Hostname: page-component-7bb8b95d7b-wpx69 Total loading time: 0 Render date: 2024-09-29T20:18:46.832Z Has data issue: false hasContentIssue false

Kefir as a therapeutic agent in clinical research: a scoping review

Published online by Cambridge University Press:  30 March 2023

Milena Klippel Bessa*
Affiliation:
Postgraduate Program in Pathology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, 90050-170, Porto Alegre, RS, Brazil
Giancarlo Rezende Bessa
Affiliation:
Universidade Feevale, ERS-239, 2755, 93525-075, Novo Hamburgo, RS, Brazil
Renan Rangel Bonamigo
Affiliation:
Postgraduate Program in Pathology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, 90050-170, Porto Alegre, RS, Brazil
*
*Corresponding author: Milena Klippel Bessa, email: [email protected]
Rights & Permissions [Opens in a new window]

Abstract

Increasing research has been conducted on the role of probiotics in disease treatment. Kefir, a safe, low-cost probiotic fermented milk drink, has been investigated in many in vitro and animal studies, although parameters for human therapeutic dose or treatment time have not yet been determined. Here we perform a scoping review of clinical studies that have used kefir as a therapeutic agent, compiling the results for perspectives to support and direct further research. This review was based on Joanna Briggs Institute guidelines, including studies on the effects of kefir-fermented milk in humans. Using the term KEFIR, the main international databases were searched for studies published in English, Spanish or Portuguese until 9 March 2022. A total of 5835 articles were identified in the four databases, with forty-four eligible for analysis. The research areas were classified as metabolic syndrome and type 2 diabetes, gastrointestinal health/disorders, maternal/child health and paediatrics, dentistry, oncology, women’s and geriatric health, and dermatology. The many study limitations hampered generalisation of the results. The small sample sizes, methodological variation and differences in kefir types, dosage and treatment duration prevented clear conclusions about its benefits for specific diseases. We suggest using a standard therapeutic dose of traditionally prepared kefir in millilitres according to body weight, making routine consumption more feasible. The studies showed that kefir is safe for people without serious illnesses.

Type
Review Article
Copyright
© The Author(s), 2023. Published by Cambridge University Press on behalf of The Nutrition Society

Introduction

The idea that gut health and microbiota balance directly affect the homeostasis of human organ systems is gaining increasing acceptance. Accordingly, foods are being researched as therapeutic agents in search of benefits that help maintain the body in a healthy state. The human gut is home to trillions of microbes, and their impact on human health has been extensively studied(Reference Walker, Vlamakis and Lee1). In addition to being an important digestive organ, the intestine is the largest immune organ in the human body(Reference Pu, Lin and Gao2). Numerous studies on the influence of intestinal microbiota have shown the importance of diet for wellbeing. Interactions between intestinal microorganisms and a series of health problems have been described(Reference Yue, Cai and Xiao3Reference Dabke, Hendrick and Devkota6).

Gut microbiota refers to the microbes that reside in the human gastrointestinal tract(Reference Strandwitz7). It contributes to the integrity of the intestinal epithelial barrier by maintaining cell junctions and promoting damage repair, as well as by helping protect the host against pathogenic microorganisms, neutralising the effects of toxins and/or drugs and providing essential metabolites/vitamins(Reference Pereira, Coco and Ton8,Reference Zheng, Xu and Jiang9) . It also improves nutrient bioavailability and modulates the intestinal epithelium’s absorption capacity(Reference Iebba, Totino and Gagliardi10). Research on gut microbiota is ongoing in a number of fields, including dermatology(Reference Yu, Dunaway and Champer11), endocrinology(Reference Li, Li and Li12,Reference Qi, Yun and Pang13) , dentistry(Reference Kitamoto and Kamada14,Reference de Oliveira, Lourenco and Colombo15) and oncology(Reference Gori, Inno and Belluomini16,Reference Routy, Gopalakrishnan and Daillere17) .

While the inherited genome is essentially stable over the host’s lifetime, the microbiome is immensely diverse(Reference Mosca, Leclerc and Hugot18,Reference Pasolli, Asnicar and Manara19) , dynamic(Reference Lloyd-Price, Mahurkar and Rahnavard20), and responsive to external stimuli, which increases its potential as a target for therapeutic intervention. The composition of the microbiota is influenced by host genotype, environment and diet(Reference Iebba, Totino and Gagliardi10).

Intestinal dysbiosis refers to changes in the quantitative and qualitative composition of our commensal microbes, which can alter host microbial interaction and contribute to an inflammatory disease state, which is associated with the development of many non-communicable diseases(Reference Byndloss and Baumler21). One current strategy for treating dysbiosis is the use of probiotics in an effort to recover microbial diversity and disturbed gut microbiota.

Probiotics refers to microorganisms that, when administered in adequate amounts, confer health benefits to the host(Reference Plaza-Diaz, Ruiz-Ojeda and Gil-Campos22Reference Hill, Guarner and Reid25). A probiotic must be able to survive under gastrointestinal conditions (acidic pH, enzymes, bile salts, etc.), have the ability to adhere to the intestinal mucosa, antagonise pathogens and stimulate the immune system(Reference Plaza-Diaz, Ruiz-Ojeda and Gil-Campos22,Reference Hill, Guarner and Reid25) . Among their different mechanisms of action, the following stand out: colonisation and normalisation of disturbed intestinal microbial communities in children and adults, competitive exclusion of pathogens and production of bacteriocins, modulation of faecal enzyme activities associated with bile salt metabolism, inactivation of carcinogens and other xenobiotics, production of short-chain and branched-chain fatty acids (which have broad effects in the gut and peripheral tissue by interacting with short-chain fatty acid receptors, which modulate tissue sensitivity to insulin), cell adhesion and mucin production, immune system modulation and interaction with the brain–gut axis by regulating endocrine and neurological function(Reference Plaza-Diaz, Ruiz-Ojeda and Gil-Campos22,Reference Vieira, Fukumori and Ferreira26) . Prebiotics, foods that contain non-digestible fibre, stimulate the growth and activity of beneficial microorganisms(Reference Ceapa, Wopereis and Rezaiki27), and include human milk oligosaccharides, inulin, fructooligosaccharides and galactooligosaccharides(Reference Quigley28).

Kefir is fermented milk produced by the action of bacteria and yeast that symbiotically associate in kefir grains(Reference de Oliveira Leite, Miguel and Peixoto29). This slightly effervescent and foamy beverage originates from the action of the natural microbiota present in these grains(Reference Weschenfelder, Wiest and Carvalho30), which consists of an inert matrix of polysaccharides and proteins(Reference Farag, Jomaa and El-Wahed31). This matrix is densely populated by species of lactic acid bacteria, acetic acid bacteria and yeast(Reference Leite, Miguel and Peixoto32).

The large number of microorganisms in kefir and their microbial interactions as well as the bioactive compounds that result from microbial metabolism and the benefits associated with this beverage make kefir a natural probiotic. The microbial composition of kefir can vary, being influenced by region of origin, duration of use, substrate and management techniques(Reference Weschenfelder33). However, kefir grains generally contain relatively stable and specific microbiota, always including a predominance of certain Lactobacillus species.(Reference de Oliveira Leite, Miguel and Peixoto29,Reference Stiemsma, Nakamura and Nguyen34) .

Numerous in vitro and animal studies have demonstrated the beneficial action of milk fermented by kefir grains and its nutraceutical potential, which includes anti-inflammatory(Reference Chen, Hung and Yen35Reference Seo, Park and Ko37), antioxidant(Reference Ghoneum, Abdulmalek and Pan38,Reference Azizi, Kumar and Yeap39) , anticancer(Reference Azizi, Kumar and Yeap39,Reference Sharifi, Moridnia and Mortazavi40) , antimicrobial(Reference Al-Mohammadi, Ibrahim and Moustafa41Reference Bolla, Carasi and Bolla Mde43), antidiabetic(Reference Hadisaputro, Djokomoeljanto and Judiono44), antihypertensive(Reference Brasil, Silva-Cutini and Moraes45Reference Klippel, Duemke and Leal48) and anti-hypercholesterolaemic properties(Reference Liu, Wang and Chen49,Reference Wang, Xu and Xi50) . These effects can be attributed to probiotic microorganisms and the wide diversity of bioactive compounds produced during the fermentation process(Reference Azizi, Kumar and Yeap39,Reference Rosa, Dias and Grzeskowiak51) . However, clinical research on these benefits is still in its infancy, complicated by the heterogeneity of dosages and forms of administration, making it difficult, for example, to compile results through meta-analysis. Thus, the present scoping review aimed to assess clinical studies that have tested kefir as a therapeutic agent for diseases or health conditions, identifying therapeutic patterns and adverse effects. From this analysis, the data were organised to point out strategies and present perspectives for future research.

Methodology

This is a scoping review of human studies on the effects of milk fermented by kefir grains as a therapeutic agent for diseases of organic systems or to improve patient health conditions. This review was structured according to Joanna Briggs Institute guidelines(52). Scoping reviews are useful when studies in a topic are heterogeneous, as they map and summarise existing evidence and identify possible knowledge gaps to direct future research(52). A search was performed in the main international databases: PubMed, Web of Science, Embase and Scopus, using the term KEFIR. The inclusion criteria were prospective clinical studies, with kefir as the object of the study, published in English, Spanish or Portuguese until 9 March 2022. Duplicated studies and those using water kefir were excluded, since it was not the focus of this review. The inclusion and exclusion criteria were identified by reading the abstracts and, occasionally, if there were any doubts, by reading the full article. Study eligibility was determined by two independent researchers, and a third arbitrated in cases of disagreement. The search results were managed using EndNote.

The titles and abstracts of all articles were evaluated for potential relevance according to the inclusion and exclusion criteria. Data from the selected clinical studies were extracted and summarised regarding the objective, materials and methods (design, dose and intervention time), sample/population, comparison parameters between groups, adverse effects and clinical outcome. The selection process for relevant studies is shown in Fig. 1.

Fig. 1. Flowchart of article inclusion.

Results

The term kefir resulted in a wide range of studies in the selected databases. The vast majority, however, concerned laboratory research on strains of microorganisms in kefir or were in vitro or animal studies testing its disease-fighting properties or its ability to maintain a healthy state. A total of 5835 articles were identified in the four databases, and forty-four were eligible for analysis after applying the inclusion and exclusion criteria and removing duplicates. The selection process for relevant studies is shown in Fig. 1.

