• Ei tuloksia

Food ingredients in human health: Ecological and metabolic perspectives implicating gut microbiota function

N/A
N/A
Info
Lataa
Protected

Academic year: 2022

Jaa "Food ingredients in human health: Ecological and metabolic perspectives implicating gut microbiota function"

Copied!
51
0
0

Kokoteksti

(1)

UEF//eRepository

DSpace https://erepo.uef.fi

Rinnakkaistallenteet Terveystieteiden tiedekunta

2020

Food ingredients in human health:

Ecological and metabolic perspectives implicating gut microbiota function

Wu, Qinglong

Elsevier BV

Tieteelliset aikakauslehtiartikkelit

© 2020 Elsevier BV

CC BY-NC-ND https://creativecommons.org/licenses/by-nc-nd/4.0/

http://dx.doi.org/10.1016/j.tifs.2020.04.007

https://erepo.uef.fi/handle/123456789/8233

Downloaded from University of Eastern Finland's eRepository

(2)

Journal Pre-proof

Food ingredients in human health: Ecological and metabolic perspectives implicating gut microbiota function

Qinglong Wu, Tingtao Chen, Hani El-Nezami, Tor C. Savidge

PII: S0924-2244(20)30436-2

DOI: https://doi.org/10.1016/j.tifs.2020.04.007 Reference: TIFS 2820

To appear in: Trends in Food Science & Technology Received Date: 24 October 2019

Revised Date: 30 March 2020 Accepted Date: 11 April 2020

Please cite this article as: Wu, Q., Chen, T., El-Nezami, H., Savidge, T.C., Food ingredients in human health: Ecological and metabolic perspectives implicating gut microbiota function, Trends in Food Science & Technology (2020), doi: https://doi.org/10.1016/j.tifs.2020.04.007.

This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

© 2020 Published by Elsevier Ltd.

(3)

Review

Food ingredients in human health: Ecological and metabolic perspectives implicating gut microbiota function

Qinglong Wua,b, Tingtao Chenc, Hani El-Nezamid,e,*, Tor C. Savidgea,b,*

a Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA

b Texas Children’s Microbiome Center, Department of Pathology, Texas Children’s Hospital, Houston, TX 77030, USA

c Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330031, China

d School of Biological Sciences, The University of Hong Kong, Pokfulam, Hong Kong S.A.R., China

e Institute of Public Health and Clinical Nutrition, University of Eastern Finland, FI-70211 Kuopio, Finland

* Correspondence: Tor.Savidge@bcm.edu (T.C.S.) and elnezami@hku.hk (H.E.N.)

(4)

Highlights

1) Dietary imbalance associated with consumption of processed foods alters gut microbiome homeostasis

2) Perturbation of gut microbiome composition and function is mediated by specific food ingredients

3) Technical recommendations for evaluating food ingredient effects on gut microbiome 4) Potential strategies for reversing diet/food ingredient-induced gut dysbiosis

5) Consider gut microbiome as part of safety assessments of food ingredients

(5)

Abstract 1

Background: Dietary imbalance and harmful food substances are well established risk factors 2

that can adversely impact human health. The gut microbiome is emerging as a new metabolic 3

organ that can be serendipitously linked to these poor dietary outcomes. Modern manufactured 4

foods and process ingredients can significantly alter gut microbiome composition and function, 5

leading investigators to conclude that disruption of host-microbiome commensalism is a key 6

mechanism in human disease linked to imbalanced diets or processed foods.

7

Scope and Approach: In this review, we highlight disease-associated perturbations and 8

precision manipulation of the human gut microbiome in the context of food and nutrition. We 9

detail technical recommendations for evaluating food ingredient effects on gut microbiome 10

composition and function, following Koch’s principles of commensal postulates to evaluate 11

whether reported findings can be implicated in disease causation. Strategies for minimizing 12

and/or reversing diet/food ingredient-induced gut dysbiosis linked with human disease are also 13

considered.

14

Key Findings and Conclusions: Current findings should encourage us to reevaluate the potential 15

health hazards of modern food ingredients and processed foods by considering the 16

involvement of the gut microbiome through well-established pipelines for deciphering the 17

causality of a perturbated gut microbiome in disease induction.

18

Keywords: gut microbiome; diet; food ingredients; microbiome metabolism; host health 19

(6)

1. Introduction 20

From the standpoint of modern food science and technology, our diet commonly involves 21

mixing natural ingredients (agricultural products and by-products) with artificial substances 22

(preservatives, decorative food additives, fortifying agents, etc.) to improve the quality and 23

functionality of processed foods during manufacture (Carocho, Barreiro, Morales, & Ferreira, 24

2014; Carocho, Morales, & Ferreira, 2015). Termed “processed foods”, these popular modern 25

diets are frequently implicated in long-term consequences on human health and development 26

(Botelho, Araújo, & Pineli, 2018; Monteiro, Moubarac, Cannon, Ng, & Popkin, 2013; Moodie et 27

al., 2013; Stuckler & Nestle, 2012). For the purpose of this review, food and diet are collectively 28

considered as ingredients ingested by an individual and processed by the gastrointestinal (GI) 29

tract. The GI tract has naturally evolved to digest and absorb the foods that we eat by facilitating 30

coordinated transit and autonomic contractions that mix dietary ingredients with digestive fluids 31

secreted by the gut, gallbladder and pancreas. Food substances are also exposed to the 32

consortia of microbes (including bacteria, fungi and viruses), termed the microbiome, that aid in 33

nutrient digestion and biotransformation (Sonnenburg & Bäckhed, 2016).

34

Over the last two decades, tremendous effort has been invested in elucidating the role of 35

nutrient-microbiome cross-talk with the host, often termed functional foods. These nutrient-host- 36

microbiome interactions have co-evolved over millennia, for example by developing tolerant 37

host defense systems, i.e. innate (Thaiss, Zmora, Levy, & Elinav, 2016), adaptive (Cullender et 38

al., 2013), and mucosal immunity (Mcdermott & Huffnagle, 2014) to avoid exacerbated immune 39

responses to dietary components in genetically susceptible individuals. In infancy, this field 40

emerged from studies identifying adverse mucosal immune responses, for example to dietary 41

gluten (Galipeau & Verdu, 2015; Sapone et al., 2011) or transient cow’s milk protein allergy in 42

young children (Walker-Smith, 2003). Since these early pioneering studies, our understanding 43

of nutrient-host-microbiome commensalism has moved in leaps and bounds to now implicate 44

(7)

specific microbe-derived metabolite classes in diverse human diseases, for example, succinate 45

for colonic epithelial gluconeogenesis (De Vadder et al., 2016), prevention of diabetes by short- 46

chain fatty acids (SCFAs) (Wen & Wong, 2017; Zhao et al., 2018), and bile acid homeostasis 47

linked to errors in host metabolism (Shapiro, Kolodziejczyk, Halstuch, & Elinav, 2018;

48

Wahlström, Sayin, Marschall, & Bäckhed, 2016). Moreover, the association of altered gut 49

microbiome composition with host phenotypes, i.e. obesity (Christensen, Roager, Astrup, &

