Next Article in Journal
Bifidobacterium breve and Bifidobacterium longum Attenuate Choline-Induced Plasma Trimethylamine N-Oxide Production by Modulating Gut Microbiota in Mice
Previous Article in Journal
Vitamin D and Parkinson’s Disease
Previous Article in Special Issue
Targeting the Gut Microbiota of Vertically HIV-Infected Children to Decrease Inflammation and Immunoactivation: A Pilot Clinical Trial
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Relationship of Diet to Gut Microbiota and Inflammatory Biomarkers in People with HIV

1
Department of Nutrition and Food Science, Faculty of Pharmacy, Universidad Complutense de Madrid, 28040 Madrid, Spain
2
Department of Infectious Diseases, Hospital Universitario Ramón y Cajal and IRYCIS, 28034 Madrid, Spain
3
CIBERINFEC, Instituto de Salud Carlos III, 28029 Madrid, Spain
4
FISABIO-Salud Pública, 46020 Valencia, Spain
5
CIBERESP, Instituto de Salud Carlos III, 28029, Madrid, Spain
6
Bioinformatics Unit, Hospital Universitario Ramón y Cajal and IRYCIS, 28034 Madrid, Spain
7
Research Group VALORNUT-UCM (920030), Universidad Complutense de Madrid, 28040 Madrid, Spain
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Nutrients 2022, 14(6), 1221; https://doi.org/10.3390/nu14061221
Submission received: 3 February 2022 / Revised: 8 March 2022 / Accepted: 11 March 2022 / Published: 14 March 2022
(This article belongs to the Special Issue Nutritional Supplementation in People with HIV)

Abstract

:
While changes in microbiome composition have been associated with HIV, the effect of diet and its potential impact on inflammation remains unclear. Methods: Twenty-seven people living with HIV (PWH) on antiretroviral therapy (ART) were studied. A comprehensive dietary analysis was performed and two types of dietary patterns were determined. We explored the associations of each dietary pattern with gut microbiota and plasma inflammatory biomarkers. Results: We appreciated two dietary patterns, Mediterranean-like (MEL) and one Western-like (WEL). Compared to participants with the WEL pattern, participants with MEL pattern showed higher abundance of Lachnospira (p-value = 0.02) and lower levels of the inflammatory biomarkers D-dimer (p-value = 0.050) and soluble TNF-alpha receptor 2 (sTNFR2) (p-value = 0.049). Men who have sex with men (MSM) with MEL pattern had lower abundance of Erysipelotrichaceae (p-value < 0.001) and lower levels of D-dimer (p-value = 0.026) than MSM with WEL pattern. Conclusion: MEL pattern favours Lachnospira abundance, and protects against Erysipelotrichaceae abundance and higher levels of the inflammatory biomarkers D-dimer and sTNFR2, precursors of inflammatory processes in HIV-infected patients. Our study contributes to understanding the determinants of a healthier diet and its connections with gut microbiota and inflammation.

Graphical Abstract

1. Introduction

Interactions between altered gut mucosa and bacteria during HIV infection appear to contribute to chronic immune dysfunction and correlate with the route of HIV transmission [1,2]. However, further study is needed to understand how nutritional interventions might improve gut dysbiosis, especially during chronic diseases.
Scientific evidence has associated the more Western dietary pattern, high in saturated fatty acids, cholesterol and sugars and low in fibre and micronutrients, with various inflammatory metabolic and chronic gastrointestinal tract conditions, whereas diets rich in fibre, vitamins and antioxidants have beneficial effects on gut homeostasis by increasing microbial diversity and inducing a regulatory environment [3,4,5,6]. Although diet and dietary habits have been extensively studied in healthy people or people with different pathologies, studies referring to diet in HIV-infected patients are very scarce [6,7].
On the other hand, some studies have found differences in the intestinal microbiota of HIV-infected patients by transmission risk group and have suggested that these differences could be attributed to diet [1,8].
We aimed to analyse the dietary patterns and dietary quality of a group of 27 people living with HIV (PWH), and correlate the nutritional parameters with the gut microbiota composition and inflammatory biomarkers.

2. Materials and Methods

2.1. Study Design and Sample Collection

We conducted a cross-sectional study. Participants were recruited at the HIV unit of the Ramón y Cajal University Hospital in Madrid, Spain, between October 2017 and February 2018. Participant contact took place at the HIV unit of the hospital, where physicians informed participants about the details of the study beforehand.
The protocol was approved by the Ethics Committee of the Hospital Ramón y Cajal (Ref. 030/20) and all participants signed an informed consent form.
Stool and blood samples were collected on the same day that the dietary questionnaires were completed.
Inclusion criteria were confirmed HIV infection, age of 18 years or more, and antiretroviral therapy (ART) with undetectable plasma HIV RNA during at least 48 weeks. Exclusion criteria were failure to provide signed informed consent, acute and intercurrent health problems.

2.2. Dietary Data

A three-day dietary record [9], including two working days and one weekend day, was used to collect all food, beverages and food supplements taken by the participant during that time period.
The questionnaire was completed by the participants. To ensure that the questionnaire was accurately completed, participants were provided with written detailed instructions on how all information was to be recorded, including food and dish ingredients (where possible), cooking techniques, brand names of products and quantities, which could be recorded in home measurements.
To minimize errors, all data were reviewed by the research group’s nutritionists to identify unrealistic servings or fluid intake or any errors in recording.
All dietary information was processed with the DIAL software version 3.0.0.12 (Alce Ingeniería, Madrid, Spain), which uses data from the Spanish Food Composition Tables [10]. The observed energy intake, the caloric profile of the macronutrients in the participants’ diets, as well as the intake of vitamins and minerals were obtained through this programme [11].

2.3. Gut Microbiota

2.3.1. Purification of Nucleic Acids

Faecal samples from participants were stored in Omnigene Gut kits (DNA Genotek, Kanata, ON, Canada). Faecal samples were divided into aliquots and cryopreserved at −80 °C until use.

2.3.2. Amplification of the 16 S rRNA Gene

Total DNA was extracted from faecal samples on the MagNA Pure LC Instrument robotic workstation (Roche, Basel, Switzerland) using the MagNA Pure LC III DNA isolation kit (Bacteria, Fungi) (Roche). Total DNA was quantified with a Qubit fluorometer (ThermoFisher, Waltham, MA, USA). For each sample, regions V3–V4 of the 16 S rRNA gene were amplified and amplicon libraries were constructed following Illumina instructions (Illumina, San Diego, CA, USA) [12]. Sequencing was performed using the V3 kit (2 × 300 cycles) with MiSeq sequencer (Illumina, San Diego, CA, USA) at the Sequencing and Bioinformatics Service FISABIO, Valencia, Spain. We obtained an average of 62,939 united 16 S rRNA sequences per sample.

2.3.3. Pre-Processing and Quality Control

All sequences used in this analysis passed quality control, where the length and quality of reads were filtered using trimmomatic v0.33. To standardise the number of reads in the diversity analyses, sub-sampling methods (seqkit, sample subcommand) were used, which was performed based on the minimum number of reads per sample.