Clinical studies published in English (but not Portuguese or Spanish) on kefir as a therapeutic agent can be found beginning in 2002, and this number has shown a growing trend, reaching eight in 2021, as shown in Fig. 2. Turkey, Iran, and the United States have produced the most clinical studies, followed by Brazil and Taiwan, as illustrated in Fig. 3. Most studies are single-centre clinical trials, with some crossover trials, one case series and one case report, all prospective studies. The mean sample size was 45·9 (standard deviation 28·7) participants.

Fig. 2. Year of publication of English-language studies on kefir as a therapeutic agent. *Until 9 March 2022.

Fig. 3. Number of articles according to country of origin (absolute number, percentage).

The mean intervention time could not be calculated, since it was presented in days, weeks, months, medication cycles, continuous use or single dose, or was not daily, as in O’Brien et al. (Reference O’Brien, Stewart and Forney53), in which it was administered twice a week for 15 weeks. The study population also varied, although none of the subjects had serious diseases or severe comorbidities, which we considered exclusion criteria.

Table 1 (Reference St-Onge, Farnworth and Savard54Reference Tunay and Taş96) presents the population, sample size, therapeutic dosage, kefir preparation type, intervention time and main findings of each study. Two pairs of publications (Ostadrahimi et al. (Reference Tunay and Taş68) and Alihosseini et al. (Reference Alihosseini, Moahboob and Farrin72); Fathi et al. (Reference Fathi, Faghih and Zibaeenezhad70) and Fathi et al. (Reference Fathi, Ghodrati and Zibaeenezhad71)) used the same population to investigate different outcomes.

Table 1. Overview of included studies

n, sample number; i.p., industrial preparation; h.p., home preparation; u.t., unspecified type; AD, Alzheimer’s disease; ALT, alanine aminotransferase; BDC-LDL, baseline diene conjugates of low-density lipoprotein; Beta2-GPI-oxLDL, oxLDL-β2 glycoprotein I; BMD, bone mineral density; BMI, body mass index; CRP, C-reactive protein; ECOG, Eastern Cooperative Oncology Group; ESR, erythrocyte sedimentation rate; FBG, fasting blood glucose; FMT, faecal microbiota transplantation; GI, glycaemic index; HbA1c, glycated haemoglobin; Hgb, haemoglobin; HOMA-IR, insulin resistance Homeostasis Model Assessment; IBD, inflammatory bowel disease; LI, lactose intolerance; MDA, malondialdehyde; MetS, metabolic syndrome; oxLDL, oxidised low-density lipoprotein; OxS, oxidative stress; PBG, postprandial blood glucose levels; QoL, quality of life; STAW, staggered and tapered antibiotic withdrawal; T2DM, type 2 diabetes mellitus; TOS, total oxidant status; WC, waist circumference. Triple therapy* consists of lansoprazole (30 mg), amoxicillin (1000 mg) and clarithromycin (500 mg).

The largest number of clinical studies with kefir was for the prevention or treatment of metabolic and gastrointestinal diseases, and their results were usually positive. Other fields of knowledge in which kefir was tested included maternal/child health and paediatrics, dentistry, oncology, women’s and geriatric health, and dermatology (Table 2).

Table 2. Areas of clinical research on kefir

ATB, antibiotic; IBD, inflammatory bowel disease; MetS, metabolic syndrome; QT, chemotherapy; T2DM, type 2 diabetes mellitus.

Discussion

The discussion of the results will be presented according to the field in which kefir was studied in humans as a therapeutic agent.

Metabolic syndrome and type 2 diabetes

Metabolic syndrome is considered a pro-thrombotic and pro-inflammatory state characterised by a set of clinical findings that include central and abdominal obesity, systemic hypertension, insulin resistance and atherogenic dyslipidaemia, including elevated inflammatory cytokine activity(Reference Aziz, Whitehead and Palsson97,Reference Samson and Garber98) . Therapeutic options involve dietary control, regular exercise and pharmacological treatment for dyslipidaemia, hypertension and hyperglycaemia(Reference Samson and Garber98).

Gut microbes influence host metabolic balance by modulating energy absorption, intestinal motility, appetite, glucose and lipid metabolism, and hepatic fat storage(Reference Festi, Schiumerini and Eusebi99). Dysbiosis favours the translocation of bacterial fragments and can lead to systemic inflammation and insulin resistance(Reference Festi, Schiumerini and Eusebi99,Reference Li, Zhou and Gan100) . Administration of pre- and probiotics can reduce low-grade inflammation and improve intestinal barrier integrity, aiding metabolic balance. Zonulin, a family of proteins associated with the permeability of the intestinal barrier(Reference Fasano101), is a physiological modulator of the tight intercellular junctions of the intestinal epithelium, and the loss of barrier function secondary to its up-regulation can lead to an uncontrolled influx of food and microbial antigens. Pražnikar et al. (Reference Praznikar, Kenig and Vardjan85) studied the effects of supplementing asymptomatic overweight adults with milk or kefir for 3 weeks, finding that kefir supplementation led to lower serum zonulin levels than milk.

Bellikci-Koyu et al. (Reference Bellikci-Koyu, Sarer-Yurekli and Akyon82) and Da Silva Ghizi et al. (Reference Da Silva Ghizi, De Almeida Silva and De Andrade Moraes95) tested the effects of regular kefir consumption in people with metabolic syndrome. The intervention period in both studies was 12 weeks and they used similar dosages, although the kefir preparation methods differed. The bioactive peptide profile of milk kefir is completely different from that of raw milk, including 236 unique peptides produced during the fermentation process, many with antihypertensive (angiotensin-converting enzyme inhibitors), antimicrobial, immunomodulatory, antioxidant and antithrombotic properties(Reference Ebner, Asci Arslan and Fedorova46). Bellikci-Koyu et al. (Reference Bellikci-Koyu, Sarer-Yurekli and Akyon82) found no significant differences between milk and kefir groups, while Da Silva Ghizi et al. (Reference Da Silva Ghizi, De Almeida Silva and De Andrade Moraes95) considered the origin of their fermented beverage key to their more expressive findings than Bellikci-Koyu et al. (Reference Bellikci-Koyu, Sarer-Yurekli and Akyon82). Da Silva Ghizi et al. used milk inoculated with kefir grains, which contains more than thirty live bacteria species and twelve yeast and fungi species in a complex symbiotic system, a culture that synthesises several bioactive compounds during the fermentation process, such as organic acids, bioactive peptides, bacteriocins and exopolysaccharides(Reference Azizi, Kumar and Yeap39,Reference Ebner, Asci Arslan and Fedorova46,Reference Rosa, Dias and Grzeskowiak51) . Industrial kefir production, which usually involves commercial starter cultures, allows standardisation of this process, although it results in loss of cell viability. Commercial kefir contains a smaller number and variety of microorganisms, especially yeasts(Reference Ebner, Asci Arslan and Fedorova46,Reference Gonzalez-Orozco, Garcia-Cano and Jimenez-Flores102) . Moreover, in fermented food production (including kefir), the backslopping technique is used to increase production by up to fifty times, maintaining the physico-chemical characteristics and nutritional values, but decreasing the microbiological diversity and producing some consistency deficiencies(Reference Farag, Jomaa and El-Wahed31). In a randomised, double-blind, placebo-controlled clinical study, Da Silva Ghizi et al. (Reference Da Silva Ghizi, De Almeida Silva and De Andrade Moraes95) found significant improvement in several clinical parameters in the kefir group compared with controls (who received homemade curd prepared with the same type of milk as the kefir group), including blood pressure, lipid profile blood, fasting glucose and oxidised LDL, which led to lower cardiovascular risk according to Framingham scores. Abd-Alwahab and Al-Dulaimi(Reference Abd-Alwahab and Al-Dulaimi78) administered grain-fermented kefir drink to volunteers, finding significant improvement in the lipid profile, although their control group received water instead of milk, which prevents direct comparison of the results.

One hypothesis about kefir’s mechanism of action on serum cholesterol levels is the deconjugation of bile acids by Lactobacillus spp., while yeasts increase bile acid discharge, which in turn increases cholesterol expenditure during production(Reference Abd-Alwahab and Al-Dulaimi78,Reference Brashears, Gilliland and Buck103) . In addition, colonic microbiota can metabolise dietary and endogenous cholesterol, reducing it mainly to coprostanol (5β-colestan-3β-ol) and coprostanone, both through low intestinal absorption and elimination through faeces(Reference Lye, Rusul and Liong104,Reference Ooi and Liong105) . Higher intake of calcium, which is abundant (about 0·12/100 g) in kefir, can also positively affect the serum lipid profile(Reference Ditscheid, Keller and Jahreis106Reference Denke, Fox and Schulte108). St-Onge et al. (Reference St-Onge, Farnworth and Savard54), Fathi et al. (Reference Fathi, Ghodrati and Zibaeenezhad71), Pražnikar et al. (Reference Praznikar, Kenig and Vardjan85), Ostadrahimi et al. (Reference Ostadrahimi, Taghizadeh and Mobasseri68), Gezginc and Maranci(Reference Gezginc and Maranci79) and Bashiti and Zabut(Reference Bashiti and Zabut84) investigated kefir’s effects on the lipid profile, but found different results, probably a consequence of the studies’ different methodologies.

The conflicting outcomes of these studies can be explained, at least in part, by the heterogeneity of their populations, especially the floor (baseline) effect (i.e. the lower the baseline level of an outcome, the less likely a treatment will result in a reduction in that outcome level), which was evident in studies such as that by Gezginc and Maranci(Reference Gezginc and Maranci79), who tested the influence of kefir on the lipid profile of healthy young people.

Hosainzadegn and Hosainzadegan(Reference Hosainzadegn and Hosainzadegan92) reported a case of marked improvement in glycated hemoglobin (HbA1c) levels (from 7·9 to 7·1) and a 4 kg weight loss in a woman with type 2 diabetes after 3 months of using kefir prepared at home. Clinical studies by Judiono et al. (Reference Judiono, Hadisaputro and Indranila67), Ostradahimi et al. (Reference Ostadrahimi, Taghizadeh and Mobasseri68), Alihosseini et al. (Reference Alihosseini, Moahboob and Farrin72), Bellikci-Koyu et al. (Reference Bellikci-Koyu, Sarer-Yurekli and Akyon82), Bashiti and Zabut(Reference Bashiti and Zabut84) and Praznikar et al. (Reference Praznikar, Kenig and Vardjan85) were included in a systematic review with meta-analysis(Reference Salari, Ghodrat and Gheflati109) of randomised clinical trials that assessed the effects of kefir drink on glycaemic control. The authors found significant improvement in fasting glucose and insulin levels in participants who consumed kefir, but no significant difference in HbA1c levels. The results of this meta-analysis, however, can be questioned due to the heterogeneity of study populations, dosages, and intervention times.