50

Hjorth, 2018; Vangay et al., 2018), diabetes (Vatanen et al., 2018; Zhao et al., 2018), 51

hypertension and cardiovascular disease (Brown & Hazen, 2018; Marques, Mackay, & Kaye, 52

2018; Wilck et al., 2017), inflammatory bowel diseases (Khalili et al., 2018; Zhu et al., 2018), 53

and cancer (Goodman & Gardner, 2018; Shono & Van Den Brink, 2018; Zitvogel, Ayyoub, 54

Routy, & Kroemer, 2016), has been proposed as pathogenic and in some cases is supported by 55

Koch's commensal postulates (Neville, Forster, & Lawley, 2018; Zhao, 2013). Through these 56

studies, dysbiosis has become a central finding in many human diseases with tentative causes 57

linked to poorly balanced diets, unexpected chemicals/ingredients or other environmental 58

exposures. Consumption of different formulated foods and diets might lead to microbiome 59

changes in altered host health status (Botelho et al., 2018). For example, the dysbiosis 60

associated with high-fat or high-sugar diets (even from natural ingredients) is implicated in 61

obesity (Schulz et al., 2014; Turnbaugh et al., 2006). Biotransformation of foods by gut bacteria 62

is even reported to generate disease-causing metabolites, as suggested for L-carnitine 63

catabolism to trimethylamine-N-oxide (TMAO); this bacterial metabolite is derived primarily from 64

red meat and although it may promote bacterial osmolyte health benefits, is linked with 65

development of atherosclerosis in susceptible individuals by altering host cholesterol transport 66

(Koeth et al., 2013; Ussher, Lopaschuk, & Arduini, 2013). This finding is notable in the context 67

of food ingredients which are accumulating evidence of their capacity to adversely alter gut 68

microbiome composition and function in a manner that could impact host health.

69

(8)

In this review, we highlight recent links between specific food ingredients and gut dysbiosis, 70

and we provide technical recommendations for studying these effects. We also consider 71

potential strategies for reconstituting a healthy gut microbiome after dietary perturbation. We 72

conclude that consideration of the gut microbiome is important when evaluating food safety, as 73

well as nutritional intervention and management.

74

2. Perturbation of the human gut microbiome by imbalanced diet formulations and food 75

ingredients 76

Despite known lifestyle effects (including diet) on the assembly of distinct gut microbiome 77

communities in humans (Jha et al., 2018), it is now widely recognized that global food trade has 78

minimized such differences in populations consuming similar types of food. Modern food 79

manufacture among countries tends to be standardized, especially in its use of food additives to 80

prolong shelf life, enhance flavors and improve bio-functionality of manufactured foods (Carocho 81

et al., 2014). This has led to an emerging concern that frequent exposure to conserved food 82

additives or ingredients may disrupt gut microbiome homeostasis, thereby increasing host 83

disease susceptibility (Figure 1). Representative studies of food ingredient effects on gut 84

microbiome are summarized in Table 1.

85

2.1. Diet imbalance 86

Through natural symbiotic design to facilitate efficient food digestion, the GI tract offers 87

residence to trillions of commensal microorganisms that are shaped by our dietary influences.

88

Drastic shifts in gut microbiome composition are evident following long-term changes in diets 89

and are potentially associated with disease development. For example, in an adult French 90

cohort of 33,343 participants, an increase in the proportion of ultra-processed foods in the diet 91

was associated with a higher risk of developing functional gastrointestinal disorders, such as 92

(9)

irritable bowel syndrome and functional dyspepsia (Schnabel et al., 2018); but further studies 93

are needed to support causation by linking these longitudinal microbiome compositional shifts 94

with disease induction and restoring clinical health benefits through dietary intervention, ideally 95

in blinded placebo controlled cross-over trials. In further support of this notion, long-term 96

consumption of Western diet (high consumption of red meat and processed foods, etc.), but not 97

Mediterranean diet (high proportion of vegetables and whole grains, etc.), is widely recognized 98

as a risk factor in chronic inflammatory disorders such as Crohn’s disease and ulcerative colitis;

99

plausible mechanisms are linked to gut dysbiosis and enhanced intestinal permeability 100

triggering pro-inflammatory development (Khalili et al., 2018).

101

Seasonal recovery of the gut microbiome in Hadza hunter-gatherers of Tanzania is also 102

supportive of dietary impact on human microbiome patterns, being distinctly different from 103

urbanized individuals representing 18 different populations in 16 countries (Smits et al., 2017).

104

These findings are strongly supportive of dietary alterations that can significantly induce 105

microbial adaptation linked to human health. This phenomenon is exemplified by Hmong and 106

Karen populations from Thailand after immigration to the US, and involves microbiome 107

enterotype switching from Prevotella to Bacteroides after Westernization. Notably, this 108

enterotype switch is functionally linked with decreases in microbial enzymes required for plant 109

fiber degradation and loss of microbiome diversity, which are features associated with 110

development of obesity across generations (Vangay et al., 2018); Knights and colleagues 111

reasoned this microbiome shift might be linked to dietary changes. In addition to these 112

population-scale longitudinal studies, short-term (within days) intake of entirely animal-based 113

foods enriches bile-tolerant Alistipes and Bacteroides while decreasing the abundance of 114

Eubacterium and Ruminococcus that are capable of catabolizing plant polysaccharides (David 115

et al., 2014). These long- and short-term investigations on human subjects demonstrate that 116

dietary imbalance promotes gut dysbiosis and is associated with higher risk or susceptibility to 117

(10)

disease development. Thus, different formulations of food ingredients in the diet can 118

dramatically influence the composition and metabolic potential of the gut microbiome.

119

2.2. Non-caloric artificial sweeteners 120

Dietary non-caloric/nutritive artificial sweeteners (NAS) such as saccharin and sucralose 121

have been Generally Recognized As Safe (GRAS) by the US Food and Drug Administration 122

(FDA), and are widely used for manufacturing diverse processed/packaged foods which are 123

consumed by millions of type II diabetics and obese individuals without increasing caloric intake 124

(Suez, Korem, Zilberman-Schapira, Segal, & Elinav, 2015). However, to date there has been no 125

detailed investigation on the association between NAS consumption and gut microbiome 126

composition. In 2014, it was reported that glucose intolerance developed in mice fed 127

commercial saccharin, sucralose and aspartame when compared with mice fed isocaloric 128

sucrose and glucose; this finding models long-term NAS consumption being associated with 129

impaired glucose tolerance in humans. High resolution shotgun metagenomic analysis of human 130

fecal microbiome transplanted (FMT) germ-free mice identified NAS-enriched gut commensals 131

such as Bacteroides vulgatus as potential causative targets among the altered gut microbiome 132

(Bokulich & Blaser, 2014; Suez et al., 2014). Interestingly, administration of ciprofloxacin, 133

metronidazole and vancomycin to NAS-fed mice suppressed the glycemic response to NAS 134

suggesting elimination of causative bacteria by these antimicrobial agents (Bokulich & Blaser, 135

2014). Similar findings are evident in humans where responders showing elevated glycemic 136

response to NAS consumption demonstrate gut microbiome changes, whereas a proportion of 137

non-responder subjects maintaining a normal glycemic response to short-term NAS 138

consumption demonstrated insignificant changes in gut microbiota community (Suez et al., 139

2014). Since this dysbiosis may represent a compensatory microbiome response to elevated 140

systemic glucose levels rather than being causative, it would be interesting to explore whether 141

transfaunation of gut microbiome communities from responders versus non-responders into 142

(11)

germ-free models supports the transmissive glycemic phenotype. Nevertheless, these studies 143

highlight the dynamic association of gut microbiome communities with personalized glycemic 144

phenotypes (Zmora, Zeevi, Korem, Segal, & Elinav, 2016). Further support of nutraceutical 145

impact on the gut microbiome is provided by a recent study highlighting that sucralose 146

consumption induces liver inflammation in C57BL/6 mice and is associated with fecal 147

proinflammatory metabolite shifts (Bian et al., 2017). When considering the extensive 148

consumption of NAS, especially by individuals with metabolic syndrome, it becomes clear that 149

this class of foods needs to be re-assessed for impact on the microbiome and long-term health 150

consequences.

