Hostname: page-component-76fb5796d-22dnz Total loading time: 0 Render date: 2024-04-27T23:58:21.646Z Has data issue: false hasContentIssue false

Modulation of gut microbiota by diet and probiotics: potential approaches to prevent gestational diabetes mellitus

Published online by Cambridge University Press:  13 June 2023

Marisa Carreira Cruz
Affiliation:
School of Health Sciences, Polytechnic of Leiria, Leiria, Portugal
Sarah Azinheiro
Affiliation:
Center for Innovative Care and Health Technology, Polytechnic of Leiria, Leiria, Portugal
Sónia Gonçalves Pereira*
Affiliation:
Center for Innovative Care and Health Technology, Polytechnic of Leiria, Leiria, Portugal
*
Corresponding author: Sónia Gonçalves Pereira; Email: sonia.pereira@ipleiria.pt

Abstract

Gestational diabetes mellitus (GDM) is a rising global health problem that affects approximately 6% of pregnant women. Lifestyle interventions, particularly diet, and exercise are the first-line treatment, followed by pharmacotherapy, but with associated side effects to both mother and offspring. Modulation of gut microbiota may help prevent or manage GDM. Some gut bacterial groups associated with GDM are also associated with inflammatory biomarkers and gut dysbiosis. Available literature reports that low-glycaemic index diet reduces maternal fasting and 2-hour postprandial glucose and maintains a beneficial gut bacterial composition. Pre- and probiotics can aid GDM therapy by modulating gut microbiota to eubiotic status and improving glucose metabolism. Probiotics as adjuvant GDM therapy should consider bacterial strains, dosage, and treatment duration. Limitations in their use require further studies to develop specific probiotic-based GDM supplement therapy that impacts glycaemic control and inflammatory status by reducing fasting plasma glucose, insulin resistance, and improving lipid profiles of pregnant women.

Type
Mini Review
Creative Commons
Creative Common License - CCCreative Common License - BYCreative Common License - NCCreative Common License - ND
This is an Open Access article, distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivatives licence (http://creativecommons.org/licenses/by-nc-nd/4.0), which permits non-commercial re-use, distribution, and reproduction in any medium, provided that no alterations are made and the original article is properly cited. The written permission of Cambridge University Press must be obtained prior to any commercial use and/or adaptation of the article.
Copyright
© The Author(s), 2023. Published by Cambridge University Press in association with The Nutrition Society

Introduction

Gestational diabetes mellitus (GDM), defined as glucose intolerance that results in hyperglycaemia with first recognition during pregnancy (Eades et al., Reference Eades, Cameron and Evans2017; Hasain et al., Reference Hasain, Mokhtar, Kamaruddin, Mohamed Ismail, Razalli, Gnanou and Raja Ali2020), is an increasing public health concern with a rising prevalence of 5.4% in European and 7.6% in North American pregnant women (Casagrande et al., Reference Casagrande, Linder and Cowie2018). GDM impacts maternal-foetal health causing both short- and long-term adverse effects, including higher risk of preeclampsia and caesarean delivery for mothers; and macrosomia, preterm birth, respiratory distress, and shoulder dystocia for foetuses (HAPO Study Cooperative Research Group et al., Reference Metzger, Lowe, Dyer, Trimble, Chaovarindr, Coustan, Hadden, McCance, Hod, McIntyre, Oats, Persson, Rogers and Sacks2008; Schneider et al., Reference Schneider, Hoeft, Freerksen, fischer, Roehrig, Yamamoto and Maul2011; Wendland et al., Reference Wendland, Torloni, Falavigna, Trujillo, Dode, Campos, Duncan and Schmidt2012; Kc et al., Reference Kc, Shakya and Zhang2015; Billionnet et al., Reference Billionnet, Mitanchez, Weill, Nizard, Alla, Hartemann and Jacqueminet2017). More than half of pregnant women who develop GDM have at least one risk factor: >25 years old, body mass index (BMI) > 30 kg/m2, history of impaired glucose tolerance, previous pregnancies with GDM or macrosomia, multiple pregnancies, family history of type 2 diabetes, among others (Lefkovits et al., Reference Lefkovits, Stewart and Murphy2019). GDM diagnosis is made in the first trimester if a fasting glucose test returns a plasma glucose value ≥92 and <126 mg/dL or if returning a normal value, through an oral glucose tolerance test (OGTT) made at 24–28 weeks of gestation if a fasting plasma glucose value ≥92 mg/dL or ≥180 mg/dL at one hour or ≥153 mg/dL at 2 hours (Wendland et al., Reference Wendland, Torloni, Falavigna, Trujillo, Dode, Campos, Duncan and Schmidt2012). Appropriate management of hyperglycaemia combined with lifestyle interventions, diet, and exercise prevents maternal obesity and GDM (Koivusalo et al., Reference Koivusalo, Rönö, Klemetti, Roine, Lindström, Erkkola, Kaaja, Pöyhönen-Alho, Tiitinen, Huvinen, Andersson, Laivuori, Valkama, Meinilä, Kautiainen, Eriksson and Stach-Lempinen2016; Wang et al., Reference Wang, Wei, Zhang, Zhang, Xu, Sun, Su, Zhang, Liu, Feng, Shou, Guelfi, Newnham and Yang2017). Other approaches are also available when diet and exercise alone are insufficient to control blood glucose levels, including pharmacological (insulin and/or oral hypoglycaemic agents, such as metformin) and non-pharmacological therapies (probiotics; Simmons, Reference Simmons2015). Pharmacotherapy has benefits for glucose control, especially insulin since it is unable to cross the placental barrier. However, this approach may result in side effects, including hypertensive disorders, when using insulin (Brown et al., Reference Brown, Grzeskowiak, Williamson, Downie and Crowther2017) and diarrhoea, abdominal pain, and headache, when using metformin (Dodd et al., Reference Dodd, Grivell, Deussen and Hague2018). Probiotics are defined as “live microorganisms which when administered in adequate amounts confer a health benefit on the host” (Food and Agricultural Organization [FAO] of the United Nations and World Health Organization [WHO], 2001] and thus may be part of a non-pharmacological therapy. Gut microbiota, the group of living microorganisms colonising the gastrointestinal tract, is linked to human metabolism regulation. Alterations in gut microbiota composition are associated with metabolic diseases such as obesity, type 2 diabetes, and metabolic syndrome (Pascale et al., Reference Pascale, Marchesi, Marelli, Coppola, Luzi, Govoni, Giustina and Gazzaruso2018) and can play an essential role in modulating insulin resistance (IR) and inflammatory response in GDM (Kuang et al., Reference Kuang, Lu, Li, Li, Yuan, He, Chen, Xiao, Shen, Qiu, Wu, Hu, Wu, Li, Chen, Deng, Papasian, Xia and Qiu2017). Diet and probiotics can affect gut microbiota composition (Wen and Duffy, Reference Wen and Duffy2017; Ponzo et al., Reference Ponzo, Fedele, Goitre, Leone, Lezo, Monzeglio, Finocchiaro, Ghigo and Bo2019,Reference Ponzo, Ferrocino, Zarovska, Amenta, Leone, Monzeglio, Rosato, Pellegrini, Gambino, Cassader, Ghigo, Cocolin and Bob) and therefore can be a potential associative therapy for GDM if applied simultaneously.

The aim of this literature review was to compare the results of studies reporting the efficacy of both approaches (probiotics and diet) to prevent and manage GDM during pregnancy and to clarify if they could be potential therapeutic adjuvants for GDM treatment.

Current status of knowledge

The role of human gut microbiota in homeostasis

The human gastrointestinal tract is colonised by a collection of approximately 100 trillion microorganisms (Lozupone et al., Reference Lozupone, Stombaugh, Gordon, Jansson and Knight2012), whose composition is determined by host genetics, diet, immune responses, and the environment (Lynch and Pedersen, Reference Lynch and Pedersen2016). The development of our gut microbiota begins immediately at birth, being initially undifferentiated and progressively shaped by three major factors – type of delivery, breastfeeding, and weaning (Nicholson et al., Reference Nicholson, Holmes, Kinross, Burcelin, Gibson, Jia and Pettersson2012). In adult life, the gut microbiota has similar patterns between individuals at higher phylogenetic levels, being mainly composed of five bacteria phyla – Bacteroidetes, Firmicutes, Proteobacteria, Actinobacteria, and Verrucomicrobia (Tremaroli and Bäckhed, Reference Tremaroli and Bäckhed2012). Firmicutes and Bacteroidetes are the most prevalent, representing more than 90% of the overall intestinal microbiota (Shen et al., Reference Shen, Obin and Zhao2013). At a genus level, some of the most frequently found bacteria are Bifidobacterium, Lactobacillus, Bacteroides, Clostridium, Escherichia, Streptococcus, and Ruminococcus (Conlon and Bird, Reference Conlon and Bird2015). At a species level, the variability is much higher, with healthy individuals having highly diverse gut microbiota (Flint et al., Reference Flint, Duncan, Scott and Louis2015). This complex micro-ecosystem holds a synbiotic relationship between each other (the microbes) and the host, who provides them a balanced environment to live while microbes provide the host with several benefits through a variety of functions such as modulating the intestinal barrier (Natividad and Verdu, Reference Natividad and Verdu2013), producing bioactive compounds like short-chain fatty acids (SCFA; Topping and Clifton, Reference Topping and Clifton2001), maintaining host immune system (Gensollen et al., Reference Gensollen, Iyer, Kasper and Blumberg2016), and protecting the host against ingested pathogens (Bäumler and Sperandio, Reference Bäumler and Sperandio2016). Alterations in the intestinal microbiota and its metabolic pathways, termed dysbiosis (major changes in the resident microbial community with impairments for the host; Clemente et al., Reference Clemente, Ursell, Parfrey and Knight2012), may contribute to the development of several chronic diseases, such as inflammatory bowel disease, multiple sclerosis, arthritis, and allergic inflammation (Kamada et al., Reference Kamada, Seo, Chen and Núñez2013). Hence, it is important to maintain the balance of our gut microbiota composition to ensure homeostasis. Gut microbial fermentation of non-digestible foods (dietary fibres and resistant starches) maintains bowel health (Canfora et al., Reference Canfora, Jocken and Blaak2015) and leads to the production of SCFA, mainly butyrate, acetate, and propionate, via diverse biochemical pathways (Topping and Clifton, Reference Topping and Clifton2001). These metabolites can be used as energy sources by colonic epithelial cells or absorbed into the bloodstream (den Besten et al., Reference den Besten, van Eunen, Groen, Venema, Reijngoud and Bakker2013). In the colon lumen, acetate, and propionate, mainly produced by Bacteroidetes (LeBlanc et al., Reference LeBlanc, Chain, Martín, Bermúdez-Humarán, Courau and Langella2017), release peptide YY (PPY) and glucagon-like peptide (GLP-1), which influence satiety and bowel transit; whereas butyrate, mainly produced by Firmicutes (LeBlanc et al., Reference LeBlanc, Chain, Martín, Bermúdez-Humarán, Courau and Langella2017), inhibits histone deacetylases (HDACs) and a specific G protein-coupled receptor (GPR109A), conferring anti-inflammatory effects (Koh et al., Reference Koh, de Vadder, Kovatcheva-Datchary and Bäckhed2016), apart from modulating the expression of tight junction protein and mucins (Canfora et al., Reference Canfora, Jocken and Blaak2015). Additionally, SCFA also plays an important role in blood glucose level regulation and glucose homeostasis by inhibiting glycolysis and stimulating lipogenesis or gluconeogenesis, along with managing excessive production of cholesterol and conferring anti-carcinogenic action (Pascale et al., Reference Pascale, Marchesi, Marelli, Coppola, Luzi, Govoni, Giustina and Gazzaruso2018). Further studies are needed to understand the impact of the different compounds produced by gut microbiome in the metabolism of both mother and offspring, and elucidate which microorganisms are responsible for these changes.

Modification of gut microbiota during pregnancy

Pregnancy imposes a great number of physiological adaptations. Currently, there is no specific definition of healthy gut microbiota; however, it is known that its composition is highly diverse in healthy individuals (Meijnikman et al., Reference Meijnikman, Gerdes, Nieuwdorp and Herrema2018). Studies that explored the gut microbiota composition of healthy pregnant women found some important changes associated with an increase in maternal body weight by fat deposition and new dietary habits that culminate in a progressive increase in the food intake, essential for the foetus growth (Reference di Simone, Santamaria Ortiz, Specchia, Tersigni, Villa, Gasbarrini, Scambia and D’Ippolitodi Simone et al., 2020). Physiological changes are also part of a normal pregnancy, with maternal tissues becoming increasingly resistant to insulin in approximately 50–60% of women with normal glucose tolerance or with GDM (Kampmann et al., Reference Kampmann, Knorr, Fuglsang and Ovesen2019). In addition to weight gain and adiposity, pregnant women have significantly higher leptin, insulin and IR, cholesterol, and glycated haemoglobin levels as compared with nonpregnant women (Collado et al., Reference Collado, Isolauri, Laitinen and Salminen2008). In the pregnancy first trimester, the gut microbiota is expected to be similar to that of a healthy nonpregnant woman, classified into different enterotypes, considering three different groups of bacteria: one enterotype characterised by the presence of Bacteroides; another enterotype by higher proportion of Prevotella; and a third enterotype dominated by Ruminococcus (di Simone et al., Reference di Simone, Santamaria Ortiz, Specchia, Tersigni, Villa, Gasbarrini, Scambia and D’Ippolito2020). Between the first and the third trimester, gut microbiota changes substantially decreasing its diversity, with increased proportions of Proteobacteria, commonly associated with inflammation, while the number of butyrate-producing bacteria with anti-inflammatory effects decreases (Koren et al., Reference Koren, Goodrich, Cullender, Spor, Laitinen, Kling Bäckhed, Gonzalez, Werner, Angenent, Knight, Bäckhed, Isolauri, Salminen and Ley2012).