Weight control and exercise are also involved in the prevention and treatment of metabolic syndrome. Studies by O’Brien et al. (Reference O’Brien, Stewart and Forney53), Gölünük et al. (Reference Gölünük, Öztaşan and Sözen73) and Lee et al. (Reference Lee, Jhang and Lee88) investigated the effects of kefir on post-exercise oxidative stress parameters. The samples of these studies were healthy young people, although the intervention time, kefir type, quantity and tested parameters differed greatly. Kefir improved total oxidant status levels (an indicator of oxidative stress)(Reference Gölünük, Öztaşan and Sözen73), did not change C-reactive protein levels (which increased in the control group)(Reference O’Brien, Stewart and Forney53), improved exercise endurance, reduced lactic acid production after exercise, accelerated recovery(Reference Lee, Jhang and Lee88) and did not significantly affect the other parameters.

Regarding weight loss, Fathi et al. (Reference Fathi, Faghih and Zibaeenezhad70) and Hosainzadegn and Hosainzadegan(Reference Hosainzadegn and Hosainzadegan92) found positive results with kefir, unlike other studies that measured anthropometric parameters such as weight, body mass index and waist circumference(Reference O’Brien, Stewart and Forney53,Reference Lee, Jhang and Lee88,Reference Da Silva Ghizi, De Almeida Silva and De Andrade Moraes95) . Caferoglu and Aytekin Sahin(Reference Caferoglu and Aytekin Sahin94) investigated whether adding kefir to a low or high glycaemic index meal would affect participant appetite and food intake, finding that kefir can help limit appetite and energy intake for high glycaemic index meals but not low glycaemic index meals(Reference Caferoglu and Aytekin Sahin94).

Gastrointestinal health/disorders

The gastrointestinal tract and its microbiome provide unique metabolic functions to the host and are critical to maintaining health. The abundance of different species of microorganisms, their function and their interaction with organic systems have been the focus of numerous medical studies(Reference Hills, Pontefract and Mishcon110). Metagenomics and analysis of twin data have shown that environmental factors, such as diet and domestic cohabitation, far outweigh heritable genetic contributions to gut microbiota composition and function(Reference Rothschild, Weissbrod and Barkan111).

Inflammatory bowel disease is characterised by chronic and relapsing inflammation of the gastrointestinal tract and includes 2 chronic idiopathic inflammatory diseases: Crohn’s disease and ulcerative colitis(Reference Yilmaz, Dolar and Ozpinar83,Reference Sairenji, Collins and Evans112,Reference Flynn and Eisenstein113) . These heterogeneous and complex immune disorders of the gastrointestinal tract share many common clinical features but differ in inflamed areas and are treated differently(Reference Hills, Pontefract and Mishcon110,Reference Flynn and Eisenstein113,Reference Lee and Chang114) . Considered a serious and debilitating condition that affects general health and quality of life, inflammatory bowel disease has been associated with intestinal dysbiosis, which decreases microbial biodiversity and slows or stops important functions of intestinal barrier integrity and immune system regulation, which results in inflammation and increased immune response(Reference Lee and Chang114). In addition, mucolytic and pathogenic bacteria are also increased, which leads to degradation of the mucosal barrier and greater pathogen penetration into intestinal tissues(Reference Lee and Chang114,Reference Chassaing and Darfeuille-Michaud115) .

Probiotics and faecal microbial transplantation, both aimed at reintroducing beneficial microbes into dysbiotic guts, are currently being used to treat inflammatory bowel disease(Reference Lee and Chang114,Reference Jeong, Kim and Son116) . Since kefir contains a diverse range of microorganisms, many of which have already been studied as probiotics(Reference Slattery, Cotter and O’Toole117), it has promise as an alternative treatment for intestinal dysbiosis. Wang(Reference Wang, Zaydi and Lin87) found that 21 d of freeze-dried (industrialised) kefir had positive effects on gastrointestinal symptoms, such as abdominal pain and bloating, and increased abundance of bifidobacteria. Forejt et al. (Reference Forejt, Šimůnek and Brázdová57) found significant improvement in gastrointestinal discomfort and lower Enterococcus faecalis after 2 weeks of kefir therapy in a sample of women. Figler et al. (Reference Figler, Mozsik and Schaffer56) investigated how consuming two types of kefir influenced the levels of primary probiotic Streptococcus, Lactobacillus and Bifidobacterium among the total number of microbes, finding that these populations increased in both groups after 4 weeks, with a more expressive increase in the intervention group (Biofir® kefir). In contrast, Sepp(Reference Sepp, Smidt and Štšepetova80) found increased diversity of Lactobacillus spp. after 8 weeks only in the group who received kefir (industrialised) with added L. fermentum ME-3. The studies’ samples, however, were healthy people with no diagnosed gastrointestinal disease, and different intervention times were used. In a sample of patients with Crohn’s disease, Yilmaz et al. (Reference Yilmaz, Dolar and Ozpinar83) found that regular kefir use can improve both symptoms and short-term quality of life and has a positive effect on biochemical parameters (such as haemoglobin, C-reactive protein and erythrocyte sedimentation rate).

Kefir has also been studied as an adjuvant treatment in two other gastrointestinal pathologies, Helicobacter pylori infection and recurrent Clostridioides difficile infection. Among patients who received kefir, Bekar et al. (Reference Bekar, Yilmaz and Gulten61) found a lower prevalence of side effects, such as headache, nausea, diarrhoea and abdominal pain, in a group that received a triple therapy consisting of lansoprazole (30 mg), amoxicillin (1000 mg) and clarithromycin (500 mg). This may have important implications for increasing treatment adherence. In a case series of patients at risk of recurrent Clostridioides difficile infection, Bakken(Reference Bakken64) used continuous kefir ingestion in association with a regimen of staggered and tapered antibiotic withdrawal, finding the same efficacy as faecal microbial transplantation.

Microorganisms present in kefir, such as Lactococcus spp., Lactobacillus spp. and some strains of Kluyveromyces spp. hydrolyse lactose to glucose and galactose(Reference Leite, Leite and Del Aguila118). During the fermentation process, milk proteins are also extensively hydrolysed, releasing functional peptides and improving digestibility(Reference Dallas, Citerne and Tian119). Two studies tested kefir for digestibility in healthy adults with poor digestion(Reference Hertzler and Clancy55) or lactose intolerance(Reference Mądry, Krasińska and Woźniewicz63), both using a single intervention dose. They concluded that milk fermented with kefir, as well as yogurt, causes less severe discomfort than normal milk.

Turan et al. (Reference Turan, Dedeli and Bor66) and Maki et al. (Reference Maki, Matsukawa and Matsuduka77) studied the effects of kefir on chronic intestinal constipation, a highly prevalent condition for which diet is a possible treatment(Reference Aziz, Whitehead and Palsson97). The first study enrolled patients who met Rome II criteria for chronic constipation, without metabolic or structural disorders that could be responsible for the disease; the second enrolled patients with some physical or mental disability who were admitted to a Japanese hospital. In addition to the different study populations, the type and amount of kefir used were completely different. Turan et al. (Reference Turan, Dedeli and Bor66) used a higher dose of kefir (250 ml twice per day) for 28 d, while Maki et al. (Reference Maki, Matsukawa and Matsuduka77) used lyophilised kefir at a much lower dose (about 1/8 of Turan et al.) for 84 d. Turan et al. found that kefir supplementation was associated with a statistically significant decrease in laxative use, increased defecation frequency, higher bowel satisfaction scores and shortened colonic transit times. Maki et al. (Reference Maki, Matsukawa and Matsuduka77) found that kefir worked better in patients with non-severe chronic constipation and suggested identifying individuals who could benefit from its use. Clinical trials with larger sample sizes and a control group are highly recommended to determine kefir’s benefits in preventing and/or treating these gastrointestinal disorders.

Maternal/child health and paediatrics

Intestinal bacterial colonisation in the first years of life (generally the first three) has a major impact on the immune system, and the immunological influences of microbiota during this specific window can determine resistance or susceptibility to diseases, affecting the host’s health throughout life(Reference Kim and Kim120Reference Gensollen and Blumberg122). Breast milk is a critical factor in the development and composition of intestinal microbiota in neonates. One possible origin of the bacteria in this fluid, many of which are potentially probiotic, is the maternal gastrointestinal tract via bacterial translocation through the lymphatic system(Reference Stinson, Sindi and Cheema123). Tunay et al. (Reference Tunay and Taş96) tracked transmission of bacteria unique to kefir grains (Lactobacillus kefiranofaciens, Lentiactobacillus kefiri, Lentiactobacillus parakefiri) in breast milk and the faeces of newborns, finding that these microorganisms were transmitted to milk through maternal consumption of kefir, resulting in infant intestinal colonisation. Kurt et al. assessed the effects of milk kefir in nursing mothers in relation to the carbohydrate profile of their breast milk(Reference Kurt, Gökırmaklı and Guzel-Seydim91). The authors detected a trend towards more carbohydrates, including galactoligosaccharides (structures with prebiotic properties), in the milk of mothers who consumed kefir. They also reported that neither the mothers nor their infants experienced abdominal discomfort from using kefir. While a good body of evidence about the effects of probiotics on paediatric populations already exists(Reference Guo, Goldenberg and Humphrey124Reference Jiang, Ni and Liu126), studies about the benefits of kefir in children are still incipient.

Merenstein et al. (Reference Merenstein, Foster and D’Amico60) tested kefir’s effects on prevention of antibiotic-associated diarrhoea, a disease with a high morbidity rate and low treatment adherence(Reference Turck, Bernet and Marx127). The sample consisted of children aged between 1 and 5 years who received antibiotics to treat upper airway infections. Although they found no difference in diarrhoea rates between the intervention and control groups, they did find different absolute numbers of diarrhoea incidents in children aged 3–5 years (14% in the control group versus 6% in the kefir group) and in boys (32% in the control group versus 24% in the kefir group, compared with a 2% difference among girls in these groups). The differences in absolute numbers of diarrhoea incidents in the older groups were quite expressive (57% higher in the placebo group), which suggests that a study with a larger sample in this population could help elucidate the promising role of kefir for preventing antibiotic-associated diarrhoea. The authors further reported that patient safety was excellent, as expected for a food.