151

2.3. Nutritive sweeteners 152

Trehalose is a non-reducing and temperature-stable disaccharide making it a preferred 153

baking ingredient. Its use as a stabilizer is now also extended to thousands of other common 154

foods, including ground beef and ice cream. Although the microbiome is reported to utilize 155

trehalose, the metabolic advantage that this disaccharide confers to epidemic strains of the 156

pathogen Clostridioides difficile has drawn a lot of clinical and press attention (Collins et al., 157

2018). Two epidemic ribotypes (RT027 and RT078) of C. difficile are able to utilize levels of 158

trehalose which are expected from the consumption of trehalose-rich food products, promoting a 159

selective advance for pathogen colonization and virulence in the intestine. This finding is 160

interesting since the emergence of these epidemic strains co-occurred around the time that 161

trehalose was introduced in bulk as a food supplement around the world. The pathogenic fitness 162

displayed by epidemic C. difficile strains was linked to two independently acquired mutations 163

that confer an enhanced ability to utilize trehalose in the diet. Specifically, in RT027 strains a 164

conserved L1721 amino acid substitution in TreR, a repressor of the treAR operon and an 165

effector of the phosphotransferase system for trehalose translocation was identified. As a result, 166

the elevated induction of the treA gene increased pathogen sensitivity to dietary trehalose. In 167

(12)

RT078 strains, a horizontally acquired gene cluster consisting of a second copy of TreA and 168

TreR, and a trehalose-specific phosphotransferase system IIBC component confer enhanced 169

trehalose catabolism for energy generation needed for bacterial expansion (Collins et al., 2018).

170

When considering the high trehalose content of modern Western diets, these findings raise a 171

concern that epidemic C. difficile (RT027 and RT078) infections will remain prevalent until food 172

manufacturing practices are changed (Britton & Young, 2014; Collins et al., 2018). Recently, 173

degradation-resistant trehalose analogues (trehalase inhibitors) have been synthesized that 174

block trehalose utilization by hypervirulent C. difficile strains (Danielson et al., 2019), but use of 175

analogues as co-additives needs further evaluation in terms of safety. Importantly, these 176

findings set precedent that other GRAS-granted dietary sweeteners might confer a fitness 177

advantage to some certain infectious pathogens.

178

More recently, lactose, the natural sugar found in raw milk and processed dairy products, 179

has been reported to be associated with the expansion of Enterococcus faecium, but not other 180

enterococci in the large intestine of patients who underwent allogeneic hematopoietic stem cell 181

transplantation (allo-HSCT); such enterococcal expansion exacerbated graft-versus-host 182

disease (GVHD) and patient mortality in both human and mouse models (Stein-Thoeringer et al., 183

2019). It is well known that various antibiotics including vancomycin have been used to 184

eliminate opportunistic pathogens during the conditioning process prior to stem cell infusion and 185

even during cell engraftment; this treatment selectively enriches vancomycin-resistant 186

enterococci including Enterococcus faecium in the GI (Taur et al., 2018). Not surprisingly, allo- 187

HSCT patients with lactase deficiency showed significantly higher abundance of Enterococcus 188

compared with lactose absorbers (Stein-Thoeringer et al., 2019). To overcome such potentially 189

detrimental microbiome responses associated with lactose consumption, dietary manipulation 190

resulting in lactose depletion decreased enterococcal abundance and attenuated GVHD-related 191

mortality (Stein-Thoeringer et al., 2019). Lactose is a common dietary component in Western 192

(13)

diets, and consumption of lactose-rich diet favors dysbiosis with outgrowth of Enterococcus 193

faecium. Instead of using the Leloir metabolic pathway for exclusively processing glucose 194

moieties from lactose, this organism adopts a tagatose-6-phosphate (T6P) pathway for 195

catabolizing lactose thus utilizing both glucose and galactose moieties of lactose (Wu, Cheung, 196

& Shah, 2015). However, our early genetic survey indicated that not all strains of Enterococcus 197

faecium and Enterococcus faecalis possess intact T6P metabolic pathways, and the T6P 198

pathway in Enterococcus faecium could be either plasmid-encoded or present in its 199

chromosome (Wu et al., 2015). Thus, further investigation of the functional analysis of the T6P 200

pathway in clinically relevant strains of Enterococcus faecium isolated from allo-HSCT patients 201

are merited to profile such differences.

202

2.4. Dietary emulsifying agents 203

Dietary emulsifiers are detergent-like agents that are common ingredients in modern 204

processed foods. Although these agents are approved by the FDA, their influence on the gut 205

microbiome remains unclear. In 2015, carboxymethylcellulose and polysorbate-80 were shown 206

to alter gut microbiome composition in mice, associated with a significant reduction in the alpha- 207

diversity metric namely observed Operational Taxonomic Units (OTUs) that was linked with 208

blooming of Akkermansia muciniphila and inflammation-promoting Clostridium perfringens.

209

Transfaunation of the emulsifier-induced dysbiotic microbiota in germ-free mice induced low- 210

grade gut inflammation and promoted metabolic syndrome with increased adiposity and 211

dysglycaemia linking causation of these disease states with emulsifier consumption and impact 212

on the host microbiome (Chassaing et al., 2015). To investigate emulsifier effects directly on the 213

microbiome, a simulator of the human intestinal microbial ecosystem lacking host influence was 214

used: carboxymethylcellulose and polysorbate-80 not only increased alpha- and beta-diversity 215

(UniFrac distance) metrics in human fecal microbiome communities, but also induced microbial 216

gene expression resulting in higher levels of pro-inflammatory lipopolysaccharide and flagellin 217

(14)

FliC (Chassaing, Van De Wiele, De Bodt, Marzorati, & Gewirtz, 2017). Microbiome perturbations 218

caused by carboxymethylcellulose and polysorbate-80 are also reported to drive alterations in 219

proliferation and apoptosis pathways promoting colon carcinogenesis (Viennois, Merlin, Gewirtz, 220

& Chassaing, 2017). Similarly, carrageenan – a sulfated polysaccharide – is reported to induce 221

intestinal inflammation associated with a decreased relative abundance of Akkermansia 222

muciniphila in mice (Shang et al., 2017). However, further causation studies are required to 223

demonstrate the importance of Akkermansia muciniphila as an anti-inflammatory commensal in 224

response to dietary carrageenan. Since other dietary emulsifiers, for example glycerol 225

monolaurate, are also reported to induce gut dysbiosis, metabolic syndrome and low-grade 226

inflammation, even in a low-fat diet mouse model (Jiang et al., 2018), further evaluation of 227

detrimental emulsifier-host-microbiome interactions are merited.