2.3.4. 16 S RNA Gene Analysis

The 16 S rRNA gene amplicon data were analysed using the Kraken taxonomic sequence classifier (v2.0.7-beta, paired-end option) [13,14], which examines the k-mers within a query sequence and uses the information within those k-mers to query a database. That database maps k-mers to the lowest common ancestor of all genomes known to contain a given k-mer. Taxonomic information on 16 S rDNA sequences was obtained using the Silva ribosomal RNA database (version 132) [15] available on Kraken 2 web [16]. After assigning taxonomic labels to the sequence reads, the Operational Taxonomic Units (OTU) table was extracted using Pavian version.

2.4. Inflammatory Biomarkers

A fasting venous blood sample was collected from each participant and plasma levels of 8 inflammatory biomarkers were determined from the cryopreserved plasma by immunoassay in triplicate. Tumour soluble necrosis factor sTNFR2 (DRT200, R & D Systems, Bio-Techne Corporation, Minneapolis, MN, USA), C-reactive protein (CRP) (DCRP00, Quantikine ELISA kit, R & D Systems, Minneapolis, MN, USA), sCD14 (AbClonal, Wuhan, China), sCD163 (AbClonal, Wuhan, China), FABP2/IFABP (Boster Biological Technology, Wuhan, China), D-dimers (Ray Biotech, Norcross, GA, USA), LTA (Abbexa, Cambridge, UK), LBP (Boster Biolo-gical Technology, Wuhan, China).

2.5. Statistical Analysis

Results are presented as mean ± standard deviation (SD) or as re-counts and proportions in the case of categorical variables. Differences between the different dietary patterns were considered statistically significant if p < 0.05. To study the normality of the different variables within the sample and within the different analysis groups, the Shapiro–Wilk test recommended for N < 50 was used. Comparison of data between dietary patterns was performed using the Student’s t-test for two independent samples, in the case of normally distributed variables, and the Mann–Whitney U-test, in the case of non-parametric variables. For nominal variables, the chi-square test (χ2) was used.
Dietary patterns were obtained by performing a K-means cluster statistical analysis, prefixing the number of clusters to 2, and standardising (z-scores) the intake variables of 14 food groups measured in total grams per day.
Spearman correlations were used to assess the relationship of the different nutritional variables with gut microbiota and inflammatory biomarkers. The statistical programme IBM SPSS Inc. Version 20.0 (Armonk, NY, USA) was used for the analysis of the results and R software (R Core Team, Vienna, Austria) for the correlation analyses between MEDDQI and bacterial counts (libraries corrplot) and for the rest graphics Orange Data Mining Version 3.30 (University of Ljubljana, Ljubljana, Slovenia) and Ms Office Profesional Plus 2016 Version 21.12 (Microsoft, Redmond, WA, USA).

2.6. MED-DQI Dietary Quality Index

To assess the dietary quality of the dietary patterns, the value of the MED-DQI dietary quality index was calculated for each pattern, and for each participant, as it is considered one of the most suitable indices for assessing the quality of the Mediterranean diet in adults [17,18,19]. The calculation was made taking into account the following components: % of saturated fatty acids in relation to total energy, cholesterol (mg), grams of meat, mL of olive oil, grams of fish and grams of fruit and vegetables.
Each nutrient or food group was assigned three scores (0, 1 and 2) based on the recommended guidelines.
MED-DQI scores between 1–4 are considered good, between 5–7 medium good, between 8–10 medium poor and 11–14 poor.

3. Results

3.1. Description of the General Population

The final sample analysed was 27 participants, 18 of whom were men who had sex with men (MSM), 6 ex-injection drug users (IDU) and 3 heterosexuals (HTX) with a mean age of 48.5 years and of Caucasian, Latin American and Sub-Saharan African origin, all participants were on triple ART and only six participants had taken antibiotics in the 6 months prior to the start of the study (the last intake being 14 weeks before the start of the study) and mean BMI was 25.19 (Table 1). Two dietary patterns were identified as a result of the k-means cluster statistical analysis: the Mediterranean-like pattern (MEL) and the Western-like pattern (WEL).

3.2. Dietary Patterns and Dietary Quality Assessment (MED-DQI)

After processing the dietary information, daily intake values (total grams per day) were obtained for 14 food groups (Table 2).
Two dietary patterns were identified as a result of the k-means cluster statistical analysis: the Mediterranean-like pattern (MEL) and the Western-like pattern (WEL). Figure 1 shows the resulting heatmap of the z-score variables for the 14 food groups.
The MEL pattern was characterised by a higher consumption of vegetables, cereals and legumes, and a lower consumption of sugars, white and red meat and potatoes compared to WEL pattern. The values of the final cluster centres are shown in Table A1.
MEL pattern was characterised by a lower intake of saturated fatty acids (SFA) and a high intake of total dietary fibre (33.99 ± 13.45 g/day vs. 22.23 ± 5.84 g/day of WEL pattern; p-value = 0.015), and a higher caloric intake from carbohydrates (Table A2). Regarding the contribution of vitamins, antioxidants and fatty acids, a higher intake of folic acid, biotin, total tocopherols, total carotenes, β-carotenes, and a higher oxygen radical absorbance capacity (ORAC) index was observed (Table A3), as well as a lower intake of stearic acid (5.31 ± 1.98 g/d vs. 8.23 ± 3.81 g/d; p-value = 0.044) and arachidonic acid (0.16 ± 0.13 g/d vs. 0.31 ± 0.20 g/d; p-value = 0.015).
To assess the dietary quality of the standards, the mean value of the MED-DQI quality index was calculated (Table A2), showing differences, with a mean score of 4.5 for the MEL dietary standard, recognising it as a good-medium good diet, compared to a mean score of 7.5 for the WEL dietary standard, considering it as a medium-poor diet.

3.3. Factors Associated with the Risk Group

72% of MSM participants followed MEL dietary pattern compared to 28% of MSM participant who followed WEL dietary pattern. The rest of the non-MSM participants (HTX and IDU) were more evenly distributed, with 44% following MEL pattern and 56% following WEL pattern.

3.4. Association between Dietary Pattern and Gut Microbiota

After searching for possible associations between dietary pattern and gut microbiota, we found increased 5-fold Lachnospira abundance associated with MEL pattern (p-value = 0.02) (Figure 2 and Table A4).
Possible associations between MED-DQI dietary quality index scores and gut microbiota were analysed in PWH (Figure 3 and Table A5), showing a negative correlation between MED-DQI with Lachnospira (Rho = −0.42; p-value = 0.028) and a positive correlation with Bacteroides (Rho = 0.48; p-value = 0.012). Lower MED-DQI scores indicate better quality diet and are associated with higher abundance of Lachnospira, and lower levels of Bacteroides.
We explored the possible correlation of Lachnospira and Bacteroides with nutrients and foods in (Figure 4 and Figure 5). We observed a positive correlation between Lachnospira and cellulose (Rho = 0.49 and p-value = 0.008), fructose (Rho = 0.49 and p-value = 0.009), dietary fibre (Rho = 0.48 and p-value = 0.012), and glucose (Rho = 0.41 and p-value = 0.034). We observed a negative correlation between Bacteroides and glucose (Rho = −0.51 and p-value = 0.006), malic acid (Rho = −0,46 and p-value = 0.015), citric acid (Rho = −0.41 and p-value = 0.032), olive oil (Rho = −0.39 and p-value = 0.044) and vegetables (Rho = −0.38 and p-value = 0.048).