De Araújo et al. studied the effects of lyophilised kefir on the clinical parameters of wheezing among infants aged 6–24 months and on cytokine expression via T-helper 1 and T-regulatory cells(Reference De Araujo, De Lorena and Montenegro75), finding no significant decrease in the clinical parameters of wheezing, only a trend towards lower recurrence (perhaps the sample was too small to demonstrate significance). However, they suggested that this probiotic mixture triggers immunomodulation due to the production of T-helper 1 and T-regulatory cell cytokines, including IL-10 and IL-12, since there they increased significantly in the intervention group. Intestinal bacteria contribute to proper development of the immune system in the first years of life, and the intestinal flora and its metabolites (such as short-chain fatty acids) actively participate in the proliferation and differentiation of B cells and T cells, inducing a protective antibody response(Reference Nguyen, Himes and Martinez128).

Dentistry

The human mouth harbours a complex microbiome whose imbalance can lead to dental caries and periodontal disease(Reference Mosaddad, Tahmasebi and Yazdanian129). When metabolising carbohydrates, cariogenic microorganisms produce lactic, formic, acetic and propionic acids, which decrease the mouth’s pH to below 5·5, resulting in demineralisation of enamel hydroxyapatite crystals and proteolytic breakdown of the hard tissue structure of the teeth. Streptococcus mutans is the most important bacterial species related to oral health(Reference Dashper, Mitchell and Le Cao130); it is more abundant in disease and is the main bacteria involved in early childhood caries(Reference Patidar, Sogi and Singh131). Three included studies analysed the effects of kefir on the salivary count of S. mutans. Nevertheless, the results differed depending on the population, intervention time and dosage. The results showed that kefir was as effective as sodium fluoride for reducing salivary S. mutans counts in young adults(Reference Ghasempour, Sefdgar and Moghadamnia65). Daily consumption of kefir and the use of probiotic toothpaste decreased salivary microbial colonisation in orthodontic patients(Reference Alp and Baka76); probiotic products together with dental restorations effectively reduced S. mutans in children aged 8–12 years(Reference Reddy, Madhu and Punithavathy89).

Oncology

In vitro and animal studies have found positive results with probiotic dietary products like kefir in the prevention and treatment of various types of cancer(Reference Sharifi, Moridnia and Mortazavi40,Reference Fatahi, Soleimani and Afrough132,Reference Rafie, Golpour Hamedani and Ghiasvand133) . Studies by Topuz et al. (Reference Topuz, Derin and Can58) and Can et al. (Reference Can, Topuz and Derin59) explored the use of kefir in very similar populations, using the same dose in almost identical intervention times, although their objectives differed. Topuz et al. measured serum levels of pro-inflammatory cytokines, the antimicrobial effects of kefir and the mucositis rate in patients with colorectal cancer receiving chemotherapy (5-fluorouracil or oral fluoropyrimidine), but found no statistically significant differences between the intervention and control groups. Can et al. (Reference Can, Topuz and Derin59) explored kefir’s ability to prevent treatment-related gastrointestinal symptoms and its effects on the quality of life of cancer patients. They detected no differences in quality-of-life indices and reported more gastrointestinal complaints, but found better sleep quality in patients who used kefir. The amount of fermented milk used in the intervention may explain the increased complaints, since the ingestion of 250 ml of any liquid can be uncomfortable and increase symptoms such as nausea, vomiting and diarrhoea. Furthermore, the control group did not receive any type of similar liquid.

Smoak et al. (Reference Smoak, Harman and Flores93) studied patients with cancer who had undergone chemotherapy or radiotherapy in the previous 2 years and were enrolled in an exercise programme at the University of Northern Colorado Cancer Rehabilitation Institute. The intervention group drank approximately 240 ml of kefir up to 30 min after the exercise sessions, which took place three times a week for 12 weeks. The control group performed the same exercises but received no placebo. The kefir group improved in lean body mass, depression symptoms, fatigue, gastric discomfort and a biomarker of intestinal dysbiosis, which suggests that including kefir as part of a post-exercise diet can have significant psychological and physical benefits for cancer survivors(Reference Smoak, Harman and Flores93).

Women’s and geriatric health

Özcan et al. (Reference Ozcan, Oskay and Bodur81) studied perimenopause middle-aged women, in whom hormonal changes can lead to sleep and mood disturbances, sexual problems and, in the long term, decreased bone density(Reference Gracia and Freeman134,Reference Potter, Schrager and Dalby135) . The authors used the Women’s Health Insomnia Rating Scale, the Menopause-Specific Quality of Life Questionnaire and the Beck Depression Inventory to determine whether kefir benefitted post-menopausal women suffering from sleep disorders. After 1 month of intervention there was significant improvement in the first two parameters but not in Beck Depression Inventory results, which showed a non-significant trend towards improvement. Despite the study’s small sample size, the authors found kefir to be a non-pharmacological alternative for minimising some of the discomforts of the menopausal transition period.

Tu et al. (Reference Tu, Chen and Tung69) studied patients with osteoporosis, a disorder especially prevalent in postmenopausal women and older men, characterised by decreased bone mass and increased fracture risk(Reference Lane, Russell and Khan136). Therapy for patients with osteoporosis includes non-pharmacological measures (exercise, adequate calcium intake, etc.), medications to increase bone density and improve bone strength, and strategies to reduce the risk of falling(Reference Lane, Russell and Khan136,137) . In their controlled, parallel, double-blind intervention study, Tu et al. (Reference Tu, Chen and Tung69) divided participants into an intervention group, which received 1600 mg of freeze-dried milk kefir plus CaCO3 (1500 mg) daily, while the control group received the same amount of CaCO3 plus placebo (1600 mg of freeze-dried unfermented raw milk) daily for 6 months. By 6 months, the intervention had promoted short-term changes in bone turnover markers and greater increases in hip bone mineral density.

In the field of neurodegenerative diseases, Ton et al. (Reference Ton, Campagnaro and Alves86) conducted an uncontrolled clinical investigation to explore the antioxidant effects of milk fermented by kefir grains in patients with Alzheimer’s disease. Alzheimer’s disease causes progressive functional and cognitive decline in older adults and is the most common cause of dementia worldwide(Reference Lopez and Kuller138). From a pathological point of view, it is characterised by extracellular deposition of β-amyloid peptides and intracellular accumulation of hyperphosphorylated and aggregated hyperphosphorylated tau (a protein abundant in neurons), forming neurofibrillary tangles(Reference Querfurth and LaFerla139). The central mediators in the pathogenesis of Alzheimer’s disease are oxidative stress and neuroinflammation(Reference Kinney, Bemiller and Murtishaw140Reference Chen and Zhong142). Numerous studies have shown that gut microbiota play an important role in brain function(Reference Megur, Baltriukiene and Bukelskiene143Reference Doifode, Giridharan and Generoso145). The brain and microbiota communicate through a complex bidirectional connection known as the ‘microbiota–gut–brain axis’, which involves the immune system, neuroendocrine mechanisms, tryptophan metabolism, vagus nerve and enteric nervous system.(Reference Yue, Cai and Xiao3,Reference Megur, Baltriukiene and Bukelskiene143,Reference Cryan, O’Riordan and Cowan146) . Lipopolysaccharides (endotoxins) and bacterial amyloids synthesised by the gut microbiota can activate brain immune response and lead to neuroinflammation(Reference Megur, Baltriukiene and Bukelskiene143,Reference Jiang, Li and Huang144) . Neuroinflammatory cytokines may compromise β-amyloid clearance, leading to its accumulation in the brain(Reference Pereira, Coco and Ton8,Reference Alasmari, Alshammari and Alasmari147) . Ton et al. (Reference Ton, Campagnaro and Alves86) evaluated the benefits of kefir supplementation for 90 d at a dose of 2 ml/kg/d on cognitive function and biomarkers of oxidative stress, inflammation and cell damage in older adults with Alzheimer’s disease. They found improvement in every test (memory, visual–spatial function and abstraction skills, executive and language functions, constructive skills and attentive function), a protective effect for mitochondria, and cytoprotective and anti-apoptotic action, whose effects slow neurodegeneration.

Dermatology

The skin and gut appear to share a series of indirect bidirectional metabolic pathways known as the ‘gut–skin axis’(Reference Alves, Gregorio and Baby90). Patients with atopic dermatitis, a highly prevalent inflammatory skin disease worldwide, present with intestinal and cutaneous dysbiosis(Reference Kim and Kim120,Reference Watanabe, Narisawa and Arase148,Reference Fieten, Totte and Levin149) . This condition of microbiome imbalance, along with skin barrier dysfunction, immune dysregulation and environmental risk factors, contributes to disease onset and the atopic march(Reference Yu, Dunaway and Champer11,Reference Lee, Lee and Park150) . In their controlled crossover intervention study, Alves et al. (Reference Alves, Gregorio and Baby90) compared the effects of kefir ingestion (grain-fermented at home) on the skin of adults with and without atopic dermatitis. The primary outcomes were transepidermal water loss and stratum corneum hydration in all participants and the severity scoring of atopic dermatitis (SCORAD) index in patients with atopic dermatitis. Significant improvements were found in both groups, including a significant SCORAD index decrease in the atopic dermatitis group.

Kefir production and dosage

Commercially produced kefir models may contain different species of Lactobacillus than those produced with the inoculation of the grains, which can make an important difference, because species exclusive to kefir grains such as L. kefiranofaciens and L. kefiri have already demonstrated beneficial health effects(Reference Carasi, Racedo and Jacquot151Reference Chen, Hsiao and Hong154). Furthermore, commercial kefir samples usually do not contain acetic acid bacteria, which are abundant in traditionally prepared kefir(Reference Dobson, O’Sullivan and Cotter155Reference Walsh, Crispie and Kilcawley157). Another noteworthy difference between commercially and traditionally prepared kefir is the variety of yeasts, radically smaller in commercial models(Reference Marsh, O’Sullivan and Hill156). Bourrie et al. (Reference Bourrie, Cotter and Willing158,Reference Bourrie, Ju and Fouhse159) demonstrated the impact of these microbial differences on kefir’s ability to improve metabolic parameters in obese mice.

