of cancers, cardiovascular diseases, diabetes, osteoporosis and neurodegenerative diseases. Here we present knowledge about the biological effects of plant polyphenols in the context of relevance to human health.

Cardona et al published a review Benefits of polyphenols on gut microbiota and implications in human health Journal of Nutritional Biochemistry. Aug 2013;24(8):1415-22. doi: 10.1016/j.jnutbio.2013.05.001 focuses on the reciprocal interactions between the gut microbiota and polyphenols, the mechanisms of action and the consequences of these interactions on human health.The biological properties of dietary polyphenols are greatly dependent on their bioavailability that, in turn, is largely influenced by their degree of polymerization. The gut microbiota play a key role in modulating the production, bioavailability and, thus, the biological activities of phenolic metabolites, particularly after the intake of food containing high-molecular-weight polyphenols. In addition, evidence is emerging on the activity of dietary polyphenols on the modulation of the colonic microbial population composition or activity. However, although the great range of health-promoting activities of dietary polyphenols has been widely investigated, their effect on the modulation of the gut ecology and the two-way relationship "polyphenols ↔ microbiota" are still poorly understood. Only a few studies have examined the impact of dietary polyphenols on the human gut microbiota, and most were focused on single polyphenol molecules and selected bacterial populations.

van Duynhoven et al published Metabolic fate of polyphenols in the human superorganism in Proceedings of the National Academy of Sciences of U. S. A. Mar 2011 15;108 Suppl 1(Suppl 1):4531-8. doi: 10.1073/pnas.1000098107. Epub 2010 Jun 25.

Dietary polyphenols are components of many foods such as tea, fruit, and vegetables and are associated with several beneficial health effects although, so far, largely based on epidemiological studies. The intact forms of complex dietary polyphenols have limited bioavailability, with low circulating levels in plasma. A major part of the polyphenols persists in the colon, where the resident microbiota produce metabolites that can undergo further metabolism upon entering systemic circulation. Unraveling the complex metabolic fate of polyphenols in this human superorganism requires joint deployment of in vitro and humanized mouse models and human intervention trials. Within these systems, the variation in diversity and functionality of the colonic microbiota can increasingly be captured by rapidly developing microbiomics and metabolomics technologies. Furthermore, metabolomics is coming to grips with the large biological variation superimposed on relatively subtle effects of dietary interventions. In particular when metabolomics is deployed in conjunction with a longitudinal study design, quantitative nutrikinetic signatures can be obtained. These signatures can be used to define nutritional phenotypes with different kinetic characteristics for the bioconversion capacity for polyphenols. Bottom-up as well as top-down approaches need to be pursued to link gut microbial diversity to functionality in nutritional phenotypes and, ultimately, to bioactivity of polyphenols. This approach will pave the way for personalization of nutrition based on gut microbial functionality of individuals or populations.

SEMSAR related enteric eubiosis is associated with inflammaging soothing decline in hostile triglycerides and also of inflammaging markers like C-reactive protein

Tzounis et al published Prebiotic evaluation of cocoa-derived flavanols in healthy humans by using a randomized, controlled, double-blind, crossover intervention study in American Journal of Clinical Nutrition. 2011 Jan;93(1):62-72. doi: 10.3945/ajcn.110.000075 demonstrating that consumption of cocoa flavanols tend to positively impact the growth of select gut microflora in humans, which suggests the potential prebiotic benefits associated with the dietary inclusion of flavanol-rich foods. The absorption of cocoa flavanols in the small intestine is limited, and the majority of the flavanols reach the large intestine where they may be metabolized by resident microbiota. We assessed the prebiotic potential of cocoa flavanols in a randomized, double-blind, crossover, controlled intervention study. Twenty-two healthy human volunteers were randomly assigned to either a high-cocoa flavanol (HCF) group (494 mg cocoa flavanols/d) or a low-cocoa flavanol (LCF) group (23 mg cocoa flavanols/d) for 4 wk. This was followed by a 4-wk washout period before volunteers crossed to the alternant arm. Fecal samples were recovered before and after each intervention, and bacterial numbers were measured by fluorescence in situ hybridization. A number of other biochemical and physiologic markers were measured.