228

2.5. Salt 229

Dietary salt (sodium chloride) is a ubiquitous component in modern diets. High salt 230

consumption is one of the causative factors contributing to cognitive impairment (Faraco et al., 231

2018, 2019) and cardiovascular diseases driven by hypertension (Mozaffarian et al., 2014).

232

However, the impact of this common food additive on the gut microbiome has not been well 233

characterized. Recently, a high salt diet was shown to alter mouse fecal microbiome community 234

composition with decreased relative abundances of Lactobacillus, especially Lb. murinus which 235

recovered after introduction of normal salt chow (Wilck et al., 2017); interestingly, administration 236

of Lb. murinus to high salt-challenged mice reduced the levels of hypertension-contributing TH17 237

lymphocytes (Wilck et al., 2017). In an open-label clinical pilot study, 14 days of increased salt 238

intake depleted several species of Lactobacillus including Lb. casei group, Lb. plantarum and Lb.

239

brevis as assessed via shotgun metagenomic analysis; high dietary salt also lead to elevated 240

nocturnal systolic and diastolic blood pressure, and reduced circulating TH17 lymphocytes 241

(Wilck et al., 2017). Although mouse and human differences exist in the specific Lactobacillus 242

(15)

sp. that are adversely impacted by a high salt diet, it does link Lactobacillus as a genus with 243

blood pressure abnormalities. This association could be due to altered host physiology caused 244

by high salt, for example intestinal transit is commonly linked with blood pressure and 245

Lactobacillus abundance. However, causation is indicated by a recent meta-analysis of nine 246

human trials showing that consumption of probiotic Lactobacillus reduces human systolic and 247

diastolic blood pressure (Khalesi, Sun, Buys, & Jayasinghe, 2014). Potentially, altered 248

microbiome function could mediate these changes in host physiology through modulation of 249

vagal nerve and immune signaling. For example, a high-salt diet administrated to mice 250

decreased the production of protective microbiome-derived short-chain fatty acids e.g. butyrate 251

and altered gut immune homeostasis involving enhanced pro-inflammatory gene expression 252

which exacerbated experimental colitis in mice. Transfaunation of this high salt diet-associated 253

microbiome into germfree mice demonstrated that changes in community composition are 254

dependent on continuous dietary salt exposure and that changes may be transient (Miranda et 255

al., 2018).

256

2.6. Dietary soluble fiber and carbohydrates 257

Soluble and insoluble dietary fibers are generally thought to be beneficial to human health 258

via sustaining a healthy gut microbiome and gastrointestinal homeostasis (Makki, Deehan, 259

Walter, & Bäckhed, 2018). Western diet which is typically a low-fiber diet shown to deplete gut 260

commensal bacteria that are capable of producing key physiological metabolites such as short- 261

chain fatty acids (SCFAs) (Deehan & Walter, 2016; Makki et al., 2018). In other words, dietary 262

fiber is vital for SCFAs production by the healthy colonic microbiome. Consumption of fiber-rich 263

diet also demonstrates other potentially notable health-promoting effects including improved 264

glucose tolerance through regulating the ratio of Bacteroides/Prevotella (Kovatcheva-Datchary 265

et al., 2015), facilitating clearance of C. difficile via possible link to elevated levels of SCFAs 266

(Hryckowian et al., 2018), and reducing risk of hepatocellular carcinoma (Yang et al., 2019).

267

(16)

There are a few exceptions to this consensus, for example Vijay-Kumar and the team 268

demonstrated that soluble fibers (inulin, pectin and fructo-oligosaccharides), but not insoluble 269

fiber counterpart cellulose, induced hepatocellular carcinoma in a subset of Toll-like receptor 5 270

(TLR5)-null mice, a model which is deficient in immune responses to bacterial flagellin (Singh et 271

al., 2018). They found that diet enriched in soluble fibers promoted γ-Proteobacteria relative 272

abundance as a potential pathogenic source of bacterial flagellin and lipopolysaccharides. The 273

soluble fiber diet also enriched for fermenters of the Clostridium XIVa cluster that was 274

associated with high levels of fecal butyrate and serum secondary bile acids linked to liver 275

fibrosis disease progression (Singh et al., 2018). Hepatocellular carcinoma was not observed in 276

germ-free, wild-type, and metronidazole-treated TLR5 deficient mice fed soluble fiber, which the 277

authors claim support a role for microbial-derived mediators in the disease pathogenesis (Singh 278

et al., 2018). It is also possible that the microbiome community shift represents a compensatory 279

response to liver injury and further work is needed to delineate disease causation. The impact of 280

food on secondary bile acids is an area of intense research currently, especially since colonic 281

7α-dehydroxylating bacteria which convert primary to secondary bile acids are also emerging as 282

potentially important modulators of human disease, including infectious colitis (Buffie et al., 2015;

283

Savidge & Sorg, 2019).

284

In addition to the unexpected side effects of soluble fiber (long-chain polysaccharides) 285

mentioned above, soluble short-chain carbohydrates are reported to exacerbate diseases such 286

as irritable bowel disease. Diets containing a high proportion of FODMAPs (Fermentable 287

Oligosaccharides, Disaccharides, Monosaccharides, And Polyols), such as in a normal Western 288

diet, can trigger gastrointestinal symptoms in patients with irritable bowel syndrome and have 289

been linked with altered microbiome fermentation (McIntosh et al., 2017; Shepherd, Lomer, &

290

Gibson, 2013). Symptom relief and improved quality of life can be achieved in a proportion of 291

irritable bowel syndrome patients on a diet that is low in FODMAPs (Camilleri & Acosta, 2014;

292

Shepherd et al., 2013). Much public attention has been levied on this dietary management as 293

(17)

this disease impacts up to 20% of the western population (Oshima & Miwa, 2015).

294

Carbohydrate-trigged diseases are likely highly dependent on microbiome activity, but 295

identifying the culprit species and disease mechanisms have been difficult. Another important 296

consideration here that applies broadly to the analysis of fecal specimens from human subjects 297

is that these may represent poor surrogates to investigate a disease process that happens in 298

the small intestine or at gut mucosal surfaces.

299

As shown in Table 1, food ingredients may demonstrate specific microbiome alterations 300

which are not in common across studies. One explanation for such diverse changes could be 301

linked to the metabolic traits of gut bacteria on those food ingredients, i.e. competitive growth 302

fitness conferred by specific ingredients such as simple sugars; however, for food ingredient- 303

induced depletion of beneficial commensal bacteria, indirect influences driven by alterations in 304

host and dysbiosis-associated chemical niches should not be ignored. Importantly, many 305

experimental validation studies transferred ingredient-induced microbiome communities into 306

germ-free or conventional mouse model and recapitulate the phenotype confirming the 307

microbiome dysbiosis-driven disease outcome. Although this highlights the potential 308

cooperation among microbes for phenotype induction, few of these studies have confirmed 309

disease causality of individual microbes associated with disease phenotypes. This merits further 310

effort to decipher microbe-microbe interactions to establish disease causation.