On the basis of these animal studies and on the analysed intervention studies, our suggestion is to use a therapeutic dose pattern in millilitres of traditionally prepared kefir (kefir grains inoculated in milk), due to its greater microbiological complexity and potential bioactive compounds.

The randomised, double-blind, placebo-controlled trial by Da Silva Ghizi et al. (Reference Da Silva Ghizi, De Almeida Silva and De Andrade Moraes95) was the only one to use individualised dose per kilogram (1·6 ml/kg for males and 1·9 ml/kg for females), calculated on the basis of studies by Reagan-Shaw et al. (Reference Reagan-Shaw, Nihal and Ahmad160) and Rosa et al. (Reference Diniz Rosa, Gouveia Peluzio do and Pérez Bueno161). This dose was close to the lowest standardised doses used in other studies that showed benefits. So, we suggest this individualised dose to be used in future studies avoiding underdose/overdose error. For example, a dose of 1·6 ml/kg for a man weighing 90 kg would be 144 ml of kefir per day, making routine consumption much more viable. When such individualisation is not possible, doses of 100–200 ml/d are suggested.

These doses were determined by the perception that a minimum amount necessary is better tolerated by people and is more economically viable, increasing the probability of the consumption of this food to become a habit and its possible benefits to extend over time. Such doses would also make the methodology of future studies closer to the reality of the population. Very high amounts of kefir can hamper the viability of longer-term use due to palatability and maintenance costs, even when prepared at home. Daily doses of 500 or 600 ml, used in some studies, are not feasible long term for the general population, thus preventing any possible benefits.

Conclusions

Despite a good body of evidence and interesting and promising findings in several areas of research, the included studies involved many limitations and their results cannot be generalised. The small sample sizes, methodological differences and varying kefir types, dosage and therapy times prevent us from drawing clear conclusions about its benefits for specific diseases. However, this review indicates fruitful paths for further research on kefir, facilitating the compilation of data and strengthening the results of future meta-analyses.

We suggest using a daily therapeutic dose pattern in millilitres of traditionally prepared kefir according to body weight (1·6 ml/kg for males and 1·9 ml/kg for females) or, alternatively, doses of 100–200 ml/d.

The studies included in this review found kefir to be a safe food for people without serious illnesses. However, further research is necessary before generalising this to people with severe disabilities or more advanced diseases. A healthy, balanced diet is fundamental for quality of life and the prevention of numerous diseases. Easy access to the initial culture (kefir grains), often available by donation, makes this healthy food a viable nutritional alternative for low-income populations, guaranteeing, at the very least, an optimal source of bioactive compounds and essential nutrients.

Financial support

This study received no specific grants from funding agencies, commercial enterprises or non-profit organisations.

Conflict of interest

None.

Authorship

M.K.B.: conception and design, analysis and interpretation, data collection, writing the article, critical revision of the article, final approval of the article, overall responsibility.

G.R.B.: analysis and interpretation, data collection, critical revision of the article, final approval of the article, overall responsibility.

R.R.B.: critical revision of the article, final approval of the article, overall responsibility.

All authors have read and approved the final version of the article.