Results: Compared with the consumption of the LCF drink, the daily consumption of the HCF drink for 4 wk significantly increased the bifidobacterial (P < 0.01) and lactobacilli (P < 0.001) populations but significantly decreased clostridia counts (P < 0.001). These microbial changes were paralleled by significant reductions in plasma triacylglycerol (P < 0.05) and C-reactive protein (P < 0.05) concentrations. Furthermore, changes in C-reactive protein concentrations were linked to changes in lactobacilli counts (P < 0.05, R(2) = -0.33 for the model). These in vivo changes were closely paralleled by cocoa flavanol-induced bacterial changes in mixed-batch culture experiments.

Enteric eubiotic impact of red wine polyphenols

Queipo-Ortuño et al published Influence of red wine polyphenols and ethanol on the gut microbiota ecology and biochemical biomarkers in American Journal of Clinical Nutrition. 2012 Jun;95(6):1323-34. doi: 10.3945/ajcn.111.027847. concluding that red wine consumption can significantly modulate the growth of select gut microbiota in humans, which suggests possible prebiotic benefits associated with the inclusion of red wine polyphenols in the diet.

Background: Few studies have investigated the effect of dietary polyphenols on the complex human gut microbiota, and they focused mainly on single polyphenol molecules and select bacterial populations.

Objective: The objective was to evaluate the effect of a moderate intake of red wine polyphenols on select gut microbial groups implicated in host health benefits.

Design: Ten healthy male volunteers underwent a randomized, crossover, controlled intervention study. After a washout period, all of the subjects received red wine, the equivalent amount of de-alcoholized red wine, or gin for 20 d each. Total fecal DNA was submitted to polymerase chain reaction(PCR)-denaturing gradient gel electrophoresis and real-time quantitative PCR to monitor and quantify changes in fecal microbiota. Several biochemical markers were measured.

Results: The dominant bacterial composition did not remain constant over the different intake periods. Compared with baseline, the daily consumption of red wine polyphenol for 4 wk significantly increased the number of Enterococcus, Prevotella, Bacteroides, Bifidobacterium, Bacteroides uniformis, Eggerthella lenta, and Blautia coccoides-Eubacterium rectale groups (P < 0.05). In parallel, systolic and diastolic blood pressures and triglyceride, total cholesterol, HDL cholesterol, and C-reactive protein concentrations decreased significantly (P < 0.05). Moreover, changes in cholesterol and C-reactive protein concentrations were linked to changes in the bifidobacteria number.

Probiosis (eubiosis) promoters or sidekicks. Polyphenols can’t be digested efficiently by human cells, but they are efficiently broken down by the gut microbiota. The end result from SEMDAR is that of improving health outcomes related to inflammaging associated conditions and mortality..

10. Probiotic Supplement to be consumed only with medical prescription.

The biological contents of the probiotic supplements tend not to colonize the gut on long term basis but bring about their beneficial effects via modifying the pH and other aspects of the gut chemistry that helps with SEMDAR