Studies comparing the intestinal microbiota between healthy-weight pregnant women and overweight and/or obese pregnant women were also considered. Gomez-Arango et al. (Reference Gomez-Arango, Barrett, McIntyre, Callaway, Morrison and Dekker Nitert2016) indicate that the ratio between phyla Firmicutes and Bacteroidetes was approximately 3:1 when analysing stool samples of both overweight and obese pregnant women, showing a slightly higher abundance of Firmicutes in obese women. These results were linked with the higher expression of enzymes engaged in the digestion of polysaccharide, highlighting that more energy can be obtained from the same diet (Cani, Reference Cani2013). Some correlations between specific taxa and pregnancy variables were observed. Lachnospiraceae and Ruminococcaceae families (both from Firmicutes phyla) were strongly correlated with leptin and positively associated with BMI. Bacteroidaceae relates with ghrelin that, in turn, was negatively associated with BMI, and positively correlated with Rikenellaceae (both from Bacteroidetes phyla). Collinsella positively correlated with insulin levels and triglycerides while Coprococcus (butyrate producer) correlated with gastric-inhibitory polypeptide (GIP), an incretin hormone (Gomez-Arango et al., Reference Gomez-Arango, Barrett, McIntyre, Callaway, Morrison and Dekker Nitert2016). When gestational weight gain (GWG) is excessive, pregnant women have an increased risk of developing GDM, obesity, metabolic syndrome (Carreno et al., Reference Carreno, Clifton, Hauth, Myatt, Roberts, Spong, Varner, Thorp, Mercer, Peaceman, Ramin, Carpenter, Sciscione, Tolosa, Saade and Sorokin2012; Gilmore et al., Reference Gilmore, Klempel-Donchenko and Redman2015) and delivering a baby larger-for-gestational age (Carreno et al., Reference Carreno, Clifton, Hauth, Myatt, Roberts, Spong, Varner, Thorp, Mercer, Peaceman, Ramin, Carpenter, Sciscione, Tolosa, Saade and Sorokin2012; Ferraro et al., Reference Ferraro, Barrowman, Prud’homme, Walker, Wen, Rodger and Adamo2012; Kim et al., Reference Kim, Sharma, Sappenfield, Wilson and Salihu2014). In this situation, gut microbiota is associated with lower α-diversity (DiGiulio et al., Reference DiGiulio, Callahan, McMurdie, Costello, Lyell, Robaczewska, Sun, Goltsman, Wong, Shaw, Stevenson, Holmes and Relman2015), and the presence of Eisenbergiella, Lactobacillus (Crusell et al., Reference Crusell, Hansen, Nielsen, Allin, Rühlemann, Damm, Vestergaard, Rørbye, Jørgensen, Christiansen, Heinsen, Franke, Hansen, Lauenborg and Pedersen2018), Blautia, Ruminoccocus, and Feacalibacterium (Stanislawski et al., Reference Stanislawski, Dabelea, Wagner, Sontag, Lozupone and Eggesbø2017) genus and Escherichia coli (Santacruz et al., Reference Santacruz, Collado, García-Valdés, Segura, Martín-Lagos, Anjos, Martí-Romero, Lopez, Florido, Campoy and Sanz2010) species. On the other hand, Bifidobacterium and Akkermansia muciniphila (Collado et al., Reference Collado, Isolauri, Laitinen and Salminen2008; Santacruz et al., Reference Santacruz, Collado, García-Valdés, Segura, Martín-Lagos, Anjos, Martí-Romero, Lopez, Florido, Campoy and Sanz2010), along with Christensenella and Alistipes (Crusell et al., Reference Crusell, Hansen, Nielsen, Allin, Rühlemann, Damm, Vestergaard, Rørbye, Jørgensen, Christiansen, Heinsen, Franke, Hansen, Lauenborg and Pedersen2018), are associated with the opposite trend. Additionally, overweight and obese pregnant women were reported to have higher levels of Bacteroides (Collado et al., Reference Collado, Isolauri, Laitinen and Salminen2008), Staphylococcus (Collado et al., Reference Collado, Isolauri, Laitinen and Salminen2008; Santacruz et al., Reference Santacruz, Collado, García-Valdés, Segura, Martín-Lagos, Anjos, Martí-Romero, Lopez, Florido, Campoy and Sanz2010) as well as Enterobacteriaceae and E. coli (Santacruz et al., Reference Santacruz, Collado, García-Valdés, Segura, Martín-Lagos, Anjos, Martí-Romero, Lopez, Florido, Campoy and Sanz2010).

Gut microbiota modifications during GDM pregnancy

Gut microbiota is involved in human metabolism regulation but can also contribute to the pathogenesis of many diseases, including GDM (Chwalba and Otto-Buczkowska, Reference Chwalba and Otto-Buczkowska2017; Ponzo et al., Reference Ponzo, Fedele, Goitre, Leone, Lezo, Monzeglio, Finocchiaro, Ghigo and Bo2019a,Reference Ponzo, Ferrocino, Zarovska, Amenta, Leone, Monzeglio, Rosato, Pellegrini, Gambino, Cassader, Ghigo, Cocolin and Bob). During gestation, adjustments in the women’s glucose metabolism occur to ensure proper glucose levels to warrant foetal growth and development combined with appropriate maternal nutrition (Angueira et al., Reference Angueira, Ludvik, Reddy, Wicksteed, Lowe and Layden2015). In early gestation, fasting blood glucose (FBG) levels decrease, possibly due to dilution effects (caused by an increased plasma volume), increased glucose uptake by the placenta (Lain and Catalano, Reference Lain and Catalano2007; Angueira et al., Reference Angueira, Ludvik, Reddy, Wicksteed, Lowe and Layden2015), and inadequate hepatic glucose production (Lain and Catalano, Reference Lain and Catalano2007). These levels remain constant in the second trimester and increase during the last trimester (Angueira et al., Reference Angueira, Ludvik, Reddy, Wicksteed, Lowe and Layden2015). In a regular pregnancy’s last trimester, maternal insulin sensitivity declines, which is considered to be advantageous to support foetal development with increased energy requirements at this stage (Koren et al., Reference Koren, Goodrich, Cullender, Spor, Laitinen, Kling Bäckhed, Gonzalez, Werner, Angenent, Knight, Bäckhed, Isolauri, Salminen and Ley2012). In a severe GDM pregnancy, insulin decreases an additional 40% (relatively to a healthy gestation), leading to glucose intolerance (Lain and Catalano, Reference Lain and Catalano2007). To balance these alterations, and due to a decreased capacity of insulin to suppress lipolysis, an increase in free fatty acids, hepatic gluconeogenesis, and severe IR occurs (Taddei et al., Reference Taddei, Cortez, Mattar, Torloni and Daher2018).

In recent years, more information on the correlation between GDM and gut microbiota has become available, demonstrating a distinct microbiota profile associated with this pathology (Kuang et al., Reference Kuang, Lu, Li, Li, Yuan, He, Chen, Xiao, Shen, Qiu, Wu, Hu, Wu, Li, Chen, Deng, Papasian, Xia and Qiu2017; Mokkala et al., Reference Mokkala, Houttu, Vahlberg, Munukka, Rönnemaa and Laitinen2017; Crusell et al., Reference Crusell, Hansen, Nielsen, Allin, Rühlemann, Damm, Vestergaard, Rørbye, Jørgensen, Christiansen, Heinsen, Franke, Hansen, Lauenborg and Pedersen2018; Ferrocino et al., Reference Ferrocino, Ponzo, Gambino, Zarovska, Leone, Monzeglio, Goitre, Rosato, Romano, Grassi, Broglio, Cassader, Cocolin and Bo2018; Hasan et al., Reference Hasan, Aho, Pereira, Paulin, Koivusalo, Auvinen and Eriksson2018; Cortez et al., Reference Cortez, Taddei, Sparvoli, Ângelo, Padilha, Mattar and Daher2019; Liu et al., Reference Liu, Pan, Lv, Yang, Zhang, Chen, Lv and Sun2015; Ye et al., Reference Ye, Zhang, Wang, Chen, Gu, Wang, Leng, Gu and Xie2019; Ma et al., Reference Ma, You, Huang, Long, Zhang, Guo, Zhang, Wu, Xiao and Tan2020; Zheng et al., Reference Zheng, Xu, Huang, Yan, Chen, Zhang, Tian, Liu, Yuan, Liu, Luo, Guo, Song, Zhang, Liang, Qin and Li2020). At a genus level, increased abundance of Klebsiella (Kuang et al., Reference Kuang, Lu, Li, Li, Yuan, He, Chen, Xiao, Shen, Qiu, Wu, Hu, Wu, Li, Chen, Deng, Papasian, Xia and Qiu2017), Collinsella (Collado et al., Reference Collado, Isolauri, Laitinen and Salminen2008; Santacruz et al., Reference Santacruz, Collado, García-Valdés, Segura, Martín-Lagos, Anjos, Martí-Romero, Lopez, Florido, Campoy and Sanz2010; Carreno et al., Reference Carreno, Clifton, Hauth, Myatt, Roberts, Spong, Varner, Thorp, Mercer, Peaceman, Ramin, Carpenter, Sciscione, Tolosa, Saade and Sorokin2012; Koren et al., Reference Koren, Goodrich, Cullender, Spor, Laitinen, Kling Bäckhed, Gonzalez, Werner, Angenent, Knight, Bäckhed, Isolauri, Salminen and Ley2012; Jost et al., Reference Jost, Lacroix, Braegger and Chassard2014; Kim et al., Reference Kim, Sharma, Sappenfield, Wilson and Salihu2014; Priyadarshini et al., Reference Priyadarshini, Thomas, Reisetter, Scholtens, Wolever, Josefson and Layden2014; DiGiulio et al., Reference DiGiulio, Callahan, McMurdie, Costello, Lyell, Robaczewska, Sun, Goltsman, Wong, Shaw, Stevenson, Holmes and Relman2015; Gomez-Arango et al., Reference Gomez-Arango, Barrett, McIntyre, Callaway, Morrison and Dekker Nitert2016; Stanislawski et al., Reference Stanislawski, Dabelea, Wagner, Sontag, Lozupone and Eggesbø2017; Aatsinki et al., Reference Aatsinki, Uusitupa, Munukka, Pesonen, Rintala, Pietilä, Lahti, Eerola, Karlsson and Karlsson2018; Crusell et al., Reference Crusell, Hansen, Nielsen, Allin, Rühlemann, Damm, Vestergaard, Rørbye, Jørgensen, Christiansen, Heinsen, Franke, Hansen, Lauenborg and Pedersen2018; Ferrocino et al., Reference Ferrocino, Ponzo, Gambino, Zarovska, Leone, Monzeglio, Goitre, Rosato, Romano, Grassi, Broglio, Cassader, Cocolin and Bo2018; Meijnikman et al., Reference Meijnikman, Gerdes, Nieuwdorp and Herrema2018; Smid et al., Reference Smid, Ricks, Panzer, Mccoy, Azcarate-Peril, Keku and Boggess2018; Crusell et al., Reference Crusell, Hansen, Nielsen, Allin, Rühlemann, Damm, Vestergaard, Rørbye, Jørgensen, Christiansen, Heinsen, Franke, Hansen, Lauenborg and Pedersen2018), Rothia (Angueira et al., Reference Angueira, Ludvik, Reddy, Wicksteed, Lowe and Layden2015; Chwalba and Otto-Buczkowska, Reference Chwalba and Otto-Buczkowska2017; Mokkala et al., Reference Mokkala, Houttu, Vahlberg, Munukka, Rönnemaa and Laitinen2017; Crusell et al., Reference Crusell, Hansen, Nielsen, Allin, Rühlemann, Damm, Vestergaard, Rørbye, Jørgensen, Christiansen, Heinsen, Franke, Hansen, Lauenborg and Pedersen2018; Taddei et al., Reference Taddei, Cortez, Mattar, Torloni and Daher2018; Liu et al., Reference Liu, Pan, Lv, Yang, Zhang, Chen, Lv and Sun2015; Ponzo et al., Reference Ponzo, Fedele, Goitre, Leone, Lezo, Monzeglio, Finocchiaro, Ghigo and Bo2019a,Reference Ponzo, Ferrocino, Zarovska, Amenta, Leone, Monzeglio, Rosato, Pellegrini, Gambino, Cassader, Ghigo, Cocolin and Bob; Zheng et al., Reference Zheng, Xu, Huang, Yan, Chen, Zhang, Tian, Liu, Yuan, Liu, Luo, Guo, Song, Zhang, Liang, Qin and Li2020), Eubacterium (Kuang et al., Reference Kuang, Lu, Li, Li, Yuan, He, Chen, Xiao, Shen, Qiu, Wu, Hu, Wu, Li, Chen, Deng, Papasian, Xia and Qiu2017), Ruminococcus (Cortez et al., Reference Cortez, Taddei, Sparvoli, Ângelo, Padilha, Mattar and Daher2019), Blautia (Crusell et al., Reference Crusell, Hansen, Nielsen, Allin, Rühlemann, Damm, Vestergaard, Rørbye, Jørgensen, Christiansen, Heinsen, Franke, Hansen, Lauenborg and Pedersen2018; Ye et al., Reference Ye, Zhang, Wang, Chen, Gu, Wang, Leng, Gu and Xie2019; Zheng et al., Reference Zheng, Xu, Huang, Yan, Chen, Zhang, Tian, Liu, Yuan, Liu, Luo, Guo, Song, Zhang, Liang, Qin and Li2020), Prevotella (Cortez et al., Reference Cortez, Taddei, Sparvoli, Ângelo, Padilha, Mattar and Daher2019; Zheng et al., Reference Zheng, Xu, Huang, Yan, Chen, Zhang, Tian, Liu, Yuan, Liu, Luo, Guo, Song, Zhang, Liang, Qin and Li2020), Parabacteroides (Kuang et al., Reference Kuang, Lu, Li, Li, Yuan, He, Chen, Xiao, Shen, Qiu, Wu, Hu, Wu, Li, Chen, Deng, Papasian, Xia and Qiu2017; Dong et al., Reference Dong, Han, Duan, Lin, Li and Liu2020), Eisenbergiella and Tyzzerella (Ma et al., Reference Ma, You, Huang, Long, Zhang, Guo, Zhang, Wu, Xiao and Tan2020), and a reduced richness in Akkermansia (Cortez et al., Reference Cortez, Taddei, Sparvoli, Ângelo, Padilha, Mattar and Daher2019), Marvinbryantia, Acetivibrio, Anaerosporobacter (Jost et al., Reference Jost, Lacroix, Braegger and Chassard2014), and Roseburia (Kuang et al., Reference Kuang, Lu, Li, Li, Yuan, He, Chen, Xiao, Shen, Qiu, Wu, Hu, Wu, Li, Chen, Deng, Papasian, Xia and Qiu2017; Cortez et al., Reference Cortez, Taddei, Sparvoli, Ângelo, Padilha, Mattar and Daher2019) were described in patients with GDM compared with normoglycemic women (Figure 1). Mokkala et al. (Reference Mokkala, Houttu, Vahlberg, Munukka, Rönnemaa and Laitinen2017) reported changes in the gut microbiota before GDM diagnosis, indicating that higher abundance of Ruminococcaceae in the intestine, a family of bacteria important for starch digestion in the large intestine and production of SCFAs (Oriá et al., Reference Oriá, Empadinhas and Malva2020), may be associated with an increased probability to develop GDM (Mokkala et al., Reference Mokkala, Houttu, Vahlberg, Munukka, Rönnemaa and Laitinen2017).