311

Long-term frequent consumption of food ingredients-containing foods and diets could be 312

cause diverse gut microbiome dysbiosis resulting in disease induction and susceptibility.

313

Modern food manufacturing should rethink the content of food additives and re-innovate such 314

process would improve food quality. Considering ingredient-specific induction of microbiome 315

alterations, strategies to reverse such dysbiosis might vary but the replacement with healthy gut 316

microbiome should be a universal and effective approach to recover healthy state. However, 317

microbiota replacement seems to be not a broadly acceptable way at current stage because our 318

(18)

knowledge on gut microbiome is still expanding and more extensive investigations are still 319

needed to illustrate microbial dark matter of gut microbiome. Thus, the best preventative 320

strategy so far is to follow certified dieting guidelines to avoid excessive consumption of food 321

additives. More precisely, we need to know what we are feeding, and thus microbiome 322

monitoring should aid such decision on diet choice and disease treatment.

323

3. Association to causation – technical recommendations for evaluating the effects of 324

food ingredients on host health with a focus on the gut microbiome 325

Numerous next generation sequencing techniques are established to profile the human 326

gut microbiome, especially targeted and shotgun metagenomic sequencing which are culture- 327

independent strategies. An important feature of these methods is that they are able to detect or 328

assemble a large proportion of uncultured or unknown microbes in the host GI tract beyond our 329

current culture collections and databases. Thus, the term “culturomics”, a conventional culture- 330

dependent method using varying nutrients for isolation and culture, has been introduced to 331

advance this field (Browne et al., 2016; Lagier et al., 2018, 2016). This labor-intensive approach 332

identified over 200 previously “unculturable” species from the human GI tract (Lagier et al., 333

2016), and generated an archive of gut bacteria representing 90% of GI bacterial species 334

commonly identified using next generation sequencing (Browne et al., 2016). These single 335

bacterial isolates can even be assembled together to perform functional dynamic assays of key 336

human microbiome consortia identified (Samuel, Rowedder, Braendle, Félix, & Ruvkun, 2016).

337

Although these culture-dependent efforts are meaningful, microbe-microbe and host-microbe 338

interactions still need to be considered when performing food/nutrition-associated microbiome 339

studies, such as toxicology of food additives and evaluation of nutritional interventions. To best 340

achieve this type of approach, we present a basic experimental workflow incorporating three 341

typical models for microbiome research as illustrated in Figure 2.

342

(19)

3.1. Consider the resilience and individuality of the human gut microbiome 343

The temporal stability of the human gut microbiome as measured by its taxonomic 344

composition over time has been unclear for a long time. Recent longitudinal studies have shed 345

some important new light on this question: in a Chinese longitudinal pilot study, bi-directional 346

plasticity and resilience of the human gut microbiome were observed across a longer time scale 347

(6 months) and was associated with multiple dietary shifts (Liu et al., 2019). Similar 348

observations were re-captured in another longitudinal (more than one year) and geographically 349

diverse study on Hadza hunter-gatherers in Tanzania showing annual cyclical reconfiguration of 350

microbiome community compositions that are linked to seasonal availability of different types of 351

food (Smits et al., 2017). Geography also shows a strong association with host microbiome 352

variation as demonstrated in a Chinese regional trial composing over 7,000 individuals from 14 353

districts in Guangdong province (He et al., 2018). Notably, ethnic origins contribute to the inter- 354

individual dissimilarities in gut microbiome composition in Amsterdam, Netherlands revealed in 355

a trial of over 2,000 participants (Deschasaux et al., 2018); these reports demonstrate the 356

contribution of geography and ethnicity (i.e. genetics) in explaining microbiome variation (Gupta, 357

Paul, & Dutta, 2017), but variation is also influenced by diet and lifestyle indicating the 358

importance of understanding the personalized microbiome in terms of individuality. Large 359

pediatric and adult cohort studies of healthy populations, including MetaHIT, Human Microbiome 360

Project, American Gut Project and TwinsUK, endorse these findings by demonstrating 361

population-scale stratification based on distinct microbiome community clusters termed as 362

“enterotypes” (Arumugam et al., 2011; Costea et al., 2017). In adults, this variation is 363

contributed to by genetics and epigenetics, although the basis for enterotyping an individual 364

based on their gut microbiome composition remains controversial. Further, enterotypes have not 365

been reported in infants, and the TEDDY cohort, a large longitudinal study composing over 900 366

infants with follow-up from 3 to 46 months of age, showed that the early assembly of the infant 367

(20)

microbiome underwent distinct developmental, transitional and stable phases (Stewart et al., 368

2018). Infant microbiome composition was largely associated with breastfeeding, while birth 369

mode, geography and household exposure also contributed to microbial variation (Stewart et al., 370

2018). These population scale studies have thought us that the individuality of the human gut 371

microbiome is associated with age, diet, and environmental exposures. These are important 372

lessons since we need to consider host genetics and epigenetics when assessing the impact of 373

food ingredients on the human gut microbiome, as well as microbiome-based interventions for 374

personalized nutrition and disease management (Christensen et al., 2018; He et al., 2018).

375

In the context of personalized nutritional management, a pioneering study measured host 376

glycemic responses to dietary carbohydrates such as bread (Zeevi et al., 2015). Elevated post- 377

meal blood glucose levels are normally a major risk factor for developing type II diabetes, yet 378

postprandial glycemic responses among individuals consuming the same identical meals are 379

highly variable (Zeevi et al., 2015). One major factor to be considered for such variability is the 380

individuality of the gut microbiome (Korem et al., 2017), which may be considered a significant 381

metabolic organ in personalized nutrition (Bashiardes, Godneva, Elinav, & Segal, 2018). Such 382

an approach potentially has significant implications for general published guidelines on diet and 383

lifestyle aimed at educating populations on healthy food choices. Because an individual’s gut 384

microbiome may require deviations from general dietary guidelines to accommodate an 385

optimized lifestyle based on diet (David et al., 2014; Deschasaux et al., 2018; Jha et al., 2018;

386

Petersen et al., 2017), geography (Gupta et al., 2017; He et al., 2018; Yatsunenko et al., 2012), 387

age (Yatsunenko et al., 2012) and environmental conditions (Worthmann et al., 2017), this 388

provides new challenges to nutritionists providing dietary recommendations. This becomes 389

especially taxing with patients seeking clinical advice on microbiome profiling provided by 390

emerging commercial vendors, who often refer the client back to their own physician for correct 391

interpretation. A pragmatic approach is needed, with clinical associations with certain microbial 392

(21)

taxa being recognized as such until causation can be demonstrated using the approaches 393

outlined in Figure 2.

394

3.2. In vitro bioreactor studies 395

In vitro models have been extensively used to monitor the taxonomic and functional 396

changes in microbiome communities perturbed by food or specific nutrients. Different types of 397

bioreactors have been developed to maintain the complex structure and diversity of a 398

microbiome community. A well-designed model is the Simulator of the Human Intestinal 399

Microbial Ecosystem (SHIME), a 5-step multi-chamber bioreactor, which was developed two 400

decades ago for mimicking the progressive digestive processes of the stomach, small intestine 401

and colon (Molly, Vande Woestyne, & Verstraete, 1993). The spatiotemporal model enables a 402

complex and stable microbiome community structure to form, and a specific model termed M- 403

SHIME also incorporated an unstirred mucus layer for adhering microbiome biofilm (Van den 404

Abbeele et al., 2012). A more simplistic model – Mini-BioReactor Arrays (MBRAs), was 405

developed for rapid cultivation of microbiome communities from human fecal specimens 406

(Auchtung, Robinson, & Britton, 2015). This MBRAs model enables medium throughput studies, 407

which have been applied to investigate for example trehalose effects on C. difficile growth in 408

complex microbiota communities (Collins et al., 2018). A similar model strategy was used to 409

establish symbiotic (prebiotic and probiotic combination) mechanisms that drive antimicrobial 410

activity against C. difficile (Steiner, 2010). However, significant limitations using these in vitro 411

models include lack of host influences e.g. antibodies or antimicrobial, or limited experimental 412

throughput especially in SHIME models which provide a more stable microbiome community 413

structure (Mcdermott & Huffnagle, 2014; Pabst, Cerovic, & Hornef, 2016; Zeng et al., 2016).

414

Although this in vitro approach provides a clearer understanding of the perturbations that 415

directly target gut microbiome structure-function relationships, host-microbiome interactions 416

should not be neglected since these may exert profound effects on host physiology.

417

(22)

3.3. Ex vivo or in vivo studies 418

Ex vivo approaches are not extensively reported for gut microbiome studies. When this 419

strategy is adopted, it usually involves time-series fecal cultivation in combination with dietary 420

perturbations (Collins et al., 2018; Lamendella et al., 2018). Results from this type of study 421

design should be interpreted with caution since the stability of the microbiome community 422

structure during experimental anaerobic cultivation is uncertain and not well controlled.

423

An advantage of using animal models (typically mouse) for studying the gut microbiome is 424

that perturbation can be controlled using standardized feed and housing conditions. A 425

translational limitation that needs to be considered is the major species difference between 426

murine and human microbiome community composition and function. To overcome this, 427

significant effort has been levied to generate humanized models by introducing an individual’s 428

gut microbiome communities into the germ-free mice, a process termed transfaunation (Collins, 429

Auchtung, Schaefer, Eaton, & Britton, 2015; Faith et al., 2010). However, this model often does 430

not contain the same diversity and structure of human gut microbiome and emerging data 431

indicates that certain host responses e.g. immune responses are not appropriately developed in 432

the presence of human-derived microbial species as these have not evolutionary adapted to the 433

mouse gut environment or genetic determinants (Zhou, Chow, Fleming, & Oh, 2019).

434

3.4. Human cohort studies 435

Generally, clinical trials should be carried out when expected outcomes are validated in 436

preclinical models, but with major microbiome compositional differences existing across species 437

that possibly complicate experimental findings, exceptions might be applied for GRAS-granted 438

or Qualified Presumption of Safety (QPS)-listed food ingredients and probiotics. The 439

observation of region-dependent microbiome differences among age-matched human 440

individuals has been linked to diets contributing to rapid transit of the gut microbiome (David et 441

(23)

al., 2014; Deschasaux et al., 2018; He et al., 2018; Yatsunenko et al., 2012). Hence, any pilot 442

study involving human subjects for microbiome-based assessment needs to control for or 443

document dietary differences. A diet provided by the clinical coordinator with a detailed food and 444

symptoms diary is the preferred documentation for such clinical studies since recall memory is 445

often reported to provide inaccurate information. Although representing the gold standard, new 446

electronic applications are assisting with dietary assessment and confer some advantages since 447

time stamped entries of snacks and meals may provide a more accurate approach to food 448

questionnaires and diaries as these provide gross-level estimates. Smartphone applications, for 449

example MyFitnessPal provides automated energy and macro- and micronutrient intake values 450

based on foods consumed (Teixeira, Voci, Mendes-Netto, & da Silva, 2018). Longitudinal 451

measurements of dietary and gut microbiome data, ideally as a double-blinded cross-over or 452

parallel design trials, may assist in powering the study and to control for individual specific 453

responses. Data analysis of responder versus non-responder phenotypes should be performed 454

separately and blindly to avoid possible interpretation bias.

455

3.5. Analytical methods for profiling microbiome composition and inferring metabolic 456

function 457

Several strategies as shown in Figure 3 are used for profiling the gut microbiome and 458

include: (1) targeted metagenomic sequencing, i.e., 16S rDNA amplicon or full-length 459

sequencing; (2) shotgun (deep or shallow) metagenomic sequencing; (3) shotgun 460

metatranscriptomic sequencing; (4) mass spectrometry-based metaproteomic analysis. Among 461

these high-throughput assays, targeted metagenomics – commonly referred to as 16S rDNA- 462

based amplicon sequencing is a more affordable approach for gut microbiome analysis.

463

Amplicon sequence data has been generated for thousands of projects, but each project is 464

designed with little consideration to preference for selecting variable 16S rDNA region that are 465

commonly used i.e. V4, V1-V3, V3-V5 or V6-V9. Sequencing strategies are often influenced by 466

(24)

large studies such as Human Microbiome, Earth Microbiome and American Gut Projects since 467

validations have been conducted before the production phase of these studies. However, the 468

influence of amplicon length (after quality trimming), sequence orientation and 16S region on 469

taxonomic calls has not been benchmarked systematically. To ensure the same observations on 470

gut microbiome features are validated by previously published data, meta-analysis of gut 471

microbiome-associated studies might be performed via a closed-reference analysis strategy 472

allowing comparisons and concatenation of data (Wang et al., 2018), however, attention need to 473

be exercised when using such an approach: 1) quality filtering and trimming of amplicon 474

sequences should adopt expected error-based methods but not average quality-based filtering 475

(Edgar & Flyvbjerg, 2015); 2) for the sake of accuracy default similarity (97%) for clustering 476

amplicon sequences might be avoided since there is no strong evidence to support the even 477

distribution of 3% dissimilarity in nucleotides confined to the nine variable regions of the 16S 478

rRNA gene that are sequenced (Mysara et al., 2017; Nguyen, Warnow, Pop, & White, 2016;

479

Yang, Wang, & Qian, 2016), thus more stringent clustering or sequence denoising for meta- 480

analysis may generate more accurate profiles; 3) methods of taxonomic classification for the 481

feature sequences or representative sequences of OTUs should be benchmarked in detail for 482

selecting the confidence threshold for controlling misclassification and overclassification (Edgar, 483

2018; Murali, Bhargava, & Wright, 2018); 4) appropriate choice of reference database is 484

important for accurate taxonomy, for example Greengenes and SILVA databases do not have 485

precisely curated databases because those collections are usually derived from GenBank and 486

European Nucleotide Archives where world-wide users can deposit sequence data (Edgar, 487

2018). An authoritative database from the training set of Ribosomal Database Project (RDP), 488

NCBI 16S rRNA RefSeq Targeted Loci Project or Genome Taxonomy Database (GTDB) is 489

commonly recommended but users should be cautious in relying exclusively on these 490

databases. Another strategy that is emerging is applying exact amplicon sequence variants 491

(ASVs) introduced by DADA2 for the meta-analysis (Callahan et al., 2016). Moreover, multiple 492

(25)

copies (ranging from 5 to 10) of prokaryotic ribosomal RNA operons in a single genome are 493

commonly found (Stoddard, Smith, Hein, Roller, & Schmidt, 2015); ideally, individual 494

microbiome OTUs and ASVs profiles should be corrected for 16S copy number so that this is 495

matched to the ribosomal RNA database. A major challenge in conducting this type of 496

bioinformatics correction is that short amplicon sequences of 16S rRNA gene do not offer 497

confident calls at the rank of species because those amplicons may be derived from either 498

known species or uncultured bacteria sharing the same sequence identity of the sequenced 499

region of 16S rRNA gene. Even with these considerations, microbiome profiling bias might still 500

be there and this merits future bioinformatics and statistical efforts (McLaren, Willis, & Callahan, 501

2019). Importantly, microbiome community profiles built on calculating relative abundances do 502

not reflect the biomass of the microbial consortia, thus total 16S copy numbers should be 503

assessed, i.e. real-time quantitative PCR with bacterial universal primers for the same DNA 504

extracts.

505

Since 16S rDNA amplicon sequencing does not offer high resolution to taxon, especially at 506

species rank, shotgun metagenomic sequencing becomes a useful strategy to provide potential 507

whole-genome information for precise taxonomic profiling of the gut microbiome. In general, two 508

types of analysis have been performed as illustrated in Figure 3B: 1) read-mapping based 509

analysis: filtered and trimmed sequence reads are mapped to the curated reference microbial 510

genomes or a set of marker genes per bacterial genomes by using sequence alignment or k- 511

mers spectrum, this approach is adopted in many popular analysis packages – MetaPhlAn, 512

DIAMOND, Kraken and CLARK (McIntyre et al., 2017); 2) de novo assembly-based analysis:

513

individual or concatenated sequencing reads from all samples are first assembled prior to 514

taxonomic assignment and functional annotations. This type of analysis includes many 515

packages but the comprehensive Anvi’o platform is useful for downstream analysis (Eren et al., 516

2015). For de novo assembly, curations must be performed for misassemblies and misbinning 517

(26)

of metagenomic contigs; sometimes, authoritative genome databases are required to guide 518

those processes depending on the programs. Timely documentation from large and 519

comprehensive benchmarking studies on analytical strategies, performance of programs, and 520

databases have been generated (McIntyre et al., 2017; Sczyrba et al., 2017), and these 521

comparisons provide valuable information to users for selecting analysis strategies of shotgun 522

metagenomics.

523

Shotgun metagenomics is able to provide not only precise taxonomy but also the potential 524

functional dynamics of the gut microbiome. Although meta-transcriptomics is a preferable 525

approach providing better understanding on functional potential (Abu-Ali et al., 2018), unbiased 526

label-free metaproteome analysis of fecal specimens or biopsies also provides potential at 527

protein level to probe microbiome functional capacity (Zhang et al., 2016). A potential concern 528

using this latter technique is protein detection may be biased to abundant proteins as these are 529

more easily profiled; detection limits are associated with: 1) invalid methods for protein 530

extraction with high yields; 2) simple or lack of fractionation of tryptic peptides; 3) constraints 531

from database-driven peptide identification, while de novo peptide sequencing for tandem mass 532

spectra is still prone to errors (Muth & Renard, 2018). As such, paired metagenomics- 533

assembled genomes together with public protein databases including UniProt and Unified 534

Human Gastrointestinal Protein (UHGP) (Almeida et al., 2019) are commonly recommended for 535

the database search strategy during metaproteome analysis (Heyer et al., 2017).

536

4. Potential strategies for overcoming gut dysbiosis caused by specific food ingredients 537

Globalization of the food industry contributes to the wide spread consumption of common 538

food ingredients around the world. It is important for government agencies to rapidly update 539

public guidelines once GRAS status of specific food ingredients is revoked after re-evaluation of 540

possible impact on human health. To apply such guidelines to dietary effects on the human 541

(27)

microbiome, we must first understand how to define a “normal microbiome”, especially since 542

dysbiosis is a frequent manifestation without an associated health outcome. Recently, there is 543

increasing demand for manipulations and interventions that may counter and/or correct 544

dysbiosis. Often, little consideration is given to possible compensatory responses of the 545

microbiome being beneficial to the host, and thus the term dysbiosis may be misleading as it 546

implies a detrimental outcome to the host. For this reason, we and others emphasize the 547

importance of establishing disease causation before considering microbiome manipulation as a 548

form of therapy. If causation can be established, two strategies including ecological modelling 549

and non-modelling approaches may be considered for discovering effector strains that could be 550

considered to restore a healthy microbiome (Figure 4).

551

4.1. Dietary intervention 552

A healthy diet is key to maintaining gut microbiome homeostasis by supplying appropriate 553

nutrients and cofactors to benefit microbial function. Patients with type 2 diabetes (T2D) have a 554

dysbiosis that is linked to a deficiency in short-chain fatty acids (SCFAs) production (Tolhurst et 555

al., 2012); a high-fiber diet, demonstrated in a randomized clinical trial to selectively enrich 15 556

species with acetate or/and butyrate production potential. This finding was associated with 557

improved glucose homeostasis via triggering of intestinal glucagon-like peptide-1 production 558

that that enhances pancreatic insulin secretion in T2D participants (Zhao et al., 2018). This 559

benefit was validated in a mouse model with dysregulation of glucose homeostasis by 560

administrating acetate-producing Bifidobacterium pseudocatenulatum (Zhao et al., 2018).

561

Moreover, a specialized diet containing acetate and butyrate provided protection against insulitis 562

in non-obese diabetic mice, a model that is prone to developing human T1D-like symptoms 563

(Mariño et al., 2017; Wen & Wong, 2017). This intervention has also been applied to lactose- 564

intolerant individuals administrated short-chain galacto-oligosaccharides that enriched lactose- 565

metabolizing commensal bacteria, which in turn improved clinical outcomes of host lactose 566

(28)

digestion and tolerance (Azcarate-Peril et al., 2017). Another conceptual study demonstrated in 567

mice showing that SCFAs generated from gut bacteria via fermenting microbiota-accessible 568

carbohydrates (MACs), i.e. dietary plant polysaccharides, reduced the fitness of C. difficile in the 569

gut (Hryckowian et al., 2018); such a dietary strategy might be appropriate in combination with 570

fecal microbiota transplant or the treatment and management of patients with complicated 571

recurrent CDI. The future holds promise to develop precise and personalized dietary 572

management using computational modelling, i.e. supervised machine learning, to identify 573

human gut microbiome functional features that are associated with clinical metadata; this type of 574

an approach has been demonstrated to aid in optimizing dietary interventions for patients with 575

Crohn’s disease, prediabetes and T2D-diabetes (Bauer & Thiele, 2018; Zeevi et al., 2015).

576

Loss of specific gut commensal bacteria is common in dysbiosis with the reduction of new 577

and often unexplored species; thus, recovery of a healthy gut microbiome is a time-consuming 578

process via dietary intervention and management. Such manipulations are important for us to 579

better understand the role of dysbiosis in clinical practice, but may not be a first-line strategy, 580

especially in patients requiring fast symptom relief from severe illness, such as cancer and 581

infectious diseases.

582

4.2. Fecal microbiome transplantation 583

Fecal microbiome transplantation (FMT) is originally designed as investigational therapy for 584

treating patients with gut dysbiosis-associated recurrent C. difficile infection (rCDI) (Britton &

585

Young, 2014). FMT is not an FDA-approved procedure and is being explored for treating 586

diseases with potentially serious health consequences, especially in the critically ill or 587

immunodeficient patient. Recent adverse events associated with FMT transfer of antibiotic- 588

resistant pathogens has prompted the FDA to restrict its use to clinical trials that have an 589

approved investigational new drug (IND) status (Kim & Gluck, 2019). Generally regarded as a 590

(29)

treatment after multiple recurrent disease episodes in CDI patients, a major reason for adopting 591

FMT is that conventional antibiotic treatment worsens or does not reverse the dysbiotic 592

microbiome (Kassam, Lee, Yuan, & Hunt, 2013). Unlike many other diseases, there is strong 593

evidence to support microbiota causation in rCDI and this justifies its use as investigational 594

microbiota therapy, especially since CDI can be rapidly fatal in these patients (Britton & Young, 595

2014; Buffie et al., 2015; Seekatz & Young, 2014).

596

FMT use has also recently been extended to the treatment of other gut dysbiosis-associated 597

diseases with mixed outcomes, including some efficacy in Crohn’s disease and ulcerative colitis 598

(Moayyedi et al., 2015; Paramsothy et al., 2017), improved cognition in cirrhosis with recurrent 599

hepatic encephalopathy (Bajaj et al., 2018, 2017), and decreased GVHD-related mortality in 600

recipients of allogeneic hematopoietic stem cell transplantation (Shono et al., 2016; Shono &

601

Van Den Brink, 2018; Taur et al., 2018). Use of immune checkpoint inhibitors for treating 602

patients with numerous cancer types may induce serious immune-associated adverse effects, 603

such as colitis in a subset of patients (Wang et al., 2018); but colitis may be abrogated by FMT 604

to restore regulatory T-cell function in cancer management (Wang et al., 2018). Several of these 605

clinical trials demonstrate rapid microbiome reconstitution following FMT and improved 606

symptoms. However, other studies demonstrate that FMT is contraindicated and should be used 607

with caution because the well reported safety concerns on the transfer of infectious agents (Kim 608

& Gluck, 2019; Zuo et al., 2018).

609

4.3. Targeted microbial therapeutics – next-generation probiotics 610

Targeted microbial therapeutics are being developed to remove or inhibit certain features of 611

the dysbiotic microbiome, i.e. antimicrobial resistance genes and infectious pathogens. As 612

illustrated in Figure 4, ecological modelling and non-modelling approaches might prove useful 613

in the discovery of effector strains during dietary interventions and FMT; those effector strains 614

(30)

would be potential probiotics for precisely reversing host disease states. Recently, such an 615

approach was used to identify Clostridium scindens as candidate probiotic to protect against C.

616

difficile infection via secondary bile acid-mediated resistance and tryptophan-derived antibiotics 617

(Buffie et al., 2015; Kang et al., 2019). Other examples include consumption of probiotic Bacillus 618

subtilis to abolish gut colonization of pathogenic Staphylococcus aureus via fengycins-trigged 619

inhibition of S. aureus quorum sensing (Piewngam et al., 2018); reduced intestinal inflammation 620

through tungstate-mediated microbiota editing that specifically inhibits activity of gut 621

Enterobacteriaceae enriched during colonic inflammation (Byndloss et al., 2017; Litvak, 622

Byndloss, & Bäumler, 2018; Zhu et al., 2018); oral antibiotic administration also sustained 623

intestinal T cell dysfunction in the host even after FMT-based microbiome reconstitution, but the 624

immune defect was spared after supplying exogenous butyrate to the host (Scott et al., 2018);

625

Bacteroides ovatus alleviated gut inflammation induced by lipopolysaccharide and dextran 626

sodium sulfate (Ihekweazu et al., 2019; Tan, Zhao, Zhang, Zhai, & Chen, 2019); Bacteroides 627

uniformis with expanded glycolytic capability exhibited pre-clinical efficacy on obesity-associated 628

metabolic and immune dysfunction (Benítez-Páez, Gómez del Pulgar, & Sanz, 2017);

629

Clostridium butyricum also demonstrated potential to attenuate acute colitis via regulating TLR2- 630

MyD88 anti-inflammatory responses (Kanai, Mikami, & Hayashi, 2015); supplementation of 631

ethanol-depleted Akkermansia muciniphila conferred protection against ethanol-associated gut 632

leakiness by increasing mucus thickness and elevating tight-junction protein expression 633

(Grander et al., 2017, 2018). Interestingly, a mixture of probiotics suppressed the growth of 634

hepatocellular carcinoma tumors by modulating the gut microbiome regulation of Th17 cell 635

differentiation and IL-17 cytokine bioavailability in the tumor microenvironment (Li et al., 2016).

636

Over the last decade, newly-identified commensal strains with probiotic potential have been 637

identified and include Bacteroides xylanisolvens, Bacteroides ovatus, Bacteroides dorei, 638

Bacteroides fragilis, Faecalibacterium prausnitzii, Eubacterium hallii, Eubacterium rectale, 639

Anaerostipes hadrus and Roseburia hominis, demonstrating efficacy against cancer, gut 640

Viittaukset

LIITTYVÄT TIEDOSTOT

The most important food source for breeding female wolverines was human- and wolf- killed moose (55%), while the diet also comprised mountain hare (13%), domestic pig (13%) and

Late gestation diet supplementation of resin acid-enriched composition increases sow colostrum immunoglobulin G content, piglet colostrum intake and improve sow gut microbiota..

This study aimed at validating a questionnaire used to inquire about food allergies in children, at researching associations between maternal diet during pregnancy and lactation

However, the following concerns are related to circulating endotoxins as markers of barrier function (Teixeira et al., 2012a): 1) Modifications in gut microbiota may

A majority (85 %) of participants were students, of whom 77 % attended university and 23 % were enrolled in vocational education. Together, they represented a wide variety of

In papers II and III, multi-level analyses were used to take into account the sampling strategy, which was performed based on school classes and pre- schools. In paper II, only a

Metabolic stress in obesity – The impact of high-fat diet and food ingredients on adipose tissue inflammation and satiety.. Kuopio: University of

The growing awareness of the importance of sensory factors in food choice, and the scientific evidence on the diet – health associations, have stimulated research into sensory