Mary Ellen Sanders published Impact of probiotics on colonizing microbiota of the gut in Journal of Clinical Gastroenterol. Nov 2011;45 Suppl:S115-9. doi: 10.1097/MCG.0b013e318227414a. Although precise mechanisms responsible for all demonstrations of probiotic health benefits are not known, many lines of evidence suggest that probiotics function through direct or indirect impact on colonizing microbiota of the gut. Probiotics can directly influence colonizing microbes through multiple mechanisms, including the production of inhibitory compounds (bacteriocins, short chain fatty acids, and others), by producing substrates that might promote the growth of colonizing microbes (secreted exopolysaccharides, vitamins, fatty acids, sugars from undigested carbohydrates and others), and by promoting immune responses against specific microbes. Indirectly, probiotics can influence colonizing microbes by inhibiting attachment through stimulated mucin production, reinforcing gut barrier effects, and downregulation of gut inflammation, thereby promoting microbes that are associated with a healthier gut physiology. Although the value of targeted changes in populations of gut bacteria is a matter of debate, increased levels of Bifidobacterium and Lactobacillus in the gut correlate with numerous health endpoints. Microbiota changes due to probiotic intake include increased numbers of related phylotypes, decreasing pathogens and their toxins, altering bacterial community structure to enhance evenness, stabilizing bacterial communities when perturbed (eg, with antibiotics), or promoting a more rapid recovery from a perturbation. Further research will provide insight into the degree of permanence of probiotic-induced changes, although research to date suggests that continued probiotic consumption is needed for sustained impact.

There does exist some evidence that supports that probiotics tend to help SEMDAR in certain diseases.

Kristensen et al published a review Alterations in fecal microbiota composition by probiotic supplementation in healthy adults: a systematic review of randomized controlled trials published in Genome Medicine . 2016 May 10;8(1):52. doi: 10.1186/s13073-016-0300-5 concluding that a good review of literature demonstrates a lack of evidence for impact of probiotics on fecal microbiota composition in healthy adults. Future studies would benefit from pre-specifying the primary outcome and transparently reporting the results including effect sizes, confidence intervals, and P values as well as providing a clear distinction of between-group and within-group comparisons.

Background: The effects of probiotic supplementation on fecal microbiota composition in healthy adults have not been well established. We aimed to provide a systematic review of the potential evidence for an effect of probiotic supplementation on the composition of human fecal microbiota as assessed by high-throughput molecular approaches in randomized controlled trials (RCTs) of healthy adults.

Methods: The survey of peer-reviewed papers was performed on 17 August 2015 by a literature search through PubMed, SCOPUS, and ISI Web of Science. Additional papers were identified by checking references of relevant papers. Search terms included healthy adult, probiotic, bifidobacterium, lactobacillus, gut microbiota, fecal microbiota, intestinal microbiota, intervention, and (clinical) trial. RCTs of solely probiotic supplementation and placebo in healthy adults that examined alteration in composition of overall fecal microbiota structure assessed by shotgun metagenomic sequencing, 16S ribosomal RNA sequencing, or phylogenetic microarray methods were included. Independent collection and quality assessment of studies were performed by two authors using predefined criteria including methodological quality assessment of reports of the clinical trials based on revised tools from PRISMA/Cochrane and by the Jadad score.

Seven RCTs investigating the effect of probiotic supplementation on fecal microbiota in healthy adults were identified and included in the present systematic review. The quality of the studies was assessed as medium to high. Still, no effects were observed on the fecal microbiota composition in terms of α-diversity, richness, or evenness in any of the included studies when compared to placebo. Only one study found that probiotic supplementation significantly modified the overall structure of the fecal bacterial community in terms of β-diversity when compared to placebo.

Probiotics improve the performance of gut microbiome bacteria function, as well as the types of chemicals like neurotransmitters, endocrines and other products they release into the enteric submucosa and then into blood stream which impacts the main nervous system the enteric nervous system and regulate the rate of inflammaging..

Probiotics have not been shown to materially alter the composition and the chemical output of the microbiota in fit and healthy people but they do have a limited role in in sick elderly with Non-communicable Disease (NCD) and can help with the restitution component of SEMDAR.

Eloe-Fadrosh et al published Functional dynamics of the gut microbiome in elderly people during probiotic consumption in mBio 2015 Apr 14;6(2):e00231-15. doi: 10.1128/mBio.00231-15.