3.5. Association between Dietary Pattern and Inflammatory Biomarkers

To further investigate the potential role of the diet on systemic makers of clinical progression, we analysed the associations between the dietary pattern and inflammatory biomarkers. We found that participants with a MEL pattern, exhibited reduced D-dimer (p-value = 0.050) and sTNFR2 (p-value = 0.049) levels compared to those with a WEL pattern (Figure 6 and Table A6).

3.6. Association between Dietary Pattern and Gut Microbiota in MSM

Additionally, significant differences were observed in the levels of Erysipelotrichaceae (p-value = <0.001) in MSM participants with MEL pattern versus MSM participants with WEL pattern, being more abundant in the latter, accounting for an approximate two-fold increase (Figure 7 and Table A7).
We explored in the whole MSM the possible correlation of Erysipelotrichaceae with nutrients and foods in (Figure 8). We observed a negative correlation with vitamin E (Rho = −0.9 and p-value = 0.037), fish (Rho = −0.76 and p-value < 0.001), zeaxanthin (Rho = −0.55 and p-value = 0.019), and a positive correlation with meat (Rho = 0.75 and p-value < 0.001).
In MSM with MEL pattern (Figure 9), we observed a negative correlation with the carotenoide zeaxanthin (Rho = −0.61 and p-value = 0.027), fish (Rho = −0.58 and p-value = 0.038) and with eicosanpentanoic Acid (Rho = −0.55 and p-value = 0.049).
In contrast, in MSM with WEL pattern (Figure 10) we observed a negative correlation with vitamin E (Rho = −0.9 and p-value = 0.037), fruit (Rho = −0.9 and p-value = 0.037) and dietary Fibre (Rho = −0.9 and p-value = 0.037).

3.7. Association between Dietary Pattern and Inflammatory Biomarkers in MSM

Analysing the data by risk group, we observed that in MSM participants with MEL pattern, there were s differences in D-dimer (p-value = 0.026) with mean levels almost half of those found in participants with WEL pattern (Figure 11 and Table A8).

4. Discussion

In this study in PWH, we found evidence of an intersection between dietary habits, the microbiota and systemic inflammation. We found that compared to participants with a WEL pattern, those with MEL pattern exhibited increased abundance of Lachnospira and lower levels of D-dimers and sTNFR2, and among MSM also a lower Erysipelotrichaceae abundance and lower D-dimers. While it is now assumed that Western and Mediterranean dietary patterns and dietary fibre intake play an important role in modulating gut microbiota [20,21,22,23,24,25], the information in the setting of specific chronic diseases such as HIV remains scarce.
Lachnospira genus is a producer of short-chain fatty acids (SCFA), especially butyrate production, which generates energy for colonocytes, which will induce enterocyte binding proteins, promoting intestinal barrier function and favouring the increase of intestinal regulatory T cells that mitigate inflammation [26]. Our findings regarding the association between total fibre intake and Lachnospira abundance are in keep with previous reports outside HIV infection [27,28]. In our study, a higher abundance of Lachnospira and dietary fibre intake was associated with a MEL dietary pattern, and a positive correlation was found between Lachnospira and dietary fibre. In addition, a higher carbohydrate intake (%) was observed in the MEL versus WEL pattern, although no correlation of the latter with Lachnospira was observed.
There was also a negative correlation between the MED-DQI with Lachnospira and a positive correlation with Bacteroides. This latter result is consistent with numerous published studies in the general population indicating that Bacteroides dominated microbiota [20,29], that is associated with long-term dietary patterns, and associated with a higher abundance of animal proteins and fats characteristic of the Western pattern [30,31].
In an exploratory analysis in PWH of this study, we observed a positive correlation between Lachnospira and digestive carbohydrates like fructose and glucose and non-digestive carbohydrates like dietary fibre. We also observed a negative correlation between Bacteroides and glucose, organic acids like malic and citric acid, olive oil, and vegetables.
Furthermore, we found a lower Erysipelotrichaceae abundance in MSM with MEL pattern. Although until recently, the Erysipelotrichaceae family of bacteria was not considered to play a significant role in health and disease. However, an increasing number of studies have been published attributing a potential role in numerous diseases and inflammatory processes [32] and linking it to a high-fat Western diet [33]. Notably, one study found that the relative abundance of Erysipelotrichix, a bacterium belonging to the Erysipelotrichaceae family, correlated positively with levels sTNFR2 [32] in PWH on ART. Other studies in animal models showed a positive correlation between against the abundance of Erysipelotrichaceae levels and cholesterol metabolites [34,35] and that supplementation with antioxidants (quercetin), which are typically abundant in the MEL pattern, inhibited the growth of against the abundance of Erysipelotrichaceae [36].
In an exploratory analysis restricted to MSM in this study we observed a negative correlation between vitamin E, zeaxanthin and fish consumption with Erysipelotrichaceae abundance and a positive correlation with meat. For the case of MSM with MEL we observed there is a negative correlation with fish and eicosapentaonic acid (EPA) w-3 fatty acids found in oily fish with anti-inflammatory action and is clinically relevant beneficial [37,38] and with the carotenoide zeanxanthin. MSM with WEL pattern showed a strong negative correlation of Erysipelotrichaceae with antioxidants intake like vitamin E, dietary fibre and fruit consumption.
Importantly, we found differences in the inflammatory biomarkers D-dimmer and sTNFR2 among PWH according to the dietary pattern followed, with levels in MEL being much lower than in WEL. sTNFR2 is a proinflammatory cytokine that is used as a marker of atherogenesis and is increased in PWH despite effective ART [37], as well as in other pro-inflammatory diseases [39,40,41]. D-dimer is a marker of fibrin degradation and its levels are often increased in PWH and denote a pre-thrombotic state that may lead to clinical thrombosis [42]. Importantly, higher levels of inflammatory biomarkers during treated HIV have consistently been linked to an excess risk of comorbidities during HIV treatment and suggested as a contributing risk factor [43]. As chronic inflammation has become a stubbornly elusive target in HIV, pursuing the effect of dietary interventions deserve further investigations. Our findings linking the MEL pattern with microbiome shifts and lower inflammation provide further support to recent intervention studies showing that adherence to the Mediterranean diet or particular components such as extra virgin olive oil is a feasible intervention to mitigate chronic inflammation in PWH [44,45,46].
Our study also has several limitations that must be taken into considerations when interpreting our results. First, we only assessed a limited number of participants, so we might have lacked statistical power to detect differences between groups in some analyses. Second, the observational design prevented us to assess causal relationships. However, in the context of the current state of the art, we can safely assume that dietary changes should explain the differences in the microbiome composition and inflammatory biomarkers, instead of the opposite.

5. Conclusions

In this observational study in PWH, compared to WEL pattern, MEL pattern was characterised by a higher intake of vegetables, cereals and legumes and a high intake of fibre, as well as a lower intake of sugars, meat and potatoes. MEL pattern determined higher Lachnospira abundance and a better score on the MED-DQI food quality index, which correlated directly with Lachnospira abundance and indirectly with Bacteroides abundance. In addition, MSM with MEL pattern exhibited lower abundance of Erysipelotrichaceae than MSM with a WEL pattern, a family of bacteria implicated in numerous inflammatory processes and which is more prevalent in Western diets characterised by a high intake of saturated fat and cholesterol and a low intake of dietary fibre, vitamins and antioxidants. Finally, MEL pattern was associated with decreased levels of D-dimer and sTNFR2, suggesting beneficial effects on long-term clinical outcomes.
Our study supports the notion that the dietary interventions should be further pursued as a feasible target to decrease inflammation in treated HIV. Larger studies with larger sample sizes assessing dietary interventions are justified to further inform the best strategies to improve chronic inflammation in PWH.