Figure 1. Possible composition of the gut microbiota in pregnant women with GDM according to the available literature.

There is evidence suggesting associations between some bacterial taxa and GDM indicators. Desulfovibrio, reported as a GDM biomarker (Crusell et al., Reference Crusell, Hansen, Nielsen, Allin, Rühlemann, Damm, Vestergaard, Rørbye, Jørgensen, Christiansen, Heinsen, Franke, Hansen, Lauenborg and Pedersen2018), is also a known lipopolysaccharide (LPS) producer, which is one of the strongest inducers of inflammation (Cani et al., Reference Cani, Osto, Geurts and Everard2012), associated with IR (Kim et al., Reference Kim, Keogh and Clifton2018) and leading to intestinal barrier damage (Sanchez-Alcoholado et al., Reference Sanchez-Alcoholado, Castellano-Castillo, Jordán-Martínez, Moreno-Indias, Cardila-Cruz, Elena, Muñoz-Garcia, Queipo-Ortuño and Jimenez-Navarro2017; Zhang et al., Reference Zhang, Han, Chen, Li, Silva-Zolezzi, Parés, Wang and Qin2018a). Collinsella has been related to higher scores in the Homeostasis Model Assessment for Insulin Resistance (HOMA-IR) scale and insulin levels (Ferrocino et al., Reference Ferrocino, Ponzo, Gambino, Zarovska, Leone, Monzeglio, Goitre, Rosato, Romano, Grassi, Broglio, Cassader, Cocolin and Bo2018). Blautia was associated with glucose intolerance (Egshatyan et al., Reference Egshatyan, Kashtanova, Popenko, Tkacheva, Tyakht, Alexeev, Karamnova, Kostryukova, Babenko, Vakhitova and Boytsov2016) and unfavourable metabolic profile in high BMI patients (Crusell et al., Reference Crusell, Hansen, Nielsen, Allin, Rühlemann, Damm, Vestergaard, Rørbye, Jørgensen, Christiansen, Heinsen, Franke, Hansen, Lauenborg and Pedersen2018; Ye et al., Reference Ye, Zhang, Wang, Chen, Gu, Wang, Leng, Gu and Xie2019). Prevotella was positively associated with increased LPS and gut inflammation mediated by pro-inflammatory cytokines (Alves et al., Reference de Brito Alves, de Oliveira, Carvalho, Cavalcante, Pereira Lira, Nascimento, Magnani, Vidal, Braga and de Souza2019).

Although such associations have been established, subjacent mechanisms concerning host and microbiota interactions remain unidentified (Angueira et al., Reference Angueira, Ludvik, Reddy, Wicksteed, Lowe and Layden2015). Also, there are some controversial results since Megasphera and Eggerthella were reported to be enriched in normoglycaemic controls in some studies (Stanislawski et al., Reference Stanislawski, Dabelea, Wagner, Sontag, Lozupone and Eggesbø2017; Wen and Duffy, Reference Wen and Duffy2017) while in others both genera were described to be enriched in the GDM group (Zheng et al., Reference Zheng, Xu, Huang, Yan, Chen, Zhang, Tian, Liu, Yuan, Liu, Luo, Guo, Song, Zhang, Liang, Qin and Li2020). Additionally, at a phylum level, Actinobacteria was found simultaneously to be increased (Crusell et al., Reference Crusell, Hansen, Nielsen, Allin, Rühlemann, Damm, Vestergaard, Rørbye, Jørgensen, Christiansen, Heinsen, Franke, Hansen, Lauenborg and Pedersen2018) and decreased in GDM cohorts (DiGiulio et al., Reference DiGiulio, Callahan, McMurdie, Costello, Lyell, Robaczewska, Sun, Goltsman, Wong, Shaw, Stevenson, Holmes and Relman2015; Cortez et al., Reference Cortez, Taddei, Sparvoli, Ângelo, Padilha, Mattar and Daher2019).

In an attempt to outline available information in a more visual format, Figure 1 presents the possible composition of a pregnant woman with GDM gut microbiota, mainly at the genus level, separating those currently described to be increased from those currently described as being decreased in these patients.

Finally, GDM mother’s offspring gut microbiota have differences in α-richness compared with neonates of mothers without GDM, with a higher abundance of Actinobacteria, associated with higher levels of fasting glucose, and reduction in Bacteroides abundance. At a genus level, an increase of opportunistic pathogens, such as Escherichia and Parabacteroides, and a decrease in probiotic Lactobacillus were observed in GDM mother’s offspring (Su et al., Reference Su, Nie, Shao, Duan, Jiang, Wang, Xing, Sun, Liu and Xu2018; Ponzo et al., Reference Ponzo, Ferrocino, Zarovska, Amenta, Leone, Monzeglio, Rosato, Pellegrini, Gambino, Cassader, Ghigo, Cocolin and Bo2019b). Therefore, the influence of the mother’s gut microbiota composition, blood glucose, BMI, and dietary intake on the gut microbiota of their offspring needs to be further investigated.

Interactions between diet and gut microbiota during pregnancy

The microbial community living in the gut highly depends on the host’s diet, one of the most significant contributors to the modulation of intestinal microbiota and human health (Hasan and Yang, Reference Hasan and Yang2019). Among nutrients, the effects of complex carbohydrates (CHO) are the best studied, having an important impact on microbiota composition (Gentile and Weir, Reference Gentile and Weir2018). It has been reported that diets rich in CHO can modify the gut microbial composition in only a few days or weeks, because in the large intestine there is an intensive fermentative activity of resistant starch and fibres, creating a set of metabolites produced by bacteria – the human metabolome – that can be detected in the host faeces, urine, and blood that can pass the intestinal barrier (Flint et al., Reference Flint, Duncan, Scott and Louis2015). Dietary fibre is the energy source for commensal SCFA-producing bacteria and its fermentation has the potential to decrease postprandial blood glucose, insulin responses and, decrease cholesterol absorption. Dietary patterns marked by low-fibre intake inhibit the growth of SCFA-producing bacteria and enable the development of other bacterial strains that use glycoproteins as energy, leading to harmful effects on the gut barrier (Gomez-Arango et al., Reference Gomez-Arango, Barrett, Wilkinson, Callaway, McIntyre, Morrison and Dekker Nitert2017). A different study also showed that, in early pregnancy, diets associated with higher ingestion of fibres, like vegetarian diets, resulted in a diminished abundance of Collinsella (lactate producing bacteria, instead of SCFA, associated with IR), as well as an increased richness of Roseburia (a butyrate producing bacteria) when compared with the gut microbiota of early pregnant women with omnivore diets (Barret et al., Reference Barrett, Gomez-Arango, Wilkinson, McIntyre, Callaway, Morrison and Dekker Nitert2018). Additionally, Prevotella was found to be enriched in maternal microbiota related to diets with higher CHO intake, whereas Ruminococcus was more enriched in the cohorts ingesting vegetal protein and fat diets (García-Mantrana et al., Reference García-Mantrana, Selma-Royo, González, Parra-Llorca, Martínez-Costa and Collado2020). Furthermore, in overweight and obese pregnancies higher fibre intake increased intestinal microbiota richness, while greater fat intake (saturated fatty acids, monounsaturated fatty acids, and n-6 polyunsaturated fatty acids) decreased its richness (Röytiö et al., Reference Röytiö, Mokkala, Vahlberg and Laitinen2017).

Current literature also reports the association between pro-inflammatory bacteria and dietary patterns. Proteobacteria were associated with a diet abundant in vitamin D, retinol, and mono-unsaturated fat in normal-weight pregnant women, whereas vitamin E was associated with the opposite trend (Mandal et al., Reference Mandal, Godfrey, McDonald, Treuren, Bjørnholt, Midtvedt, Moen, Rudi, Knight, Brantsæter, Peddada and Eggesbø2016). One study implemented routine dietary counselling according to International Federation of Gynecology and Obstetrics (FIGO) guidelines (Hod et al., Reference Hod, Kapur, Sacks, Hadar, Agarwal, di Renzo, Roura, McIntyre, Morris and Divakar2015) to a cohort of 41 overweight pregnant women with GDM, where participants completed a 3-day food record and it was found that only one-third accomplished the recommendations, with two-third consuming a diet rich in fat and low in fibre (Ferrocino et al., Reference Ferrocino, Ponzo, Gambino, Zarovska, Leone, Monzeglio, Goitre, Rosato, Romano, Grassi, Broglio, Cassader, Cocolin and Bo2018). Adherent participants, whose diet was poor in saturated and total fats, showed a decreased abundance of Bacteroides in their gut microbiota composition, which is associated with high-fat animal-based diets (David et al., Reference David, Maurice, Carmody, Gootenberg, Button, Wolfe, Ling, Devlin, Varma, Fischbach, Biddinger, Dutton and Turnbaugh2014). In conclusion, food and its constituents are crucial in regulating host health and disease since they can modulate the environment in which gut microbes live, as well as their diversity and metabolism.

Medical nutrition therapy in GDM

Most GDM women are diagnosed between 24 and 28 weeks of pregnancy, when there is already an increased maternal IR that tends to be higher each week (Farabi and Hernandez, Reference Farabi and Hernandez2019). Medical nutrition therapy (MNT), defined as an individualised nutritional plan developed between the woman and the dietitian (American Diabetes Association Professional Practice Committee, 2022), can be an effective first-line therapy to treat GDM. It should be a food plan that provides adequate calories for both mother and foetus, based on nutrition assessment and the control of the amount and distribution of CHO in the diet, in order to achieve adequate nourishment and normoglycaemia without ketosis and also to improve glycaemic control (Reader, Reference Reader2007; Moreno-Castilla et al., Reference Moreno-Castilla, Hernandez, Bergua, Alvarez, Arce, Rodriguez, Martinez-Alonso, Iglesias, Mateu, Santos, Pacheco, Blasco, Martin, Balsells, Aranda and Mauricio2013). For a successful treatment, individual and personalised dietary counselling and prescription should be assumed by a registered dietitian (or universal equivalent) for all pregnant women diagnosed with GDM (Duarte-Gardea et al., Reference Duarte-Gardea, Gonzales-Pacheco, Reader, Thomas, Wang, Gregory, Piemonte, Thompson and Moloney2018).

The major focus of MNT is to lower postprandial plasma glucose levels, either by adjusting CHO distribution or by altering the glycaemic load (GL; Hernandez et al., Reference Hernandez, Anderson, Chartier-Logan, Friedman and Barbour2013). The first steps of diet manipulation were taken during the 1950s and 60s, focusing on CHO restriction, with approximately 40% of total daily calories (Hernandez, Reference Hernandez2016), considering the principle that it could help lower postprandial glucose and prevent foetal hyperinsulinemia (Mulla, Reference Mulla2016). Currently, it is recommended by the Institute of Medicine (United States of America – USA) a minimum of 175 g CHO/day for pregnant women, equivalent to 35% of a 2000 calories diet, with an extra 45 g compared with non-pregnant women, since an average of 33g glucose/day are required to support foetal brain development and functioning (Trumbo et al., Reference Trumbo, Schlicker, Yates and Poos2002), and also 71 g of protein and 28 g of fibre. Low-CHO diets remain the conventional diet therapy for GDM in some countries. Hence, more studies are needed to construct solid evidence about CHO restriction diets (Trumbo et al., Reference Trumbo, Schlicker, Yates and Poos2002; Moreno-Castilla et al., Reference Moreno-Castilla, Mauricio and Hernandez2016; Mulla, Reference Mulla2016). Moderation seems to be key, since proportions greater than 55% CHO are associated with increased postprandial plasma glucose (Filardi et al., Reference Filardi, Panimolle, Crescioli, Lenzi and Morano2019).