References

Walker, RL, Vlamakis, H, Lee, JWJ, et al. (2021) Population study of the gut microbiome: associations with diet, lifestyle, and cardiometabolic disease. Genome Med 13, 188.CrossRefGoogle ScholarPubMed
Pu, Q, Lin, P, Gao, P, et al. (2021) Gut microbiota regulate gut-lung axis inflammatory responses by mediating ILC2 compartmental migration. J Immunol 207, 257267.CrossRefGoogle ScholarPubMed
Yue, Q, Cai, M, Xiao, B, et al. (2022) The microbiota–gut–brain axis and epilepsy. Cell Mol Neurobiol 42, 439453.CrossRefGoogle ScholarPubMed
Bonaz, B, Bazin, T & Pellissier, S. (2018) The vagus nerve at the interface of the microbiota-gut-brain axis. Front Neurosci 12, 49.CrossRefGoogle ScholarPubMed
Anand, S & Mande, SS (2018) Diet, microbiota and gut–lung connection. Front Microbiol 9, 2147.CrossRefGoogle ScholarPubMed
Dabke, K, Hendrick, G & Devkota, S. (2019) The gut microbiome and metabolic syndrome. J Clin Invest 129, 40504057.CrossRefGoogle ScholarPubMed
Strandwitz, P (2018) Neurotransmitter modulation by the gut microbiota. Brain Res 1693, 128133.CrossRefGoogle ScholarPubMed
Pereira, TMC, Coco, LZ, Ton, AMM, et al. (2021) The emerging scenario of the gut–brain axis: the therapeutic actions of the new actor kefir against neurodegenerative diseases. Antioxidants (Basel) 10, 1845.CrossRefGoogle ScholarPubMed
Zheng, H, Xu, P, Jiang, Q, et al. (2021) Depletion of acetate-producing bacteria from the gut microbiota facilitates cognitive impairment through the gut–brain neural mechanism in diabetic mice. Microbiome 9, 145.CrossRefGoogle ScholarPubMed
Iebba, V, Totino, V, Gagliardi, A, et al. (2016) Eubiosis and dysbiosis: the two sides of the microbiota. New Microbiol 39, 112.Google ScholarPubMed
Yu, Y, Dunaway, S, Champer, J, et al. (2020) Changing our microbiome: probiotics in dermatology. Br J Dermatol 182, 3946.CrossRefGoogle ScholarPubMed
Li, R, Li, Y, Li, C, et al. (2020) Gut microbiota and endocrine disorder. Adv Exp Med Biol 1238, 143164.CrossRefGoogle ScholarPubMed
Qi, X, Yun, C, Pang, Y, et al. (2021) The impact of the gut microbiota on the reproductive and metabolic endocrine system. Gut Microbes 13, 121.CrossRefGoogle ScholarPubMed
Kitamoto, S & Kamada, N (2022) Periodontal connection with intestinal inflammation: microbiological and immunological mechanisms. Periodontol 2000 89, 142153.CrossRefGoogle ScholarPubMed
de Oliveira, AM, Lourenco, TGB & Colombo, APV (2022) Impact of systemic probiotics as adjuncts to subgingival instrumentation on the oral–gut microbiota associated with periodontitis: a randomized controlled clinical trial. J Periodontol 93, 3144.CrossRefGoogle ScholarPubMed
Gori, S, Inno, A, Belluomini, L, et al. (2019) Gut microbiota and cancer: How gut microbiota modulates activity, efficacy and toxicity of antitumoral therapy. Crit Rev Oncol Hematol 143, 139147.CrossRefGoogle ScholarPubMed
Routy, B, Gopalakrishnan, V, Daillere, R, et al. (2018) The gut microbiota influences anticancer immunosurveillance and general health. Nat Rev Clin Oncol 15, 382396.CrossRefGoogle ScholarPubMed
Mosca, A, Leclerc, M & Hugot, JP (2016) Gut microbiota diversity and human diseases: should we reintroduce key predators in our ecosystem? Front Microbiol 7, 455.CrossRefGoogle ScholarPubMed
Pasolli, E, Asnicar, F, Manara, S, et al. (2019) Extensive unexplored human microbiome diversity revealed by over 150,000 genomes from metagenomes spanning age, geography, and lifestyle. Cell 176, 649662 e620.CrossRefGoogle ScholarPubMed
Lloyd-Price, J, Mahurkar, A, Rahnavard, G, et al. (2017) Strains, functions and dynamics in the expanded human microbiome project. Nature 550, 6166.CrossRefGoogle ScholarPubMed
Byndloss, MX & Baumler, AJ (2018) The germ-organ theory of non-communicable diseases. Nat Rev Microbiol 16, 103110.CrossRefGoogle ScholarPubMed
Plaza-Diaz, J, Ruiz-Ojeda, FJ, Gil-Campos, M, et al. (2019) Mechanisms of action of probiotics. Adv Nutr 10, S49S66.CrossRefGoogle ScholarPubMed
Tamime, AY (2002) Fermented milks: a historical food with modern applications-a review. Eur J Clin Nutr 56(Suppl 4), S2S15.CrossRefGoogle Scholar
Brunser, O (2017) Probiotics: innocuousness, prevention and risks. Rev Chil Pediatr 88, 534540.CrossRefGoogle ScholarPubMed
Hill, C, Guarner, F, Reid, G, et al. (2014) Expert consensus document. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat Rev Gastroenterol Hepatol 11, 506514.CrossRefGoogle ScholarPubMed
Vieira, AT, Fukumori, C & Ferreira, CM (2016) New insights into therapeutic strategies for gut microbiota modulation in inflammatory diseases. Clin Transl Immunology 5, e87.CrossRefGoogle ScholarPubMed
Ceapa, C, Wopereis, H, Rezaiki, L, et al. (2013) Influence of fermented milk products, prebiotics and probiotics on microbiota composition and health. Best Pract Res Clin Gastroenterol 27, 139155.CrossRefGoogle Scholar
Quigley, EMM (2019) Prebiotics and probiotics in digestive health. Clin Gastroenterol Hepatol 17, 333344.CrossRefGoogle ScholarPubMed
de Oliveira Leite, AM, Miguel, MA, Peixoto, RS, et al. (2013) Microbiological, technological and therapeutic properties of kefir: a natural probiotic beverage. Braz J Microbiol 44, 341349.CrossRefGoogle ScholarPubMed
Weschenfelder, S, Wiest, JM & Carvalho, HHC (2013) Anti-Escherichia coli activity in traditional kefir and kefir whey. Rev Inst Latic “Cândido Tostes” 64, 4855.Google Scholar
Farag, MA, Jomaa, SA, El-Wahed, AA, et al. (2020) The many faces of kefir fermented dairy products: quality characteristics, flavour chemistry, nutritional value, health benefits, and safety. Nutrients 12, 346.CrossRefGoogle ScholarPubMed
Leite, AM, Miguel, MA, Peixoto, RS, et al. (2015) Probiotic potential of selected lactic acid bacteria strains isolated from Brazilian kefir grains. J Dairy Sci 98, 36223632.CrossRefGoogle ScholarPubMed
Weschenfelder, S (2009) Characterization of traditional kefir on physicalchemical composition, sensorial characteristic and anti-Escherichia coli activity. Master’s thesis, Universidade Federal do Rio Grande do Sul (UFRGS).Google Scholar
Stiemsma, LT, Nakamura, RE, Nguyen, JG, et al. (2020) Does consumption of fermented foods modify the human gut microbiota? J Nutr 150, 16801692.CrossRefGoogle ScholarPubMed
Chen, HL, Hung, KF, Yen, CC, et al. (2019) Kefir peptides alleviate particulate matter <4 µm (PM4.0)-induced pulmonary inflammation by inhibiting the NF-κB pathway using luciferase transgenic mice. Sci Rep 9, 11529.CrossRefGoogle Scholar
Carasi, P, Racedo, SM, Jacquot, C, et al. (2015) Impact of kefir derived Lactobacillus kefiri on the mucosal immune response and gut microbiota. J Immunol Res 2015, 361604.CrossRefGoogle ScholarPubMed
Seo, MK, Park, EJ, Ko, SY, et al. (2018) Therapeutic effects of kefir grain Lactobacillus-derived extracellular vesicles in mice with 2,4,6-trinitrobenzene sulfonic acid-induced inflammatory bowel disease. J Dairy Sci 101, 86628671.CrossRefGoogle ScholarPubMed
Ghoneum, M, Abdulmalek, S & Pan, D (2020) Reversal of age-associated oxidative stress in mice by PFT, a novel kefir product. Int J Immunopathol Pharmacol 34, 2058738420950149.CrossRefGoogle ScholarPubMed
Azizi, NF, Kumar, MR, Yeap, SK, et al. (2021) Kefir and its biological activities. Foods 10, 1210.CrossRefGoogle ScholarPubMed
Sharifi, M, Moridnia, A, Mortazavi, D, et al. (2017) Kefir: a powerful probiotics with anticancer properties. Med Oncol 34, 183.CrossRefGoogle ScholarPubMed
Al-Mohammadi, AR, Ibrahim, RA, Moustafa, AH, et al. (2021) Chemical constitution and antimicrobial activity of kefir fermented beverage. Molecules 26, 2635.CrossRefGoogle ScholarPubMed
Rodrigues, KL, Caputo, LR, Carvalho, JC, et al. (2005) Antimicrobial and healing activity of kefir and kefiran extract. Int J Antimicrob Agents 25, 404408.CrossRefGoogle ScholarPubMed
Bolla, PA, Carasi, P, Bolla Mde, L, et al. (2013) Protective effect of a mixture of kefir-isolated lactic acid bacteria and yeasts in a hamster model of Clostridium difficile infection. Anaerobe 21, 2833.CrossRefGoogle Scholar
Hadisaputro, S, Djokomoeljanto, RR, Judiono, , et al. (2012) The effects of oral plain kefir supplementation on proinflammatory cytokine properties of the hyperglycemia Wistar rats induced by streptozotocin. Acta Med Indones 44, 100104.Google ScholarPubMed
Brasil, GA, Silva-Cutini, MA, Moraes, FSA, et al. (2018) The benefits of soluble non-bacterial fraction of kefir on blood pressure and cardiac hypertrophy in hypertensive rats are mediated by an increase in baroreflex sensitivity and decrease in angiotensin-converting enzyme activity. Nutrition 51–52, 6672.CrossRefGoogle ScholarPubMed
Ebner, J, Asci Arslan, A, Fedorova, M, et al. (2015) Peptide profiling of bovine kefir reveals 236 unique peptides released from caseins during its production by starter culture or kefir grains. J Proteomics 117, 4157.CrossRefGoogle ScholarPubMed
Friques, AG, Arpini, CM, Kalil, IC, et al. (2015) Chronic administration of the probiotic kefir improves the endothelial function in spontaneously hypertensive rats. J Transl Med 13, 390.CrossRefGoogle ScholarPubMed
Klippel, BF, Duemke, LB, Leal, MA, et al. (2016) Effects of kefir on the cardiac autonomic tones and baroreflex sensitivity in spontaneously hypertensive rats. Front Physiol 7, 211.CrossRefGoogle ScholarPubMed
Liu, JR, Wang, SY, Chen, MJ, et al. (2006) Hypocholesterolaemic effects of milk-kefir and soyamilk-kefir in cholesterol-fed hamsters. Br J Nutr 95, 939946.CrossRefGoogle ScholarPubMed
Wang, Y, Xu, N, Xi, A, et al. (2009) Effects of Lactobacillus plantarum MA2 isolated from Tibet kefir on lipid metabolism and intestinal microflora of rats fed on high-cholesterol diet. Appl Microbiol Biotechnol 84, 341347.CrossRefGoogle ScholarPubMed
Rosa, DD, Dias, MMS, Grzeskowiak, LM, et al. (2017) Milk kefir: nutritional, microbiological and health benefits. Nutr Res Rev 30, 8296.CrossRefGoogle ScholarPubMed
JBI Manual for Evidence Synthesis (2020) Chapter 11: Scoping reviews. https://jbi-global-wiki.refined.site/space/MANUAL/4687342/Chapter+11%3A+Scoping+reviews (accessed October 2021).Google Scholar
O’Brien, KV, Stewart, LK, Forney, LA, et al. (2015) The effects of postexercise consumption of a kefir beverage on performance and recovery during intensive endurance training. J Dairy Sci 98, 74467449.CrossRefGoogle ScholarPubMed
St-Onge, MP, Farnworth, ER, Savard, T, et al. (2002) Kefir consumption does not alter plasma lipid levels or cholesterol fractional synthesis rates relative to milk in hyperlipidemic men: a randomized controlled trial [ISRCTN10820810]. BMC Complement Altern Med 2, 1.CrossRefGoogle ScholarPubMed
Hertzler, SR & Clancy, SM. (2003) Kefir improves lactose digestion and tolerance in adults with lactose maldigestion. J Am Diet Assoc 103, 582587.CrossRefGoogle ScholarPubMed
Figler, M, Mozsik, G, Schaffer, B, et al. (2006) Effect of special Hungarian probiotic kefir on faecal microflora. World J Gastroenterol 12, 11291132.CrossRefGoogle ScholarPubMed
Forejt, M, Šimůnek, J, Brázdová, Z, et al. (2007) The influence of regular consumption of kefir beverage on the incidence of Enterococcus faecalis in the human stool. Scr Med (Brno) 80, 279286.Google Scholar
Topuz, E, Derin, D, Can, G, et al. (2008) Effect of oral administration of kefir on serum proinflammatory cytokines on 5-FU induced oral mucositis in patients with colorectal cancer. Invest New Drugs 26, 567572.CrossRefGoogle ScholarPubMed
Can, G, Topuz, E, Derin, D, et al. (2009) Effect of kefir on the quality of life of patients being treated for colorectal cancer. Oncol Nurs Forum 36, E335E342.CrossRefGoogle ScholarPubMed
Merenstein, DJ, Foster, J & D’Amico, F (2009) A randomized clinical trial measuring the influence of kefir on antibiotic-associated diarrhea: the measuring the influence of kefir (MILK) study. Arch Pediatr Adolesc Med 163, 750754.CrossRefGoogle ScholarPubMed
Bekar, O, Yilmaz, Y & Gulten, M (2011) Kefir improves the efficacy and tolerability of triple therapy in eradicating Helicobacter pylori . J Med Food 14, 344347.CrossRefGoogle ScholarPubMed
Kullisaar, T, Shepetova, J, Zilmer, K, Songisepp, E, Rehema, A, Mikelsaar, M & Zilmer, M (2011) An antioxidant probiotic reduces postprandial lipemia and oxidative stress. Cent Eur J Biol 6, 3240.Google Scholar
Mądry, E, Krasińska, B, Woźniewicz, M, et al. (2011) Tolerance of different dairy products in subjects with symptomatic lactose malabsorption due to adult type hypolactasia. Prz Gastroenterol 6, 310315.Google Scholar
Bakken, JS (2014) Staggered and tapered antibiotic withdrawal with administration of kefir for recurrent Clostridium difficile infection. Clin Infect Dis 59, 858861.CrossRefGoogle ScholarPubMed
Ghasempour, M, Sefdgar, SA, Moghadamnia, AA, et al. (2014) Comparative study of kefir yogurt-drink and sodium fluoride mouth rinse on salivary mutans streptococci. J Contemp Dent Pract 15, 214217.Google ScholarPubMed
Turan, I, Dedeli, O, Bor, S, et al. (2014) Effects of a kefir supplement on symptoms, colonic transit, and bowel satisfaction score in patients with chronic constipation: a pilot study. Turk J Gastroenterol 25, 650656.CrossRefGoogle ScholarPubMed
Judiono, J, Hadisaputro, S, Indranila, KS, et al. (2014) Effects of clear kefir on biomolecular aspects of glycemic status of type 2 diabetes mellitus (T2DM) patients in Bandung, West Java [study on human blood glucose, c peptide and insulin]. FFHD 4, 340348.CrossRefGoogle Scholar
Ostadrahimi, A, Taghizadeh, A, Mobasseri, M, et al. (2015) Effect of probiotic fermented milk (kefir) on glycemic control and lipid profile in type 2 diabetic patients: a randomized double-blind placebo-controlled clinical trial. Iran J Public Health 44, 228237.Google ScholarPubMed
Tu, MY, Chen, HL, Tung, YT, et al. (2015) Short-term effects of kefir-fermented milk consumption on bone mineral density and bone metabolism in a randomized clinical trial of osteoporotic patients. PLoS One 10, e0144231.CrossRefGoogle Scholar
Fathi, Y, Faghih, S, Zibaeenezhad, MJ, et al. (2016) Kefir drink leads to a similar weight loss, compared with milk, in a dairy-rich non-energy-restricted diet in overweight or obese premenopausal women: a randomized controlled trial. Eur J Nutr 55, 295304.CrossRefGoogle ScholarPubMed
Fathi, Y, Ghodrati, N, Zibaeenezhad, MJ, et al. (2017) Kefir drink causes a significant yet similar improvement in serum lipid profile, compared with low-fat milk, in a dairy-rich diet in overweight or obese premenopausal women: A randomized controlled trial. J Clin Lipidol 11, 136146.CrossRefGoogle ScholarPubMed
Alihosseini, N, Moahboob, SA, Farrin, N, et al. (2017) Effect of probiotic fermented milk (kefir) on serum level of insulin and homocysteine in type 2 diabetes patients. Acta Endocrinol (Buchar) 13, 431436.CrossRefGoogle ScholarPubMed
Gölünük, SB, Öztaşan, N, Sözen, H, et al. (2017) Effects of traditional fermented beverages on some blood parameters in aerobic exercises. Biomed Res 28, 94759480.Google Scholar
Diken, H, Oguz, Z, Kaya, H, et al. (2017) Effect of kefir consumption on erythrocyte osmotic fragility and some haemetological parameters in smokers and non-smokers. Acta Physiol 221, 60.Google Scholar
De Araujo, GV, De Lorena, VMB, Montenegro, SML, et al. (2017) Probiotics as an adjunct for the treatment of recurrent wheezing in infants and effects on expression of T-helper 1 and regulatory T cytokines. J Funct Foods 35, 398407.CrossRefGoogle Scholar
Alp, S & Baka, ZM (2018) Effects of probiotics on salivary Streptecoccus mutans and Lactobacillus levels in orthodontic patients. Am J Orthod Dentofacial Orthop 154, 517523.CrossRefGoogle ScholarPubMed
Maki, R, Matsukawa, M, Matsuduka, A, et al. (2018) Therapeutic effect of lyophilized, kefir-fermented milk on constipation among persons with mental and physical disabilities. Jpn J Nurs Sci 15, 218225.CrossRefGoogle ScholarPubMed
Abd-Alwahab, WI & Al-Dulaimi, FK (2018) Effects of kefir as a probiotic on total lipid profile and activity of aspartate amino transferase and alanine amino transferase in serum of human. Biochem Cell Arch 18, 411414.Google Scholar
Gezginc, Y & Maranci, C (2018) Effect of fermented food consumption on biochemical parameters and adipokines levels. Progr Nutr 20, 642647.Google Scholar
Sepp, E, Smidt, I, Štšepetova, J, et al. (2018) The effect of Lactobacillus fermentum ME-3 on the intestinal microbiota and urine polyamines content: a double-blind placebo-controlled pilot trial. J Funct Foods 48, 430438.CrossRefGoogle Scholar
Ozcan, H, Oskay, U & Bodur, AF (2019) Effects of kefir on quality of life and sleep disturbances in postmenopausal women. Holist Nurs Pract 33, 207213.CrossRefGoogle ScholarPubMed
Bellikci-Koyu, E, Sarer-Yurekli, BP, Akyon, Y, et al. (2019) Effects of regular kefir consumption on gut microbiota in patients with metabolic syndrome: a parallel-group, randomized, controlled study. Nutrients 11, 2089.CrossRefGoogle ScholarPubMed
Yilmaz, I, Dolar, ME & Ozpinar, H (2019) Effect of administering kefir on the changes in fecal microbiota and symptoms of inflammatory bowel disease: a randomized controlled trial. Turk J Gastroenterol 30, 242253.CrossRefGoogle ScholarPubMed
Bashiti, TA & Zabut, BM (2019) Effect of probiotic fermented milk (Kefir) on some blood biochemical parameters among newly diagnosed type 2 diabetic adult males in Gaza governorate. Curr Res Nutr Food Sci 7, 568575.CrossRefGoogle Scholar
Praznikar, ZJ, Kenig, S, Vardjan, T, et al. (2020) Effects of kefir or milk supplementation on zonulin in overweight subjects. J Dairy Sci 103, 39613970.CrossRefGoogle ScholarPubMed
Ton, AMM, Campagnaro, BP, Alves, GA, et al. (2020) Oxidative stress and dementia in Alzheimer’s patients: effects of synbiotic supplementation. Oxid Med Cell Longev 2020, 2638703.CrossRefGoogle ScholarPubMed
Wang, MC, Zaydi, AI, Lin, WH, et al. (2020) Putative probiotic strains isolated from kefir improve gastrointestinal health parameters in adults: a randomized, single-blind, placebo-controlled study. Probiotics Antimicrob Proteins 12, 840850.CrossRefGoogle ScholarPubMed
Lee, MC, Jhang, WL, Lee, CC, et al. (2021) The effect of kefir supplementation on improving human endurance exercise performance and antifatigue. Metabolites 11.CrossRefGoogle ScholarPubMed
Reddy, S, Madhu, V, Punithavathy, R et al. (2021) Comparative evaluation of efficacy of kefir milk probiotic curd and probiotic drink on streptococcus mutans in 8–12-year-old children: an in vivo study. Int J Clin Pediatr Dent 14, 120127.CrossRefGoogle ScholarPubMed
Alves, E, Gregorio, J, Baby, AR et al. (2021) Homemade kefir consumption improves skin condition-a study conducted in healthy and atopic volunteers. Foods 10, 2794.CrossRefGoogle ScholarPubMed
Kurt, TT, Gökırmaklı, Ç & Guzel-Seydim, ZB (2021) Effects of kefir consumption on carbohydrate profile of mother’s milk. FFHD 11, 473483.CrossRefGoogle Scholar
Hosainzadegn, H & Hosainzadegan, M (2021) Traditional probiotic (kefir) effects on glycated hemoglobin A level and weight of an indexed diabetic patient. Int J Prev Med 12, 139.Google ScholarPubMed
Smoak, P, Harman, N, Flores, V, et al. (2021) Kefir is a viable exercise recovery beverage for cancer survivors enrolled in a structured exercise program. Med Sci Sports Exerc 53, 20452053.CrossRefGoogle Scholar
Caferoglu, Z & Aytekin Sahin, G (2021) The effects of kefir in mixed meals on appetite and food intake: a randomized cross-over trial. Rev Nutr 34, e190174.CrossRefGoogle Scholar
Da Silva Ghizi, AC, De Almeida Silva, M, De Andrade Moraes, FS, et al. (2021) Kefir improves blood parameters and reduces cardiovascular risks in patients with metabolic syndrome. PharmaNutrition 16, 100266.CrossRefGoogle Scholar
Tunay, RT & Taş, TK (2022) Verticle transmission of unique bacterial strains from mother to infant via consuming natural kefir. Int Dairy J 126, 105251.CrossRefGoogle Scholar
Aziz, I, Whitehead, WE, Palsson, OS, et al. (2020) An approach to the diagnosis and management of Rome IV functional disorders of chronic constipation. Expert Rev Gastroenterol Hepatol 14, 3946.CrossRefGoogle Scholar
Samson, SL & Garber, AJ (2014) Metabolic syndrome. Endocrinol Metab Clin North Am 43, 123.CrossRefGoogle ScholarPubMed
Festi, D, Schiumerini, R, Eusebi, LH, et al. (2014) Gut microbiota and metabolic syndrome. World J Gastroenterol 20, 1607916094.CrossRefGoogle ScholarPubMed
Li, HY, Zhou, DD, Gan, RY, et al. (2021) Effects and mechanisms of probiotics, prebiotics, synbiotics, and postbiotics on metabolic diseases targeting gut microbiota: a narrative review. Nutrients 13, 3211CrossRefGoogle ScholarPubMed
Fasano, A (2020) All disease begins in the (leaky) gut: role of zonulin-mediated gut permeability in the pathogenesis of some chronic inflammatory diseases. F1000Res 9, F1000 Faculty Rev-69.CrossRefGoogle ScholarPubMed
Gonzalez-Orozco, BD, Garcia-Cano, I, Jimenez-Flores, R, et al. (2022) Invited review: milk kefir microbiota—direct and indirect antimicrobial effects. J Dairy Sci 105, 37033715.CrossRefGoogle ScholarPubMed
Brashears, MM, Gilliland, SE & Buck, LM (1998) Bile salt deconjugation and cholesterol removal from media by Lactobacillus casei . J Dairy Sci 81, 21032110.CrossRefGoogle ScholarPubMed
Lye, HS, Rusul, G & Liong, MT (2010) Removal of cholesterol by lactobacilli via incorporation and conversion to coprostanol. J Dairy Sci 93, 13831392.CrossRefGoogle ScholarPubMed
Ooi, LG & Liong, MT (2010) Cholesterol-lowering effects of probiotics and prebiotics: a review of in vivo and in vitro findings. Int J Mol Sci 11, 24992522.CrossRefGoogle ScholarPubMed