Author Contributions

Conceptualization, M.M., B.N. and S.S.-V.; formal analysis, M.M., A.T., V.F.L., B.N. and S.S.-V.; investigation, M.M., A.T., E.M., N.M., M.J.G., R.R., F.D., J.A.P.-M., V.F.L., J.D., S.M., B.N. and S.S.-V.; supervision, V.F.L., B.N. and S.S.-V.; writing—original draft, M.M. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the Instituto de Salud Carlos III and Fondos FEDER, Acción Estratégica en Salud (AC17/00019, PI18/00154, ICI20/00058 and PI21/00041). S.S-V. has been funded by a grant from the Spanish Society of Microbiology and Infectious Diseases (SEIMC) and by the Spanish Ministry of Science and Innovation- Instituto de Salud Carlos III and Fondos FEDER (BA21/00017).

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki, and approved by the Ethics Committee) of Ethics Committee of the Hospital Ramón y Cajal (Ref. 030/20) on 16 January 2017.)

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The data presented in this study are available upon reasonable request to the corresponding author. Some restrictions might apply due to ethics limitations.

Conflicts of Interest

The authors declare no conflict of interest.

Appendix A

Table A1. Final cluster centres.
Table A1. Final cluster centres.
Variables
(z-Scores)
MELWELp-Value
Z_Dairy products0.1−0.20.409
Z_Fish−0.10.10.728
Z_Eggs0.2−0.30.181
Z_Sugar−0.30.50.030
Z_Olive oil0.2−0.30.253
Z_Nuts 0.2−0.40.135
Z_ Potatoes−0.50.80.000
Z_Fruits0.2−0.30.230
Z_Vegetables0.3−0.60.025
Z_Cereals and legumes0.4−0.70.005
Z_White meat−0.40.70.002
Z_Red meat−0.30.50.039
Z_Aperitives−0.20.50.085
Z_Alcohol−0.10.20.535
The values of the final cluster centres are presented. The p-value of the ANOVA table is presented. The p-values < 0.05 that allow us to infer the most significant variables when constructing the clusters are marked in bold.
Table A2. Energy intake, calorie profile of nutrients and MED-DQI.
Table A2. Energy intake, calorie profile of nutrients and MED-DQI.
MEL (n = 17)WEL (n = 10)Total (n = 27)p-Value
Energy intake (kcal/d) (t)2412.71 ± 512.192396.40 ± 423.622406.67 ± 472.890.933
Proteins (g/d)97.32 ± 23.99111.87 ± 41.81102.71 ± 31.790.706
Proteins (%TEI)16.23 ± 3.4418.42 ± 3.8617.04 ± 3.690.209
Carbohydrates (g/d)257.00 ± 89.81214.70 ± 42.48241.33 ± 77.600.315
Carbohydrates (%TEI) (t)45.22 ± 8.0338.47 ± 8.5342.72 ± 8.710.049
Dietary fibre (g/d) (t)33.99 ± 13.4522.23 ± 5.8429.63 ± 12.520.015
Lipids (g/d) (t)97.31 ± 25.79107.46 ± 35.70101.07 ± 29.580.400
Lipids (%TEI) (t)36.44 ± 7.2439.49 ± 7.0437.57 ± 7.190.296
SFA (%TEI) (t)10.12 ± 2.9712.91 ± 2.8811.15 ± 3.190.025
PUFA (%TEI)5.39 ± 2.275.37 ± 1.125.38 ± 1.900.393
MUFA (%TEI) (t)17.22 ± 4.2317.64 ± 3.8317.37 ± 4.020.797
Omega 3 (%TEI)0.80 ± 0.440.80 ± 0.480.80 ± 0.440.763
Cholesterol (mg/d) (t)312.59 ± 131.66402.80 ± 155.91346.00 ± 145.090.121
MED-DQI (t)4.53 ± 1.847.50 ± 1.435.63 ± 2.22<0.001
Results are presented as mean ± SD. Significant differences between dietary patterns (p-value < 0.05) are indicated in bold. (t): p-value calculated with t-test for independent samples. Remaining variable p-value calculated with Mann–Whitney U test. (%EI): % Total Energy Intake; SFA: Saturated Fatty Acids; PUFA: Polyunsaturated Fatty Acids; MFA: Monounsaturated Fatty Acids. For the calculation of the MED-DQI, the following variables were taken into account: % SFA, cholesterol (mg), grams of meat, ml of olive oil, grams of fish and grams of fruit and vegetables.
Table A3. Vitamins y antioxidants.
Table A3. Vitamins y antioxidants.
MEL (n = 17)WEL (n = 10)Total (n = 27)p-Value
Ascorbic Acid (t) (mg/d)218.93 ± 134.09135.93 ± 73.63188.19 ± 120.870.085
Pantothenic Acid (t) (mg/d)6.28 ± 1.236.13 ± 1.576.22 ± 1.340.790
B12 (µg/d)5.74 ± 2.319.89 ± 10.547.27 ± 6.770.183
Biotine (t) (µg/d)38.82 ± 17.7323.87 ± 10.1333.28 ± 16.820.023
Folic Acid (t) (µg/d)407.29 ± 141.28283.70 ± 85.03361.52 ± 135.960.019
Niacin (mg/d)39.57 ± 10.6149.12 ± 22.3643.11 ± 16.260.407
Riboflavin (t) (mg/d)1.81 ± 0.581.80 ± 0.641.81 ± 0.590.952
Thiamine (mg/d)1.56 ± 0.401.80 ± 0.941.65 ± 0.650.980
Vitamin B6 (t) (mg/d)2.51 ± 0.692.82 ± 1.142.63 ± 0.870.386
Vitamin A (t) (µg/d)1121.06 ± 542.52840.40 ± 569.891017.11 ± 559.130.214
Vitamin D (µg/d)3.49 ± 3.865.77 ± 10.784.34 ± 7.120.860
Vitamin E (mg/d)13.73 ± 9.258.64 ± 3.0411.84 ± 7.880.056
Vitamin K (µg/d)190.04 ± 130.66122.05 ± 76.11164.86 ± 116.750.175
Total Carotens (t) (µg/d)4822.53 ± 2817.072702.60 ± 1863.214037.37 ± 2678.360.045
Alpha-Carotens (µg/d)627.31 ± 618.89359.73 ± 356.75528.21 ± 545.070.160
Beta-Carotens (t) (µg/d)4097.06 ± 2432.032265.10 ± 1713.013418.56 ± 2338.450.047
Beta-Cryptoxanthin (µg/d)528.78 ± 926.80157.20 ± 180.27391.16 ± 757.150.802
Lutein (µg/d)1075.44 ± 899.76868.33 ± 986.07998.73 ± 919.320.393
Zeaxanthin (µg/d)34.25 ± 58.6114.50 ± 26.1926.94 ± 49.460.310
Total Tocopherols (mg/d)10.69 ± 9.474.14 ± 1.948.26 ± 8.180.014
