ABSTRACT
High-fat diet (HFD) is well known to impact various aspects of gut health and has been associated with many diseases and inflammation. However, the impact of HFD feeding on HIV-1 rectal transmission has not yet been well addressed. With an increasing threat of HIV-1 infection in men who have sex with men (MSM), where the rectal route is the primary mode of infection, it is imperative to understand the impact of HFD on gut microbiota and inflammation and consequently, its effect on HIV-1 rectal transmission. Here, we utilized our double humanized bone marrow, liver, thymus (dHu-BLT) mouse model to assess the impact of HFD feeding on the host’s susceptibility to HIV-1 rectal transmission. We found that feeding an HFD successfully altered the gut microbial composition within 3 weeks in the dHu-BLT mouse model. In addition, levels of inflammatory mediators, specifically IL-12p70, IP-10, ICAM-1, and fecal calprotectin, were significantly higher in HFD-fed mice compared to control mice on a regular chow diet. We also observed that significantly different inflammatory markers (IL-12p70 and ICAM-1) were negatively correlated with the number of observed ASVs, Shannon diversity, and Faith’s diversity in the HFD-fed group. Notably, when repeatedly challenged with a low dose of HIV-1 via a rectal route, mice receiving an HFD were significantly more susceptible to HIV-1 rectal infection than control mice. Together, these results underscore the impact of HFD feeding on the gut microbiota and inflammation and suggest the significance of diet-induced gut microbial dysbiosis and inflammation in promoting viral infection.
IMPORTANCE
HFD induces gut microbial dysbiosis and inflammation and has been associated with many infections and disease progression; however, its impact on HIV-1 rectal transmission is largely unknown. Given the increasing threat of HIV-1 incidence in men who have sex with men (MSM), it has become crucial to comprehend the impact of factors associated with gut health, like HFD consumption, on host susceptibility to HIV-1 rectal transmission. This is particularly important since anal intercourse remains the primary mode of HIV transmission within the MSM group. In this study, utilizing our unique mouse model, featuring both the human immune system and gut microbiota, we showed that HFD feeding led to gut microbial dysbiosis, induced inflammation, and increased HIV-1 rectal transmission. Collectively, our study highlights the significant impact of HFD on gut microbiota and inflammation and suggests an HFD consumption as a potential risk factor for promoting HIV-1 rectal susceptibility.
KEYWORDS: HIV-1 rectal transmission, HFD, MSM, inflammatory markers, gut microbial dysbiosis, double humanized BLT mice
INTRODUCTION
New HIV-1 infections have been declining over the past two decades globally; however, their incidence in the men who have sex with men (MSM) population has continued to expand (1, 2). Since the onset of the HIV epidemic, MSM have been disproportionately affected by this virus. In the United States, 68% of HIV-1 new infections diagnosed in 2020 were from MSM (3). Recent studies have revealed unique compositional characteristics of the gut microbiome in MSM compared to that of men who have sex with women (MSW) regardless of HIV infection (4–6). Such alterations in the gut microbiome have been linked with increased immune activation and inflammatory markers, as well as enhanced susceptibility to HIV-1 transmission in MSM (7–9). Within the MSM population, these microbial variations, coupled with potential injury from receptive anal intercourse, may create a microenvironment that facilitates HIV-1 rectal transmission. In addition, compared to the proximal colonic section, the most distal section of the rectum is populated with greater concentrations of HIV target cells such as activated CD4+ T cells, macrophages, and dendritic cells, further fueling the risk of HIV transmission in MSM (10). Despite these links, further studies are needed to better understand how factors associated with gut health can disrupt gut homeostasis through microbial perturbation, metabolic inflammation, and immune modulation to influence rectal HIV-1 transmission.
Diets high in fat have profound effects on the gut microbiota and intestinal barrier functions. An HFD is well known to induce gut microbial dysbiosis that leads to increased microbial translocation and metabolic inflammation (11–13). Excessive dietary fats are increasingly linked to heightened intestinal permeability through multiple mechanisms, including changes in tight junction proteins, production of barrier-disrupting bile acids, stimulation of pro-inflammatory signaling cascades and cytokines, and induction of oxidative stress and apoptosis (14–16). In addition, studies in animal models have demonstrated that HFD feeding alters the phenotype of immune cells and enhances systemic immune activation and inflammation (17–20). Furthermore, HFD feeding has been linked to accelerated disease progression, including infection of Simian immunodeficiency virus (SIV), a close relative of HIV, in rhesus macaques (20, 21). Collectively, the combination of compromised intestinal barrier functions, gut microbial dysbiosis, and elevated levels of mucosal inflammation induced by an HFD may represent a risk factor for HIV-1 rectal transmission.
In this study, we assessed the impact of HFD feeding on the host’s susceptibility to HIV-1 rectal transmission utilizing our established double humanized bone-marrow, liver, thymus (dHu-BLT) mouse model featuring both an engrafted human immune system and a human fecal microbiota (22). We fed one group of dHu-BLT mice with an HFD to induce gut microbial dysbiosis and inflammation, while other dHu-BLT mice received a regular chow diet to serve as controls. We examined the effect of HFD feeding on both the gut microbiota and systemic immune activation and inflammation. Furthermore, we investigated the impact of HFD consumption on host susceptibility to HIV-1 rectal transmission. HFD feeding quickly changed the gut microbiome profile and increased both peripheral blood levels of inflammatory biomarkers and fecal calprotectin levels compared to controls. When repeatedly challenged with a low dose of HIV-1 via the rectal route, we found that HFD feeding exacerbated HIV-1 rectal transmission. This study provides useful insight into the impact of the HFD on the gut microbiota and systemic inflammation and suggests HFD consumption as a potential risk factor for HIV-1 rectal transmission and possibly a significant contributor to the increased incidence of HIV-1 infection in MSM.
RESULTS
Human donor fecal microbiota successfully engrafted in dHu-BLT mice
Pre-treatment with a cocktail of broad-spectrum antibiotics has become a widely adopted practice for depleting the resident gut microbiota in mice prior to transplanting a donor microbiota (22–24). Treating Hu-BLT mice with ampicillin, metronidazole, neomycin, and vancomycin dramatically altered the gut microbial profiles of mice (Fig. S1), including a significant rise in the abundance of Pseudomonadota (formerly Proteobacteria). Following 14 days of antibiotic treatment, we engrafted an unbiased human donor sample (Donor mix) created by mixing an equal portion of three different healthy human donor samples (Fig. S2). We next compared the proportion of ASVs shared between the donor mix and the mouse gut microbiota before antibiotic treatment and 1 week after FMT. The average proportion of human donor mix ASVs shared with mice microbiotas was less than 2% before antibiotic treatment, while it was 33.91% after 1 week of FMT (Fig. 1). Furthermore, 23 ASVs from the donor mix were found in all dHu-BLT mice 1 week after FMT (Fig. S3; Table S10), suggesting successful engraftment of a portion of human fecal microbiota members in our double humanized mouse model.
HFD induced gut microbial dysbiosis in dHu-BLT mice
HFD is well documented to induce alterations in gut microbial composition and promote intestinal damage and inflammation (11–13, 16, 25–27). To assess the impact of an HFD on an engrafted human microbiota in our dHu-BLT mice, we first examined common metrics for alpha diversity (Observed ASVs, Shannon diversity index, and Faith’s phylogenetic diversity) and beta diversity (Bray-Curtis dissimilarity and weighted UniFrac). One-week post-fecal microbiota transplantation (FMT), no significant differences in alpha and beta diversity metrics were observed between the two sets of dHu-BLT mice under investigation (Fig. 2). However, within a week of feeding an HFD to one set of dHu-BLT mice, a noticeable shift in microbial diversity was evident (Fig. S4). After 3 weeks of HFD feeding (prior to the first HIV-1 rectal challenge), all evaluated alpha metrics were significantly different between HFD-fed mice and controls. Both Faith’s phylogenetic diversity and the number of observed ASVs were significantly lower for HFD mice compared to controls, while the Shannon diversity index was significantly higher. Also, in Bray-Curtis dissimilarity and weighted UniFrac beta diversity metrics, both diet groups clustered separately (Fig. 2).
We further investigated differences in the microbial compositions of the individual mice from each diet group 3 weeks after initiating HFD feeding. First, we measured the relative abundance of different phyla in each mouse. The majority of HFD-fed mice had a relatively higher abundance of Bacteroidota, Desulfobacterota, Pseudomonadota (formerly Proteobacteria), and Verrucomicrobiota compared to controls (Fig. 3). We then searched for differentially abundant taxa using Analysis of Composition of Microbiomes with Bias Correction (ANCOM-BC). Four phyla— Cyanobacteriota, Desulfobacterota, Bacillota (formerly Firmicutes), and Patescibacteria—were differentially abundant between dietary treatments (Fig. S5). At the family-level resolution, 35 taxa and 89 taxa at the genus-level resolution were identified as differentially abundant between groups (Fig. 4). The highest log2 fold change at the family level was observed in the increase in Acidaminococcaceae and decrease in [Eubacterium]_coprostanoligenes_group. Notably, common pathobionts such as Staphylococcaceae were only present in the HFD-fed mice, and beneficial taxa like Lactobacillaceae were observed in reduced abundance as compared to the control mice. In addition, Uncultured_Coriobacteriales, Coriobacteriales_Incertae_Sedis, Unclassified_Bacilli_RF39, Oxalobacteraceae, Saccharimonadaceae, and Selenomonadaceae were absent in the HFD group while Halomonadaceae and [Clostridium]_methylpentosum_group were absent in the controls. Similar to family level analysis, the majority of genera were reduced in abundance in HFD-fed mice as compared to controls, including unclassified_[Eubacterium]_coprostanoligenes group and [Eubacterium]_ventriosum_group and Ruminococcaceae_CAG-352 group. There were significant increases in the abundance of Lactococcus [detected only in HFD-fed mice; likely a dietary contribution (28)], Phascolarctobacterium, and Coprobacillus. Furthermore, 135 taxa with species-level resolution were found to be differentially abundant between diet groups (Table S4). Together, these data demonstrate significant alterations in the engrafted human microbiota upon HFD intervention.