Ditscheid, B, Keller, S & Jahreis, G (2005) Cholesterol metabolism is affected by calcium phosphate supplementation in humans. J Nutr 135, 16781682.CrossRefGoogle ScholarPubMed
Lorenzen, JK & Astrup, A (2011) Dairy calcium intake modifies responsiveness of fat metabolism and blood lipids to a high-fat diet. Br J Nutr 105, 18231831.CrossRefGoogle ScholarPubMed
Denke, MA, Fox, MM & Schulte, MC (1993) Short-term dietary calcium fortification increases fecal saturated fat content and reduces serum lipids in men. J Nutr 123, 10471053.Google ScholarPubMed
Salari, A, Ghodrat, S, Gheflati, A, et al. (2021) Effect of kefir beverage consumption on glycemic control: a systematic review and meta-analysis of randomized controlled clinical trials. Complement Ther Clin Pract 44, 101443.CrossRefGoogle ScholarPubMed
Hills, RD Jr, Pontefract, BA, Mishcon, HR, et al. (2019) Gut microbiome: profound implications for diet and disease. Nutrients 11, 1613.CrossRefGoogle ScholarPubMed
Rothschild, D, Weissbrod, O, Barkan, E, et al. (2018) Environment dominates over host genetics in shaping human gut microbiota. Nature 555, 210215.CrossRefGoogle ScholarPubMed
Sairenji, T, Collins, KL & Evans, DV (2017) An update on inflammatory bowel disease. Prim Care 44, 673692.CrossRefGoogle ScholarPubMed
Flynn, S & Eisenstein, S (2019) Inflammatory bowel disease presentation and diagnosis. Surg Clin North Am 99, 10511062.CrossRefGoogle ScholarPubMed
Lee, M & Chang, EB (2021) Inflammatory bowel diseases (IBD) and the microbiome-searching the crime scene for clues. Gastroenterology 160, 524537.CrossRefGoogle ScholarPubMed
Chassaing, B & Darfeuille-Michaud, A (2011) The commensal microbiota and enteropathogens in the pathogenesis of inflammatory bowel diseases. Gastroenterology 140, 17201728.CrossRefGoogle ScholarPubMed
Jeong, DY, Kim, S, Son, MJ, et al. (2019) Induction and maintenance treatment of inflammatory bowel disease: a comprehensive review. Autoimmun Rev 18, 439454.CrossRefGoogle ScholarPubMed
Slattery, C, Cotter, PD & O’Toole, PW (2019) Analysis of health benefits conferred by lactobacillus species from kefir. Nutrients 11, 1252.CrossRefGoogle ScholarPubMed
Leite, AM, Leite, DC, Del Aguila, EM, et al. (2013) Microbiological and chemical characteristics of Brazilian kefir during fermentation and storage processes. J Dairy Sci 96, 41494159.CrossRefGoogle ScholarPubMed
Dallas, DC, Citerne, F, Tian, T, et al. (2016) Peptidomic analysis reveals proteolytic activity of kefir microorganisms on bovine milk proteins. Food Chem 197, 273284.CrossRefGoogle ScholarPubMed
Kim, JE & Kim, HS (2019) Microbiome of the skin and gut in atopic dermatitis (AD): understanding the pathophysiology and finding novel management strategies. J Clin Med 8, 444.CrossRefGoogle ScholarPubMed
Nie, P, Li, Z, Wang, Y, et al. (2019) Gut microbiome interventions in human health and diseases. Med Res Rev 39, 22862313.CrossRefGoogle ScholarPubMed
Gensollen, T & Blumberg, RS (2017) Correlation between early-life regulation of the immune system by microbiota and allergy development. J Allergy Clin Immunol 139, 10841091.CrossRefGoogle ScholarPubMed
Stinson, LF, Sindi, ASM, Cheema, AS, et al. (2021) The human milk microbiome: who, what, when, where, why, and how? Nutr Rev 79, 529543.CrossRefGoogle Scholar
Guo, Q, Goldenberg, JZ, Humphrey, C, et al. (2019) Probiotics for the prevention of pediatric antibiotic-associated diarrhea. Cochrane Database Syst Rev 4, CD004827.Google ScholarPubMed
Depoorter, L & Vandenplas, Y (2021) Probiotics in pediatrics. A review and practical guide. Nutrients 13, 2176.CrossRefGoogle Scholar
Jiang, W, Ni, B, Liu, Z, et al. (2020) The role of probiotics in the prevention and treatment of atopic dermatitis in children: an updated systematic review and meta-analysis of randomized controlled trials. Paediatr Drugs 22, 535549.CrossRefGoogle ScholarPubMed
Turck, D, Bernet, JP, Marx, J, et al. (2003) Incidence and risk factors of oral antibiotic-associated diarrhea in an outpatient pediatric population. J Pediatr Gastroenterol Nutr 37, 2226.Google Scholar
Nguyen, QN, Himes, JE, Martinez, DR, et al. (2016) The impact of the gut microbiota on humoral immunity to pathogens and vaccination in early infancy. PLoS Pathog 12, e1005997.CrossRefGoogle ScholarPubMed
Mosaddad, SA, Tahmasebi, E, Yazdanian, A, et al. (2019) Oral microbial biofilms: an update. Eur J Clin Microbiol Infect Dis 38, 20052019.CrossRefGoogle ScholarPubMed
Dashper, SG, Mitchell, HL, Le Cao, KA, et al. (2019) Temporal development of the oral microbiome and prediction of early childhood caries. Sci Rep 9, 19732.CrossRefGoogle ScholarPubMed
Patidar, D, Sogi, S, Singh, V, et al. (2018) Salivary levels of Streptococcus mutans and Streptococcus sanguinis in early childhood caries: an in vivo study. J Indian Soc Pedod Prev Dent 36, 386390.Google ScholarPubMed
Fatahi, A, Soleimani, N & Afrough, P (2021) Anticancer activity of kefir on glioblastoma cancer cell as a new treatment. Int J Food Sci 2021, 8180742.CrossRefGoogle ScholarPubMed
Rafie, N, Golpour Hamedani, S, Ghiasvand, R, et al. (2015) Kefir and cancer: a systematic review of literatures. Arch Iran Med 18, 852857.Google Scholar
Gracia, CR & Freeman, EW (2018) Onset of the menopause transition: the earliest signs and symptoms. Obstet Gynecol Clin North Am 45, 585597.CrossRefGoogle ScholarPubMed
Potter, B, Schrager, S, Dalby, J, et al. (2018) Menopause. Prim Care 45, 625641.CrossRefGoogle ScholarPubMed
Lane, JM, Russell, L & Khan, SN (2000) Osteoporosis. Clin Orthop Relat Res 372, 139150.CrossRefGoogle Scholar
The North American Menopause Society (2021) Management of osteoporosis in postmenopausal women: the 2021 position statement of The North American Menopause Society. Menopause 28, 973997.CrossRefGoogle Scholar
Lopez, OL & Kuller, LH (2019) Epidemiology of aging and associated cognitive disorders: prevalence and incidence of Alzheimer’s disease and other dementias. Handb Clin Neurol 167, 139148.CrossRefGoogle ScholarPubMed
Querfurth, HW & LaFerla, FM (2010) Alzheimer’s disease. N Engl J Med 362, 329344.CrossRefGoogle ScholarPubMed
Kinney, JW, Bemiller, SM, Murtishaw, AS, et al. (2018) Inflammation as a central mechanism in Alzheimer’s disease. Alzheimers Dement (N Y) 4, 575590.CrossRefGoogle ScholarPubMed
Luca, M, Di Mauro, M, Di Mauro, M, et al. (2019) Gut microbiota in Alzheimer’s disease, depression, and type 2 diabetes mellitus: the role of oxidative stress. Oxid Med Cell Longev 2019, 4730539.Google ScholarPubMed
Chen, Z & Zhong, C (2014) Oxidative stress in Alzheimer’s disease. Neurosci Bull 30, 271281.CrossRefGoogle ScholarPubMed
Megur, A, Baltriukiene, D, Bukelskiene, V, et al. (2020) The microbiota–gut–brain axis and Alzheimer’s disease: neuroinflammation is to blame? Nutrients 13, 37.CrossRefGoogle ScholarPubMed
Jiang, C, Li, G, Huang, P, et al. (2017) The gut microbiota and Alzheimer’s disease. J Alzheimers Dis 58, 115.CrossRefGoogle ScholarPubMed
Doifode, T, Giridharan, VV, Generoso, JS, et al. (2021) The impact of the microbiota-gut-brain axis on Alzheimer’s disease pathophysiology. Pharmacol Res 164, 105314.CrossRefGoogle ScholarPubMed
Cryan, JF, O’Riordan, KJ, Cowan, CSM, et al. (2019) The microbiota–gut–brain axis. Physiol Rev 99, 18772013.CrossRefGoogle ScholarPubMed
Alasmari, F, Alshammari, MA, Alasmari, AF, et al. (2018) Neuroinflammatory cytokines induce amyloid beta neurotoxicity through modulating amyloid precursor protein levels/metabolism. Biomed Res Int 2018, 3087475.CrossRefGoogle ScholarPubMed
Watanabe, S, Narisawa, Y, Arase, S, et al. (2003) Differences in fecal microflora between patients with atopic dermatitis and healthy control subjects. J Allergy Clin Immunol 111, 587591.CrossRefGoogle ScholarPubMed
Fieten, KB, Totte, JEE, Levin, E, et al. (2018) Fecal microbiome and food allergy in pediatric atopic dermatitis: a cross-sectional pilot study. Int Arch Allergy Immunol 175, 7784.CrossRefGoogle ScholarPubMed
Lee, SY, Lee, E, Park, YM, et al. (2018) Microbiome in the gut–skin axis in atopic dermatitis. Allergy Asthma Immunol Res 10, 354362.CrossRefGoogle ScholarPubMed
Carasi, P, Racedo, SM, Jacquot, C, et al. (2015) Impact of kefir derived Lactobacillus kefiri on the mucosal immune response and gut microbiota. J Immunol Res 2015, 361604.CrossRefGoogle ScholarPubMed
Kim, D-H, Jeong, D, Kang, I-B, et al. (2017) Dual function of Lactobacillus kefiri DH5 in preventing high-fat-diet-induced obesity: direct reduction of cholesterol and upregulation of PPARα in adipose tissue. Mol Nutr Food Res 61, 1700252.CrossRefGoogle ScholarPubMed
Chen, YP & Chen, MJ (2013) Effects of Lactobacillus kefiranofaciens M1 isolated from kefir grains on germ-free mice. PLoS One 8, 74677477.Google ScholarPubMed
Chen, YP, Hsiao, PJ, Hong, WS, et al. (2012) Lactobacillus kefiranofaciens M1 isolated from milk kefir grains ameliorates experimental colitis in vitro and in vivo . J Dairy Sci 95, 6374.CrossRefGoogle ScholarPubMed
Dobson, A, O’Sullivan, O, Cotter, PD, et al. (2011) High-throughput sequence-based analysis of the bacterial composition of kefir and an associated kefir grain. FEMS Microbiol Lett 320, 5662.CrossRefGoogle Scholar
Marsh, AJ, O’Sullivan, O, Hill, C, et al. (2013) Sequencing-based analysis of the bacterial and fungal composition of kefir grains and milks from multiple sources. PLoS One 8, e69371.CrossRefGoogle ScholarPubMed
Walsh, AM, Crispie, F, Kilcawley, K, et al. (2016) Microbial succession and flavor production in the fermented dairy beverage Kefir. mSystems 1, e0005216.CrossRefGoogle ScholarPubMed
Bourrie, BCT, Cotter, PD & Willing, BP (2018) Traditional kefir reduces weight gain and improves plasma and liver lipid profiles more successfully than a commercial equivalent in a mouse model of obesity. J Funct Foods, 46, 2937.CrossRefGoogle Scholar
Bourrie, BCT, Ju, T, Fouhse, JM, et al. (2021) Kefir microbial composition is a deciding factor in the physiological impact of kefir in a mouse model of obesity. Br J Nutr 125, 129138.CrossRefGoogle Scholar
Reagan-Shaw, S, Nihal, M & Ahmad, N (2008) Dose translation from animal to human studies revisited. FASEB J 22, 659661.CrossRefGoogle ScholarPubMed
Diniz Rosa, D, Gouveia Peluzio do, MC, Pérez Bueno, T, et al. (2014) Evaluation of the subchronic toxicity of kefir by oral administration in Wistar rats. Nutr Hosp 29, 13521359.Google ScholarPubMed
Figure 0

Fig. 1. Flowchart of article inclusion.

Figure 1

Fig. 2. Year of publication of English-language studies on kefir as a therapeutic agent. *Until 9 March 2022.

Figure 2

Fig. 3. Number of articles according to country of origin (absolute number, percentage).

Figure 3

Table 1. Overview of included studies

Figure 4

Table 2. Areas of clinical research on kefir