ORAC (µmol TE/100g)20,352.12 ± 11,973.5212,652.80 ± 10,515.4417,500.52 ± 11,868.280.040
Results are presented as mean ± SD. Significant differences between dietary patterns (p < 0.05) are indicated in bold. (t): p-value calculated with t-test for independent samples. Rest of variable p-value calculated with Mann–Whitney U test.
Table A4. Number of readings of the main bacterial taxa in the gut microbiota by dietary patterns.
Table A4. Number of readings of the main bacterial taxa in the gut microbiota by dietary patterns.
MEL (n = 17)WEL (n = 10)Total (n = 27)p-Value
Acidaminococcus261.88 ± 393.77596.60 ± 1286.51385.85 ± 833.950.687
Actinobacteria892.71 ± 1174.411641.20 ± 2681.401169.93 ± 1863.670.547
Bacteroides1102.06 ± 1640.531845.00 ± 1941.951377.22 ± 1759.340.209
Bifidobacteriaceae358.94 ± 555.44680.30 ± 1151.12477.96 ± 820.700.725
Erysipelotrichaceae (t)954.82 ± 461.541284.60 ± 886.841076.96 ± 655.490.296
Faecalibacterium1295.35 ± 737.271420.00 ± 1064.011341.52 ± 854.490.688
Fusobacteria15.06 ± 54.6661.70 ± 184.9632.33 ± 119.190.302
Lachnospira420.35 ± 703.0483.70 ± 105.89295.67 ± 579.210.025
Lactobacillales391.24 ± 559.82260.30 ± 615.35342.74 ± 572.790.581
Prevotella (t)4227.12 ± 3394.932965.50 ± 3118.453759.85 ± 3293.070.346
Proteobacteria829.94 ± 903.30523.20 ± 298.87716.33 ± 745.540.616
Ruminococcaceae (t)3858.53 ± 1269.854041.50 ± 1587.123926.30 ± 1368.340.744
Succinivibrio505.29 ± 937.23134.30 ± 218.95367.89 ± 768.430.286
Results are presented as mean ± SD. Significant differences between dietary patterns (p-value < 0.05) are indicated in bold. (t): p-value calculated with t-test for independent samples. Rest of variable p-value calculated with Mann–Whitney U test.
Table A5. MED-DQI correlations vs. gut microbiota.
Table A5. MED-DQI correlations vs. gut microbiota.
MicrobiotaRho Spearmanp-Value
Acidaminococcus0.100.64
Actinobacteria0.130.520
Bacteroides0.390.044
Bifidobacteriaceae0.060.780
Erysipelotrichaceae0.030.882
Faecalibacterium0.040.852
Fusobacteria0.350.074
Lachnospira−0.420.028
Lactobacillales0.050.812
Prevotella−0.310.117
Proteobacteria−0.050.817
Ruminococcaceae−0.040.847
Succinivibrio−0.220.270
Table A6. Inflammatory biomarkers (ng/mL) by dietary patterns.
Table A6. Inflammatory biomarkers (ng/mL) by dietary patterns.
MEL (n = 17)WEL (n = 10)Total (n = 27)p-Value
sCD143.88 ± 3.133.88 ± 3.133.88 ± 3.130.960
sCD163174.96 ± 148.80174.96 ± 148.80174.96 ± 148.800.651
LTA0.04 ± 0.040.06 ± 0.120.05 ± 0.080.436
LBP(t)442.43 ± 182.75424.84 ± 246.65435.92 ± 204.170.834
FABP2/IFABP4.84 ± 3.224.80 ± 2.754.82 ± 3.000.763
CRP814.76 ± 1530.21517.92 ± 530.74704.82 ± 1248.920.939
D-Dimer352,974.12 ± 407,711.97826,040.00 ± 744,196.27528,183.70 ± 590,083.900.050
sTNFR2 (t)2.89 ± 1.844.56 ± 2.313.51 ± 2.150.049
Results are presented as mean ± SD. Significant differences between dietary patterns (p-value < 0.05) are indicated in bold. (t): p-value calculated with t-test for independent samples. Rest of variable p-value calculated with Mann–Whitney U test.
Table A7. Number of readings of the main bacterial taxa in the gut microbiota by dietary patterns in MSM.
Table A7. Number of readings of the main bacterial taxa in the gut microbiota by dietary patterns in MSM.
MEL (n = 13)WEL (n = 5)p-Value
Acidaminococcus145.77 ± 268.2678.00 ± 162.210.586
Actinobacteria1043.62 ± 1309.01333.40 ± 334.510.218
Bacteroides594.08 ± 1271.51307.40 ± 324.500.521
Bifidobacteriaceae420.69 ± 624.0485.60 ± 146.410.126
Erysipelotrichaceae (t)1079.23 ± 426.112085.60 ± 338.600.000
Faecalibacterium1392.08 ± 789.871070.40 ± 433.360.588
Fusobacteria18.85 ± 62.585.20 ± 4.210.157
Lachnospira310.38 ± 286.1495.60 ± 94.670.104
Lactobacillales224.31 ± 435.6256.60 ± 72.480.324
Prevotella5229.69 ± 3145.835653.60 ± 1890.020.882
Proteobacteria911.92 ± 1025.15675.60 ± 350.160.961
Ruminococcaceae (t)3950.85 ± 1088.713853.40 ± 995.030.864
Succinivibrio660.54 ± 1029.68267.00 ± 252.660.348
Results are presented as mean ± SD. Significant differences between dietary patterns (p-value < 0.05) are indicated in bold. (t): p-value calculated with t-test for independent samples. Rest of variable p-value calculated with Mann–Whitney U test.
Table A8. Inflammatory biomarkers (ng/mL) by dietary patterns in MSM.
Table A8. Inflammatory biomarkers (ng/mL) by dietary patterns in MSM.
MEL (n = 13)WEL (n = 5)p-Value
sCD143.43 ± 2.701.96 ± 0.560.402
sCD163153.03 ± 146.9353.20 ± 19.210.218
LTA0.04 ± 0.050.01 ± 0.010.061
LBP(t)422.31 ± 200.39395.63 ± 169.490.796
FABP2/IFABP5.06 ± 3.454.09 ± 2.390.805
CRP893.83 ± 1718.58505.13 ± 515.960.960
D-dimer300,237.95 ± 433,966.181,193,933.33 ± 908,252.170.026
sTNFR22.69 ± 2.084.60 ± 2.970.183
Results are presented as mean ± SD. Significant differences between dietary patterns (p-value < 0.05) are indicated in bold. (t): p-value calculated with t-test for independent samples. Rest of variable p-value calculated with Mann–Whitney U test.