HFD feeding elevated inflammatory markers and fecal calprotectin levels in dHu-BLT mice
Inflammation and immune activation at the mucosal portal of virus entry have been associated with increased susceptibility to HIV infection and virus shedding (29–31). Inflammation can compromise mucosal epithelial barrier integrity and function and recruit HIV-1 target cells, thereby creating a favorable niche for virus transmission and promoting productive infection (32, 33). To understand the role of HFD on inflammation and immune activation and, in turn, on the risk of HIV-1 rectal transmission, we measured multiple inflammatory markers and activated T-cell populations. Of the 20 different plasma inflammatory mediators measured (Table S5), 15 were detected in the majority of the samples. Levels of IL-12p70, IP-10, and ICAM-1 were significantly different between the diet groups (Fig. 5A). In addition, the level of human fecal calprotectin, an important marker for inflammation in the gastrointestinal tract, was significantly higher in HFD-fed mice versus controls (Fig. 5B). However, the population of activated T cells measured in peripheral blood was not statistically different between dietary treatments (Fig. S6). Together, these results suggest ongoing inflammation both locally and systemically in HFD-fed mice.
Inflammatory markers (IL-12p70 and ICAM-1) altered by HFD feeding were negatively correlated with alpha diversity metrics
The gut microbiota influences a variety of host physiological processes, and changes in gut microbial composition, particularly reduced alpha diversity, have been linked to markers of metabolic dysfunction (34, 35). We, therefore, sought to test for correlations of microbial diversity with inflammatory markers observed to be significantly altered by HFD feeding. Notably, all the alpha diversity indices evaluated in our study were negatively correlated with the level of IL-12p70 (Observed ASVs: P < 0.001, r = −0.62; Shannon: P < 0.05, r = −0.45; Faith’s diversity: P < 0.01, r = −0.66) and ICAM-1 (Observed ASVs: P < 0.05, r = −0.54; Shannon: P < 0.05, r = −0.41; Faith’s diversity: P < 0.05, r = −0.54) in mice-fed HFD (Fig. 6A). By contrast, only IP-10 in the control mice (Fig. 6B) was negatively correlated with the alpha diversity matrices evaluated (Observed ASVs: P < 0.05, r = −0.09; Shannon: P < 0.05, r = −0.14; Faith’s diversity: P < 0.01, r = −0.31) in our study. Collectively, these associations suggest that a reduction in microbial alpha diversity could potentially be a contributing factor to the heightened inflammation observed in HFD-fed mice.
HFD feeding increases the risk of HIV-1 rectal transmission
HFD feeding has been linked to increased host susceptibility to bacterial enteric infection (36, 37) and disease progression of SIV-infected macaques (20, 21). To evaluate the impact of HFD feeding on HIV-1 rectal infection, we rectally challenged both control chow and HFD-fed dHu-BLT mice once every 2 weeks with a repeated low dose of HIV-1. The cumulative proportion of mice that remained uninfected over four low doses of HIV-1 rectal challenges is shown in Fig. 7. All HFD-fed mice (11 out of 11) were infected after the fourth challenge (Table S9). By contrast, only 50% of the control mice (3 of 6 mice) were infected during this period (Table S9). The difference in the risk of HIV-1 rectal transmission between the two diet groups was statistically significant (P = 0.011, Mantel-Cox test). These results demonstrate that HFD feeding is associated with an increased risk of HIV-1 rectal transmission.
DISCUSSION
Several studies have shown that the risk of HIV transmission through receptive anal intercourse is higher than the risk associated with other sexual activities, such as vaginal intercourse or oral sex (38). With the rising incidence of HIV-1 infection in MSM, it is plausible that factors associated with the gastrointestinal tract may contribute to the increasing HIV-1 prevalence in this population. In the female genital tract, bacterial vaginosis and associated dysbiosis of the vaginal microbiome are considered major contributors to vaginal inflammation and increased risk of HIV-1 vaginal acquisition (39–41). However, the impact of gut microbial alteration and inflammation in HIV-1 rectal transmission has not been well established. Anatomically, compared to the vaginal mucosa, which consists of multilayered stratified squamous epithelium, rectal tissue is a single layer of columnar epithelial cells, thus making them more vulnerable to infection through mechanical trauma during sexual intercourse (38). In addition, there is growing evidence of unique compositional microbiome characteristics in MSM compared to MSW (4–6), where changes in the gut microbiome are associated with an increased risk of HIV-1 susceptibility (8). Furthermore, there is evidence of elevated expression of the HIV-1 co-receptor CCR5 in CD4+ T cells in rectosigmoid colonic biopsies from MSM versus MSW (42) and fecal bacterial communities from MSM inducing higher frequencies of CCR5+ CD4+ T cells during in vitro assays than those from MSW (43). Altogether, findings from these reports suggest potential connections among anatomical features of the lower gastrointestinal tract, gut microbial composition, and HIV-1 rectal susceptibility in MSM. Considering the increasing threat of HIV-1 incidence in MSM (1–3), where anal intercourse remains the primary mode of HIV transmission (44), it is crucial to explore the risk factors linked to gut health and their impact on HIV-1 rectal transmission.
HFD is well known to affect multiple aspects of gut health, ranging from the structural integrity of the gut to the composition of resident microbiota (11–14, 25–27, 45). Numerous diseases and inflammatory conditions have been associated with HFD consumption. In humans, diets rich in fat sources, a typical feature of Western diets, are associated with obesity, nonalcoholic fatty liver disease (NAFLD), type 2 diabetes mellitus, cardiovascular diseases, and cerebrovascular pathology, including cognitive impairment and emotional disorders (46, 47). Studies in rhesus macaques have demonstrated that a diet rich in fat accelerated SIV disease progression (20, 21). However, in terms of HIV-1, particularly in rectal transmission, the impact of an HFD and its associated microbial dysbiosis and inflammation is largely unknown. The challenge with such studies lies in the limited availability of suitable animal models where the combination of the human immune system, the human gut microbiota, and HIV-1 can be studied simultaneously with dietary interventions. Thus, the causal role of diet-induced inflammation and gut microbial dysbiosis in HIV-1 rectal transmission remains largely undefined. This study sought to close this knowledge gap by addressing the question of whether inflammation and microbial perturbation induced due to HFD feeding enhances the risk of HIV-1 rectal transmission by utilizing our established novel dHu-BLT mouse model featuring a stable human-like gut microbiome and human immune system (22).
The dHu-BLT mouse model provides a unique opportunity to study the relationships among a functional human immune system, a human-like microbiome, and HIV-1 infection. The BLT method of humanization enables robust engraftment of the human immune system within the gastrointestinal tract of BLT mice and has been extensively used for HIV rectal transmission studies (48). The human immune reconstitution of the dHu-BLT mice used in the current study was similar to that previously reported for BLT mice (49, 50). In addition, one-third of the human donor microbiota taxa were detected 1 week after FMT in the dHu-BLT mice used in the current study, suggesting the successful engraftment of a portion of the human gut microbiota as we reported previously (22, 51). To assess the impact of HFD feeding on the host’s susceptibility to HIV-1 rectal transmission, we fed one group of mice with an HFD, which is well documented to induce notable changes in gut microbial composition and inflammation. After 3 weeks of HFD feeding, all measures of alpha diversity evaluated were significantly different between the diet groups. HFD is reported to decrease common matrices for alpha diversity (28, 52), but in our study, the Shannon diversity index was higher in the HFD group compared to the controls. The difference between our observation versus those of others could be due to the mouse strain and model used in our study. A similar effect of increased Shannon diversity in different mouse strains fed with an HFD has been previously reported by others (27, 45). Another study has also reported an increase in the Shannon diversity index in mice fed with an HFD over time (26), which is consistent with our study in which the Shannon diversity index increased considerably during the time between the human microbiota transplant and 3 weeks of HFD feeding (Fig. 2A and C). We further investigated the differentially abundant taxa between the diet treatments prior to starting the HIV-1 rectal challenge. At the family level, Desulfobacterota and Bacillota (formerly Firmicutes) were differentially abundant in ANCOM-BC analysis (Fig. S5). Desulfobacterota, a sulfate-reducing species, is associated with increased immune responses, gut inflammation, and higher fecal IgA concentrations (53, 54), while the reduction of Bacillota (formerly Firmicutes) has been linked to increased systemic inflammation (55) and human inflammatory diseases like inflammatory bowel disease (IBD) (56). In addition, common pathobionts like Staphylococcaceae were only detected in mice fed an HFD prior to HIV-1 rectal challenge. Pathobionts such as Staphylococcaceae are known to prosper in an inflamed gut and support inflammation (57). By contrast, beneficial taxa such as Lactobacillaceae, which have been suggested to prevent inflammation (58, 59), were decreased in abundance during HFD feeding compared to the controls. Furthermore, some beneficial bacterial species suggested to play a protective role against inflammation such as Lactobacillus johnsonii (60), Alistipes finegoldii (61), Bacteroides xylanisolvens (62), Bacteroides uniformis (63), and Oxalobacter formigenes (64) were found in reduced abundance in the HFD group while opportunistic like Bacteroides nordii (65) were in increased abundance (Table S4) prior to HIV-1 rectal challenge. These bacteria are known to modulate the host physiology via their metabolites or by influencing inflammatory signaling pathways. For instance, Lactobacillus johnsonii L531 inhibits the TLR4/NF-κB/NLRP3 inflammasome signaling pathway to alleviate S. typhimurium-induced tight junction damage (60). Alistipes finegoldii mediates protection from dextran sodium sulfate (DSS)-induced colitis by restoring intestinal barrier function through the enhanced colonic expression of intestinal IL-22 and Reg3γ (61). Likewise, a recent investigation has demonstrated the ability of Bacteroides xylanisolvens AY11-1 to generate oligosaccharides and beneficial short-chain fatty acids through the breakdown of alginate and its derivatives (62). Moreover, this study also revealed that B. xylanisolvens attenuated mucosal damage in mice with DSS colitis (62). Consequently, the increase in these potentially pro-inflammatory bacteria and a concomitant loss of beneficial organisms may have contributed to the inflammation detected in our HFD-fed mice due to altered production of gut microbial metabolites or their influence on the host gene expression.
Consumption of an HFD has been linked to increased inflammatory markers and immune modulation (12, 17–21, 25, 46). In addition, several studies have found that elevated levels of inflammatory markers and immune activation are associated with increased HIV-1 susceptibility and infectiousness (9, 29, 30, 66, 67). In our study, we observed that IL-12p70, IP-10, and ICAM-1 were significantly higher in the HFD-fed mice compared to controls (Fig. 5A). IL-12p70 is an important immunoregulatory cytokine, primarily produced by professional antigen-presenting cells (APC) that induce Th1 cells differentiation as well as activation of natural killer cells (68). The microbial dysbiosis and immune activation prompted by our HFD could be a key stimulant of elevated IL12p70 in the HFD-fed mice as microbial products and activated T cells and APC interaction are known to directly induce IL-12 secretion (69). Furthermore, IL12p70 has been linked to tissue injury and is a major factor in the pathogenesis of Crohn’s disease (70, 71). IP-10 was another inflammatory modulator found to be significantly different between the diet groups. Like IL12p70, higher expression of IP-10 has been linked to chronic intestinal inflammation and tissue damage (72). This chemokine mediates apoptosis and facilitates bacterial and viral infection, including HIV-1 (73, 74). In addition, several studies have correlated elevated levels of IP-10 with an increased HIV-1 acquisition risk and disease pathogenesis (29, 66, 67, 75, 76). Interestingly, in our study, the HFD-fed mice were the most susceptible to low-dose HIV-1 rectal challenge and also had significantly higher levels of IP-10 compared to controls. We also observed significantly higher expression of ICAM-1, a master regulator of cellular responses in inflammation, in our HFD-fed mice. Similar results of elevated ICAM-1 expression in mice fed with an HFD have also been reported in other studies (25, 77). Like IP-10, elevated levels of ICAM have been associated with enhancing HIV-1 disease progression (78, 79). In addition, the levels of detection in plasma were out of range (<OOR) for the pro-inflammatory cytokine IL-6 in chow-fed dHu-BLT mice; however, IL-6 was detected in some of the HFD-fed mice, suggesting an ongoing inflammatory response in these animals (Table S5). Also, the cytokines IL-10 and IL-13 were detected in the majority of HFD-fed mice compared to control animals, which could be the result of a suppressive immune response initiated due to elevated inflammation from the HFD feeding. Calprotectin accounts for the majority of the cytosolic proteins in neutrophils, and its presence in feces indicates neutrophilic infiltration into the gut lumen due to an inflammatory process or infection (80). Compared to chow-fed mice, the level of human fecal calprotectin was significantly higher in the HFD-fed mice (Fig. 5B), suggesting notable ongoing inflammatory activity during HFD feeding. In recent studies, it has been demonstrated that HFD feeding alters the phenotype of immune cells. Specifically, the studies revealed an increase in the population of activated T cells, polarization of macrophages, and proliferation and activation of invasive Ly6Chigh monocytes in HFD-fed mice (17, 18). However, in our current study, we did not observe any statistically significant difference in the T-cell activation between diet treatments prior to the HIV-1 rectal challenge (Fig. S6). This result could be due to the duration of HFD feeding, which is considerably shorter than what has been published by others (17, 18).
Several studies have reported an association between altered microbiomes and inflammation (81, 82). Dysregulated microbiomes can impair epithelial barrier function and gut mucosal homeostasis (83, 84). In addition, the translocation of microbial products as a result of compromised intestinal integrity has the potential to drive systemic inflammation and disease progression (83, 84). Here, we further investigated the correlation between microbiome diversity indices with inflammatory markers that were significantly different between diet treatments. We observed that plasma levels of the inflammatory mediators IL-12p70 and ICAM-1 were negatively correlated with all the alpha diversity indices evaluated for HFD-fed mice (Fig. 5A). Species diversity plays an important role in the maintenance of intestinal homeostasis and increases in diversity have been generally associated with health (85) while reductions in diversity are often viewed as potential initiators of inflammatory reactions (86). Increased levels of inflammatory mediators observed in our HFD mice could have resulted from the loss of potentially beneficial bacteria responsible for maintaining the intestinal ecosystem or may have been due to the increased translocation of microbial products due to impaired barrier function.
Finally, our low-dose HIV-1 rectal challenge study showed that mice fed an HFD are more susceptible to an HIV-1 rectal challenge (Fig. 7). By the fourth rectal challenge, all the HFD-fed dHu-BLT mice were infected compared to only 50% of those fed a control chow diet. This result demonstrates that HFD feeding enhances HIV-1 rectal transmission. In our study, it is plausible that HFD feeding initiated some degree of pathophysiological and immunological changes to the gastrointestinal tract of dHu-BLT mice. In turn, these effects may have increased local inflammation and mucosal membrane permeability, thus leading to microbial translocation and systemic inflammation. In addition, the number of activated CD4+ T cells at the site of inflammation may have also been increased, thereby providing additional target cells to facilitate HIV-1 infection. Altogether, these various scenarios, coupled with diet-induced microbial perturbations, may have created a favorable microenvironment for successful HIV-1 transmission.
Our study has several important implications regarding the impact of HFD feeding on susceptibility to HIV-1 rectal transmission. In our current study, we extensively characterized the effects of consuming an HFD on gut microbial composition and inflammatory mediators prior to the HIV-1 rectal challenge and assessed its impact on the host’s susceptibility to HIV-1 rectal transmission. Such studies in human subjects are difficult to perform due to ethical constraints and many confounding factors such as human behavior, socioeconomic factors, variation of dietary intake, medication, and host genetics. However, in the current study, we circumvented those issues and assessed the effect of an HFD on HIV-1 rectal susceptibility in a strictly controlled environment. In addition, this study not only recapitulated the profound effects of an HFD on the gut microbiota and inflammation but also linked diet-induced gut microbial dysbiosis and inflammation in the context of viral infection. Moreover, the HFD used in our study contained high levels of fat and sucrose but had low fiber content. These features are typical of the Western diet, which may be a factor contributing to HIV-1 infection among MSM from developed nations. This diet formulation is particularly relevant in light of the increased incidence of HIV-1 among MSM from developed countries (western and central Europe and North America) (1). Furthermore, our comprehensive characterization of inflammatory mediators and T-cell activation in plasma adds additional valuable information to the current understanding of systemic immune activation and inflammation due to HFD feeding.
Although our work provides useful insight into the influence of HFD feeding on the human gut microbiota, inflammation, and HIV-1 rectal transmission, it is crucial to acknowledge some inherent limitations associated with our animal model. These limitations include the challenge of achieving the complete engraftment of the human donor microbiota in the murine gut and the anatomical dissimilarities of mice compared to the human lower gastrointestinal tract. Moreover, our assessment of gut inflammation over time relied solely on fecal calprotectin levels. Yet, the characterization of inflammatory mediators and activated immune cells at the local gut mucosa prior to the HIV-1 rectal challenge could have provided additional insight into the ongoing pathophysiological conditions at the site of transmission. Despite these limitations, our study demonstrates that consuming an HFD alters the gut microbiota, enhances inflammation, and increases HIV-1 rectal transmission in well-controlled animal model. In a broader context, this study highlights the ramifications of an altered gut microbiota and inflammation due to an HFD on the establishment of a viral infection. Importantly, our work supports further study on using either an FMT or dietary intervention to correct gut microbial dysbiosis as a prevention strategy to limit HIV-1 rectal transmission.
MATERIALS AND METHODS
Generation of hu-BLT mice
Hu-BLT mice were generated as we previously reported (49, 87). Briefly, NSG (NOD.Cg-PrkdcscidIl2rgtm1Wjl/SzJ; Cat# 005557; Jackson Laboratory, Bar Harbor, ME) female mice of 6–8 weeks were housed at UNL animal facility under specific-pathogen-free conditions. On the day of surgery, mice were subjected to sublethal irradiation at the dose of 12 cGy per gram of body weight using the RS200 X-ray irradiator (RAD Source Technologies, Inc., GA). Mice were then surgically implanted with a piece of human fetal thymic tissue fragments sandwiched between two pieces of human fetal liver tissue within the left renal capsule, followed by intravenous injection of 1.5–5 × 105 CD34+ hematopoietic stem cells isolated from human fetal liver within 6 hours of surgery. Human fetal thymus and liver tissue were procured from Advanced Bioscience Resources (Alameda, CA). Ten weeks after surgery, human immune reconstitution was measured using flow cytometry as described below.
Antibiotic treatment and human fecal microbiota transplant
Hu-BLT mice with similar human immune reconstitution were used to generate dHu-BLT mice following our previously reported method (22). Briefly, a broad-spectrum antibiotic cocktail of metronidazole (1 g/L), neomycin (1 g/L), vancomycin (0.5 g/L), and ampicillin (1 g/L) along with grape-flavored Kool-Aid was freshly prepared daily and given to hu-BLT mice for 14 days. During antibiotic treatment, cages were changed daily to limit re-inoculation of pre-existing bacteria in the mice due to coprophagic behavior. After 14 days of broad-spectrum antibiotic treatment, mice were given non-acidified autoclaved deionized water for 24 hours, followed by two oral gavage of 200 µL of human fecal microbiota mixture (Donor mix) at 24-hour intervals using an animal feeding needle (Cat# 9928B; Cadence science). The human fecal microbiota mixture (Donor mix) was prepared by mixing an equal portion of each donor sample from three different healthy human donors (two males and one female, aged 26–29 years; OpenBiome, Massachusetts, US) in an anaerobic chamber.
Human immune reconstitution and immunophenotyping
Human immune reconstitution and immunophenotypes of hu-BLT and dHu-BLT mice were measured in peripheral blood using a fluorescence-activated cell sorter (FACS) Aria II flow cytometer (BD Biosciences, San Jose, CA) and antibodies against mCD45-APC, hCD45-FITC, hCD19-PE/Cy5, hCD3-PE, hCD4-Alexa 700, hCD8a-APC-Cy7, hCD69-BV 785, hCD38-BV 421, and hHLA-DR-PE/Cy7 (catalog numbers 103111, 304006, 302210, 300408, 300526, 301016, 310932, 356618, and 307616 respectively; Biolegend, San Diego, CA). Raw data were analyzed with FlowJo (version 10.8.1; FlowJo LLC, Ashland, OR). All hu-BLT mice used in this study had >50% of human CD45+ reconstitution in the mouse peripheral blood lymphocytes analyzed (Table S8).
Experimental approach
The dHu-BLT mice generated as described above were divided into two groups. Both groups of mice were initially fed regular mouse chow (Teklad 2916; Inotiv, Madison, WI). One-week post-human fecal microbiota transplant, one group of mice (HFD group, n = 11) was switched to a diet containing 45% kcal from fat (D12451i; Research Diets, New Brunswick, NJ) to induce gut microbial dysbiosis while another group (Control chow, n = 6) continued receiving regular mouse chow. Three weeks after the HFD feeding was initiated, all dHu-BLT mice were challenged intrarectally with a low dose of transmitted/founder HIV-1 SUMA (1 × 104 TCID50 in 20 μL) once every 2 weeks under isoflurane anesthesia as reported previously (88). Infection was evaluated 2 weeks after each challenge via quantitative reverse transcription PCR (qRT-PCR). Once all HFD-fed mice were infected, uninfected mice in the control group were challenged with HIV-1 ramp-up doses (10-fold increase of TCID50 in every challenge) every 2 weeks until all mice were infected. Fecal samples and peripheral blood were collected at multiple time points to evaluate gut microbiome profiles and systemic immune activation, respectively (Fig. 8).
Quantification of plasma viral loads
Plasma viral loads were measured using qRT-PCR as described previously (89) 2 weeks after each HIV-1 rectal challenge. Briefly, a QIAamp Viral RNA mini kit (Qiagen, Cat# 52906) was used to extract viral RNA from the mouse plasma and HIV-1 analytical standards in the AcroMetrix HIV-1 panel (ThermoFisher Scientific, Cat# 950470) following the manufacturer’s protocol. Viral RNA was then quantified using a C1000 Thermal Cycler and the CFX96 Real-time system (Bio-Rad). The reaction mixture consisted of 5 µL extracted viral RNA, TaqMan Fast Virus 1-Step master mix (ThermoFisher Scientific, Cat# 4444432), forward primer (5′-GCCTCAATAAAGCTTGCCTTGA-3′), reverse primer (5’- GGGCGCCACTGCTAGAGA-3), probe (/56-FAM/CCAGAGTCA/ZEN/CACAACAGACGGGCACA/3IABkFQ/), and nuclease-free water to achieve a total volume of 20 µL.
Fecal sample collection and DNA extraction
Freshly collected mouse fecal samples were stored in 1.5 mL autoclaved Eppendorf tubes at −80°C until DNA extraction. Fecal DNA was extracted following the method described by Martinez et al. (90). Briefly, fecal samples were washed three times with ice-cold phosphate-buffered saline (Cat# SH30256.02, Hyclone) followed by physical lysis of cells using bead beater (Mini-Beadbeater-16, Biospec) in the presence of lysis buffer, 0.1 mm zirconia/silica beads (Biospecs), 10% SDS, Proteinase K (15 mg/mL, MC500B Promega), and phenol:chloroform:isoamyl alcohol (25:24:1). The upper phase was extracted twice with phenol:chloroform:isoamyl alcohol (25:24:1) followed by further purification with chloroform:isoamyl alcohol (24:1) two times. Standard ethanol precipitation was performed to recover DNA pellets, which were resuspended in 100 µL of Tris-HCL buffer (10 mM, pH 8.0). Fecal DNA from the human donor sample used in this study was similarly extracted and sequenced separately.
16S rRNA bacterial gene sequencing
V3-V4 16S rRNA amplicon gene sequencing was performed at the Genomics Core Facility of the University of Nebraska Medical Center using Illumina MiSeq utilizing the 600 cycle v3 MiSeq kit. The following primer sequences were used for the 16S library prep protocol: Forward Primer = 5′TCGTCGGCAGCGTCAGATGTGTATAAGAGACAGCCTACGGGNGGCWGCAG
Reverse Primer = 5′GTCTCGTGGGCTCGGAGATGTGTATAAGAGACAGGACTACHVGGGTATCTAATCC.
16S rRNA bacterial gene sequence analysis
Demultiplexed paired-end fastq files received from the sequencing facility were processed at the University of Nebraska Holland Computer Center Crane cluster following the DADA2 (v1.22) pipeline (91). A total of 6,787,923 reads from mouse fecal samples and 415,357 reads from human donor samples were recovered after processing in DADA2. Self-trained naïve Bayes classifier based on SILVA SSU Ref NR 99 138.1 (92) for the V3-V4 region was prepared using RESCRIPt (93) and used for taxonomic classification in QIIME2 (94). New names for the rank of bacterial phylum proposed by the International Committee on Systematics of Prokaryotes (ICSP) were adopted (95, 96). A phylogenetic tree was generated by FastTree analysis (97) using an MAFFT alignment (98) in QIIME2. Unassigned features at the phylum level, features assigned to mitochondria and chloroplast, and features only present in a single sample were removed from the analysis. A total of 6,618,547 reads and 2,082 Amplicon Sequence Variants (ASVs) were obtained after filtration for the mouse samples and used for further microbiome analysis (Fig. S7). Samples were rarified to 90% of the minimum library size without replacement for downstream analysis using different R packages in R (version 4.2.1). Differential abundance analysis was performed using ANCOMBC (v1.6.2) on the non-rarefied data (99). Taxa were considered differentially abundant if the adjusted P-value was <0.05 (False Discovery Rate, FDR). To evaluate the proportion of human donor fecal microbiota engraftment in the dHu-BLT mice, the DADA2 output of mouse and human donor samples were combined and processed similarly as described above for taxonomic assignment and filtration. A total of 7,037,798 reads and 2,310 ASVs were obtained from combined mouse and human donor samples, which were rarified to 90% of the minimum library size without replacement, and shared ASVs were then determined.
Fecal calprotectin measurement
Human fecal calprotectin (S100A8/A9) was measured from dHu-BLT mouse fecal samples in duplicate via ELISA (DY8226-05, DY008B; R&D systems, MN). Fecal samples were collected in 1.5 mL tubes and diluted with 50× of extraction buffer (0.1 M Tris, 0.15 M NaCl, 1.0 M urea, 10 mM CaCl2, 0.1 M citric acid monohydrate, 5 g/L BSA (pH 8.0)). Samples were vigorously vortexed using a vortex mixer to dissolve the fecal pellets in the extraction buffer. Homogenates were transferred into a new tube and further centrifuged for 20 minutes at 10,000 × g at 4°C. The supernatant was then used to measure the calprotectin level as per the manufacturer’s instructions.
Plasma cytokine profiles
To evaluate systemic inflammation, plasma samples were assayed in duplicate using an Inflammation 20-Plex Human ProcartaPlex Panel (Cat# EPX200-12185-901, ThermoFisher Scientific, Waltham, MA). Inflammatory mediators analyzed included the following: Cytokines: granulocyte-macrophage colony-stimulating factor (GM-CSF), interferon (IFN)-α, IFN-γ, interleukin (IL)−1α, IL-1β, IL-4, IL-6, IL-8, IL-10, IL- 12p70, IL-13, IL-17A, and tumor necrosis factor (TNF) α. Chemokines: interferon γ-inducible protein-10 (IP-10)/CXCL10, monocyte chemoattractant protein-1 (MCP-1)/CCL2, macrophage inflammatory protein (MIP)-1 alpha (MIP-1α)/CCL3, and MIP-1 beta (MIP-1β)/CCL4. Cell adhesion and inflammatory response proteins: intercellular adhesion molecule (ICAM)−1, CD62E (E-selectin), and CD62P (P-selectin). Samples were assayed on a Luminex MAGPIX instrument (Luminex Corporation, Austin, TX) according to the manufacturer’s protocol. Analytes out of range (OOR) in more than one sample in any dietary treatment were excluded from the statistical comparison.
Statistical analysis
Statistical comparison was performed using Welch’s t-test if all data passed the Shapiro-Wilk test of normality or Wilcoxon rank sums test if data from at least one group failed the normality test. In the case of beta diversity, both the Permutational Multivariate Analysis of Variance (PERMANOVA) and Permutational Analysis of Multivariate Dispersion (PERMDISP) were used to assess statistical differences between groups with 999 randomizations. All analyses were performed in R (version 4.2.1) except for the Kaplan-Meier analysis, which was performed with GraphPad Prism 5. A value of P < 0.05 was considered statistically significantly different.
ACKNOWLEDGMENTS
We thank Jianshiu Zhang, Tao Wang, Yilun Chang, and Rachel Kubik for their assistance in generating hu-BLT mice, and lab members Subhra Mandal and Miaoyun Zhao for their help and support during this study.
We also thank Kristin Beede from Dr. Ramer-Tait’s lab for her assistance with cytokine measurements, and Dirk Anderson from the Nebraska Center for Biotechnology for his assistance with flow cytometry. We also acknowledge the University of Nebraska DNA Sequencing Core, which receives partial support from the National Institute for General Medical Science (NIGMS) INBRE P20GM103427-19 grant as well as The Fred & Pamela Buffett Cancer Center Support Grant P30 CA036727.
We also thank the Institutional Animal Care Program staff at the University of Nebraska-Lincoln for their assistance with animal care.
This study is supported by the National Institutes of Health (NIH) R21 AI143405-01 to Q Li. The funders had no role in the study design, data collection and analysis, manuscript preparation, or publication decision.
S.C.L. and Q.L. designed the experiments and wrote the manuscript. S.C.L. performed the experiment and analyzed the data. A.R.T. provided input on experimental design and manuscript preparation.
Contributor Information
Qingsheng Li, Email: qli@unl.edu.
John F. Rawls, Duke University School of Medicine, Durham, North Carolina, USA
ETHICS APPROVAL
All animal procedures described in this study were conducted following the protocols approved by the Institutional Animal Care and Research Committee (IACUC) at the University of Nebraska-Lincoln (UNL). In addition, the Scientific Research Oversight Committee (SROC) at UNL has also approved the use of human embryonic stem cells and fetal tissues, which are procured from the Advanced Bioscience Resources for humanized mice studies (SROC#2016–1–002).
DATA AVAILABILITY
The data sets generated during the current study are available in the NCBI Sequence Read Archive (SRA) repository with accession number PRJNA939259. Scripts related to the current study are freely available from the corresponding author upon request.
SUPPLEMENTAL MATERIAL
The following material is available online at https://doi.org/10.1128/msystems.01322-23.
ASM does not own the copyrights to Supplemental Material that may be linked to, or accessed through, an article. The authors have granted ASM a non-exclusive, world-wide license to publish the Supplemental Material files. Please contact the corresponding author directly for reuse.
REFERENCES
- 1. UNAIDS . 2022. IN DANGER: UNAIDS global AIDS update 2022. Available from: https://www.unaids.org/en/resources/documents/2022/in-danger-global-aids-update
- 2. Beyrer C, Baral SD, van Griensven F, Goodreau SM, Chariyalertsak S, Wirtz AL, Brookmeyer R. 2012. Global epidemiology of HIV infection in men who have sex with men. Lancet 380:367–377. doi: 10.1016/S0140-6736(12)60821-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Prevention CfDCa . 2020. HIV surveillance report
- 4. Noguera-Julian M, Rocafort M, Guillén Y, Rivera J, Casadellà M, Nowak P, Hildebrand F, Zeller G, Parera M, Bellido R, et al. 2016. Gut microbiota linked to sexual preference and HIV infection. EBioMedicine 5:135–146. doi: 10.1016/j.ebiom.2016.01.032 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Armstrong AJS, Shaffer M, Nusbacher NM, Griesmer C, Fiorillo S, Schneider JM, Preston Neff C, Li SX, Fontenot AP, Campbell T, Palmer BE, Lozupone CA. 2018. An exploration of prevotella-rich microbiomes in HIV and men who have sex with men. Microbiome 6:198. doi: 10.1186/s40168-018-0580-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Tuddenham S, Koay WL, Sears C. 2020. HIV, sexual orientation, and gut microbiome interactions. Dig Dis Sci 65:800–817. doi: 10.1007/s10620-020-06110-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Li SX, Sen S, Schneider JM, Xiong K-N, Nusbacher NM, Moreno-Huizar N, Shaffer M, Armstrong AJS, Severs E, Kuhn K, Neff CP, McCarter M, Campbell T, Lozupone CA, Palmer BE, Silvestri G. 2019. Gut microbiota from high-risk men who have sex with men drive immune activation in gnotobiotic mice and in vitro HIV infection. PLoS Pathog 15:e1007611. doi: 10.1371/journal.ppat.1007611 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Chen Y, Lin H, Cole M, Morris A, Martinson J, Mckay H, Mimiaga M, Margolick J, Fitch A, Methe B, Srinivas VR, Peddada S, Rinaldo CR. 2021. Signature changes in gut microbiome are associated with increased susceptibility to HIV-1 infection in MSM. Microbiome 9:237. doi: 10.1186/s40168-021-01168-w [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Fulcher JA, Li F, Tobin NH, Zabih S, Elliott J, Clark JL, D’Aquila R, Mustanski B, Kipke MD, Shoptaw S, Gorbach PM, Aldrovandi GM. 2022. Gut dysbiosis and inflammatory blood markers precede HIV with limited changes after early seroconversion. EBioMedicine 84:104286. doi: 10.1016/j.ebiom.2022.104286 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. McElrath MJ, Smythe K, Randolph-Habecker J, Melton KR, Goodpaster TA, Hughes SM, Mack M, Sato A, Diaz G, Steinbach G, Novak RM, Curlin ME, Lord JD, Maenza J, Duerr A, Frahm N, Hladik F, Network NHVT. 2013. Comprehensive assessment of HIV target cells in the distal human gut suggests increasing HIV susceptibility toward the anus. J Acquir Immune Defic Syndr 63:263–271. doi: 10.1097/QAI.0b013e3182898392 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Hildebrandt MA, Hoffmann C, Sherrill-Mix SA, Keilbaugh SA, Hamady M, Chen YY, Knight R, Ahima RS, Bushman F, Wu GD. 2009. High-fat diet determines the composition of the murine gut microbiome independently of obesity. Gastroenterology 137:1716–24. doi: 10.1053/j.gastro.2009.08.042 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Kim KA, Gu W, Lee IA, Joh EH, Kim DH. 2012. High fat diet-induced gut microbiota exacerbates inflammation and obesity in mice via the TLR4 signaling pathway. PLoS One 7:e47713. doi: 10.1371/journal.pone.0047713 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. He C, Cheng D, Peng C, Li Y, Zhu Y, Lu N. 2018. High-fat diet induces dysbiosis of gastric microbiota prior to gut microbiota in association with metabolic disorders in mice. Front Microbiol 9:639. doi: 10.3389/fmicb.2018.00639 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Rohr MW, Narasimhulu CA, Rudeski-Rohr TA, Parthasarathy S. 2020. Negative effects of a high-fat diet on intestinal permeability: a review. Adv Nutr 11:77–91. doi: 10.1093/advances/nmz061 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Mouries J, Brescia P, Silvestri A, Spadoni I, Sorribas M, Wiest R, Mileti E, Galbiati M, Invernizzi P, Adorini L, Penna G, Rescigno M. 2019. Microbiota-driven gut vascular barrier disruption is a prerequisite for non-alcoholic steatohepatitis development. J Hepatol 71:1216–1228. doi: 10.1016/j.jhep.2019.08.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Hamilton MK, Boudry G, Lemay DG, Raybould HE. 2015. Changes in intestinal barrier function and gut microbiota in high-fat diet-fed rats are dynamic and region dependent. Am J Physiol Gastrointest Liver Physiol 308:G840–51. doi: 10.1152/ajpgi.00029.2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Kiran S, Kumar V, Murphy EA, Enos RT, Singh UP. 2021. High fat diet-induced CD8+ T cells in adipose tissue mediate macrophages to sustain low-grade chronic inflammation. Front Immunol 12:680944. doi: 10.3389/fimmu.2021.680944 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Boroumand P, Prescott DC, Mukherjee T, Bilan PJ, Wong M, Shen J, Tattoli I, Zhou Y, Li A, Sivasubramaniyam T, Shi N, Zhu LY, Liu Z, Robbins C, Philpott DJ, Girardin SE, Klip A. 2022. Bone marrow adipocytes drive the development of tissue invasive Ly6Chigh monocytes during obesity. Elife 11:e65553. doi: 10.7554/eLife.65553 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Daharsh L, Ramer-Tait AE, Li Q. 2020. Alterations in the gut microbiome by HIV-1 infection or a high-fat diet associates with systemic immune activation and inflammation in double humanized-BLT mice. bioRxiv. doi: 10.1101/2020.03.16.993956 [DOI] [Google Scholar]
- 20. He T, Xu C, Krampe N, Dillon SM, Sette P, Falwell E, Haret-Richter GS, Butterfield T, Dunsmore TL, McFadden WM, Martin KJ, Policicchio BB, Raehtz KD, Penn EP, Tracy RP, Ribeiro RM, Frank DN, Wilson CC, Landay AL, Apetrei C, Pandrea I. 2019. High-fat diet exacerbates SIV pathogenesis and accelerates disease progression. J Clin Invest 129:5474–5488. doi: 10.1172/JCI121208 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Mansfield KG, Carville A, Wachtman L, Goldin BR, Yearley J, Li W, Woods M, Gualtieri L, Shannon R, Wanke C. 2007. A diet high in saturated fat and cholesterol accelerates simian immunodeficiency virus disease progression. J Infect Dis 196:1202–1210. doi: 10.1086/521680 [DOI] [PubMed] [Google Scholar]
- 22. Daharsh L, Zhang J, Ramer-Tait A, Li Q. 2019. A double humanized BLT-mice model featuring a stable human-like gut microbiome and human immune system. J Vis Exp. doi: 10.3791/59773 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Tan J, Gong J, Liu F, Li B, Li Z, You J, He J, Wu S. 2022. Evaluation of an antibiotic cocktail for fecal microbiota transplantation in mouse. Front Nutr 9:918098. doi: 10.3389/fnut.2022.918098 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Bokoliya SC, Dorsett Y, Panier H, Zhou Y. 2021. Procedures for fecal microbiota transplantation in murine microbiome studies. Front Cell Infect Microbiol 11:711055. doi: 10.3389/fcimb.2021.711055 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Chan J, Telang R, Kociszewska D, Thorne PR, Vlajkovic SM. 2022. A high-fat diet induces low-grade cochlear inflammation in CD-1 mice. IJMS 23:5179. doi: 10.3390/ijms23095179 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Tang W, Pan L, Cheng J, Wang X, Zheng L, Wang S, Zhou Y, Wang H. 2022. High-fat-diet-induced gut microbiome changes in mice. Stress Brain 2:17–30. doi: 10.26599/SAB.2022.9060012 [DOI] [Google Scholar]
- 27. Xiao L, Sonne SB, Feng Q, Chen N, Xia Z, Li X, Fang Z, Zhang D, Fjære E, Midtbø LK, et al. 2017. High-fat feeding rather than obesity drives Taxonomical and functional changes in the gut microbiota in mice. Microbiome 5:43. doi: 10.1186/s40168-017-0258-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Bisanz JE, Upadhyay V, Turnbaugh JA, Ly K, Turnbaugh PJ. 2019. Meta-analysis reveals reproducible gut microbiome alterations in response to a high-fat diet. Cell Host & Microbe 26:265–272. doi: 10.1016/j.chom.2019.06.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Liebenberg LJP, Masson L, Arnold KB, Mckinnon LR, Werner L, Proctor E, Archary D, Mansoor LE, Lauffenburger DA, Abdool Karim Q, Abdool Karim SS, Passmore J-AS. 2017. Genital-systemic chemokine gradients and the risk of HIV acquisition in women. J Acquir Immune Defic Syndr 74:318–325. doi: 10.1097/QAI.0000000000001218 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Masson L, Passmore J-AS, Liebenberg LJ, Werner L, Baxter C, Arnold KB, Williamson C, Little F, Mansoor LE, Naranbhai V, Lauffenburger DA, Ronacher K, Walzl G, Garrett NJ, Williams BL, Couto-Rodriguez M, Hornig M, Lipkin WI, Grobler A, Abdool Karim Q, Abdool Karim SS. 2015. Genital inflammation and the risk of HIV acquisition in women. Clin Infect Dis 61:260–269. doi: 10.1093/cid/civ298 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Jaspan HB, Liebenberg L, Hanekom W, Burgers W, Coetzee D, Williamson AL, Little F, Myer L, Coombs RW, Sodora D, Passmore JA. 2011. Immune activation in the female genital tract during HIV infection predicts mucosal CD4 depletion and HIV shedding. J Infect Dis 204:1550–1556. doi: 10.1093/infdis/jir591 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Arnold KB, Burgener A, Birse K, Romas L, Dunphy LJ, Shahabi K, Abou M, Westmacott GR, McCorrister S, Kwatampora J, Nyanga B, Kimani J, Masson L, Liebenberg LJ, Abdool Karim SS, Passmore J-AS, Lauffenburger DA, Kaul R, McKinnon LR. 2016. Increased levels of inflammatory cytokines in the female reproductive tract are associated with altered expression of proteases, mucosal barrier proteins, and an influx of HIV-susceptible target cells. Mucosal Immunol 9:194–205. doi: 10.1038/mi.2015.51 [DOI] [PubMed] [Google Scholar]
- 33. Passmore J-AS, Jaspan HB, Masson L. 2016. Genital inflammation, immune activation and risk of sexual HIV acquisition. Curr Opin HIV AIDS 11:156–162. doi: 10.1097/COH.0000000000000232 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Hijazi K, Morrison RW, Mukhopadhya I, Martin B, Gemmell M, Shaw S, Santoro F. 2020. Oral bacterial diversity is inversely correlated with mucosal inflammation. Oral Dis 26:1566–1575. doi: 10.1111/odi.13420 [DOI] [PubMed] [Google Scholar]
- 35. Zouiouich S, Loftfield E, Huybrechts I, Viallon V, Louca P, Vogtmann E, Wells PM, Steves CJ, Herzig K-H, Menni C, Jarvelin M-R, Sinha R, Gunter MJ. 2021. Markers of metabolic health and gut microbiome diversity: findings from two population-based cohort studies. Diabetologia 64:1749–1759. doi: 10.1007/s00125-021-05464-w [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Las Heras V, Clooney AG, Ryan FJ, Cabrera-Rubio R, Casey PG, Hueston CM, Pinheiro J, Rudkin JK, Melgar S, Cotter PD, Hill C, Gahan CGM. 2019. Short-term consumption of a high-fat diet increases host susceptibility to Listeria monocytogenes infection. Microbiome 7:7. doi: 10.1186/s40168-019-0621-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Agus A, Denizot J, Thévenot J, Martinez-Medina M, Massier S, Sauvanet P, Bernalier-Donadille A, Denis S, Hofman P, Bonnet R, Billard E, Barnich N. 2016. Western diet induces a shift in microbiota composition enhancing susceptibility to adherent-invasive E. coli infection and intestinal inflammation. Sci Rep 6:19032. doi: 10.1038/srep19032 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Caputo V, Libera M, Sisti S, Giuliani B, Diotti RA, Criscuolo E. 2023. The initial interplay between HIV and mucosal innate immunity. Front Immunol 14:1104423. doi: 10.3389/fimmu.2023.1104423 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Gosmann C, Anahtar MN, Handley SA, Farcasanu M, Abu-Ali G, Bowman BA, Padavattan N, Desai C, Droit L, Moodley A, Dong M, Chen Y, Ismail N, Ndung’u T, Ghebremichael MS, Wesemann DR, Mitchell C, Dong KL, Huttenhower C, Walker BD, Virgin HW, Kwon DS. 2017. Lactobacillus-deficient cervicovaginal bacterial communities are associated with increased HIV acquisition in young South African women. Immunity 46:29–37. doi: 10.1016/j.immuni.2016.12.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Taha TE, Hoover DR, Dallabetta GA, Kumwenda NI, Mtimavalye LA, Yang LP, Liomba GN, Broadhead RL, Chiphangwi JD, Miotti PG. 1998. Bacterial vaginosis and disturbances of vaginal flora: association with increased acquisition of HIV. AIDS 12:1699–1706. doi: 10.1097/00002030-199813000-00019 [DOI] [PubMed] [Google Scholar]
- 41. McClelland RS, Lingappa JR, Srinivasan S, Kinuthia J, John-Stewart GC, Jaoko W, Richardson BA, Yuhas K, Fiedler TL, Mandaliya KN, Munch MM, Mugo NR, Cohen CR, Baeten JM, Celum C, Overbaugh J, Fredricks DN. 2018. Evaluation of the association between the concentrations of key vaginal bacteria and the increased risk of HIV acquisition in African women from five cohorts: a nested case-control study. Lancet Infect Dis 18:554–564. doi: 10.1016/S1473-3099(18)30058-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Coleman SL, Neff CP, Li SX, Armstrong AJS, Schneider JM, Sen S, Fennimore B, Campbell TB, Lozupone CA, Palmer BE. 2020. Can gut microbiota of men who have sex with men influence HIV transmission Gut Microbes 11:610–619. doi: 10.1080/19490976.2019.1700756 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Yamada E, Martin CG, Moreno-Huizar N, Fouquier J, Neff CP, Coleman SL, Schneider JM, Huber J, Nusbacher NM, McCarter M, Campbell TB, Lozupone CA, Palmer BE. 2021. Intestinal microbial communities and Holdemanella isolated from HIV+/- men who have sex with men increase frequencies of lamina propria CCR5+ CD4+ T cells. Gut Microbes 13:1997292. doi: 10.1080/19490976.2021.1997292 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Kelley CF, Haaland RE, Patel P, Evans-Strickfaden T, Farshy C, Hanson D, Mayer K, Lennox JL, Brooks JT, Hart CE. 2011. HIV-1 RNA Rectal shedding is reduced in men with low plasma HIV-1 RNA viral loads and is not enhanced by sexually transmitted bacterial infections of the rectum. J Infect Dis 204:761–767. doi: 10.1093/infdis/jir400 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Hassan AM, Mancano G, Kashofer K, Fröhlich EE, Matak A, Mayerhofer R, Reichmann F, Olivares M, Neyrinck AM, Delzenne NM, Claus SP, Holzer P. 2019. High-fat diet induces depression-like behaviour in mice associated with changes in microbiome, neuropeptide Y, and brain metabolome. Nutr Neurosci 22:877–893. doi: 10.1080/1028415X.2018.1465713 [DOI] [PubMed] [Google Scholar]
- 46. Duan Y, Zeng L, Zheng C, Song B, Li F, Kong X, Xu K. 2018. Inflammatory links between high fat diets and diseases. Front Immunol 9:2649. doi: 10.3389/fimmu.2018.02649 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. López-Taboada I, González-Pardo H, Conejo NM. 2020. Western diet: implications for brain function and behavior. Front Psychol 11:564413. doi: 10.3389/fpsyg.2020.564413 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Wahl A, Victor Garcia J. 2014. The use of BLT humanized mice to investigate the immune reconstitution of the gastrointestinal tract. J Immunol Methods 410:28–33. doi: 10.1016/j.jim.2014.06.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Li Q, Tso FY, Kang G, Lu W, Li Y, Fan W, Yuan Z, Destache CJ, Wood C. 2015. Early initiation of antiretroviral therapy can functionally control productive HIV-1 infection in humanized-BLT mice. J Acquir Immune Defic Syndr 69:519–527. doi: 10.1097/QAI.0000000000000687 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Zhang J, Lohani SC, Cheng Y, Wang T, Guo L, Kim WK, Gorantla S, Li Q. 2021. Human microglia extensively reconstitute in humanized-BLT mice with human interleukin-34 transgene and support HIV-1 brain infection. Front. Immunol 12:672415. doi: 10.3389/fimmu.2021.672415 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Daharsh L, Ramer-Tait AE, Li Q. 2019. Stable engraftment of a human gut bacterial microbiome in double humanized BLT-mice. bioRxiv. doi: 10.1101/749093 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Campbell CL, Yu R, Li F, Zhou Q, Chen D, Qi C, Yin Y, Sun J. 2019. Modulation of fat metabolism and gut microbiota by resveratrol on high-fat diet-induced obese mice. Diabetes Metab Syndr Obes 12:97–107. doi: 10.2147/DMSO.S192228 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Figliuolo VR, Dos Santos LM, Abalo A, Nanini H, Santos A, Brittes NM, Bernardazzi C, de Souza HSP, Vieira LQ, Coutinho-Silva R, Coutinho CMLM. 2017. Sulfate-reducing bacteria stimulate gut immune responses and contribute to inflammation in experimental colitis. Life Sci 189:29–38. doi: 10.1016/j.lfs.2017.09.014 [DOI] [PubMed] [Google Scholar]
- 54. Rajput M, Momin T, Singh A, Banerjee S, Villasenor A, Sheldon J, Paudel P, Rajput R. 2023. Determining the association between gut microbiota and its metabolites with higher intestinal immunoglobulin A response. Vet Anim Sci 19:100279. doi: 10.1016/j.vas.2022.100279 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Samaddar A, van Nispen J, Armstrong A, Song E, Voigt M, Murali V, Krebs J, Manithody C, Denton C, Ericsson AC, Jain AK. 2022. Lower systemic inflammation is associated with gut firmicutes dominance and reduced liver injury in a novel ambulatory model of parenteral nutrition. Ann Med 54:1701–1713. doi: 10.1080/07853890.2022.2081871 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Matsuoka K, Kanai T. 2015. The gut microbiota and inflammatory bowel disease. Semin Immunopathol 37:47–55. doi: 10.1007/s00281-014-0454-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Santoro A, Ostan R, Candela M, Biagi E, Brigidi P, Capri M, Franceschi C. 2018. Gut microbiota changes in the extreme decades of human life: a focus on centenarians. Cell Mol Life Sci 75:129–148. doi: 10.1007/s00018-017-2674-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Park JS, Choi JW, Jhun J, Kwon JY, Lee BI, Yang CW, Park SH, Cho ML. 2018. Lactobacillus acidophilus improves intestinal inflammation in an acute colitis mouse model by regulation of Th17 and treg cell balance and fibrosis development. J Med Food 21:215–224. doi: 10.1089/jmf.2017.3990 [DOI] [PubMed] [Google Scholar]
- 59. Aghamohammad S, Sepehr A, Miri ST, Najafi S, Pourshafie MR, Rohani M. 2022. Anti-inflammatory and immunomodulatory effects of Lactobacillus spp. as a preservative and therapeutic agent for IBD control. Immun Inflamm Dis 10:e635. doi: 10.1002/iid3.635 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Chen S, Li Y, Chu B, Yuan L, Liu N, Zhu Y, Wang J. 2021. Lactobacillus johnsonii L531 alleviates the damage caused by Salmonella typhimurium via inhibiting TLR4, NF-κB, and NLRP3 inflammasome signaling pathways. Microorganisms 9:1983. doi: 10.3390/microorganisms9091983 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Fischer F, Romero R, Hellhund A, Linne U, Bertrams W, Pinkenburg O, Eldin HS, Binder K, Jacob R, Walker A, Stecher B, Basic M, Luu M, Mahdavi R, Heintz-Buschart A, Visekruna A, Steinhoff U. 2020. Dietary cellulose induces anti-inflammatory immunity and transcriptional programs via maturation of the intestinal microbiota. Gut Microbes 12:1–17. doi: 10.1080/19490976.2020.1829962 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Fu T, Wang Y, Ma M, Dai W, Pan L, Shang Q, Yu G. 2023. Isolation of alginate-degrading bacteria from the human gut microbiota and discovery of Bacteroides xylanisolvens AY11-1 as a novel anti-colitis probiotic bacterium. Nutrients 15:1352. doi: 10.3390/nu15061352 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Fabersani E, Portune K, Campillo I, López-Almela I, la Paz SM, Romaní-Pérez M, Benítez-Páez A, Sanz Y. 2021. Bacteroides uniformis CECT 7771 alleviates inflammation within the gut-adipose tissue axis involving TLR5 signaling in obese mice. Sci Rep 11:11788. doi: 10.1038/s41598-021-90888-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Duncan SH, Richardson AJ, Kaul P, Holmes RP, Allison MJ, Stewart CS. 2002. Oxalobacter formigenes and its potential role in human health. Appl Environ Microbiol 68:3841–3847. doi: 10.1128/AEM.68.8.3841-3847.2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Sun Z, Song ZG, Liu C, Tan S, Lin S, Zhu J, Dai FH, Gao J, She JL, Mei Z, Lou T, Zheng JJ, Liu Y, He J, Zheng Y, Ding C, Qian F, Zheng Y, Chen YM. 2022. Gut microbiome alterations and gut barrier dysfunction are associated with host immune homeostasis in COVID-19 patients. BMC Med 20:24. doi: 10.1186/s12916-021-02212-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Kahle EM, Bolton M, Hughes JP, Donnell D, Celum C, Lingappa JR, Ronald A, Cohen CR, de Bruyn G, Fong Y, Katabira E, McElrath MJ, Baeten JM, Partners in Prevention HSV/HIV Transmission Study Team . 2015. Plasma cytokine levels and risk of HIV type 1 (HIV-1) transmission and acquisition: a nested case-control study among HIV-1-serodiscordant couples. J Infect Dis 211:1451–1460. doi: 10.1093/infdis/jiu621 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Lajoie J, Juno J, Burgener A, Rahman S, Mogk K, Wachihi C, Mwanjewe J, Plummer FA, Kimani J, Ball TB, Fowke KR. 2012. A distinct cytokine and chemokine profile at the genital mucosa is associated with HIV-1 protection among HIV-exposed seronegative commercial sex workers. Mucosal Immunol 5:277–287. doi: 10.1038/mi.2012.7 [DOI] [PubMed] [Google Scholar]
- 68. Trinchieri G. 2003. Interleukin-12 and the regulation of innate resistance and adaptive immunity. Nat Rev Immunol 3:133–146. doi: 10.1038/nri1001 [DOI] [PubMed] [Google Scholar]
- 69. McDyer JF, Wu CY, Seder RA. 1998. The regulation of IL-12: its role in infectious, autoimmune, and allergic diseases. J Allergy Clin Immunol 102:11–15. doi: 10.1016/s0091-6749(98)70047-8 [DOI] [PubMed] [Google Scholar]
- 70. Monteleone G, MacDonald TT, Wathen NC, Pallone F, Pender SL. 1999. Enhancing lamina propria Th1 cell responses with interleukin 12 produces severe tissue injury. Gastroenterology 117:1069–1077. doi: 10.1016/s0016-5085(99)70391-4 [DOI] [PubMed] [Google Scholar]
- 71. Fuss IJ, Becker C, Yang Z, Groden C, Hornung RL, Heller F, Neurath MF, Strober W, Mannon PJ. 2006. Both IL-12P70 and IL-23 are synthesized during active crohn’s disease and are down-regulated by treatment with anti-IL-12 P40 monoclonal antibody. Inflamm Bowel Dis 12:9–15. doi: 10.1097/01.mib.0000194183.92671.b6 [DOI] [PubMed] [Google Scholar]
- 72. Mayer L, Sandborn WJ, Stepanov Y, Geboes K, Hardi R, Yellin M, Tao X, Xu LA, Salter-Cid L, Gujrathi S, Aranda R, Luo AY. 2014. Anti-IP-10 antibody (BMS-936557) for ulcerative colitis: a phase II randomised study. Gut 63:442–450. doi: 10.1136/gutjnl-2012-303424 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Liu M, Guo S, Hibbert JM, Jain V, Singh N, Wilson NO, Stiles JK. 2011. CXCL10/IP-10 in infectious diseases pathogenesis and potential therapeutic implications. Cytokine Growth Factor Rev 22:121–130. doi: 10.1016/j.cytogfr.2011.06.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Lane BR, King SR, Bock PJ, Strieter RM, Coffey MJ, Markovitz DM. 2003. The C-X-C chemokine IP-10 stimulates HIV-1 replication. Virology 307:122–134. doi: 10.1016/S0042-6822(02)00045-4 [DOI] [PubMed] [Google Scholar]
- 75. Ploquin MJ, Madec Y, Casrouge A, Huot N, Passaes C, Lécuroux C, Essat A, Boufassa F, Jacquelin B, Jochems SP, et al. 2016. Elevated basal pre-infection CXCL10 in plasma and in the small intestine after infection are associated with more rapid HIV/SIV disease onset. PLoS Pathog 12:e1005774. doi: 10.1371/journal.ppat.1005774 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Jiao Y, Zhang T, Wang R, Zhang H, Huang X, Yin J, Zhang L, Xu X, Wu H. 2012. Plasma IP-10 is associated with rapid disease progression in early HIV-1 infection. Viral Immunol. 25:333–337. doi: 10.1089/vim.2012.0011 [DOI] [PubMed] [Google Scholar]
- 77. Brake DK, Smith EO, Mersmann H, Smith CW, Robker RL. 2006. ICAM-1 expression in adipose tissue: effects of diet-induced obesity in mice. Am J Physiol Cell Physiol 291:C1232–9. doi: 10.1152/ajpcell.00008.2006 [DOI] [PubMed] [Google Scholar]
- 78. Kondo N, Melikyan GB. 2012. Intercellular adhesion molecule 1 promotes HIV-1 attachment but not fusion to target cells. PLoS One 7:e44827. doi: 10.1371/journal.pone.0044827 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Yu X, Shang H, Jiang Y. 2020. ICAM-1 in HIV infection and underlying mechanisms. Cytokine 125:154830. doi: 10.1016/j.cyto.2019.154830 [DOI] [PubMed] [Google Scholar]
- 80. Pathirana WGW, Chubb SP, Gillett MJ, Vasikaran SD. 2018. Faecal calprotectin. Clin Biochem Rev 39:77–90. [PMC free article] [PubMed] [Google Scholar]
- 81. Jiao N, Baker SS, Nugent CA, Tsompana M, Cai L, Wang Y, Buck MJ, Genco RJ, Baker RD, Zhu R, Zhu L. 2018. Gut microbiome may contribute to insulin resistance and systemic inflammation in obese rodents: a meta-analysis. Physiol Genomics 50:244–254. doi: 10.1152/physiolgenomics.00114.2017 [DOI] [PubMed] [Google Scholar]
- 82. Brandsma E, Kloosterhuis NJ, Koster M, Dekker DC, Gijbels MJJ, van der Velden S, Ríos-Morales M, van Faassen MJR, Loreti MG, de Bruin A, Fu J, Kuipers F, Bakker BM, Westerterp M, de Winther MPJ, Hofker MH, van de Sluis B, Koonen DPY. 2019. A proinflammatory gut microbiota increases systemic inflammation and accelerates atherosclerosis. Circ Res 124:94–100. doi: 10.1161/CIRCRESAHA.118.313234 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Alam A, Neish A. 2018. Role of gut microbiota in intestinal wound healing and barrier function. Tissue Barriers 6:1539595. doi: 10.1080/21688370.2018.1539595 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Yu LC-H. 2018. Microbiota dysbiosis and barrier dysfunction in inflammatory bowel disease and colorectal cancers: exploring a common ground hypothesis. J Biomed Sci 25:79. doi: 10.1186/s12929-018-0483-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Lloyd-Price J, Abu-Ali G, Huttenhower C. 2016. The healthy human microbiome. Genome Med 8:51. doi: 10.1186/s13073-016-0307-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Gong D, Gong X, Wang L, Yu X, Dong Q. 2016. Involvement of reduced microbial diversity in inflammatory bowel disease. Gastroenterol Res Pract 2016:6951091. doi: 10.1155/2016/6951091 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Wang LX, Kang G, Kumar P, Lu W, Li Y, Zhou Y, Li Q, Wood C. 2014. Humanized-BLT mouse model of Kaposi’s sarcoma-associated herpesvirus infection. Proc Natl Acad Sci U S A 111:3146–3151. doi: 10.1073/pnas.1318175111 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Sun M, Li Y, Yuan Z, Lu W, Kang G, Fan W, Li Q. 2016. VRC01 antibody protects against vaginal and rectal transmission of human immunodeficiency virus 1 in Hu-BLT mice. Arch Virol 161:2449–2455. doi: 10.1007/s00705-016-2942-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Yuan Z, Kang G, Ma F, Lu W, Fan W, Fennessey CM, Keele BF, Li Q. 2016. Recapitulating cross-species transmission of simian immunodeficiency virus SIVcpz to humans by using humanized BLT mice. J Virol 90:7728–7739. doi: 10.1128/JVI.00860-16 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Martínez I, Wallace G, Zhang C, Legge R, Benson AK, Carr TP, Moriyama EN, Walter J. 2009. Diet-induced metabolic improvements in a hamster model of hypercholesterolemia are strongly linked to alterations of the gut microbiota. Appl Environ Microbiol 75:4175–4184. doi: 10.1128/AEM.00380-09 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Callahan BJ, McMurdie PJ, Rosen MJ, Han AW, Johnson AJA, Holmes SP. 2016. DADA2: high-resolution sample inference from Illumina amplicon data. Nat Methods 13:581–583. doi: 10.1038/nmeth.3869 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Quast C, Pruesse E, Yilmaz P, Gerken J, Schweer T, Yarza P, Peplies J, Glöckner FO. 2013. The SILVA ribosomal RNA gene database project: improved data processing and web-based tools. Nucleic Acids Res. 41:D590–6. doi: 10.1093/nar/gks1219 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Robeson MS, O’Rourke DR, Kaehler BD, Ziemski M, Dillon MR, Foster JT, Bokulich NA, Pertea M. 2021. Rescript: reproducible sequence taxonomy reference database management. PLoS Comput Biol 17:e1009581. doi: 10.1371/journal.pcbi.1009581 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Bolyen E, Rideout JR, Dillon MR, Bokulich NA, Abnet CC, Al-Ghalith GA, Alexander H, Alm EJ, Arumugam M, Asnicar F, et al. 2019. Reproducible, interactive, scalable and extensible microbiome data science using QIIME 2. Nat Biotechnol 37:852–857. doi: 10.1038/s41587-019-0209-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Oren A, Garrity GM. 2021. Valid publication of the names of forty-two phyla of prokaryotes. Int J Syst Evol Microbiol 71:005056. doi: 10.1099/ijsem.0.005056 [DOI] [PubMed] [Google Scholar]
- 96. Göker M, Oren A. 2023. Valid publication of four additional phylum names. Int J Syst Evol Microbiol 73:006024. doi: 10.1099/ijsem.0.006024 [DOI] [PubMed] [Google Scholar]
- 97. Price MN, Dehal PS, Arkin AP. 2010. FastTree 2--Approximately maximum-likelihood trees for large alignments. PLoS One 5:e9490. doi: 10.1371/journal.pone.0009490 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Katoh K, Misawa K, Kuma K, Miyata T. 2002. MAFFT: a novel method for rapid multiple sequence alignment based on fast fourier transform. Nucleic Acids Res 30:3059–3066. doi: 10.1093/nar/gkf436 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Lin H, Peddada SD. 2020. Analysis of compositions of microbiomes with bias correction. Nat Commun 11:3514. doi: 10.1038/s41467-020-17041-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
The data sets generated during the current study are available in the NCBI Sequence Read Archive (SRA) repository with accession number PRJNA939259. Scripts related to the current study are freely available from the corresponding author upon request.