A mechanistic understanding of the purported health benefits conferred by consumption of probiotic bacteria has been limited by our knowledge of the resident gut microbiota and its interaction with the host. Here, we detail the impact of a single-organism probiotic, Lactobacillus rhamnosus GG ATCC 53103 (LGG), on the structure and functional dynamics (gene expression) of the gut microbiota in a study of 12 healthy individuals, 65 to 80 years old. The analysis revealed that while the overall community composition was stable as assessed by 16S rRNA profiling, the transcriptional response of the gut microbiota was modulated by probiotic treatment. Comparison of transcriptional profiles based on taxonomic composition yielded three distinct transcriptome groups that displayed considerable differences in functional dynamics. The transcriptional profile of LGG in vivo was remarkably concordant across study subjects despite the considerable interindividual nature of the gut microbiota. However, we identified genes involved in flagellar motility, chemotaxis, and adhesion from Bifidobacterium and the dominant butyrate producers Roseburia and Eubacterium whose expression was increased during probiotic consumption, suggesting that LGG may promote interactions between key constituents of the microbiota and the host epithelium. These results provide evidence for the discrete functional effects imparted by a specific single-organism probiotic and challenge the prevailing notion that probiotics substantially modify the resident microbiota within nondiseased individuals in an appreciable fashion.

A review of 63 studies was inclusive evidence on the efficacy of probiotics supplements with their role being limited only to those with compromised or injured enteric microbiome.

(53Trusted Source).

Lynne McFarland published a review Use of probiotics to correct dysbiosis of normal microbiota following disease or disruptive events: a systematic review in BMJ Open . 2014 Aug 25;4(8):e005047. doi: 10.1136/bmjopen-2014-005047 http://DOI: 10.1136/bmjopen-2014-005047 Concluding that the ability to assess the degree of dysbiosis improvement is dependent on the enrolled population and the timing of microbiological assays. The functional claim for correcting dysbiosis is poorly supported for most probiotic strains and requires further research.

Objective: To assess the evidence for the claim probiotics can correct dysbiosis of the normal microbiota resulting from disease or disruptive events.

Setting: Systematic review of published clinical trials of patients receiving a probiotic intervention for the prevention or treatment of various diseases.

Data sources: Sources searched (1985-2013): PubMed, EMBASE, Cochrane Database of Systematic Reviews, CINAHL, AMED and ISI Web of Science. Three on-line clinical trial registries were searched: Cochrane Central Register of Controlled trials, MetaRegister of Controlled Trials and National Institutes of Health.

Review methods: Included studies were randomised clinical trials of probiotic interventions having microbiological assays. Studies were evaluated following Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines for specific probiotic strains. A standard data extraction form was used to collect the raw data.

Outcome measures: The primary outcome is the degree of microbiota correction by specific probiotic strains. Secondary outcome was the association between the degree of dysbiosis correction and clinical efficacy.

Results: The review of the literature found three distinct study designs: model A (restoration) assayed patients enrolled with a healthy, undisturbed microbiota and then assayed postdisruptive event and probiotic therapy; model B (alteration) assayed patients with pre-existing disrupted microbiota and then postprobiotic therapy; model C (no dysbiosis) assayed volunteers with no disruptive event prebiotic and postprobiotic. From a total of 63 trials, 83% of the probiotic products using model A restored the microbiota, 56% using model B improved the microbiota and only 21% using model C had any effect on microbiota. Clinical efficacy was more commonly associated with strains capable of restoration of the normal microbiota.

6. Breastfeed for at Least Six Months

A baby’s microbiota begins to properly develop at birth. However, some recent studies suggest that babies may be exposed to some bacteria before birth (32Trusted Source).

During the first two years of life, an infant’s microbiota is continuously developing and rich in beneficial Bifidobacteria, which can digest the sugars in breast milk (33Trusted Source).

Many studies have shown that infants who are formula fed have an altered microbiota that has fewer Bifidobacteria than infants who are breastfed (33Trusted Source, 34Trusted Source, 35Trusted Source).

Breastfeeding is also associated with lower rates of allergies, obesity and other diseases that may be due to differences in the gut microbiota (36Trusted Source).

BOTTOM LINE:

Breastfeeding helps an infant develop a healthy microbiota, which may help protect against certain diseases in later life.