References

  1. Zhou, Y.; Ou, Z.; Tang, X.; Zhou, Y.; Xu, H.; Wang, X.; Li, K.; He, J.; Du, Y.; Wang, H.; et al. Alterations in the gut microbiota of patients with acquired immune deficiency syndrome. J. Cell. Mol. Med. 2018, 22, 2263–2271. [Google Scholar] [CrossRef] [Green Version]
  2. Klatt, N.R.; Manuzak, J.A. Microbial Effects on Immunity in HIV: Virus, Gender or Sexual Preference Induced? EBioMedicine 2018, 31, 7–8. [Google Scholar] [CrossRef] [Green Version]
  3. Singh, R.K.; Chang, H.-W.; Yan, D.; Lee, K.M.; Ucmak, D.; Wong, K.; Abrouk, M.; Farahnik, B.; Nakamura, M.; Zhu, T.H.; et al. Influence of diet on the gut microbiome and implications for human health. J. Transl. Med. 2017, 15, 73. [Google Scholar] [CrossRef] [Green Version]
  4. Asnicar, F.; Berry, S.E.; Valdes, A.M.; Nguyen, L.H.; Piccinno, G.; Drew, D.A.; Leeming, E.; Gibson, R.; Le Roy, C.; Khatib, H.A.; et al. Microbiome connections with host metabolism and habitual diet from 1098 deeply phenotyped individuals. Nat. Med. 2021, 27, 321–332. [Google Scholar] [CrossRef]
  5. Siracusa, F.; Schaltenberg, N.; Villablanca, E.J.; Huber, S.; Gagliani, N. Dietary habits and intestinal immunity: From food intake to CD4+TH cells. Front. Immunol. 2019, 10, 3177. [Google Scholar] [CrossRef]
  6. Hernández, D.H. Gut microbiota and grain fiber: Evidence and practical recommendations. Nutr. Hosp. 2021, 38, 13–16. [Google Scholar] [CrossRef]
  7. Noguera-Julian, M.; Rocafort, M.; Guillén, Y.; Rivera, J.; Casadellà, M.; Nowak, P.; Hildebrand, F.; Zeller, G.; Parera, M.; Bellido, R.; et al. Gut Microbiota Linked to Sexual Preference and HIV Infection. EBioMedicine 2016, 5, 135–146. [Google Scholar] [CrossRef] [Green Version]
  8. Armstrong, A.J.S.; Shaffer, M.; Nusbacher, N.M.; Griesmer, C.; Fiorillo, S.; Schneider, J.M.; Preston Neff, C.; Li, S.X.; Fontenot, A.P.; Campbell, T.; et al. An exploration of Prevotella-rich microbiomes in HIV and men who have sex with men. Microbiome 2018, 6, 198. [Google Scholar] [CrossRef]
  9. Ortega, R.M.; Requejo, A.M.; López-Sobaler, A.M. Registro de consumo de alimentos y bebidas. In Nutriguía. Manual de Nutrición Clínica En Atención Primaria; Ortega, R.M., Requejo, A.M., Eds.; Editorial Médica Panamericana: Madrid, Spain, 2015. [Google Scholar]
  10. Ortega, R.; López-Sobaler, A.; Requejo, A.; Andrés, P. La Composición de los Alimentos: Herramienta Básica para la Valoración Nutricional; Editorial Complutense: Madrid, Spain, 2010. [Google Scholar]
  11. Ortega, R.M.; Requejo, A.M.; Navia, B. Ingestas Diarias Recomendadas de Energía y Vitaminas para Población Española [Internet]: Madrid, Spain. 2014. Available online: https://www.ucm.es/data/cont/docs/980-2014-09-24-Tablas%20IR%20para%20seminarios.pdf (accessed on 5 February 2020).
  12. Klindworth, A.; Pruesse, E.; Schweer, T.; Peplies, J.; Quast, C.; Horn, M.; Glöckner, F.O. Evaluation of general 16S ribosomal RNA gene PCR primers for classical and next-generation sequencing-based diversity studies. Nucleic Acids Res. 2013, 41, e1. [Google Scholar] [CrossRef]
  13. Wood, D.E.; Salzberg, S.L. Kraken: Ultrafast metagenomic sequence classification using exact alignments. Genome Biol. 2014, 15, R46. [Google Scholar] [CrossRef] [Green Version]
  14. Lu, J.; Salzberg, S.L. Ultrafast and accurate 16S rRNA microbial community analysis using Kraken 2. Microbiome 2020, 8, 124. [Google Scholar] [CrossRef] [PubMed]
  15. Quast, C.; Pruesse, E.; Yilmaz, P.; Gerken, J.; Schweer, T.; Yarza, P.; Peplies, J.; Glöckner, F.O. The SILVA ribosomal RNA gene database project: Improved data processing and web-based tools. Nucleic Acids Res. 2012, 41, D590–D596. [Google Scholar] [CrossRef]
  16. Kraken. Available online: http://ccb.jhu.edu/software/kraken/ (accessed on 5 February 2020).
  17. Gerber, M. Qualitative methods to evaluate Mediterranean diet in adults. Public Health Nutr. 2006, 9, 147–151. [Google Scholar] [CrossRef] [PubMed]
  18. Olza, J.; de Victoria, E.M.; Aranceta-Bartrina, J.; González-Gross, M.; Ortega, R.M.; Serra-Majem, L.; Varela-Moreiras, G.; Gil, Á. Adequacy of critical nutrients affecting the quality of the Spanish diet in the ANIBES study. Nutrients 2019, 11, 2328. [Google Scholar] [CrossRef] [Green Version]
  19. Milà-Villarroel, R.; Bach-Faig, A.; Puig, J.; Puchal, A.; Farran, A.; Serra-Majem, L.; Carrasco, J.L. Comparison and evaluation of the reliability of indexes of adherence to the Mediterranean diet. Public Health Nutr. 2011, 14, 2338–2345. [Google Scholar] [CrossRef] [Green Version]
  20. De Filippis, F.; Pellegrini, N.; Vannini, L.; Jeffery, I.B.; La Storia, A.; Laghi, L.; Serrazanetti, D.I.; Di Cagno, R.; Ferrocino, I.; Lazzi, C.; et al. High-level adherence to a Mediterranean diet beneficially impacts the gut microbiota and associated metabolome. Gut 2016, 65, 1812–1821. [Google Scholar] [CrossRef] [PubMed]
  21. Dahl, W.J.; Rivero Mendoza, D.; Lambert, J.M. Diet, nutrients and the microbiome. Prog. Mol. Biol. Transl. Sci. 2020, 171, 237–263. [Google Scholar] [CrossRef] [PubMed]
  22. Makki, K.; Deehan, E.C.; Walter, J.; Bäckhed, F. The Impact of Dietary Fiber on Gut Microbiota in Host Health and Disease. Cell Host Microbe 2018, 23, 705–715. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Mayengbam, S.; Lambert, J.E.; Parnell, J.A.; Tunnicliffe, J.M.; Nicolucci, A.C.; Han, J.; Sturzenegger, T.; Shearer, J.; Mickiewicz, B.; Vogel, H.J.; et al. Impact of dietary fiber supplementation on modulating microbiota–host–metabolic axes in obesity. J. Nutr. Biochem. 2019, 64, 228–236. [Google Scholar] [CrossRef]
  24. Bang, S.J.; Kim, G.; Lim, M.Y.; Song, E.J.; Jung, D.H.; Kum, J.S.; Nam, Y.-D.; Park, C.S.; Seo, D.H. The influence of in vitro pectin fermentation on the human fecal microbiome. AMB Express 2018, 8, 98. [Google Scholar] [CrossRef] [Green Version]