Considering CHO, part is categorised as complex, since their structure is resistant to digestion or even completely undigested, resulting in a slower rise in blood glucose, low-GI (Mustad et al., Reference Mustad, Huynh, López-Pedrosa, Campoy and Rueda2020). Worldwide, 10 clinical practice guidelines (in a total of 16 analysed) on CHO considerations for GDM recommend a low-GI diet (foods under 55 on the GI scale; Tsirou et al., Reference Tsirou, Grammatikopoulou, Theodoridis, Gkiouras, Petalidou, Taousani, Savvaki, Tsapas and Goulis2019), as it can reduce maternal FBG and 2-hour postprandial glucose, compared with high-GI diet. Viana et al. (Reference Viana, Gross and Azevedo2014) systematically reviewed various dietary patterns and concluded that a low-GI diet was the only one confirmed as beneficial for GDM women. Moreover, this diet may also have beneficial effects on the offspring by significantly reducing FBG, postprandial glucose levels, insulin use, and risk of macrosomia (Zhang et al., Reference Zhang, Yu, Xiao, Hu, Xin and Yu2018b; Xu and Ye, Reference Xu and Ye2020). Different types of CHO have diverse impacts on gut microbiota composition. Mardinoglu et al. (Reference Mardinoglu, Wu, Bjornson, Zhang, Hakkarainen, Räsänen, Lee, Mancina, Bergentall, Pietiläinen, Söderlund, Matikainen, Ståhlman, Bergh, Adiels, Piening, Granér, Lundbom, Williams, Romeo, Nielsen, Snyder, Uhlén, Bergström, Perkins, Marschall, Bäckhed, Taskinen and Borén2018) conducted a short-term intervention study on 10 obese individuals with non-alcoholic fat disease, in order to understand the gut microbial composition, by applying a restricted-CHO diet with increased protein for 14 days. The results show a reduction of CHO-degrading bacteria Ruminococcus, Eubacterium, Clostridium, and Bifidobacterium, and levels of SCFA, along with an increased richness in Lactococcus, Eggerthella, and Streptococcus (Mardinoglu et al., Reference Mardinoglu, Wu, Bjornson, Zhang, Hakkarainen, Räsänen, Lee, Mancina, Bergentall, Pietiläinen, Söderlund, Matikainen, Ståhlman, Bergh, Adiels, Piening, Granér, Lundbom, Williams, Romeo, Nielsen, Snyder, Uhlén, Bergström, Perkins, Marschall, Bäckhed, Taskinen and Borén2018). Another study examined the effects of a CHO-restricted diet on the gut bacteria in mice, showing a significant increase in Clostridium (bacteria that promote inflammation) and Suterella (associated with increased LPS and gut inflammation; He et al., Reference He, Wu, Hayashi, Nakano, Nakatsukasa and Tsuduki2020).

Regarding fibre intake, a study about the effects of high fibre with low-GI on gut microbiota in type 2 diabetes patients reported a decreased richness in E. coli and Enterococcus (opportunistic pathogens) and a significant increase in Bifidobacterium and Lactobacillus (beneficial bacteria) compared with the control group (Singh et al., Reference Singh, Chang, Yan, Lee, Ucmak, Wong, Abrouk, Farahnik, Nakamura, Zhu, Bhutani and Liao2017). Bifidobacterium is solidly associated with SCFA production – produced by the fermentation of microbiota-accessible carbohydrates (MACs) – decreased gut LPS levels and improved intestinal mucosal barrier. Lactobacillus is associated with anti-inflammatory and anti-carcinogenic effects, and also SCFA production (Singh et al., Reference Singh, Chang, Yan, Lee, Ucmak, Wong, Abrouk, Farahnik, Nakamura, Zhu, Bhutani and Liao2017). Additionally, the richness of Lactobacillus and Bifidobacterium are associated with the intake of prebiotic fibres (Moszak et al., Reference Moszak, Szulińska and Bogdański2020). These are indigestible fermented fibres that promote bacterial growth in the intestine with health benefits for the host, such as fructooligosaccharides (FOS), galactooligosaccharides (GOS), and inulin (Wilson and Whelan, Reference Wilson and Whelan2017). The majority of GDM guidelines recommend an augmented quantity of fibre, with some indicating approximately 28 g/day of fibres intake (Tsirou et al., Reference Tsirou, Grammatikopoulou, Theodoridis, Gkiouras, Petalidou, Taousani, Savvaki, Tsapas and Goulis2019).

DASH diet is another dietary pattern that has been studied for patients with GDM. Characterised by low-GI, low-energy density with high quantities of fibre and decreased levels of sodium, it was originally designed for patients with hypertension, but favourable effects were reported for metabolic syndrome and type 2 diabetes (Asemi et al., Reference Asemi, Samimi, Tabassi, Sabihi and Esmaillzadeh2013a,Reference Asemi, Tabassi, Samimi, Fahiminejad and Esmaillzadehb). The American Academy of Nutrition and Dietetics (USA) also considers this dietary pattern to be effective in improving both mother and foetal outcomes in GDM, including glucose tolerance, IR, glycosylated haemoglobin, insulin requirements, lipid profile and incidence of macrosomia (Duarte-Gardea et al., Reference Duarte-Gardea, Gonzales-Pacheco, Reader, Thomas, Wang, Gregory, Piemonte, Thompson and Moloney2018), HOMA-IR results and medication needs (Yamamoto et al., Reference Yamamoto, Kellett, Balsells, García-Patterson, Hadar, Solà, Gich, van der Beek, Castañeda-Gutiérrez, Heinonen, Hod, Laitinen, Olsen, Poston, Rueda, Rust, van Lieshout, Schelkle, Murphy and Corcoy2018), systolic blood pressure and lipid profiles – these last two reported on a 4-week DASH diet in patients with GDM (Asemi et al., Reference Asemi, Samimi, Tabassi, Sabihi and Esmaillzadeh2013a,Reference Asemi, Tabassi, Samimi, Fahiminejad and Esmaillzadehb). DASH shares numerous similarities with the Mediterranean diet, except for the intake of olive oil. Fruits, vegetables, and grains are the major food sources in the DASH pattern diet, containing different fibre types (Jama et al., Reference Jama, Beale, Shihata and Marques2019), which may predict its positive role in GDM nutrition, as discussed above. Although there is a lack of studies directly targeting the influence of a DASH diet on GDM women’s gut microbiota, it is already demonstrated that high adherence to DASH promotes the increased richness of SCFA-producing bacteria, such as Roseburia, whereas no adherence leads to higher urinary trimethylamine N-oxide (TMAO) levels, a microbial metabolite possibly associated with cardiovascular and neurological disorders (Filippis et al., Reference de Filippis, Pellegrini, Vannini, Jeffery, la Storia, Laghi, Serrazanetti, di Cagno, Ferrocino, Lazzi, Turroni, Cocolin, Brigidi, Neviani, Gobbetti, O’Toole and Ercolini2016). Further studies are required to better clarify the DASH diet role in gut microbiota modulation for GDM management.

Probiotics in GDM

Even though the adherence of the probiotics to the intestinal mucosal cells is challenging, which influences its effect (O’Sullivan et al., Reference O’Sullivan, Thornton, O’Sullivan and Collins1992; Kullen et al., Reference Kullen, Amann, O’Shaughnessy, O’Sullivan, Busta and Brady1997), regular use of probiotics is reported to beneficially modulate intestinal microbiota composition metabolic activities (Taylor et al., Reference Taylor, Woodfall, Sheedy, O’Riley, Rainbow, Bramwell and Kellow2017). Thus, maternal gut microbiota modulation with probiotic intervention is emerging as a safe approach capable of improving intestinal commensal bacteria and also providing beneficial effects for both mother and foetus health (Swartwout and Luo, Reference Swartwout and Luo2018; Hasain et al., Reference Hasain, Che Roos, Rahmat, Mustapa, Raja Ali and Mokhtar2021). Currently, probiotic agents are mainly produced by gram-positive bacteria such as lactic acid bacteria, namely Bifidobacterium and Lactobacillus. Its use, particularly L. casei, L. helvetica, L. acidophilus, and L. rhamnosus are associated with an improvement of some GDM biomarkers (Pereira et al., Reference de Melo Pereira, de Oliveira Coelho, Magalhães Júnior, Thomaz-Soccol and Soccol2018). Several other promising data suggest that probiotics may positively contribute to beneficial effects on glucose metabolism or even prevent GDM, either by probiotic-fortified foods or capsules (Dolatkhah et al., Reference Dolatkhah, Hajifaraji, Abbasalizadeh, Aghamohammadzadeh, Mehrabi and Mesgari Abbasi2015; Lindsay et al., Reference Lindsay, Brennan, Kennelly, Maguire, Smith, Curran, Coffey, Foley, Hatunic, Shanahan and McAuliffe2015; Ahmadi et al., Reference Ahmadi, Jamilian, Tajabadi-Ebrahimi, Jafari and Asemi2016; Jafarnejad et al., Reference Jafarnejad, Saremi, Jafarnejad and Arab2016; Karamali et al., Reference Karamali, Dadkhah, Sadrkhanlou, Jamilian, Ahmadi, Tajabadi-Ebrahimi, Jafari and Asemi2016; Wickens et al., Reference Wickens, Barthow, Murphy, Abels, Maude, Stone, Mitchell, Stanley, Purdie, Kang, Hood, Rowden, Barnes, Fitzharris and Crane2017; Badehnoosh et al., Reference Badehnoosh, Karamali, Zarrati, Jamilian, Bahmani, Tajabadi-Ebrahimi, Jafari, Rahmani and Asemi2018; Kijmanawat et al., Reference Kijmanawat, Panburana, Reutrakul and Tangshewinsirikul2019).

Seven studies assess the effect of probiotics on metabolic health in pregnant women with GDM (Table 1). Only one randomised control trial (RCT) demonstrated no impact on the metabolic health of GDM pregnant women, with the consumption of one single strain probiotic capsule, L. salivarius, in a dose of 1x10^9 CFU/day during 6 weeks. A double-blind, placebo-controlled study performed with 60 GDM pregnant women demonstrated that probiotic intervention with 3 strains, L. acidophilus, L. casei and B. bifidum, in a dose of 6x10^9 CFU/day during 6 weeks resulted in a significant decrease of FBG, insulin, HOMA-IR and HOMA for β cell function (HOMA-β) levels in the probiotic group compared with the placebo group. There were also positive results in the probiotic group concerning lipid metabolism, with considerable reductions in serum triglycerides and very low density lipoprotein (VLDL) cholesterol. Similar results were reported when administering a symbiotic supplement (probiotics plus inulin) with the same probiotic strains and doses used in Karamali et al. (Reference Karamali, Dadkhah, Sadrkhanlou, Jamilian, Ahmadi, Tajabadi-Ebrahimi, Jafari and Asemi2016) to GDM pregnant women, supplemented with 800 mg of inulin (a prebiotic fibre) during a 6-week-treatment, resulting in decreased insulin levels, and HOMA-IR and HOMA-β results, along with similar lipid outcomes when compared with placebo (Ahmadi et al., Reference Ahmadi, Jamilian, Tajabadi-Ebrahimi, Jafari and Asemi2016). Another RCT involving 70 pregnant women diagnosed with GDM showed that daily consumption of a probiotic capsule containing eight different strains, S. thermophilus, B. breve, B. longum, B. infantis, L. acidophilus, L. plantarum, L. paracasei and L. delbrueckii, in a dose of 15 × 10^9 CFU during 8 weeks, had significant differences in insulin levels and HOMA-IR; however, no changes were observed in FBG and HbA1c in the probiotic group compared with the placebo group (Jafarnejad et al., Reference Jafarnejad, Saremi, Jafarnejad and Arab2016). Data from a double-blind, placebo-controlled, and randomised study revealed that an 8-week treatment of probiotics in a 4 × 10^9 CFU/day dose with four probiotic strains, L. acidophilus, Bifidobacterium sp., S. thermophilus, and L. bulgaricus, showed reductions in FBG, HOMA-IR and GWG (Dolatkhah et al., Reference Dolatkhah, Hajifaraji, Abbasalizadeh, Aghamohammadzadeh, Mehrabi and Mesgari Abbasi2015). Moreover, a 4-week randomised double-blind and placebo-controlled study performed with a two-strain probiotic capsule containing L. acidophilus and B. bifidus in a 2 × 10^9 CFU dose in 57 GDM pregnant women, demonstrated significant improvements in glucose metabolism in the probiotic group compared with the placebo, comprising fasting plasma insulin, FBG, and HOMA-IR (Kijmanawat et al., Reference Kijmanawat, Panburana, Reutrakul and Tangshewinsirikul2019). Two other RCTs conducted with the same number of participants (n = 60, 30 in the probiotic group, 30 in placebo group) and the same three strains of probiotics, L. acidophilus, L. casei and L. bifidum, and doses (6 × 10^9 CFU/day) but with different treatment duration and type of participants, 6 weeks with GDM pregnant women (Badehnoosh et al., Reference Badehnoosh, Karamali, Zarrati, Jamilian, Bahmani, Tajabadi-Ebrahimi, Jafari, Rahmani and Asemi2018) and 12 weeks with healthy pregnant women (Ahmadi et al., Reference Ahmadi, Jamilian, Tajabadi-Ebrahimi, Jafari and Asemi2016), have both exhibited positive results in the probiotic group. In GDM pregnant women probiotic treatment significantly reduced FBG and other inflammatory biomarkers, total glutathione, high sensitivity C-reactive protein, malondialdehyde, and nitric oxide (Badehnoosh et al., Reference Badehnoosh, Karamali, Zarrati, Jamilian, Bahmani, Tajabadi-Ebrahimi, Jafari, Rahmani and Asemi2018), whereas in healthy pregnant women substantially diminished insulin levels, HOMA-IR and HOMA-β (Ahmadi et al., Reference Ahmadi, Jamilian, Tajabadi-Ebrahimi, Jafari and Asemi2016). Hasain et al. (Reference Hasain, Che Roos, Rahmat, Mustapa, Raja Ali and Mokhtar2021) conducted a meta-analysis of studies using multispecies probiotics, including Lactobacillus and Bifidobacterium, similarly to the previous studies presented. Authors reported significant reduction in different glycemic control biomarkers as FPG, fasting serum insulin, and HOMA-IR in women with GDM when taking a probiotic supplementation in a dose between 106 to 109 CFU. However no significant impact was observed when comparing the data for total cholesterol levels, which does not concur with other studies presented in this review. Further studies can help clarify this discrepancy.