  25. Myhrstad, M.C.W.; Tunsjø, H.; Charnock, C.; Telle-Hansen, V.H. Dietary Fiber, Gut Microbiota, and Metabolic Regulation-Current Status in Human Randomized Trials. Nutrients 2020, 12, 859. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Vujkovic-Cvijin, I.; Sortino, O.; Verheij, E.; Sklar, J.; Wit, F.W.; Kootstra, N.A.; Sellers, B.; Brenchley, J.M.; Ananworanich, J.; van der Loeff, M.S.; et al. HIV-associated gut dysbiosis is independent of sexual practice and correlates with noncommunicable diseases. Nat. Commun. 2020, 11, 2448. [Google Scholar] [CrossRef] [PubMed]
  27. Gorvitovskaia, A.; Holmes, S.P.; Huse, S.M. Interpreting Prevotella and Bacteroides as biomarkers of diet and lifestyle. Microbiome 2016, 4, 15. [Google Scholar] [CrossRef] [Green Version]
  28. Chen, T.; Long, W.; Zhang, C.; Liu, S.; Zhao, L.; Hamaker, B.R. Fiber-utilizing capacity varies in Prevotella- versus Bacteroides-dominated gut microbiota. Sci. Rep. 2017, 7, 2594. [Google Scholar] [CrossRef] [PubMed]
  29. Merra, G.; Noce, A.; Marrone, G.; Cintoni, M.; Tarsitano, M.G.; Capacci, A.; Lorenzo, A. De Influence of Mediterranean Diet on Human Gut Microbiota. Nutrients 2021, 13, 7. [Google Scholar] [CrossRef] [PubMed]
  30. Lim, M.Y.; Rho, M.; Song, Y.M.; Lee, K.; Sung, J.; Ko, G. Stability of gut enterotypes in Korean monozygotic twins and their association with biomarkers and diet. Sci. Rep. 2014, 4, 7348. [Google Scholar] [CrossRef] [Green Version]
  31. Wu, G.D.; Chen, J.; Hoffmann, C.; Bittinger, K.; Chen, Y.Y.; Keilbaugh, S.A.; Bewtra, M.; Knights, D.; Walters, W.A.; Knight, R.; et al. Linking long-term dietary patterns with gut microbial enterotypes. Science 2011, 334, 105–108. [Google Scholar] [CrossRef] [Green Version]
  32. Vujkovic-Cvijin, I.; Somsouk, M. HIV and the Gut Microbiota: Composition, Consequences, and Avenues for Amelioration. Curr. HIV/AIDS Rep. 2019, 16, 204–213. [Google Scholar] [CrossRef]
  33. Kaakoush, N.O. Insights into the Role of Erysipelotrichaceae in the Human Host. Front. Cell. Infect. Microbiol. 2015, 5, 84. [Google Scholar] [CrossRef] [Green Version]
  34. Dinh, D.M.; Volpe, G.E.; Duffalo, C.; Bhalchandra, S.; Tai, A.K.; Kane, A.V.; Wanke, C.A.; Ward, H.D. Intestinal microbiota, microbial translocation, and systemic inflammation in chronic HIV infection. J. Infect. Dis. 2015, 211, 19–27. [Google Scholar] [CrossRef] [Green Version]
  35. Martínez, I.; Perdicaro, D.J.; Brown, A.W.; Hammons, S.; Carden, T.J.; Carr, T.P.; Eskridge, K.M.; Walter, J. Diet-induced alterations of host cholesterol metabolism are likely to affect the gut microbiota composition in hamsters. Appl. Environ. Microbiol. 2013, 79, 516–524. [Google Scholar] [CrossRef] [Green Version]
  36. Etxeberria, U.; Arias, N.; Boqué, N.; Macarulla, M.T.; Portillo, M.P.; Martínez, J.A.; Milagro, F.I. Reshaping faecal gut microbiota composition by the intake of trans-resveratrol and quercetin in high-fat sucrose diet-fed rats. J. Nutr. Biochem. 2015, 26, 651–660. [Google Scholar] [CrossRef] [PubMed]
  37. Calder, P.C. Omega-3 fatty acids and inflammatory processes: From molecules to man. Biochem. Soc. Trans. 2017, 45, 1105–1115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Peters, B.S.; Wierzbicki, A.S.; Moyle, G.; Nair, D.; Brockmeyer, N. The Effect of a 12-Week Course of Omega-3 Polyunsaturated Fatty Acids on Lipid Parameters in Hypertriglyceridemic Adult HIV-infected Patients Undergoing HAART: A Randomized, Placebo-Controlled Pilot Trial. Clin. Ther. 2012, 34, 67–76. [Google Scholar] [CrossRef] [PubMed]
  39. Kaplan, R.C.; Landay, A.L.; Hodis, H.N.; Gange, S.J.; Norris, P.J.; Young, M.; Anastos, K.; Tien, P.C.; Xue, X.; Lazar, J.; et al. Potential cardiovascular disease risk markers among HIV-infected women initiating antiretroviral treatment. J. Acquir. Immune Defic. Syndr. 2012, 60, 359–368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Carrero, N.; Vásquez, L.; Fernández-Ávila, D.G.; Gutiérrez, J.M.; Díaz, M. Artritis reumatoide en paciente con infección por VIH. Rev. Colomb. Reumatol. 2014, 21, 109–112. [Google Scholar] [CrossRef]
  41. Jordà, F.C.I. TNF-alpha inhibitors in inflammatory bowel disease. Med. Clin. 2004, 123, 627–634. [Google Scholar] [CrossRef]
  42. Toulon, P. Hemostasis and human immunodeficiency virus (HIV) infection. Ann. Biol. Clin. 1998, 56, 153–160. [Google Scholar]
  43. Tenorio, A.R.; Zheng, Y.; Bosch, R.J.; Deeks, S.G.; Rodriguez, B.; Krishnan, S.; Hunt, P.W.; Wilson, C.; Lederman, M.M.; Landay, A.L.; et al. Soluble Markers of Inflammation & Coagulation, but not T-Cell Activation, Predict Non-AIDS- Defining Events During Suppressive Antiretroviral Therapy (ART). In Proceedings of the Conference of Retrovirus and Opportunistic Infections, Boston, MA, USA, 3–6 March 2014. [Google Scholar]
  44. Pastor-Ibáñez, R.; Blanco-Heredia, J.; Etcheverry, F.; Sánchez-Palomino, S.; Díez-Fuertes, F.; Casas, R.; Navarrete-Muñoz, M.Á.; Castro-Barquero, S.; Lucero, C.; Fernández, I.; et al. Adherence to a supplemented mediterranean diet drives changes in the gut microbiota of hiv-1-infected individuals. Nutrients 2021, 13, 1141. [Google Scholar] [CrossRef]
  45. Olalla, J.; García de Lomas, J.M.; Chueca, N.; Pérez-Stachowski, X.; De Salazar, A.; Del Arco, A.; Plaza-Díaz, J.; De la Torre, J.; Prada, J.L.; García-Alegría, J.; et al. Effect of daily consumption of extra virgin olive oil on the lipid profile and microbiota of HIV-infected patients over 50 years of age. Medicine 2019, 98, e17528. [Google Scholar] [CrossRef]
  46. Aiello, A.; Accardi, G.; Candore, G.; Carruba, G.; Davinelli, S.; Passarino, G.; Scapagnini, G.; Vasto, S.; Caruso, C. Nutrigerontology: A key for achieving successful ageing and longevity. Immun. Ageing 2016, 13, 17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Heatmap z-score variables 14 food groups.