Table 1. Studies of probiotics in GDM diagnosed women during gestation.

↔ No significant differences between probiotic and control groups; ↓significantly lower in the probiotic group compared with the control; ↑significantly higher in the probiotic group compared with the control.

HbA1c, glycosylated haemoglobin; HOMA-IR, homeostatic model of assessment of insulin resistance; HOMA-β, homeostatic model assessment for B-cell function; hs-CRP, high-sensitivity C-reactive protein; IL-6, interleukin 6; MDA, malondialdehyde; QUICKI, quantitative insulin sensitivity check index; TAC, total antioxidant capacity; TNF-α, tumour necrosis factor alpha; VLDL, very low density lipoprotein.

Current literature reveals that probiotic therapy may be an important non-pharmacological approach in terms of improving glycaemic control in pregnant women diagnosed with GDM. Probiotic mechanisms of action for treating diabetes mellitus are diverse and depend on different aspects (Khursheed et al., Reference Khursheed, Singh, Wadhwa, Kapoor, Gulati, Kumar, Ramanunny, Awasthi and Dua2019). Anti-diabetic effects emerge when the administration of probiotics is accomplished, homeostasis is recovered, along with diminished LPS levels, supporting the synthesis of different SCFA (butyrate, acetate, and propionate) on the intestine. This leads to an increased release of incretin hormones (like GLP-1), stimulating insulin secretion and delaying gastric emptying, which affects blood glucose levels and reduces intestinal permeability, by improving tight junction proteins, thus diminishing inflammation, oxidative stress, glucose intolerance, and IR (Sanchez-Alcoholado et al., Reference Sanchez-Alcoholado, Castellano-Castillo, Jordán-Martínez, Moreno-Indias, Cardila-Cruz, Elena, Muñoz-Garcia, Queipo-Ortuño and Jimenez-Navarro2017; Khursheed et al., Reference Khursheed, Singh, Wadhwa, Kapoor, Gulati, Kumar, Ramanunny, Awasthi and Dua2019).

However, additional studies are needed to clarify the underlying mechanisms of action through which probiotic therapy improve glycaemic control in GDM pregnant women and determine variables such as what strains, dosage and duration of probiotic treatment confer the highest benefits during gestation. Hsu et al. (Reference Hsu, Lin, Hou and Tain2018) reported the influence of maternal therapy with Lactobacillus casei probiotic and inulin prebiotic for hypertension treatment in rat’s offspring, showing a protective effect when a high fructose diet is administered during pregnancy and lactation. However, the mechanism behind the effect on the offspring is still not clear, as it may be due to a direct consequence of the probiotic passage through the milk or the placenta or the modification of the mother’s metabolism. Other studies evaluated the use of probiotics to manage offspring overweight. The meta-analysis developed by Wang et al. (Reference Wang, Tung, Chang, Lin and Chen2020) reveals the decrease of the newborn birth weight when women with GDM are treated with probiotics. On the other hand, the intake of probiotic by obese pregnant women has the opposite effect, increasing the newborn birth weight.

Discrepancies between studies are also present. Badehnoosh et al. (Reference Badehnoosh, Karamali, Zarrati, Jamilian, Bahmani, Tajabadi-Ebrahimi, Jafari, Rahmani and Asemi2018) and Karamali et al. (Reference Karamali, Nasiri, Taghavi Shavazi, Jamilian, Bahmani, Tajabadi-Ebrahimi and Asemi2018) observed a positive effect of 6 weeks probiotic therapy with L. acidophilus, L. casei, and B. bifidum on offspring birth weight from women with GDM, while Kijmanawat et al. (Reference Kijmanawat, Panburana, Reutrakul and Tangshewinsirikul2019) observed that supplementation with only L. acidophilus and B. bifidum during 4 weeks do not produce any effect on the infant weight. The success of the probiotic intake seems to be dependent on the type of probiotic strains used, being cocktails with higher diversity of microorganism more beneficial. The duration of treatment also appears to influence the effect of the treatment, with Wang et al. (Reference Wang, Tung, Chang, Lin and Chen2020) analysis indicating the need of at least 6 weeks to see effects.

Although proven safe and without adverse effects both for mother and offspring (Didari et al., Reference Didari, Solki, Mozaffari, Nikfar and Abdollahi2014), additional studies need to elucidate the modification not only in the mother’s metabolism and gut microbiome but also in the offspring. All of this should be taken in consideration when recommending probiotic treatment to pregnant women diagnosed with GDM.

Conclusion

Gut microbiota suffers alterations during healthy and pathological gestations like GDM. Modulating its composition through diet and probiotics can be a valid non-pharmacological preventive approach to reduce adverse GDM outcomes in both mother and offspring. Dietary management without probiotics, particularly with a low-GI approach, the currently most recommended diet for patients with GDM, showed benefits in reducing maternal FBG and 2-hour postprandial glucose, apart from being associated with beneficial gut bacteria, such as Bifidobacterium and Lactobacillus. As reviewed, probiotic supplements may ameliorate glycaemic control and inflammatory status of GDM pregnant women, demonstrating the ability to reduce fasting plasma glucose, IR, and improved lipid profiles. However, further high-quality studies are needed to verify the effectiveness of dietary interventions with probiotics as well as the definition of the bacterial strains, doses, and duration of treatment that have the best clinical significance for GDM pregnant women. Achieving this will securely increase the use of probiotic supplementation in GDM diets to better contribute to healthier GDM pregnancies and post-partum outcomes for both mother and offspring.

Author contribution

Conceptualisation and writing – original draft: M.C.C.; Writing – review and editing: S.A. and S.G.P.; Supervision: S.G.P.

Significance statement

With the increase of gestational diabetes mellitus (GDM) and the universal difficulties in the pharmacological management of diseases during pregnancy, the role of diet interventions with probiotics supplementation is incrementally becoming more important. Current literature contributes relevant information on the best approach to manage GDM through medical nutrition therapy, particularly regarding carbohydrates, lipids, and fibre intake, combined with probiotics supplementation. That is the theme that this manuscript reviews highlights, including future approaches to propel this field of work.

Disclosure statement

The authors declare no conflicts of interest.

Funding

This work was supported by the Portuguese Foundation for Science and Technology under the grants UIDB/05704/2020 for the research unit and CEECINST/00051/2018 for Sónia Gonçalves Pereira.

References

Aatsinki, A-K, Uusitupa, H-M, Munukka, E, Pesonen, H, Rintala, A, Pietilä, S, Lahti, L, Eerola, E, Karlsson, L and Karlsson, H (2018) Gut microbiota composition in mid-pregnancy is associated with gestational weight gain but not prepregnancy body mass index. Journal of Women’s Health (2002) 27(10), 12931301. https://doi.org/10.1089/jwh.2017.6488CrossRefGoogle Scholar
Ahmadi, S, Jamilian, M, Tajabadi-Ebrahimi, M, Jafari, P and Asemi, Z (2016) The effects of synbiotic supplementation on markers of insulin metabolism and lipid profiles in gestational diabetes: A randomised, double-blind, placebo-controlled trial. The British Journal of Nutrition 116(8), 13941401. https://doi.org/10.1017/S0007114516003457CrossRefGoogle ScholarPubMed
American Diabetes Association Professional Practice Committee (2022) Management of diabetes in pregnancy: Standards of medical care in diabetes. Diabetes Care 45, S232S243. https://doi.org/10.2337/dc22-S015CrossRefGoogle Scholar
Angueira, AR, Ludvik, AE, Reddy, TE, Wicksteed, B, Lowe, WL Jr and Layden, BT (2015) New insights into gestational glucose metabolism: Lessons learned from 21st century approaches. Diabetes 64(2), 327334. https://doi.org/10.2337/db14-0877CrossRefGoogle ScholarPubMed
Asemi, Z, Samimi, M, Tabassi, Z, Sabihi, SS and Esmaillzadeh, A (2013a) A randomized controlled clinical trial investigating the effect of DASH diet on insulin resistance, inflammation, and oxidative stress in gestational diabetes. Nutrition 29(4), 619624. https://doi.org/10.1016/j.nut.2012.11.020CrossRefGoogle ScholarPubMed
Asemi, Z, Tabassi, Z, Samimi, M, Fahiminejad, T and Esmaillzadeh, A (2013b) Favourable effects of the dietary approaches to stop hypertension diet on glucose tolerance and lipid profiles in gestational diabetes: A randomised clinical trial. The British Journal of Nutrition 109(11), 20242030. https://doi.org/10.1017/S0007114512004242CrossRefGoogle ScholarPubMed
Badehnoosh, B, Karamali, M, Zarrati, M, Jamilian, M, Bahmani, F, Tajabadi-Ebrahimi, M, Jafari, P, Rahmani, E and Asemi, Z (2018) The effects of probiotic supplementation on biomarkers of inflammation, oxidative stress and pregnancy outcomes in gestational diabetes. The Journal of Maternal-Fetal & Neonatal Medicine 31(9), 11281136. https://doi.org/10.1080/14767058.2017.1310193CrossRefGoogle Scholar
Barrett, HL, Gomez-Arango, LF, Wilkinson, SA, McIntyre, H, Callaway, L, Morrison, M and Dekker Nitert, M (2018) A vegetarian diet is a major determinant of gut microbiota composition in early pregnancy. Nutrients 10(7). https://doi.org/10.3390/nu10070890CrossRefGoogle Scholar
Bäumler, AJ and Sperandio, V (2016) Interactions between the microbiota and pathogenic bacteria in the gut. Nature 535(7610), 8593. https://doi.org/10.1038/nature18849CrossRefGoogle ScholarPubMed
Billionnet, C, Mitanchez, D, Weill, A, Nizard, J, Alla, F, Hartemann, A and Jacqueminet, S (2017) Gestational diabetes and adverse perinatal outcomes from 716,152 births in France in 2012. Diabetologia 60(4), 636644. https://doi.org/10.1007/s00125-017-4206-6CrossRefGoogle ScholarPubMed
Brown, J, Grzeskowiak, L, Williamson, K, Downie, MR and Crowther, CA (2017 ) Insulin for the treatment of women with gestational diabetes. Cochrane Database of Systematic Reviews. Nov 5;11(11):CD012037. https://doi.org/10.1002/14651858.cd012037.pub2 PMID: 29103210; PMCID: PMC6486160.Google ScholarPubMed
Canfora, EE, Jocken, JW and Blaak, EE (2015) Short-chain fatty acids in control of body weight and insulin sensitivity. Nature Reviews. Endocrinology 11(10), 577591. https://doi.org/10.1038/nrendo.2015.128CrossRefGoogle ScholarPubMed
Cani, PD (2013) Gut microbiota and obesity: Lessons from the microbiome. Briefings in Functional Genomics. 12(4), 381387. https://doi.org/10.1093/bfgp/elt014CrossRefGoogle ScholarPubMed
Cani, PD, Osto, M, Geurts, L and Everard, A (2012) Involvement of gut microbiota in the development of low-grade inflammation and type 2 diabetes associated with obesity. Gut Microbes 3(4), 279288. https://doi.org/10.4161/gmic.19625CrossRefGoogle ScholarPubMed
Carreno, CA, Clifton, RG, Hauth, JC, Myatt, L, Roberts, JM, Spong, CY, Varner, MW, Thorp, JM Jr, Mercer, BM, Peaceman, AM, Ramin, SM, Carpenter, MW, Sciscione, A, Tolosa, JE, Saade, GR, Sorokin, Y and Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD) Maternal-Fetal Medicine Units (MFMU) Network (2012) Excessive early gestational weight gain and risk of gestational diabetes mellitus in nulliparous women. Obstetrics and Gynecology 119(6), 12271233. https://doi.org/10.1097/AOG.0b013e318256cf1aCrossRefGoogle ScholarPubMed
Casagrande, SS, Linder, B and Cowie, CC (2018) Prevalence of gestational diabetes and subsequent type 2 diabetes among U.S. women. Diabetes Research and Clinical Practice 141, 200208. https://doi.org/10.1016/j.diabres.2018.05.010CrossRefGoogle ScholarPubMed
Chwalba, A and Otto-Buczkowska, E (2017) Participation of the microbiome in the pathogenesis of diabetes mellitus. Clinical Diabetology 6(5), 178181. https://doi.org/10.5603/DK.2017.0029CrossRefGoogle Scholar
Clemente, JC, Ursell, LK, Parfrey, LW and Knight, R (2012) The impact of the gut microbiota on human health: An integrative view. Cell 148(6), 12581270. https://doi.org/10.1016/j.cell.2012.01.035CrossRefGoogle ScholarPubMed
Collado, MC, Isolauri, E, Laitinen, K and Salminen, S (2008) Distinct composition of gut microbiota during pregnancy in overweight and normal-weight women. The American Journal of Clinical Nutrition 88(4), 894899. https://doi.org/10.1093/ajcn/88.4.894CrossRefGoogle ScholarPubMed
Conlon, MA and Bird, AR (2015) The impact of diet and lifestyle on gut microbiota and human health. Nutrients 7(1), 1744. https://doi.org/10.3390/nu7010017CrossRefGoogle Scholar
Cortez, RV, Taddei, CR, Sparvoli, LG, Ângelo, AGS, Padilha, M, Mattar, R and Daher, S (2019) Microbiome and its relation to gestational diabetes. Endocrine 64(2), 254264. https://doi.org/10.1007/s12020-018-1813-zCrossRefGoogle ScholarPubMed
Crusell, MKW, Hansen, TH, Nielsen, T, Allin, KH, Rühlemann, MC, Damm, P, Vestergaard, H, Rørbye, C, Jørgensen, NR, Christiansen, OB, Heinsen, FA, Franke, A, Hansen, T, Lauenborg, J and Pedersen, O (2018) Gestational diabetes is associated with change in the gut microbiota composition in third trimester of pregnancy and postpartum. Microbiome 6(1), 89. https://doi.org/10.1186/s40168-018-0472-xCrossRefGoogle ScholarPubMed
David, LA, Maurice, CF, Carmody, RN, Gootenberg, DB, Button, JE, Wolfe, BE, Ling, AV, Devlin, AS, Varma, Y, Fischbach, MA, Biddinger, SB, Dutton, RJ and Turnbaugh, PJ (2014) Diet rapidly and reproducibly alters the human gut microbiome. Nature 505(7484), 559563. https://doi.org/10.1038/nature12820CrossRefGoogle ScholarPubMed
de Brito Alves, JL, de Oliveira, Y, Carvalho, NNC, Cavalcante, RGS, Pereira Lira, MM, Nascimento, LCP, Magnani, M, Vidal, H, Braga, VA and de Souza, EL (2019) Gut microbiota and probiotic intervention as a promising therapeutic for pregnant women with cardiometabolic disorders: Present and future directions. Pharmacological Research 145(104252), 104252. https://doi.org/10.1016/j.phrs.2019.104252CrossRefGoogle ScholarPubMed
de Filippis, F, Pellegrini, N, Vannini, L, Jeffery, IB, la Storia, A, Laghi, L, Serrazanetti, DI, di Cagno, R, Ferrocino, I, Lazzi, C, Turroni, S, Cocolin, L, Brigidi, P, Neviani, E, Gobbetti, M, O’Toole, PW and Ercolini, D (2016) High-level adherence to a mediterranean diet beneficially impacts the gut microbiota and associated metabolome. Gut 65(11), 18121821. https://doi.org/10.1136/gutjnl-2015-309957CrossRefGoogle ScholarPubMed
de Melo Pereira, GV, de Oliveira Coelho, B, Magalhães Júnior, AI, Thomaz-Soccol, V and Soccol, CR (2018) How to select a probiotic? A review and update of methods and criteria. Biotechnology Advances 36(8), 20602076. https://doi.org/10.1016/j.biotechadv.2018.09.003CrossRefGoogle ScholarPubMed
den Besten, G, van Eunen, K, Groen, AK, Venema, K, Reijngoud, DJ and Bakker, BM (2013) The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. Journal of Lipid Research 54(9), 23252340. https://doi.org/10.1194/jlr.R036012CrossRefGoogle ScholarPubMed
di Simone, N, Santamaria Ortiz, A, Specchia, M, Tersigni, C, Villa, P, Gasbarrini, A, Scambia, G and D’Ippolito, S Recent Insights on the maternal microbiota: Impact on pregnancy outcomes. Frontiers in Immunology 11, 528202. https://doi.org/10.3389/fimmu.2020.528202CrossRefGoogle Scholar
Didari, T, Solki, S, Mozaffari, S, Nikfar, S and Abdollahi, M (2014) A systematic review of the safety of probiotics. Expert Opinion on Drug Safety 13(2), 227239. https://doi.org/10.1517/14740338.2014.872627CrossRefGoogle ScholarPubMed
DiGiulio, DB, Callahan, BJ, McMurdie, PJ, Costello, EK, Lyell, DJ, Robaczewska, A, Sun, CL, Goltsman, DSA, Wong, RJ, Shaw, G, Stevenson, DK, Holmes, SP and Relman, DA (2015) Temporal and spatial variation of the human microbiota during pregnancy. Proceedings of the National Academy of Sciences of the United States of America 112(35), 1106011065. https://doi.org/10.1073/pnas.1502875112CrossRefGoogle ScholarPubMed
Dodd, JM, Grivell, RM, Deussen, AR, Hague, WM and Cochrane Pregnancy and Childbirth Group (2018) Metformin for women who are overweight or obese during pregnancy for improving maternal and infant outcomes. Cochrane Database of Systematic Reviews 7, CD010564. https://doi.org/10.1002/14651858.CD010564.pub2Google ScholarPubMed
Dolatkhah, N, Hajifaraji, M, Abbasalizadeh, F, Aghamohammadzadeh, N, Mehrabi, Y and Mesgari Abbasi, M (2015) Is there a value for probiotic supplements in gestational diabetes mellitus? A randomized clinical trial. Journal of Health, Population, and Nutrition 33(1), 25. https://doi.org/10.1186/s41043-015-0034-9CrossRefGoogle Scholar
Dong, L, Han, L, Duan, T, Lin, S, Li, J and Liu, X (2020) Integrated microbiome–metabolome analysis reveals novel associations between fecal microbiota and hyperglycemia-related changes of plasma metabolome in gestational diabetes mellitus. RSC Advances 10(4), 20272036. https://doi.org/10.1039/C9RA07799ECrossRefGoogle ScholarPubMed
Duarte-Gardea, MO, Gonzales-Pacheco, DM, Reader, DM, Thomas, AM, Wang, SR, Gregory, RP, Piemonte, TA, Thompson, KL and Moloney, L (2018) Academy of nutrition and dietetics gestational diabetes evidence-based nutrition practice guideline. Journal of the Academy of Nutrition and Dietetics 118, 17191742. https://doi.org/10.1016/j.jand.2018.03.014CrossRefGoogle ScholarPubMed
Eades, CE, Cameron, DM and Evans, JMM (2017) Prevalence of gestational diabetes mellitus in Europe: A meta-analysis. Diabetes Research and Clinical Practice 129, 173181. https://doi.org/10.1016/j.diabres.2017.03.030CrossRefGoogle ScholarPubMed
Egshatyan, L, Kashtanova, D, Popenko, A, Tkacheva, O, Tyakht, A, Alexeev, D, Karamnova, N, Kostryukova, E, Babenko, V, Vakhitova, M and Boytsov, S (2016) Gut microbiota and diet in patients with different glucose tolerance. Endocrine Connections 5(1), 19. https://doi.org/10.1530/EC-15-0094CrossRefGoogle ScholarPubMed
Farabi, SS and Hernandez, TL (2019) Low-carbohydrate diets for gestational diabetes. Nutrients 11(8), 1737. 1https://doi.org/0.3390/nu11081737CrossRefGoogle ScholarPubMed
Ferraro, ZM, Barrowman, N, Prud’homme, D, Walker, M, Wen, SW, Rodger, M and Adamo, KB (2012) Excessive gestational weight gain predicts large for gestational age neonates independent of maternal body mass index. The Journal of Maternal-Fetal & Neonatal Medicine 25(5), 538542. https://doi.org/10.3109/14767058.2011.638953CrossRefGoogle ScholarPubMed
Ferrocino, I, Ponzo, V, Gambino, R, Zarovska, A, Leone, F, Monzeglio, C, Goitre, I, Rosato, R, Romano, A, Grassi, G, Broglio, F, Cassader, M, Cocolin, L and Bo, S (2018) Changes in the gut microbiota composition during pregnancy in patients with gestational diabetes mellitus (GDM). Scientific Reports 8(1), 12216. 1https://doi.org/0.1038/s41598-018-30735-9CrossRefGoogle ScholarPubMed
Filardi, T, Panimolle, F, Crescioli, C, Lenzi, A and Morano, S (2019) Gestational diabetes mellitus: The impact of carbohydrate quality in diet. Nutrients 11(7), 1549. https://doi.org/10.3390/nu11071549CrossRefGoogle ScholarPubMed
Flint, HJ, Duncan, SH, Scott, KP and Louis, P (2015) Links between diet, gut microbiota composition and gut metabolism. The Proceedings of the Nutrition Society 74(1), 1322. https://doi.org/10.1017/S0029665114001463CrossRefGoogle ScholarPubMed
Food and Agricultural Organization (FAO) of the United Nations, World Health Organization (WHO) (2001) A joint FAO/WHO expert consultation on the health and nutritional properties of powder milk with live lactic acid bacteria. Available at http://www.fao.org/3/a-a0512e.pdf (accessed 1 June 2023).Google Scholar
García-Mantrana, I, Selma-Royo, M, González, S, Parra-Llorca, A, Martínez-Costa, C and Collado, MC (2020) Distinct maternal microbiota clusters are associated with diet during pregnancy: Impact on neonatal microbiota and infant growth during the first 18 months of life. Gut Microbes 11(4), 962978. https://doi.org/10.1080/19490976.2020.1730294CrossRefGoogle ScholarPubMed
Gensollen, T, Iyer, SS, Kasper, DL and Blumberg, RS (2016) How colonization by microbiota in early life shapes the immune system. Science 352(6285), 539544. https://doi.org/10.1126/science.aad9378CrossRefGoogle ScholarPubMed
Gentile, CL and Weir, TL (2018) The gut microbiota at the intersection of diet and human health. Science 362(6416), 776780. https://doi.org/10.1126/science.aau5812CrossRefGoogle ScholarPubMed
Gilmore, LA, Klempel-Donchenko, M and Redman, LM (2015) Pregnancy as a window to future health: Excessive gestational weight gain and obesity. Seminars in Perinatology 39(4), 296303. https://doi.org/10.1053/j.semperi.2015.05.009CrossRefGoogle ScholarPubMed
Gomez-Arango, LF, Barrett, HL, McIntyre, HD, Callaway, LK, Morrison, M, Dekker Nitert, M and SPRING Trial Group (2016) Connections between the gut microbiome and metabolic hormones in early pregnancy in overweight and obese women. Diabetes 65(8), 22142223. https://doi.org/10.2337/db16-0278CrossRefGoogle ScholarPubMed
Gomez-Arango, LF, Barrett, HL, Wilkinson, SA, Callaway, LK, McIntyre, HD, Morrison, M and Dekker Nitert, M (2018) Low dietary fiber intake increases Collinsella abundance in the gut microbiota of overweight and obese pregnant women. Gut Microbes 9, 189201. https://doi.org/10.1080/19490976.2017.1406584CrossRefGoogle ScholarPubMed
HAPO Study Cooperative Research Group, Metzger, BE, Lowe, LP, Dyer, AR, Trimble, ER, Chaovarindr, U, Coustan, DR, Hadden, DR, McCance, DR, Hod, M, McIntyre, HD, Oats, JJ, Persson, B, Rogers, MS and Sacks, DA (2008) Hyperglycemia and adverse pregnancy outcomes. The New England Journal of Medicine 358(19), 19912002. https://doi.org/10.1056/NEJMoa0707943CrossRefGoogle Scholar
Hasain, Z, Che Roos, N, Rahmat, F, Mustapa, M, Raja Ali, R and Mokhtar, N (2021) Diet and pre-intervention washout modifies the effects of probiotics on gestational diabetes mellitus: A comprehensive systematic review and meta-analysis of randomized controlled trials. Nutrients 13(9), 3045. https://doi.org/10.3390/nu13093045CrossRefGoogle ScholarPubMed
Hasain, Z, Mokhtar, NM, Kamaruddin, NA, Mohamed Ismail, NA, Razalli, NH, Gnanou, JV and Raja Ali, RA (2020) Gut microbiota and gestational diabetes mellitus: A review of host-gut microbiota interactions and their therapeutic potential. Frontiers in Cellular and Infection Microbiology 10, 188. https://doi.org/10.3389/fcimb.2020.00188CrossRefGoogle ScholarPubMed
Hasan, S, Aho, V, Pereira, P, Paulin, L, Koivusalo, SB, Auvinen, P and Eriksson, JG (2018) Gut microbiome in gestational diabetes: A cross-sectional study of mothers and offspring 5 years postpartum. Acta Obstetricia et Gynecologica Scandinavica 97(1), 3846. https://doi.org/10.1111/aogs.13252CrossRefGoogle ScholarPubMed
Hasan, N and Yang, H (2019) Factors affecting the composition of the gut microbiota, and its modulation. PeerJ 7, e7502. https://doi.org/10.7717/peerj.7502CrossRefGoogle ScholarPubMed
He, C, Wu, Q, Hayashi, N, Nakano, F, Nakatsukasa, E and Tsuduki, T (2020) Carbohydrate-restricted diet alters the gut microbiota, promotes senescence and shortens the life span in senescence-accelerated prone mice. The Journal of Nutritional Biochemistry 78(108326), 108326. https://doi.org/10.1016/j.jnutbio.2019.108326CrossRefGoogle ScholarPubMed
Hernandez, TL (2016) Carbohydrate content in the GDM diet: Two views: View 1: Nutrition therapy in gestational diabetes: The case for complex carbohydrates. Diabetes Spectrum: A Publication of the American Diabetes Association 29(2), 8288. https://doi.org/10.2337/diaspect.29.2.82CrossRefGoogle ScholarPubMed
Hernandez, Tl, Anderson, Ma, Chartier-Logan, C, Friedman, JE and Barbour, LA (2013) Strategies in the nutritional management of gestational diabetes. Clinical Obstetrics and Gynecology 56(4), 803815. https://doi.org/10.1097/GRF.0b013e3182a8e0e5CrossRefGoogle ScholarPubMed
Hod, M, Kapur, A, Sacks, DA, Hadar, E, Agarwal, M, di Renzo, GC, Roura, LC, McIntyre, HD, Morris, JL and Divakar, H (2015) The International Federation of Gynecology and Obstetrics (FIGO) initiative on gestational diabetes mellitus: A pragmatic guide for diagnosis, management, and care. International Journal of Gynaecology and Obstetrics 131(Suppl 3), S173S211. https://doi.org/10.1016/S0020-7292(15)30033-3CrossRefGoogle ScholarPubMed
Hsu, CN, Lin, YJ, Hou, CY and Tain, YL (2018) Maternal administration of probiotic or prebiotic prevents male adult rat offspring against developmental programming of hypertension induced by high fructose consumption in pregnancy and lactation. Nutrients 10(9), 1229. https://doi.org/10.3390/nu10091229CrossRefGoogle ScholarPubMed
Jafarnejad, S, Saremi, S, Jafarnejad, F and Arab, A (2016) Effects of a multispecies probiotic mixture on glycemic control and inflammatory status in women with gestational diabetes: A randomized controlled clinical trial. Journal of Nutrition and Metabolism 2016, 5190846. https://doi.org/10.1155/2016/5190846CrossRefGoogle ScholarPubMed
Jama, HA, Beale, A, Shihata, WA and Marques, FZ (2019) The effect of diet on hypertensive pathology: Is there a link via gut microbiota-driven immunometabolism? Cardiovascular Research 115(9), 14351447. https://doi.org/10.1093/cvr/cvz091CrossRefGoogle Scholar
Jost, T, Lacroix, C, Braegger, C and Chassard, C (2014) Stability of the maternal gut microbiota during late pregnancy and early lactation. Current Microbiology 68(4), 419427. https://doi.org/10.1007/s00284-013-0491-6CrossRefGoogle ScholarPubMed
Kamada, N, Seo, S-U, Chen, GY and Núñez, G (2013) Role of the gut microbiota in immunity and inflammatory disease. Nature Reviews. Immunology 13(5), 321335. https://doi.org/10.1038/nri3430CrossRefGoogle ScholarPubMed
Kampmann, U, Knorr, S, Fuglsang, J and Ovesen, P (2019) Determinants of maternal insulin resistance during pregnancy: An updated overview. Journal of Diabetes Research 2019, 5320156. https://doi.org/10.1155/2019/5320156CrossRefGoogle ScholarPubMed
Karamali, M, Dadkhah, F, Sadrkhanlou, M, Jamilian, M, Ahmadi, S, Tajabadi-Ebrahimi, M, Jafari, P and Asemi, Z (2016) Effects of probiotic supplementation on glycaemic control and lipid profiles in gestational diabetes: A randomized, double-blind, placebo-controlled trial. Diabetes & Metabolism 42(4), 234241. https://doi.org/10.1016/j.diabet.2016.04.009CrossRefGoogle ScholarPubMed
Karamali, M, Nasiri, N, Taghavi Shavazi, N, Jamilian, M, Bahmani, F, Tajabadi-Ebrahimi, M and Asemi, Z (2018) The effects of synbiotic supplementation on pregnancy outcomes in gestational diabetes. Probiotics and Antimicrobial Proteins 10(3), 496503. https://doi.org/10.1007/s12602-017-9313-7/tables/4CrossRefGoogle ScholarPubMed
Kc, K, Shakya, S and Zhang, H (2015) Gestational diabetes mellitus and macrosomia: A literature review. Annals of Nutrition & Metabolism 66(Suppl 2), 1420. https://doi.org/10.1159/000371628CrossRefGoogle ScholarPubMed
Khursheed, R, Singh, SK, Wadhwa, S, Kapoor, B, Gulati, M, Kumar, R, Ramanunny, AK, Awasthi, A and Dua, K (2019) Treatment strategies against diabetes: Success so far and challenges ahead. European Journal of Pharmacology 862(172625), 172625. https://doi.org/10.1016/j.ejphar.2019.172625CrossRefGoogle ScholarPubMed
Kijmanawat, A, Panburana, P, Reutrakul, S and Tangshewinsirikul, C (2019) Effects of probiotic supplements on insulin resistance in gestational diabetes mellitus: A double-blind randomized controlled trial. Journal of Diabetes Investigation 10(1), 163170. https://doi.org/10.1111/jdi.12863CrossRefGoogle ScholarPubMed
Kim, YA, Keogh, JB and Clifton, PM (2018) Probiotics, prebiotics, synbiotics and insulin sensitivity. Nutrition Research Reviews 31(1), 3551. https://doi.org/ 10.1017/S095442241700018XCrossRefGoogle ScholarPubMed
Kim, SY, Sharma, AJ, Sappenfield, W, Wilson, HG and Salihu, HM (2014) Association of maternal body mass index, excessive weight gain, and gestational diabetes mellitus with large-for-gestational-age births. Obstetrics and Gynecology 123(4), 737744. https://doi.org/10.1097/AOG.0000000000000177CrossRefGoogle ScholarPubMed
Koh, A, de Vadder, F, Kovatcheva-Datchary, P and Bäckhed, F (2016) From dietary fiber to host physiology: Short-chain fatty acids as key bacterial metabolites. Cell 165(6), 13321345. https://doi.org/10.1016/j.cell.2016.05.041CrossRefGoogle ScholarPubMed
Koivusalo, SB, Rönö, K, Klemetti, MM, Roine, RP, Lindström, J, Erkkola, M, Kaaja, RJ, Pöyhönen-Alho, M, Tiitinen, A, Huvinen, E, Andersson, S, Laivuori, H, Valkama, A, Meinilä, J, Kautiainen, H, Eriksson, JG and Stach-Lempinen, B (2016) Gestational diabetes mellitus can be prevented by lifestyle intervention: The Finnish gestational diabetes prevention study (RADIEL): A randomized controlled trial. Diabetes Care 39(1), 2430. https://doi.org/10.2337/dc15-0511CrossRefGoogle ScholarPubMed
Koren, O, Goodrich, JK, Cullender, TC, Spor, A, Laitinen, K, Kling Bäckhed, H, Gonzalez, A, Werner, JJ, Angenent, LT, Knight, R, Bäckhed, F, Isolauri, E, Salminen, S and Ley, RE (2012) Host remodeling of the gut microbiome and metabolic changes during pregnancy. Cell 150(3), 470480. https://doi.org/10.1016/j.cell.2012.07.008CrossRefGoogle ScholarPubMed
Kuang, Y-S, Lu, J-H, Li, S-H, Li, JH, Yuan, MY, He, JR, Chen, NN, Xiao, WQ, Shen, SY, Qiu, L, Wu, YF, Hu, CY, Wu, YY, Li, WD, Chen, QZ, Deng, HW, Papasian, CJ, Xia, HM and Qiu, X (2017) Connections between the human gut microbiome and gestational diabetes mellitus. Gigascience 6(8), 112. https://doi.org/10.1093/gigascience/gix058CrossRefGoogle ScholarPubMed
Kullen, MJ, Amann, MM, O’Shaughnessy, MJ, O’Sullivan, DJ, Busta, FF and Brady, LJ (1997) Differentiation of ingested and endogenous Bifidobacteria by DNA fingerprinting demonstrates the survival of an unmodified strain in the gastrointestinal tract of humans. The Journal of Nutrition 127(1), 8994. https://doi.org/10.1093/jn/127.1.89CrossRefGoogle ScholarPubMed
Lain, KY and Catalano, PM (2007) Metabolic changes in pregnancy. Clinical Obstetrics and Gynecology 50(4), 938948. https://doi.org/10.1097/GRF.0b013e31815a5494CrossRefGoogle ScholarPubMed
LeBlanc, JG, Chain, F, Martín, R, Bermúdez-Humarán, LG, Courau, S and Langella, P (2017) Beneficial effects on host energy metabolism of short-chain fatty acids and vitamins produced by commensal and probiotic bacteria. Microbial Cell Factories 16(1), 79. https://doi.org/10.1186/s12934-017-0691-zCrossRefGoogle ScholarPubMed
Lefkovits, YR, Stewart, ZA and Murphy, HR (2019) Gestational diabetes. Medicine (Abingdon). 47(2), 114118. https://doi.org/10.1016/j.mpmed.2018.11.006Google Scholar
Lindsay, KL, Brennan, L, Kennelly, MA, Maguire, OC, Smith, T, Curran, S, Coffey, M, Foley, ME, Hatunic, M, Shanahan, F and McAuliffe, FM ( 2015) Impact of probiotics in women with gestational diabetes mellitus on metabolic health: A randomized controlled trial. American Journal of Obstetrics and Gynecology 212, 496.e1–e11. https://doi.org/10.1016/j.ajog.2015.02.008CrossRefGoogle ScholarPubMed
Liu, H, Pan, L-L, Lv, S, Yang, Q, Zhang, H, Chen, W, Lv, Z and Sun, J (2015) Alterations of gut microbiota and blood lipidome in gestational diabetes mellitus with hyperlipidemia. Frontiers in Physiology 10, 1015. https://doi.org/10.3389/fphys.2019.01015CrossRefGoogle Scholar
Lozupone, CA, Stombaugh, JI, Gordon, JI, Jansson, JK and Knight, R (2012) Diversity, stability and resilience of the human gut microbiota. Nature 489(7415), 220230. https://doi.org/10.1038/nature11550CrossRefGoogle ScholarPubMed
Lynch, SV and Pedersen, O (2016) The human intestinal microbiome in health and disease. The New England Journal of Medicine 375(24), 23692379. https://doi.org/10.1056/NEJMra1600266CrossRefGoogle ScholarPubMed
Ma, S, You, Y, Huang, L, Long, S, Zhang, J, Guo, C, Zhang, N, Wu, X, Xiao, Y and Tan, H (2020) Alterations in gut microbiota of gestational diabetes patients during the first trimester of pregnancy. Frontiers in Cellular and Infection Microbiology 10, 58. https://doi.org/10.3389/fcimb.2020.00058CrossRefGoogle ScholarPubMed
Mandal, S, Godfrey, KM, McDonald, D, Treuren, WV, Bjørnholt, JV, Midtvedt, T, Moen, B, Rudi, K, Knight, R, Brantsæter, AL, Peddada, SD and Eggesbø, M (2016) Fat and vitamin intakes during pregnancy have stronger relations with a pro-inflammatory maternal microbiota than does carbohydrate intake. Microbiome 4(1), 55. https://doi.org/10.1186/s40168-016-0200-3CrossRefGoogle ScholarPubMed
Mardinoglu, A, Wu, H, Bjornson, E, Zhang, C, Hakkarainen, A, Räsänen, SM, Lee, S, Mancina, RM, Bergentall, M, Pietiläinen, KH, Söderlund, S, Matikainen, N, Ståhlman, M, Bergh, PO, Adiels, M, Piening, BD, Granér, M, Lundbom, N, Williams, KJ, Romeo, S, Nielsen, J, Snyder, M, Uhlén, M, Bergström, G, Perkins, R, Marschall, HU, Bäckhed, F, Taskinen, MR and Borén, J (2018) An integrated understanding of the rapid metabolic benefits of a carbohydrate-restricted diet on hepatic steatosis in humans. Cell Metabolism 27(3), 559571.e5. https://doi.org/10.1016/j.cmet.2018.01.005CrossRefGoogle ScholarPubMed
Meijnikman, AS, Gerdes, VE, Nieuwdorp, M and Herrema, H (2018) Evaluating causality of gut microbiota in obesity and diabetes in humans. Endocrine Reviews 39(2), 133153. https://doi.org/10.1210/er.2017-00192CrossRefGoogle ScholarPubMed
Mokkala, K, Houttu, N, Vahlberg, T, Munukka, E, Rönnemaa, T and Laitinen, K (2017) Gut microbiota aberrations precede diagnosis of gestational diabetes mellitus. Acta Diabetologica 54(12), 11471149. https://doi.org/10.1007/s00592-017-1056-0CrossRefGoogle ScholarPubMed
Moreno-Castilla, C, Hernandez, M, Bergua, M, Alvarez, MC, Arce, MA, Rodriguez, K, Martinez-Alonso, M, Iglesias, M, Mateu, M, Santos, MD, Pacheco, LR, Blasco, Y, Martin, E, Balsells, N, Aranda, N and Mauricio, D (2013) Low-carbohydrate diet for the treatment of gestational diabetes mellitus: A randomized controlled trial. Diabetes Care 36(8), 22332238. https://doi.org/10.2337/dc12-2714CrossRefGoogle ScholarPubMed
Moreno-Castilla, C, Mauricio, D and Hernandez, M (2016) Role of medical nutrition therapy in the management of gestational diabetes mellitus. Current Diabetes Reports 16(4), 22. https://doi.org/10.1007/s11892-016-0717-7CrossRefGoogle ScholarPubMed
Moszak, M, Szulińska, M and Bogdański, P (2020) You are what you eat-the relationship between diet, microbiota, and metabolic disorders - a review. Nutrients 12(4), 1096. https://doi.org/10.3390/nu12041096CrossRefGoogle ScholarPubMed
Mulla, WR (2016) Carbohydrate content in the GDM diet: Two views: View 2: Low-carbohydrate diets should remain the initial therapy for gestational diabetes. Diabetes Spectrum: A Publication of the American Diabetes Association 29(2), 8991. https://doi.org/10.2337/diaspect.29.2.89CrossRefGoogle ScholarPubMed
Mustad, VA, Huynh, DTT, López-Pedrosa, JM, Campoy, C and Rueda, R (2020) The role of dietary carbohydrates in gestational diabetes. Nutrients 12(2), 385. https://doi.org/10.3390/nu12020385CrossRefGoogle ScholarPubMed
Natividad, JMM and Verdu, EF (2013) Modulation of intestinal barrier by intestinal microbiota: Pathological and therapeutic implications. Pharmacological Research 69(1), 4251. https://doi.org/10.1016/j.phrs.2012.10.007CrossRefGoogle ScholarPubMed
Nicholson, JK, Holmes, E, Kinross, J, Burcelin, R, Gibson, G, Jia, W and Pettersson, S (2012) Host-gut microbiota metabolic interactions. Science 336(6086), 12621267. https://doi.org/10.1126/science.1223813CrossRefGoogle ScholarPubMed
Oriá, RB, Empadinhas, N and Malva, JO (2020) Editorial: Interplay between nutrition, the intestinal microbiota and the immune system. Frontiers in Immunology 11, 1758. https://doi.org/0.3389/fimmu.2020.01758CrossRefGoogle ScholarPubMed
O’Sullivan, MG, Thornton, G, O’Sullivan, GC and Collins, JK (1992) Probiotic bacteria: Myth or reality? Trends in Food Science & Technology 3, 309314. https://doi.org/10.1016/s0924-2244(10)80018-4CrossRefGoogle Scholar
Pascale, A, Marchesi, N, Marelli, C, Coppola, A, Luzi, L, Govoni, S, Giustina, A and Gazzaruso, C (2018) Microbiota and metabolic diseases. Endocrine 61(3), 357371. https://doi.org/10.1007/s12020-018-1605-5CrossRefGoogle ScholarPubMed
Ponzo, V, Fedele, D, Goitre, I, Leone, F, Lezo, A, Monzeglio, C, Finocchiaro, C, Ghigo, E and Bo, S (2019a) Diet-gut microbiota interactions and gestational diabetes mellitus (GDM). Nutrients 11(2), 330. https://doi.org/10.3390/nu11020330CrossRefGoogle ScholarPubMed
Ponzo, V, Ferrocino, I, Zarovska, A, Amenta, MB, Leone, F, Monzeglio, C, Rosato, R, Pellegrini, M, Gambino, R, Cassader, M, Ghigo, E, Cocolin, L and Bo, S (2019b) The microbiota composition of the offspring of patients with gestational diabetes mellitus (GDM). PLoS One 14(12), e0226545. https://doi.org/10.1371/journal.pone.0226545CrossRefGoogle ScholarPubMed
Priyadarshini, M, Thomas, A, Reisetter, AC, Scholtens, DM, Wolever, TMS, Josefson, JL and Layden, BT (2014) Maternal short-chain fatty acids are associated with metabolic parameters in mothers and newborns. Translational Research 164(2), 153157. https://doi.org/10.1016/j.trsl.2014.01.012CrossRefGoogle ScholarPubMed
Reader, DM (2007) Medical nutrition therapy and lifestyle interventions. Diabetes Care 30, S188S193. https://doi.org/10.2337/dc07-s214CrossRefGoogle ScholarPubMed
Röytiö, H, Mokkala, K, Vahlberg, T and Laitinen, K (2017) Dietary intake of fat and fibre according to reference values relates to higher gut microbiota richness in overweight pregnant women. The British Journal of Nutrition 118(5), 343352. https://doi.org/10.1017/S0007114517002100CrossRefGoogle ScholarPubMed
Sanchez-Alcoholado, L, Castellano-Castillo, D, Jordán-Martínez, L, Moreno-Indias, I, Cardila-Cruz, P, Elena, D, Muñoz-Garcia, AJ, Queipo-Ortuño, MI and Jimenez-Navarro, M (2017) Role of gut microbiota on cardio-metabolic parameters and immunity in coronary artery disease patients with and without type-2 diabetes mellitus. Frontiers in Microbiology 8, 1936. https://doi.org/10.3389/fmicb.2017.01936CrossRefGoogle ScholarPubMed
Santacruz, A, Collado, MC, García-Valdés, L, Segura, MT, Martín-Lagos, JA, Anjos, T, Martí-Romero, M, Lopez, RM, Florido, J, Campoy, C and Sanz, Y (2010) Gut microbiota composition is associated with body weight, weight gain and biochemical parameters in pregnant women. The British Journal of Nutrition 104(1), 8392. https://doi.org/10.1017/S0007114510000176CrossRefGoogle ScholarPubMed
Schneider, S, Hoeft, B, Freerksen, N, fischer, B, Roehrig, S, Yamamoto, S and Maul, H (2011) Neonatal complications and risk factors among women with gestational diabetes mellitus: Gestational diabetes mellitus. Acta Obstetricia et Gynecologica Scandinavica 90(3), 231237. https://doi.org/10.1111/j.1600-0412.2010.01040.xGoogle ScholarPubMed
Shen, J, Obin, MS and Zhao, L (2013) The gut microbiota, obesity and insulin resistance. Molecular Aspects of Medicine 34(1), 3958. https://doi.org/10.1016/j.mam.2012.11.001CrossRefGoogle ScholarPubMed
Simmons, D (2015) Prevention of gestational diabetes mellitus: Where are we now? Diabetes, Obesity & Metabolism 17(9), 824834. https://doi.org/10.1111/dom.12495CrossRefGoogle ScholarPubMed
Singh, RK, Chang, H-W, Yan, D, Lee, KM, Ucmak, D, Wong, K, Abrouk, M, Farahnik, B, Nakamura, M, Zhu, TH, Bhutani, T and Liao, W (2017) Influence of diet on the gut microbiome and implications for human health. Journal of Translational Medicine 15(1), 73. https://doi.org/10.1186/s12967-017-1175-yCrossRefGoogle ScholarPubMed
Smid, MC, Ricks, NM, Panzer, A, Mccoy, AN, Azcarate-Peril, MA, Keku, TO and Boggess, KA (2018) Maternal gut microbiome biodiversity in pregnancy. American Journal of Perinatology 35(1), 2430. https://doi.org/10.1055/s-0037-1604412Google ScholarPubMed
Stanislawski, MA, Dabelea, D, Wagner, BD, Sontag, MK, Lozupone, CA and Eggesbø, M (2017) Pre-pregnancy weight, gestational weight gain, and the gut microbiota of mothers and their infants. Microbiome 5(1), 113. https://doi.org/10.1186/s40168-017-0332-0CrossRefGoogle ScholarPubMed
Su, M, Nie, Y, Shao, R, Duan, S, Jiang, Y, Wang, M, Xing, Z, Sun, Q, Liu, X and Xu, W (2018) Diversified gut microbiota in newborns of mothers with gestational diabetes mellitus. PLoS One 13(10), e0205695. https://doi.org/10.1371/journal.pone.0205695CrossRefGoogle ScholarPubMed
Swartwout, B and Luo, XM (2018) Implications of probiotics on the maternal-neonatal interface: Gut microbiota, immunomodulation, and autoimmunity. Frontiers in Immunology 9, 2840. https://doi.org/10.3389/fimmu.2018.02840CrossRefGoogle ScholarPubMed
Taddei, CR, Cortez, RV, Mattar, R, Torloni, MR and Daher, S (2018) Microbiome in normal and pathological pregnancies: A literature overview. American Journal of Reproductive Immunology 80, e12993. https://doi.org/10.1111/aji.12830CrossRefGoogle ScholarPubMed
Taylor, B, Woodfall, G, Sheedy, K, O’Riley, M, Rainbow, K, Bramwell, E and Kellow, N (2017) Effect of probiotics on metabolic outcomes in pregnant women with gestational diabetes: A systematic review and meta-analysis of randomized controlled trials. Nutrients 9(5), 461. https://doi.org/10.3390/nu9050461CrossRefGoogle ScholarPubMed
Topping, DL and Clifton, PM (2001) Short-chain fatty acids and human colonic function: Roles of resistant starch and nonstarch polysaccharides. Physiological Reviews 81(3), 10311064. 1https://doi.org/0.1152/physrev.2001.81.3.1031CrossRefGoogle ScholarPubMed
Tremaroli, V and Bäckhed, F (2012) Functional interactions between the gut microbiota and host metabolism. Nature 489(7415), 242249. https://doi.org/10.1038/nature11552CrossRefGoogle ScholarPubMed
Trumbo, P, Schlicker, S, Yates, AA, Poos, M and Food and Nutrition Board of the Institute of Medicine, The National Academies (2002) Dietary reference intakes for energy, carbohydrate, fiber, fat, fatty acids, cholesterol, protein and amino acids. Journal of the American Dietetic Association 102(11), 16211630. https://doi.org/10.1016/s0002-8223(02)90346-9CrossRefGoogle ScholarPubMed
Tsirou, E, Grammatikopoulou, MG, Theodoridis, X, Gkiouras, K, Petalidou, A, Taousani, E, Savvaki, D, Tsapas, A and Goulis, DG (2019) Guidelines for medical nutrition therapy in gestational diabetes mellitus: Systematic review and critical appraisal. Journal of the Academy of Nutrition and Dietetics 119(8), 13201339. https://doi.org/10.1016/j.jand.2019.04.002CrossRefGoogle ScholarPubMed
Viana, LV, Gross, JL and Azevedo, MJ (2014) Dietary intervention in patients with gestational diabetes mellitus: A systematic review and meta-analysis of randomized clinical trials on maternal and newborn outcomes. Diabetes Care 37(12), 33453355. https://doi.org/10.2337/dc14-1530CrossRefGoogle ScholarPubMed
Wang, C, Tung, YT, Chang, HC, Lin, CH and Chen, YC (2020) Effect of probiotic supplementation on newborn birth weight for mother with gestational diabetes mellitus or overweight/obesity: A systematic review and meta-analysis. Nutrients, 12(11), 3477. https://doi.org/10.3390/nu12113477CrossRefGoogle ScholarPubMed
Wang, C, Wei, Y, Zhang, X, Zhang, Y, Xu, Q, Sun, Y, Su, S, Zhang, L, Liu, C, Feng, Y, Shou, C, Guelfi, KJ, Newnham, JP and Yang, H (2017) A randomized clinical trial of exercise during pregnancy to prevent gestational diabetes mellitus and improve pregnancy outcome in overweight and obese pregnant women. American Journal of Obstetrics and Gynecology 216(4), 340351. https://doi.org/10.1016/j.ajog.2017.01.037CrossRefGoogle ScholarPubMed
Wen, L and Duffy, A (2017) Factors influencing the gut microbiota, inflammation, and type 2 diabetes. The Journal of Nutrition 147(7), 1468S1475S. https://doi.org/10.3945/jn.116.240754CrossRefGoogle ScholarPubMed
Wendland, EM, Torloni, MR, Falavigna, M, Trujillo, J, Dode, MA, Campos, MA, Duncan, BB and Schmidt, MI (2012) Gestational diabetes and pregnancy outcomes-a systematic review of the World Health Organization (WHO) and the International Association of Diabetes in pregnancy study groups (IADPSG) diagnostic criteria. BMC Pregnancy and Childbirth 12(1), 23. https://doi.org/10.1186/1471-2393-12-23CrossRefGoogle ScholarPubMed
Wickens, KL, Barthow, CA, Murphy, R, Abels, PR, Maude, RM, Stone, PR, Mitchell, EA, Stanley, TV, Purdie, GL, Kang, JM, Hood, FE, Rowden, JL, Barnes, PK, Fitzharris, PF and Crane, J (2017) Early pregnancy probiotic supplementation with lactobacillus rhamnosus HN001 may reduce the prevalence of gestational diabetes mellitus: A randomised controlled trial. The British Journal of Nutrition 117(6), 804813. https://doi.org/10.1017/S0007114517000289CrossRefGoogle ScholarPubMed
Wilson, B and Whelan, K (2017) Prebiotic inulin-type fructans and galacto-oligosaccharides: Definition, specificity, function, and application in gastrointestinal disorders: Prebiotic fructans and GOS. Journal of Gastroenterology and Hepatology 32(Suppl 1), 6468. https://doi.org/10.1111/jgh.13700CrossRefGoogle Scholar
Xu, J and Ye, S (2020) Influence of low-glycemic index diet for gestational diabetes: A meta-analysis of randomized controlled trials. The Journal of Maternal-Fetal & Neonatal Medicine 33(4), 687692. https://doi.org/10.1080/14767058.2018.1497595CrossRefGoogle ScholarPubMed
Yamamoto, JM, Kellett, JE, Balsells, M, García-Patterson, A, Hadar, E, Solà, I, Gich, I, van der Beek, EM, Castañeda-Gutiérrez, E, Heinonen, S, Hod, M, Laitinen, K, Olsen, SF, Poston, L, Rueda, R, Rust, P, van Lieshout, L, Schelkle, B, Murphy, HR and Corcoy, R (2018) Gestational diabetes mellitus and diet: A systematic review and meta-analysis of randomized controlled trials examining the impact of modified dietary interventions on maternal glucose control and neonatal birth weight. Diabetes Care 41(7), 13461361. https://doi.org/10.2337/dc18-0102CrossRefGoogle Scholar
Ye, G, Zhang, L, Wang, M, Chen, Y, Gu, S, Wang, K, Leng, J, Gu, Y and Xie, X (2019) The gut microbiota in women suffering from gestational diabetes mellitus with the failure of glycemic control by lifestyle modification. Journal Diabetes Research 2019, 6081248. https://doi.org/10.1155/2019/6081248CrossRefGoogle ScholarPubMed
Zhang, R, Han, S, Chen, G-C, Li, ZN, Silva-Zolezzi, I, Parés, GV, Wang, Y and Qin, LQ (2018a) Effects of low-glycemic-index diets in pregnancy on maternal and newborn outcomes in pregnant women: A meta-analysis of randomized controlled trials. European Journal of Nutrition 57(1), 167177. https://doi.org/10.1007/s00394-016-1306-xCrossRefGoogle ScholarPubMed
Zhang, Q, Yu, H, Xiao, X, Hu, L, Xin, F and Yu, X (2018b) Inulin-type fructan improves diabetic phenotype and gut microbiota profiles in rats. PeerJ 6, e4446. https://doi.org/10.7717/peerj.4446CrossRefGoogle ScholarPubMed
Zheng, W, Xu, Q, Huang, W, Yan, Q, Chen, Y, Zhang, L, Tian, Z, Liu, T, Yuan, X, Liu, C, Luo, J, Guo, C, Song, W, Zhang, L, Liang, X, Qin, H and Li, G (2020) Gestational diabetes mellitus is associated with reduced dynamics of gut microbiota during the first half of pregnancy. mSystems 5(2). https://journals.asm.org/doi/epub/10.1128/msystems.00109-20CrossRefGoogle ScholarPubMed
Figure 0

Figure 1. Possible composition of the gut microbiota in pregnant women with GDM according to the available literature.

Figure 1

Table 1. Studies of probiotics in GDM diagnosed women during gestation.