Figure 1. Heatmap z-score variables 14 food groups.
Nutrients 14 01221 g001
Figure 2. Gut microbiota by dietary pattern.
Figure 2. Gut microbiota by dietary pattern.
Nutrients 14 01221 g002
Figure 3. Heat map of correlations between MEDDQI and bacterial counts. The pie charts represent the magnitude of each individual Spearman Rho correlation coefficient in a color gradient from red (Rho −1) to blue (Rho +1). Correlations with a p-value < 0.05 are marked with a red asterisk.
Figure 3. Heat map of correlations between MEDDQI and bacterial counts. The pie charts represent the magnitude of each individual Spearman Rho correlation coefficient in a color gradient from red (Rho −1) to blue (Rho +1). Correlations with a p-value < 0.05 are marked with a red asterisk.
Nutrients 14 01221 g003
Figure 4. Scatter plot X—Lachnospira, Y—Nutrients/Food.
Figure 4. Scatter plot X—Lachnospira, Y—Nutrients/Food.
Nutrients 14 01221 g004
Figure 5. Scatter plot X—Bacteroides, Y—Nutrients/Food.
Figure 5. Scatter plot X—Bacteroides, Y—Nutrients/Food.
Nutrients 14 01221 g005
Figure 6. Inflammatory biomarkers by dietary pattern.
Figure 6. Inflammatory biomarkers by dietary pattern.
Nutrients 14 01221 g006
Figure 7. Erypelotrichaceae by dietary pattern in MSM.
Figure 7. Erypelotrichaceae by dietary pattern in MSM.
Nutrients 14 01221 g007
Figure 8. Scatter plot X—Erysipelotrichaceae, Y—Nutrients/Food for all MSM.
Figure 8. Scatter plot X—Erysipelotrichaceae, Y—Nutrients/Food for all MSM.
Nutrients 14 01221 g008
Figure 9. Scatter plot X—Erysipelotrichaceae, Y—Nutrients/Food for MSM-MEL.
Figure 9. Scatter plot X—Erysipelotrichaceae, Y—Nutrients/Food for MSM-MEL.
Nutrients 14 01221 g009
Figure 10. Scatter plot X—Erysipelotrichaceae, Y—Nutrients/Food for MSM-WEL.
Figure 10. Scatter plot X—Erysipelotrichaceae, Y—Nutrients/Food for MSM-WEL.
Nutrients 14 01221 g010
Figure 11. D-dimer biomarker in MSM by dietary pattern.
Figure 11. D-dimer biomarker in MSM by dietary pattern.
Nutrients 14 01221 g011
Table 1. Sample characteristics by dietary pattern.
Table 1. Sample characteristics by dietary pattern.
MEL (n = 17)WEL (n = 10)Total (n = 27)p-Value
Age (Years)46.29 ± 12.1652.20 ± 6.7548.48 ± 10.740.172
Sex, n (%) (X2) 0.303
Female1 (5.9)2 (20.0)3 (11.1)
Male16 (94.1)8 (80.0)24 (88.9)
Risk factor, n (%) (X2) 0.329
MSM13 (76.5)5 (50.0)18 (66.7)
HTX1 (5.9)2 (20.0)3 (11.1)
IDU3 (17.6)3 (30.0)6 (22.2)
Ethnicity, n (%) (X2) 0.818
Caucasian13 (76.5)8 (80.0)21 (77.8)
Latin American3 (17.6)1 (10.0)4 (14.8)
Sub-Saharan African1 (5.9)1 (10.0)2 (7.4)
Anthropometric dates
Weight (kg) (U)75.90 ± 11.2974.86 ± 12.9075.51 ± 11.670.821
Size (cm)174.71 ± 6.91170.70 ± 4.76173.22 ± 6.410.119
BMI (kg/m2) (U)24.85 ± 3.2525.75 ± 4.7925.19 ± 3.820.633
Use of antibiotic in past 6 months, n (%) (X2)3 (17.6)3 (30.0)6 (22.2)0.387
On triple ART, n (%)17 (100.0)10 (100.0)27 (100.0)0.530
INSTI-based10 (59)5 (50)15 (56.0)
NRTI-based6 (35)3 (30)9 (33.0)
PI-based1 (6)2 (20)3 (11.0)
Undetectable HIV RNA17 (100.0)10 (100.0)27 (100.0)-
Use of drugs with anti-inflammatory effects
Atorvastatine1 (6.0)0 (0)1 (6.0)-
Results are presented as mean ± SD when variables are continuous, or n (%) when categorical. Significant differences between dietary patterns (p-value < 0.05) are indicated in bold. (U): p-value calculated with the Mann-Witney U test. (X2): p-value calculated with Chi-square test. MSM: Men who have sex with men; HTX: Heterosexuals; IDU: Ex-injection drug users; BMI: Body Mass Index; ART: antiretroviral therapy.
Table 2. Intake (g/day) of each food group per dietary pattern.
Table 2. Intake (g/day) of each food group per dietary pattern.
MEL (n = 17)WEL (n = 10)Total (n = 27)p-Value
Cereals and legumes279.87 ± 100.31174.28 ± 49.27240.76 ± 98.650.001
Vegetables455.34 ± 223.45257.02 ± 179.31381.89 ± 226.670.042
Fruits (t)463.85 ± 363.98307.13 ± 218.93405.80 ± 322.590.175
Dairy products311.18 ± 195.16252.87 ± 133.03289.58 ± 174.320.547
Nuts11.75 ± 17.972.40 ± 7.598.29 ± 15.480.059
Meat (t)120.29 ± 75.29288.80 ± 156.22182.70 ± 137.160.008
White meat23.92 ± 35.64117.74 ± 103.4858.67 ± 81.360.004
Red meat96.37 ± 73.29171.06 ± 104.48124.03 ± 91.840.056
Fish106.45 ± 90.45122.02 ± 125.45112.22 ± 102.670.900
Eggs37.66 ± 48.8614.64 ± 22.3629.13 ± 42.080.189
Sugar6.51 ± 8.3044.14 ± 67.0220.45 ± 44.050.071
Olive oil (t)34.84 ± 15.9228.04 ± 13.0932.32 ± 15.050.265
Alcohol7.58 ± 12.2910.97 ± 15.498.83 ± 13.370.620
Aperitives5.91 ± 11.3828.41 ± 49.9514.24 ± 32.650.159
Potatoes2.55 ± 8.2975.00 ± 71.4029.38 ± 55.480.001
Results are presented as mean ± SD. Significant differences between dietary patterns (p-value < 0.05) are indicated in bold. (t): p-value calculated with t-test for independent samples. Other variables p-value calculated with Mann–Whitney U test.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Manzano, M.; Talavera-Rodríguez, A.; Moreno, E.; Madrid, N.; Gosalbes, M.J.; Ron, R.; Dronda, F.; Pérez-Molina, J.A.; Lanza, V.F.; Díaz, J.; et al. Relationship of Diet to Gut Microbiota and Inflammatory Biomarkers in People with HIV. Nutrients 2022, 14, 1221. https://doi.org/10.3390/nu14061221

AMA Style

Manzano M, Talavera-Rodríguez A, Moreno E, Madrid N, Gosalbes MJ, Ron R, Dronda F, Pérez-Molina JA, Lanza VF, Díaz J, et al. Relationship of Diet to Gut Microbiota and Inflammatory Biomarkers in People with HIV. Nutrients. 2022; 14(6):1221. https://doi.org/10.3390/nu14061221

Chicago/Turabian Style

Manzano, Mónica, Alba Talavera-Rodríguez, Elena Moreno, Nadia Madrid, María José Gosalbes, Raquel Ron, Fernando Dronda, José A. Pérez-Molina, Val F. Lanza, Jorge Díaz, and et al. 2022. "Relationship of Diet to Gut Microbiota and Inflammatory Biomarkers in People with HIV" Nutrients 14, no. 6: 1221. https://doi.org/10.3390/nu14061221

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop