Next Article in Journal
Lead Poisoning among Male Juveniles Due to Illegal Mining: A Case Series from South Africa
Previous Article in Journal
Postmenopausal Symptoms and Their Correlates among Saudi Women Attending Different Primary Health Centers
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Lights and Shadows of Microbiota Modulation and Cardiovascular Risk in HIV Patients

by
Pablo Villoslada-Blanco
1,
Patricia Pérez-Matute
1,2,* and
José A. Oteo
3
1
Infectious Diseases, Microbiota and Metabolism Unit, Infectious Diseases Department, Center for Biomedical Research of La Rioja (CIBIR), 26006 Logroño, La Rioja, Spain
2
Unidad Predepartamental de Enfermería, Universidad de La Rioja (UR), 26006 Logroño, La Rioja, Spain
3
Infectious Diseases Department, Hospital Universidad San Pedro-Center for Biomedical Research of La Rioja (CIBIR), 26006 Logroño, La Rioja, Spain
*
Author to whom correspondence should be addressed.
Int. J. Environ. Res. Public Health 2021, 18(13), 6837; https://doi.org/10.3390/ijerph18136837
Submission received: 31 March 2021 / Revised: 7 June 2021 / Accepted: 16 June 2021 / Published: 25 June 2021
(This article belongs to the Section Health Behavior, Chronic Disease and Health Promotion)

Abstract

:
Human immunodeficiency virus (HIV) infection is associated with premature aging and the development of aging-related comorbidities, such as cardiovascular disease (CVD). Gut microbiota (GM) disturbance is involved in these comorbidities and there is currently interest in strategies focused on modulating GM composition and/or functionality. Scientific evidence based on well-designed clinical trials is needed to support the use of prebiotics, probiotics, symbiotics, and fecal transplantation (FT) to modify the GM and reduce the incidence of CVD in HIV-infected patients. We reviewed the data obtained from three clinical trials focused on prebiotics, 25 trials using probiotics, six using symbiotics, and four using FT. None of the trials investigated whether these compounds could reduce CVD in HIV patients. The huge variability observed in the type of compound as well as the dose and duration of administration makes it difficult to adopt general recommendations and raise serious questions about their application in clinical practice.

1. Introduction

In the last few decades, the life expectancy of patients with human immunodeficiency virus (HIV) infection has significantly increased due to improvements in clinical management and, specially, the extended use of antiretroviral therapy (ART). HIV infection is considered a chronic disease and life expectancy is shorter that observed in non-infected people and the general population [1]. This shorter life expectancy is associated with premature and accelerated aging [2] as well as the continuous activation of the immune system and low-grade chronic inflammation [3]. Thus, HIV patients have higher rates of cardiovascular disease (CVD), mainly atherosclerosis, and non-acquired immune deficiency syndrome (AIDS)-defining cancers, frailty, kidney or liver diseases, and neurological complications, such as dementia, compared with uninfected individuals of the same age [4,5,6,7,8]. Recent studies have looked beyond traditional cardiovascular risk (CVR) factors and have expanded the view towards immunology, intestinal microbiome, red blood cells, and endothelial function disorders [9,10,11,12]. Thus, the presence of factors other than “classical” CVR factors requires in-depth investigation of the physiopathology of these processes in the context of HIV infection. In addition, interventions to modify both traditional and “novel” risk factors are needed in people with HIV to reduce future adverse events, counteract the consequences of accelerated aging, and improve quality of life [13].
In this context, the gut microbiota (GM) is a “novel” factor recently associated with cardiovascular events in HIV-infected patients as well as the general population. The term microbiota describes the community of microorganisms that coexists in a specific environment, including bacteria, archaea, viruses, and some unicellular eukaryotes [14]. Several studies have demonstrated that alterations in the GM play an important role in health and disease [15]. The development of next-generation sequencing technologies has allowed the identification of new markers of the physiological and pathological states of each individual, enabling an approximation and description of the GM in different medical disorders [16,17]. However, it is still unclear whether GM modifications are the cause or consequence of alterations in health and each disorder should be examined in depth. HIV infection is characterized by a set of structural changes of the gut epithelial barrier, immunological shifts, and modifications to the composition and functionality of the GM. Under normal physiological conditions, microorganisms are found in the intestinal lumen interacting with the intestinal cells in a state of symbiosis; however, when HIV infection occurs, depletion of CD4+ T lymphocytes occurs in the gut-associated lymphoid tissue. This induces rupturing of the epithelial barrier, which triggers alterations in the intestinal lumen and the composition of the microbiota (at least at the bacterial level) [18]. This dysbiosis favors the passage of microorganisms and their components to the lamina propria and, hence, to the circulation, which is known as bacterial translocation (BT), leading to subsequent intestinal and systemic inflammation [19] (Figure 1). This immune activation and persistent chronic inflammation are observed in HIV-infected patients and are strongly predictive of atherosclerosis [20] and associated with the development of non-AIDS events, which increase morbidity and mortality and substantially reduce patients’ quality of life [19,21]. ART is not able to completely reverse this situation. Preliminary studies using small numbers of patients have shown an improvement in GM composition after treatment with integrase inhibitors [18].
There is interest currently in strategies focused on reversing or modulating GM composition and/or functionality along with standard care (i.e., ART). The use of prebiotics, probiotics, symbiotics, and fecal transplantation (FT) have emerged as “novel” promising therapies to control or reverse the changes observed in GM composition in HIV infection and reduce the incidence of comorbidities, such as cardiovascular events or obesity [23]. Nevertheless, despite the good reputation of these interventions in several recurrent diseases other than HIV infection, concerns have emerged regarding the use of GM modulators in clinical practice. Thus, solid scientific evidence is required to demonstrate their use in specific populations, such as HIV-infected people. It is also essential to establish the optimal dose, route, administration frequency, and exact composition of the therapies.
The objective of the present review was to investigate whether there is strong scientific evidence (based on well-designed clinical trials) to support the use of prebiotics, probiotics, symbiotics, and FT to modify GM in HIV-infected patients.

2. Search Strategies, Inclusion/Exclusion Criteria, and Quality Assessment

A search was performed using PubMed to identify all clinical trials (using the Clinical Trial filter) including adult humans (age ≥ 19 years) that were published within the last 10 years. We limited the search to clinical trials since they are at the top of the “pyramid of scientific evidence,” just below meta-analysis and systematic reviews [24]. Meta-analyses and systematic reviews were also used to confirm the clinical trials chosen for this review. A 10-year period was chosen because 657 of the 688 results obtained from PubMed using the keywords “microbiota HIV” were published in the last 10 years, which means a large and representative proportion (95.49%) of the total investigation developed in this field. Furthermore, a search in ClinicalTrials.gov (last access: 1 March 2021) was also carried out and these studies were included and described in the present review. Following these criteria, we identified three studies concerning prebiotics, 25 concerning probiotics, six concerning symbiotics, and four concerning FT. All clinical trials analyzed passed the Critical Appraisal Skills Programme.
Eight published studies were identified using PubMed concerning the effects of prebiotics in HIV-infected patients within the last 10 years. Only five were derived from clinical trials, among which three were excluded because they were related to symbiotics and were therefore included in the symbiotics section of this review. Among the two remaining articles, only one (NCT01838915) was registered with ClinicalTrials.gov. In addition, another clinical trial (NCT04058392) appeared in this website. Table 1 shows these two clinical trials registered in ClinicalTrials.gov; however, we have discussed the results obtained from all three clinical trials that focused on prebiotics and HIV infection independently of registration on the website.
PubMed identified 35 published studies concerning probiotics in HIV-infected patients in the last 10 years. Only 15 were derived from clinical trials, among which one was excluded because it only contained data relating to symbiotics and, therefore, was incorporated in the symbiotics section of the review. Another three publications were dismissed because they did not include HIV-infected patients. We included two more clinical trials that did not appear in the PubMed search but met the inclusion criteria [25,26]. We appended one more clinical trial from the unique meta-analysis and/or systematic review published [27] that met the selection criteria and was not included previously [28]. Among the 14 remaining articles, only five were registered in ClinicalTrials.gov and a further 11 clinical trials appeared in this website. Table 2 shows all the clinical trials registered with ClinicalTrials.gov, and all 25 clinical trials that focused on probiotics and HIV infection are included in this section.
Finally, five studies concerning the effects of symbiotics (a combination of synergistically acting prebiotics and probiotics [29]) in HIV-infected patients in the last 10 years were identified using PubMed, of which only four were derived from clinical trials. Among these four studies, three were registered with ClinicalTrials.gov. Two more clinical trials were included in this website. Table 3 shows the clinical trials concerning symbiotics registered with ClinicalTrials.gov. A discussion of all six clinical trials has been included in this review. Figure 2 shows a flow chart of the study selection process for this review.

3. Microbiota Modulation to Reduce Cardiovascular Risk in HIV Patients: Scientific Evidence from Clinical Trials

3.1. Prebiotics

Administration of prebiotics in HIV patients can modify the composition of the microbiota at several taxonomical levels. However, this influence is only observed in naive patients [30], possibly because the time since diagnosis of HIV infection is shorter in naive patients compared with ART-treated patients. Thus, it is possible that virus-induced changes at the intestinal level are mild in naive patients and easier to reverse, whereas ART-treated patients could present a more established and stabilized GM. Specifically, naive HIV patients treated for six or 12 weeks with different combinations of prebiotics [short-chain galacto-oligosaccharides/long-chain fructo-oligosaccharides/pectin hydrolysate-derived acidic oligosaccharides/glutamine (scGOS/lcFOS/pAOS/glutamine)) showed significant increases in Bifidobacterium genus (2.8–15.7%, p = 0.0007) [37] and the phyla Firmicutes (including Faecalibacterium, Catenibacterium, Blautia and Eubacterium) and Actinobacteria (including Collinsella and Corynebacterium) [30]. In contrast, statistically significant decreases in Clostridium lituseburense/Clostridium histolyctum (including Clostridium perfringens and Clostridium difficile) (0.016–0.002%, p = 0.009) [37] and Eubacterium rectale/Clostridium coccoides (p = 0.035) have also been demonstrated in these patients [30]. Table 4 shows the bacteria whose relative abundance was modified by these interventions and some of their functions in health and disease.
Administration of scGOS/lcFOS/pAOS for 12 weeks was associated with a decrease in BT markers, such as sCD14 and lipopolysaccharide (LPS) in naïve patients [37]. However, no statistically significant differences were observed in these BT markers using a similar mixture of prebiotics (scGOS/lcFOS/glutamine) for six weeks [30]. These discrepancies could be due to different durations of the interventions (12 weeks vs. 6 weeks). Thus, the microbiota is stable in adulthood, and longer periods of administration are required to modify it.
Interestingly, while no changes were observed in the viral load or number of CD4+ T lymphocytes after prebiotic administration, a decrease in the activation of these lymphocytes was observed after 12-week administration of scGOS/lcFOS/pAOS (0.52%–0.27% in naive patients, p < 0.01) [37] or after a six-week intervention with scGOS/lcFOS/glutamine [30]. This suggests that, although the intervention was unable to restore the number of CD4+ T lymphocytes, it facilitated the reduction of the immune system activation, which could be beneficial for these patients, given the persistent chronic inflammation characteristics of HIV infection.
In these two studies, administration of prebiotics was safe and there were no observed modifications to hepatic or renal function [30,37]. Nevertheless, in terms of tolerability, one study reported complications (mainly diarrhea), which disappeared four weeks after the start of the intervention [37]. Administration of prebiotics in HIV patients was able to modify the composition of the microbiota and reduce the activation of CD4+ T lymphocytes, while the effects on markers of inflammation and BT remain unclear. No studies have investigated whether these changes are able to reduce the incidence of comorbidities in this population and, specifically, whether they are able to reduce CVR. Moreover, the type of compound and the duration of administration is a variable to consider when drawing conclusions and making general recommendations. Further clinical trials are required to evaluate the effects of these compounds on CVR factors in HIV-infected patients as well as follow-up studies to corroborate a significant reduction in cardiovascular events in patients who showed improvements in inflammation and intestinal dysbiosis.

3.2. Probiotics

The use of probiotics, such as Lactobacillus, Bifidobacterium, and Enterococcus, that can benefit the intestinal and immune system, may be inexpensive and clinically important as coadjutants of ART in order to reduce HIV-related morbidity and mortality [57]. The mechanisms by which probiotics may interfere with HIV were previously summarized by D’Angelo et al. [58]. In essence, probiotics can rebalance the microbiota by competing with pathogens, improving the intestinal barrier, and reducing BT, consequently decreasing systemic inflammation and restoring the mucosal immune function, lowering local inflammation [58].
Administration of probiotics in HIV-infected patients can modify the composition of the microbiota in naive and ART-treated patients. Specifically, intervention of ART-treated patients with different combinations of probiotics was associated with an increase in the relative abundance of Actinobacteria and Firmicutes phyla [250 mL/day of fermented skimmed milk supplemented with Lactobacillus rhamnosus GG (108 CFU/mL), Bifidobacterium animalis subsp. lactis B-12 (108 CFU/mL), and Lactobacillus acidophilus La-5 (107 CFU/mL) for eight weeks] [32] as well as an increase in Bifidobacterium spp. (Visbiome; twice daily for 24 weeks) [33]. A statistically significant increase in the abundance of Megamonas and Desulfovibrionales levels after 12-week administration of capsules containing 56.5 mg of living Saccharomyces boulardii were also observed [59]. In contrast, a statistically significant reduction of Bacteroidetes phylum and some species of Clostridiales and Catenibacterium were also demonstrated after the administration of 250 mL/day of fermented skimmed milk supplemented with L. rhamnosus GG (108 CFU/mL), B. animalis subsp. lactis B-12 (108 CFU/mL) and L. acidophilus La-5 (107 CFU/mL) for eight weeks [32], and after administration of capsules containing 56.5 mg of living S. boulardii, respectively [59]. In a study carried out in naive patients, a decrease in total bacterial load in stools compared with the placebo group and an increased Bifidobacterium spp. concentration along with a decrease in Clostridium spp. concentration was demonstrated after daily administration of L. rhamnosus HN001 plus Bifidobacterium lactis Bi-07 [109 CFU/mL) for 16 weeks [60]. Finally, in a study with both naive and ART-treated patients, a reduction of the orders Enterobacteriales (p = 0.018) and Erysipelotrichales (p = 0.037) entirely driven by a reduction of the Enterobacteriaceae and Erysipelotrichaceae families were observed, along with an increase in an uncultured genus from the Lachnospiraceae family (p = 0.015) and Ruminiclostridium (p = 0.023) from the Ruminicoccaceae family following administration of two capsules daily of L. rhamnosus GG (1010 CFU) for eight weeks [61]. Table 4 shows the bacteria (with different taxonomic levels) whose relative abundance was modified in response to interventions, and some of their functions in health and disease.
Concerning inflammation, twice daily administration of a 1-g packet (packet A) containing Streptococcus salivarius ssp. thermophilus (≥2.04 × 1014 CFU), Bifidobacteria represented by Bifidobacterium breve, Bifidobacterium infantis and Bifidobacterium longum (≥9.3 × 1013), L. acidophilus (≥2 billion CFU), Lactobacillus plantarum (at least 2.20 × 108 CFU), Lactobacillus casei (≥2.20 × 108 CFU), Lactobacillus delbrueckii ssp. bulgaricus (≥3 × 108 CFU) and Streptococcus faecium (≥3 × 107 CFU) for 48 weeks was not able to reduce D-dimer levels [31]. However, administration of 250 mL/day of fermented skimmed milk supplemented with L. rhamnosus GG (108 CFU/mL), B. animalis subsp. lactis B-12 (108 CFU/mL), and L. acidophilus La-5 (107 CFU/mL) for eight weeks led to reductions in levels of both D-dimer (p = 0.03) and interleukin 6 (IL-6; p = 0.06) compared with the placebo group [32]. Furthermore, 12-week administration of S. boulardii led to reduced levels of IL-6 compared with the placebo group [25]. These discrepancies could be due to the different combinations used; therefore, the positive effects of probiotics on inflammation or at least on these parameters measured [D-dimer and/or IL-6) remain unclear. In relation to the effects on BT, different combinations of probiotics (capsules containing 56.5 mg of living S. boulardii, packet A, and S. boulardii) led to decreased levels of lipopolysaccharide-binding protein (LBP) compared with that of the placebo group [25,31,59]. However, no effects were observed for sCD14 or sCD163 levels [31,32,59,61] despite the fact that sCD14 has been suggested to be a more relevant biomarker of disease progression as it reflects the host response to products of BT [62]. These results make it difficult to confirm the beneficial effects of these probiotics on BT.
On the other hand, an increase in the number of CD4+ T lymphocytes was observed in ART-treated patients after the administration of L. rhamnosus GR-1 and Lactobacillus reuteri RC-14 for 25 weeks [63]. Moreover, an increased percentage of CD4+ T lymphocytes was also observed in the probiotic group after administration of packet A [31]. Administration of Bacillus coagulans GBI-30, 6086 (Ganeden BC30; 2 × 1012 CFU) once daily for 90 days was also associated with a significant increase in the percentage of CD4+ T lymphocytes compared with the placebo group (+2.8% vs. −1.8%, p = 0.018), although the concentrations were generally unchanged in each group [28]. Nonetheless, no increase in CD4+ T lymphocytes was observed after administration of 125 mL of yoghurt fortified with L. rhamnosous GR-1 (1.23 × 109 CFU/mL) once daily for four weeks, possibly due to the shorter duration of the intervention [64]. Interestingly, a decrease was observed in the activation markers, CD38 and HLA-DR, after the administration of packet A [31] and Visbiome for 24 weeks [65].
In all these studies, the administration of probiotics was safe and showed no side effects or modifications in hepatic or renal functions [26,28,31,63,64,65,66,67].
In summary, there is some scientific evidence showing that administration of probiotics in HIV patients can modify the composition of the microbiota, increase the number of CD4 T+ lymphocytes and decrease their activation, and reduce LBP levels. Although there are more studies into probiotics than prebiotics, these studies do not address the potential effects of probiotics on CVR in patients with HIV. Moreover, it is necessary to assess whether the changes observed in markers of inflammation and BT after the ingestion of probiotics could lead to improvements in the incidence of comorbidities and, specifically, CVR, in this population. In addition, the huge variability in the type of compound used, dose, and duration administration makes it impossible to make verified and confirmed conclusions. Therefore, studies using large cohorts are required to obtain a good description of the bacterial species that are decreased in HIV-infected patients. These depleted species would be good candidates for ingestion as probiotics to restore GM, but they should pass the requirements imposed by the Food and Agriculture Organization of the United Nations and the World Health Organization to ensure that they are safe to use. Finally, the effects of these probiotics on markers of inflammation, BT, and CVR should be assessed in HIV-infected patients.

3.3. Symbiotics

The administration of symbiotics in HIV patients has been shown to modify GM composition. Specifically, 16-week administration of 10 g of agavins from Agave tequilana plus L. rhamnosus HN001 and B. lactis Bi-07 (109 CFU/mL) in naive patients led to a statistically significant decrease in total load of bacteria in stools compared with the placebo group [60]. Similarly, in another study carried out in ART-treated patients, increased levels of L. plantarum and Pediococcus pentosaceus were detected after four-week administration of Synbiotic 2000 compared with the placebo group [35]. However, administration of PMT25431 for 48 weeks in naive patients who initiated ART did not lead to significant changes in GM [34]. The functions of these bacteria in health and disease are summarized in Table 4.
In terms of inflammation, a study of naive patients demonstrated a statistically significant decrease in IL-6 levels compared with at the beginning of the intervention following administration of 10 g of agavins from A. tequilana plus L. rhamnosus HN001 and B. lactis Bi-07 (109 CFU/mL) [60]. However, administration of PMT25431 over 48 weeks in naive patients who initiated ART did not induce additional changes in IL-6 levels [34]. Moreover, there were no effects on plasma levels of TNF-α, IL-1β, IL-10, sCD14, or sCD163 in naive HIV-infected patients after the administration of 10 g of agavins from A. tequilana plus L. rhamnosus HN001 and B. lactis Bi-07 (109 CFU/mL) [60]. Administration of PMT25341 over 48 weeks showed no effects in naive patients who initiated ART [34], and no effects were observed in ART-treated patients after the administration of Synbiotic 2000 over four weeks [35].
Increased levels of CD4+ T lymphocytes were observed in naive HIV-infected patients after the administration of 10 g of agavins compared with levels at the beginning of treatment [60]. However, no effects were observed after four-week administration of Synbiotic 2000 in ART-treated patients [35]. These discrepancies could be due to different lengths of intervention, different products used, and the fact that, in the second study, patients were treated with ART and their GM may have been more established, as previously mentioned. Finally, administration of PMT25341 for 48 weeks in naive HIV-infected patients who initiated ART did not induce additional changes compared with those exerted by ART in terms of levels and activation of CD4+ T lymphocytes [34].
In all these studies, administration of symbiotics was safe and without side effects [35,36,60]. Furthermore, administration of LACTOFOS twice daily for 30 weeks in naive patients significantly reduced the incidence of diarrhea (−21.8%), nausea and/or vomiting (−28.8%), constipation (−13.2%), and dyspepsia (−24.5%) [36].
In general terms, these results showed that administration of symbiotics in HIV-infected patients modified the composition of the microbiota, while the effects on the number of CD4+ T lymphocytes and the markers of inflammation and BT were not as clear as in the case of probiotics, which is interesting considering that symbiotics are a mixture of probiotics and prebiotics. However, these discrepancies could be due to the use of different probiotics and/or different durations of administration. Similarly, as observed with prebiotics and probiotics, no studies have focused on the long-term consequences of these changes. As there is a huge variability in the type of compound, dose, and duration of administration, it is not possible to draw conclusions and make general recommendations.

3.4. Fecal Transplantation

FT is the transfer of stools from a healthy donor into a patient with a disease characterized by a significantly altered microbiome. Its objective is to restore the microbiota and, therefore, treat the disease [68]. FT has gained acceptance as a safe and effective treatment for recurrent Clostridioides difficile infection (CDI)-associated diarrhea [69,70,71,72,73]. While HIV patients treated for CDI using FT did not show any adverse events, 14% of patients with inflammatory bowel disease experienced a disease flare that required hospitalization in some cases [74]. Therefore, FT could involve an uncontrollable risk, particularly in immunocompromised patients, if donor samples are not checked [75,76,77]. Thus, more well-designed studies and clinical trials are required.
To date, four clinical trials have been registered with ClinicalTrials.gov (Table 5) and the results of one trial have been published and was identified in PubMed (NCT03008941). In this clinical trial, ART-treated patients were orally administered 10 capsules of fecal content for one week followed by six-week treatment with five capsules. A significant increase in alpha diversity was observed in these patients compared with those receiving placebo. Long-lasting effects during a 48-week follow up were also observed. Furthermore, changes in beta diversity and increased abundance of different members of the Lachnospiraceae family (Anaritides spp., Blautia spp., Dorea spp., and Fusicatenibacter spp.) and the Ruminicoccaceae family were observed in patients treated with fecal microbiota transplantation (FMT). A statistically significant decrease was also observed in intestinal fatty acid-binding protein (IFABP), which is a biomarker of intestinal injury that independently predicts mortality in treated HIV-patients [78,79]. However, levels of CD4 T+ lymphocytes, CD4/CD8 ratio, lymphocyte markers of immune activation, and plasma markers of inflammation and BT were not affected by FMT. Finally, no serious adverse events were detected during the intervention or follow up [80]. The involvement of these bacteria in health and disease is shown in Table 4.
Finally, Table 6 includes the list of “biotics” analyzed and their definition or composition.

4. Conclusions

Among all the GM modulatory strategies analysed, our review identified probiotics as the most beneficial therapy in HIV patients to improve their immune and inflammatory states. However, very few studies have focused on their effects on specific CVR markers in HIV patients or the long-term consequences of these effects on inflammation. Follow-up studies are required to determine whether the effects of probiotics on BT and inflammation lead to statistically significant reductions in cardiovascular events with improvements in inflammation, intestinal dysbiosis and CVR factors. In addition, huge variability in the type of compound, dose, and length of administration was observed among the trials, which raises serious questions about the practical value of these approaches. Long-term interventions are the most potent methods to modify the GM and reverse inflammation.
In summary, HIV-infected patients have significantly increased rates of cardiovascular events, even after controlling for traditional CVR factors and this significantly affects their quality of life and represents a significant increase in healthcare costs. The use of probiotics or other GM modulatory strategies along with the standard of care (i.e., ART) could be a good strategy to modify CVR factors in this population. However, to date, the results obtained from clinical trials are not conclusive and more prospective controlled studies with larger number of patients and with standardized dose and duration of therapies are needed for appropriate applications in clinical practice. Patient follow-up is also required to confirm the results.

Author Contributions

P.P.-M. and J.A.O. conceived the study. P.V.-B. conducted the literature research and the manuscript selection. P.P.-M. and P.V.-B. wrote the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

PVB was supported by a predoctoral grant from Consejería de Desarrollo Económico e Innovación (Government of La Rioja).

Institutional Review Board Statement

Non applicable.

Informed Consent Statement

Non applicable.

Data Availability Statement

Non applicable.

Acknowledgments

We thank Consejería de Desarrollo Económico e Innovación and Fundación Rioja Salud (Government of La Rioja).

Conflicts of Interest

The authors have no conflict of interest to declare.

References

  1. Deeks, S.G.; Lewin, S.R.; Havlir, D.V. The end of AIDS: HIV infection as a chronic disease. Lancet 2013, 382, 1523–1533. [Google Scholar] [CrossRef] [Green Version]
  2. Desai, S.N.; Landay, A.L. HIV and aging: Role of the microbiome. Curr. Opin. HIV AIDS 2018, 13, 22–27. [Google Scholar] [CrossRef] [PubMed]
  3. Desai, S.N.; Landay, A.L. Early immune senescence in HIV disease. Curr. HIV/AIDS Rep. 2010, 7, 4–10. [Google Scholar] [CrossRef] [Green Version]
  4. Triant, V.A.; Lee, H.; Hadigan, C.; Grinspoon, S.K. Increased acute myocardial infarction rates and cardiovascular risk factors among patients with human immunodeficiency virus disease. J. Clin. Endocrinol. Metab. 2007, 92, 2506–2512. [Google Scholar] [CrossRef] [PubMed]
  5. Negredo, E.; Back, D.; Blanco, J.R.; Blanco, J.; Erlandson, K.M.; Garolera, M.; Guaraldi, G.; Mallon, P.; Moltó, J.; Serra, J.A.; et al. Aging in HIV-Infected Subjects: A New Scenario and a New View. BioMed Res. Int. 2017, 2017, 58972. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Desquilbet, L.; Jacobson, L.P.; Fried, L.P.; Phair, J.P.; Jamieson, B.D.; Holloway, M.; Margolick, J.B. HIV-1 infection is associated with an earlier occurrence of a phenotype related to frailty. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 2007, 62, 1279–1286. [Google Scholar] [CrossRef] [Green Version]
  7. Choi, A.I.; Shlipak, M.G.; Hunt, P.W.; Martin, J.N.; Deeks, S.G. HIV-infected persons continue to lose kidney function despite successful antiretroviral therapy. AIDS 2009, 23, 2143–2149. [Google Scholar] [CrossRef] [Green Version]
  8. Alejos, B.; Hernando, V.; López-Aldeguer, J.; Segura, F.; Antonio Oteo, J.; Rubio, R.; Sanvisens, A.; Sobrino, P.; del Amo, J.; Coris, C. Overall and cause-specific mortality in HIV-positive subjects compared to the general population. J. Int. AIDS Soc. 2014, 17, 19711. [Google Scholar] [CrossRef]
  9. Haissman, J.M.; Knudsen, A.; Hoel, H.; Kjær, A.; Kristoffersen, U.S.; Berge, R.K.; Katzenstein, T.L.; Svardal, A.; Ueland, T.; Aukrust, P.; et al. Microbiota-Dependent Marker TMAO Is Elevated in Silent Ischemia but Is Not Associated with First-Time Myocardial Infarction in HIV Infection. J. Acquir. Immune Defic. Syndr. 2016, 71, 130–136. [Google Scholar] [CrossRef]
  10. Srinivasa, S.; Fitch, K.V.; Lo, J.; Kadar, H.; Knight, R.; Wong, K.; Abbara, S.; Gauguier, D.; Capeau, J.; Boccara, F.; et al. Plaque burden in HIV-infected patients is associated with serum intestinal microbiota-generated trimethylamine. AIDS 2015, 29, 443–452. [Google Scholar] [CrossRef] [Green Version]
  11. Al-Kindi, S.G.; Kim, C.H.; Morris, S.R.; Freeman, M.L.; Funderburg, N.T.; Rodriguez, B.; McComsey, G.A.; Dalton, J.E.; Simon, D.I.; Lederman, M.M.; et al. Elevated red cell distribution width identifies elevated cardiovascular disease risk in patients with HIV infection. J. Acquir. Immune Defic. Syndr. 2017, 74, 298–302. [Google Scholar] [CrossRef] [Green Version]
  12. Haissman, J.M.; Haugaard, A.K.; Knudsen, A.; Kristoffersen, U.S.; Seljeflot, I.; Pedersen, K.K.; Lebech, A.M.; Hasbak, P.; Kjær, A.; Ostrowski, S.R.; et al. Marker of endothelial dysfunction asymmetric dimethylarginine is elevated in HIV infection but not associated with subclinical atherosclerosis. J. Acquir. Immune Defic. Syndr. 2016, 73, 507–513. [Google Scholar] [CrossRef] [PubMed]
  13. Maggi, P.; Di Biagio, A.; Rusconi, S.; Cicalini, S.; D’Abbraccio, M.; D’Ettorre, G.; Martinelli, C.; Nunnari, G.; Sighinolfi, L.; Spagnuolo, V.; et al. Cardiovascular risk and dyslipidemia among persons living with HIV: A review. BMC Infect. Dis. 2017, 17, 551. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Lynch, S.V.; Pedersen, O. The Human Intestinal Microbiome in Health and Disease. N. Engl. J. Med. 2016, 375, 2369–2379. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Villanueva-Millán, M.J.; Pérez-Matute, P.; Oteo, J.A. Gut microbiota: A key player in health and disease. A review focused on obesity. J. Physiol. Biochem. 2015, 71, 509–525. [Google Scholar] [CrossRef] [PubMed]
  16. Waldman, A.J.; Balskus, E.P. The Human Microbiota, Infectious Disease, and Global Health: Challenges and Opportunities. ACS Infect. Dis. 2018, 4, 14–26. [Google Scholar] [CrossRef]
  17. Zhang, Y.; Lun, C.Y.; Tsui, S.K.W. Metagenomics: A newway to illustrate the crosstalk between infectious diseases and host microbiome. Int. J. Mol. Sci. 2015, 16, 26263–26279. [Google Scholar] [CrossRef] [Green Version]
  18. Villanueva-Millán, M.J.; Pérez-Matute, P.; Recio-Fernández, E.; Rosales, J.M.L.; Oteo, J.A. Differential effects of antiretrovirals on microbial translocation and gut microbiota composition of HIV-infected patients. J. Int. AIDS Soc. 2017, 20, 21526. [Google Scholar] [CrossRef] [PubMed]
  19. Zevin, A.S.; McKinnon, L.; Burgener, A.; Klatt, N.R. Microbial translocation and microbiome dysbiosis in HIV-associated immune activation. Curr. Opin. HIV AIDS 2016, 11, 182–190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Trøseid, M.; Manner, I.W.; Pedersen, K.K.; Haissman, J.M.; Kvale, D.; Nielsen, S.D. Microbial translocation and cardiometabolic risk factors in HIV infection. AIDS Res. Hum. Retrovir. 2014, 30, 514–522. [Google Scholar] [CrossRef] [Green Version]
  21. Marchetti, G.; Tincati, C.; Silvestri, G. Microbial translocation in the pathogenesis of HIV infection and AIDS. Clin. Microbiol. Rev. 2013, 26, 2–18. [Google Scholar] [CrossRef] [Green Version]
  22. Pérez-Matute, P.; Íñiguez, M.; Villanueva-Millán, M.J.; Oteo, J.A. Chapter 32–The Oral, Genital and Gut Microbiome in HIV Infection; Faintuch, J., Faintuch Therapy, Strategic Applications SBT-M and M in D, Eds.; Academic Press: Boca Raton, FL, USA, 2019; pp. 307–323. [Google Scholar]
  23. El-Far, M.; Tremblay, C.L. Gut microbial diversity in HIV infection post combined antiretroviral therapy: A key target for prevention of cardiovascular disease. Curr. Opin. HIV AIDS 2018, 13, 38–44. [Google Scholar] [CrossRef] [PubMed]
  24. Varoni, E.M.; Lodi, G.; Iriti, M. Efficacy behind activity–phytotherapeutics are not different from pharmaceuticals. Pharm. Biol. 2015, 53, 404–406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Villar-García, J.; Hernández, J.J.; Güerri-Fernández, R.; González, A.; Lerma, E.; Guelar, A.; Saenz, D.; Sorlí, L.; Montero, M.; Horcajada, J.P.; et al. Effect of Probiotics (Saccharomyces boulardii) on Microbial Translocation and Inflammation in HIV-Treated Patients. JAIDS J. Acquir. Immune Defic. Syndr. 2015, 68, 256–263. [Google Scholar] [CrossRef] [PubMed]
  26. Hummelen, R.; Changalucha, J.; Butamanya, N.L.; Cook, A.; Habbema, J.D.F.; Reid, G. Lactobacillus rhamnosus GR-1 and L. reuteri RC-14 to prevent or cure bacterial vaginosis among women with HIV. Int. J. Gynecol. Obstet. 2010, 111, 245–248. [Google Scholar] [CrossRef]
  27. Kazemi, A.; Djafarian, K.; Speakman, J.R.; Sabour, P.; Soltani, S.; Shab-Bidar, S. Effect of Probiotic Supplementation on CD4 Cell Count in HIV-Infected Patients: A Systematic Review and Meta-analysis. J. Diet. Suppl. 2018, 15, 776–788. [Google Scholar] [CrossRef]
  28. Yang, O.O.; Kelesidis, T.; Cordova, R.; Khanlou, H. Immunomodulation of antiretroviral drug-suppressed chronic HIV-1 infection in an oral probiotic double-blind placebo-controlled trial. AIDS Res. Hum. Retrovir. 2014, 30, 988–995. [Google Scholar] [CrossRef]
  29. Gibson, G.R.; Roberfroid, M.B. Dietary Modulation of the Human Colonic Microbiota: Introducing the Concept of Prebiotics. J. Nutr. 1995, 125, 1401–1412. [Google Scholar] [CrossRef]
  30. Serrano-Villar, S.; Vázquez-Castellanos, J.F.; Vallejo, A.; Latorre, A.; Sainz, T.; Ferrando-Martínez, S.; Rojo, D.; Martínez-Botas, J.; Del Romero, J.; Madrid, N.; et al. The effects of prebiotics on microbial dysbiosis, butyrate production and immunity in HIV-infected subjects. Mucosal Immunol. 2017, 10, 1279–1293. [Google Scholar] [CrossRef] [Green Version]
  31. D’Ettorre, G.; Ceccarelli, G.; Giustini, N.; Serafino, S.; Calantone, N.; De Girolamo, G.; Bianchi, L.; Bellelli, V.; Ascoli-Bartoli, T.; Marcellini, S.; et al. Probiotics reduce inflammation in antiretroviral treated, HIV-infected individuals: Results of the “Probio-HIV” clinical trial. PLoS ONE 2015, 10, e0137200. [Google Scholar] [CrossRef]
  32. Stiksrud, B.; Nowak, P.; Nwosu, F.C.; Kvale, D.; Thalme, A.; Sonnerborg, A.; Ueland, P.M.; Holm, K.; Birkeland, S.E.; Dahm, A.E.A.; et al. Reduced levels of D-dimer and changes in gut microbiota composition after probiotic intervention in HIV-infected individuals on stable ART. J. Acquir. Immune Defic. Syndr. 2015, 70, 329–337. [Google Scholar] [CrossRef] [Green Version]
  33. Ceccarelli, G.; Fratino, M.; Selvaggi, C.; Giustini, N.; Serafino, S.; Schietroma, I.; Corano Scheri, G.; Pavone, P.; Passavanti, G.; Alunni Fegatelli, D.; et al. A pilot study on the effects of probiotic supplementation on neuropsychological performance and microRNA-29a-c levels in antiretroviral-treated HIV-1-infected patients. Brain Behav. 2017, 7, e00756. [Google Scholar] [CrossRef] [PubMed]
  34. Serrano-Villar, S.; De Lagarde, M.; Vázquez-Castellanos, J.; Vallejo, A.; Bernadino, J.I.; Madrid, N.; Matarranz, M.; Diáz-Santiago, A.; Gutiérrez, C.; Cabello, A.; et al. Effects of immunonutrition in advanced human immunodeficiency virus disease: A randomized placebo-controlled clinical trial (Promaltia study). Clin. Infect. Dis. 2019, 68, 120–130. [Google Scholar] [CrossRef]
  35. Schunter, M.; Chu, H.; Hayes, T.L.; McConnell, D.; Crawford, S.S.; Luciw, P.A.; Bengmark, S.; Asmuth, D.M.; Brown, J.; Bevins, C.L.; et al. Randomized pilot trial of a synbiotic dietary supplement in chronic HIV-1 infection. BMC Complement. Altern. Med. 2012, 12, 84. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. da Silveira, E.A.; Falco, M.O.; Nery, M.W.; Turchi, M.D. Effectiveness of nutritional treatment and synbiotic use on gastrointestinal symptoms reduction in HIV-infected patients: Randomized clinical trial. Clin. Nutr. 2017, 36, 680–685. [Google Scholar] [CrossRef]
  37. Gori, A.; Rizzardini, G.; Van’T Land, B.; Amor, K.B.; Van Schaik, J.; Torti, C.; Quirino, T.; Tincati, C.; Bandera, A.; Knol, J.; et al. Specific prebiotics modulate gut microbiota and immune activation in HAART-naive HIV-infected adults: Results of the “cOPA” pilot randomized trial. Mucosal Immunol. 2011, 4, 554–563. [Google Scholar] [CrossRef] [PubMed]
  38. Lopez-Siles, M.L.; Duncan, S.H.; Garcia-Gil, L.J.; Martinez-Medina, M. Faecalibacterium prausnitzii: From microbiology to diagnostics and prognostics. ISME J. 2017, 11, 841–852. [Google Scholar] [CrossRef] [PubMed]
  39. Kuang, Y.-S.; Lu, J.-H.; Li, S.-H.; Li, J.-H.; Yuan, M.-Y.; He, J.-R.; Chen, N.-N.; Xiao, W.-Q.; Shen, S.-Y.; Qiu, L.; et al. Connections between the human gut microbiome and gestational diabetes mellitus. Gigascience 2017, 6, 1–12. [Google Scholar] [CrossRef]
  40. Liu, X.; Mao, B.; Gu, J.; Wu, J.; Cui, S.; Wang, G.; Zhao, J.; Zhang, H.; Chen, W. Blautia-a new functional genus with potential probiotic properties? Gut Microbes 2021, 13, 1–21. [Google Scholar] [CrossRef]
  41. 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]
  42. Jia, W.; Rajani, C.; Xu, H.; Zheng, X. Gut microbiota alterations are distinct for primary colorectal cancer and hepatocellular carcinoma. Protein Cell 2021, 12, 374–393. [Google Scholar] [CrossRef] [PubMed]
  43. Mukherjee, A.; Lordan, C.; Ross, R.P.; Cotter, P.D. Gut microbes from the phylogenetically diverse genus Eubacterium and their various contributions to gut health. Gut Microbes 2020, 12, 1802866. [Google Scholar] [CrossRef]
  44. Hiel, S.; Gianfrancesco, M.A.; Rodriguez, J.; Portheault, D.; Leyrolle, Q.; Bindels, L.B.; da Silveira Cauduro, C.G.; Mulders, M.D.G.H.; Zamariola, G.; Azzi, A.-S. Link between gut microbiota and health outcomes in inulin-treated obese patients: Lessons from the Food4Gut multicenter randomized placebo-controlled trial. Clin. Nutr. 2020, 39, 3618–3628. [Google Scholar] [CrossRef] [PubMed]
  45. Zhang, X.; Shen, D.; Fang, Z.; Jie, Z.; Qiu, X.; Zhang, C.; Chen, Y.; Ji, L. Human gut microbiota changes reveal the progression of glucose intolerance. PLoS ONE 2013, 8, e71108. [Google Scholar] [CrossRef] [PubMed]
  46. Seddik, H.A.; Bendali, F.; Gancel, F.; Fliss, I.; Spano, G.; Drider, D. Lactobacillus plantarum and Its Probiotic and Food Potentialities. Probiotics Antimicrob. Proteins 2017, 9, 111–122. [Google Scholar] [CrossRef]
  47. Bian, X.; Yang, L.; Wu, W.; Lv, L.; Jiang, X.; Wang, Q.; Wu, J.; Li, Y.; Ye, J.; Fang, D.; et al. Pediococcus pentosaceus LI05 alleviates DSS-induced colitis by modulating immunological profiles, the gut microbiota and short-chain fatty acid levels in a mouse model. Microb. Biotechnol. 2020, 13, 1228–1244. [Google Scholar] [CrossRef]
  48. Qin, P.; Zou, Y.; Dai, Y.; Luo, G.; Zhang, X.; Xiao, L. Characterization a Novel Butyric Acid-Producing Bacterium Collinsella aerofaciens Subsp. Shenzhenensis Subsp. Nov. Microorganisms 2019, 7, 78. [Google Scholar] [CrossRef] [Green Version]
  49. Bernard, K. The genus corynebacterium and other medically relevant coryneform-like bacteria. J. Clin. Microbiol. 2012, 50, 3152–3158. [Google Scholar] [CrossRef] [Green Version]
  50. O’Callaghan, A.; van Sinderen, D. Bifidobacteria and Their Role as Members of the Human Gut Microbiota. Front. Microbiol. 2016, 7, 925. [Google Scholar] [CrossRef] [Green Version]
  51. Hagiya, H.; Kimura, K.; Nishi, I.; Yamamoto, N.; Yoshida, H.; Akeda, Y.; Tomono, K. Desulfovibrio desulfuricans bacteremia: A case report and literature review. Anaerobe 2018, 49, 112–115. [Google Scholar] [CrossRef]
  52. Kiu, R.; Caim, S.; Alcon-Giner, C.; Belteki, G.; Clarke, P.; Pickard, D.; Dougan, G.; Hall, L.J. Preterm Infant-Associated Clostridium tertium, Clostridium cadaveris, and Clostridium paraputrificum Strains: Genomic and Evolutionary Insights. Genome Biol. Evol. 2017, 9, 2707–2714. [Google Scholar] [CrossRef]
  53. Guh, A.Y.; Kutty, P.K. Clostridioides difficile Infection. Ann. Intern. Med. 2018, 169, ITC49–ITC64. [Google Scholar] [CrossRef]
  54. Wang, Y.; Wan, X.; Wu, X.; Zhang, C.; Liu, J.; Hou, S. Eubacterium rectale contributes to colorectal cancer initiation via promoting colitis. Gut Pathog. 2021, 13, 2. [Google Scholar] [CrossRef] [PubMed]
  55. Ramirez, D.G.M. Enterobacter Infections; StatPearls. Available online: https://www.ncbi.nlm.nih.gov/books/NBK559296/ (accessed on 30 June 2020).
  56. Wexler, H.M. Bacteroides: The good, the bad, and the nitty-gritty. Clin. Microbiol. Rev. 2007, 20, 593–621. [Google Scholar] [CrossRef] [Green Version]
  57. Hummelen, R.; Vos, A.P.; Van’T Land, B.; Van Norren, K.; Reid, G. Altered host-microbe interaction in HIV: A target for intervention with pro- and prebiotics. Int. Rev. Immunol. 2010, 29, 485–513. [Google Scholar] [CrossRef] [Green Version]
  58. D’Angelo, C.; Reale, M.; Costantini, E. Microbiota and probiotics in health and HIV infection. Nutrients 2017, 9, 615. [Google Scholar] [CrossRef] [Green Version]
  59. Villar-García, J.; Güerri-Fernández, R.; Moya, A.; Gonzalez, A.; Hernandez, J.J.; Lerma, E.; Guelar, A.; Sorli, L.; Horcajada, J.P.; Artacho, A.; et al. Impact of probiotic Saccharomyces boulardii on the gut microbiome composition in HIV-treated patients: A double-blind, randomised, placebo-controlled trial. PLoS ONE 2017, 12, e0173802. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. González-Hernández, L.A.; Jave-Suarez, L.F.; Fafutis-Morris, M.; Montes-Salcedo, K.E.; Valle-Gutierrez, L.G.; Campos-Loza, A.E.; Enciso-Gómez, L.F.; Andrade-Villanueva, J.F. Synbiotic therapy decreases microbial translocation and inflammation and improves immunological status in HIV-infected patients: A double-blind randomized controlled pilot trial. Nutr. J. 2012, 11, 90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Arnbjerg, C.J.; Vestad, B.; Hov, J.R.; Pedersen, K.K.; Jespersen, S.; Johannesen, H.H.; Holm, K.; Halvorsen, B.; Fallentin, E.; Hansen, A.E.; et al. Effect of lactobacillus rhamnosus GG supplementation on intestinal inflammation assessed by PET/MRI scans and Gut microbiota composition in HIV-infected individuals. J. Acquir. Immune Defic. Syndr. 2018, 78, 450–457. [Google Scholar] [CrossRef]
  62. Sandler, N.G.; Wand, H.; Roque, A.; Law, M.; Nason, M.C.; Nixon, D.E.; Pedersen, C.; Ruxrungtham, K.; Lewin, S.R.; Emery, S.; et al. Plasma levels of soluble CD14 independently predict mortality in HIV infection. J. Infect. Dis. 2011, 203, 780–790. [Google Scholar] [CrossRef] [Green Version]
  63. Hummelen, R.; Changalucha, J.; Butamanya, N.L.; Koyama, T.E.; Cook, A.; Habbema, J.D.F.; Reid, G. Effect of 25 weeks probiotic supplementation on immune function of HIV patients. Gut Microbes 2011, 2, 80–85. [Google Scholar] [CrossRef] [Green Version]
  64. Hummelen, R.; Hemsworth, J.; Changalucha, J.; Butamanya, N.L.; Hekmat, S.; Habbema, J.D.F.; Reid, G. Effect of micronutrient and probiotic fortified yogurt on immune-function of anti-retroviral therapy naive HIV patients. Nutrients 2011, 3, 897–909. [Google Scholar] [CrossRef] [Green Version]
  65. D’Ettorre, G.; Rossi, G.; Scagnolari, C.; Andreotti, M.; Giustini, N.; Serafino, S.; Schietroma, I.; Corano Scheri, G.; Najafi Fard, S.; Trinchieri, V.; et al. Probiotic supplementation promotes a reduction in T-cell activation, an increase in Th17 frequencies, and a recovery of intestinal epithelium integrity and mitochondrial morphology in ART-treated HIV-1-positive patients. Immun. Inflamm. Dis. 2017, 5, 244–260. [Google Scholar] [CrossRef]
  66. Moroti, C.; Magri, L.F.S.; de Rezende Costa, M.; Cavallini, D.C.U.; Sivieri, K. Effect of the consumption of a new symbiotic shake on glycemia and cholesterol levels in elderly people with type 2 diabetes mellitus. Lipids Health Dis. 2012, 11, 29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Hemsworth, J.C.; Hekmat, S.; Reid, G. Micronutrient supplemented probiotic yogurt for HIV-infected adults taking HAART in London, Canada. Gut Microbes 2012, 3, 414–419. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Vindigni, S.M.; Surawicz, C.M. Fecal Microbiota Transplantation. Gastroenterol. Clin. N. Am. 2017, 46, 171–185. [Google Scholar] [CrossRef] [PubMed]
  69. 69 Britton, R.A.; Young, V.B. Role of the intestinal microbiota in resistance to colonization by Clostridium difficile. Gastroenterology 2014, 146, 1547–1553. [Google Scholar] [CrossRef] [Green Version]
  70. Kuijper, E.J.; Coignard, B.; Tüll, P.; Poxton, I.; Brazier, J.; Duerden, B.; Delmée, M.; Mastrantonio, P.; Gastmeier, P.; Barbut, F.; et al. Emergence of Clostridium difficile-associated disease in North America and Europe. Clin. Microbiol. Infect. 2006, 12 (Suppl. S6), 2–18. [Google Scholar] [CrossRef] [Green Version]
  71. Chang, J.Y.; Antonopoulos, D.A.; Kalra, A.; Tonelli, A.; Khalife, W.T.; Schmidt, T.M.; Young, V.B. Decreased Diversity of the Fecal Microbiome in Recurrent Clostridium difficile–Associated Diarrhea. J. Infect. Dis. 2008, 197, 435–438. [Google Scholar] [CrossRef] [Green Version]
  72. Gough, E.; Shaikh, H.; Manges, A.R. Systematic review of intestinal microbiota transplantation (fecal bacteriotherapy) for recurrent clostridium difficile infection. Clin. Infect. Dis. 2011, 53, 994–1002. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Cammarota, G.; Masucci, L.; Ianiro, G.; Bibbò, S.; Dinoi, G.; Costamagna, G.; Sanguinetti, M.; Gasbarrini, A. Randomised clinical trial: Faecal microbiota transplantation by colonoscopy vs. vancomycin for the treatment of recurrent Clostridium difficile infection. Aliment. Pharmacol. Ther. 2015, 41, 835–843. [Google Scholar] [CrossRef]
  74. Kelly, C.R.; Ihunnah, C.; Fischer, M.; Khoruts, A.; Surawicz, C.; Afzali, A.; Aroniadis, O.; Barto, A.; Borody, T.; Giovanelli, A.; et al. Fecal microbiota transplant for treatment of clostridium difficile infection in immunocompromised patients. Am. J. Gastroenterol. 2014, 109, 1065–1071. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Kassam, Z.; Lee, C.H.; Yuan, Y.; Hunt, R.H. Fecal microbiota transplantation for clostridium difficile infection: Systematic review and meta-analysis. Am. J. Gastroenterol. 2013, 108, 500–508. [Google Scholar] [CrossRef]
  76. Smits, L.P.; Bouter, K.E.C.; De Vos, W.M.; Borody, T.J.; Nieuwdorp, M. Therapeutic potential of fecal microbiota transplantation. Gastroenterology 2013, 145, 946–953. [Google Scholar] [CrossRef] [PubMed]
  77. Petrof, E.O.; Khoruts, A. From stool transplants to next-generation microbiota therapeutics. Gastroenterology 2014, 146, 1573–1582. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Tenorio, A.R.; Zheng, Y.; Bosch, R.J.; Krishnan, S.; Rodriguez, B.; Hunt, P.W.; Plants, J.; Seth, A.; Wilson, C.C.; Deeks, S.G.; et al. Soluble markers of inflammation and coagulation but not T-cell activation predict non-AIDS-defining morbid events during suppressive antiretroviral treatment. J. Infect. Dis. 2014, 210, 1248–1259. [Google Scholar] [CrossRef] [Green Version]
  79. Hunt, P.W.; Sinclair, E.; Rodriguez, B.; Shive, C.; Clagett, B.; Funderburg, N.; Robinson, J.; Huang, Y.; Epling, L.; Martin, J.N.; et al. Gut epithelial barrier dysfunction and innate immune activation predict mortality in treated HIV infection. J. Infect. Dis. 2014, 210, 1228–1238. [Google Scholar] [CrossRef] [Green Version]
  80. Serrano-Villar, S.; Talavera-Rodríguez, A.; Gosalbes, M.J.; Madrid, N.; Pérez-Molina, J.A.; Elliott, R.J.; Navia, B.; Lanza, V.F.; Vallejo, A.; Osman, M. Fecal microbiota transplantation in HIV: A pilot placebo-controlled study. Nat. Commun. 2021, 12, 1–13. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Under normal physiological conditions, microorganisms in the intestinal lumen interact with intestinal cells in a state of symbiosis; however, when HIV infection occurs, depletion of CD4+ T lymphocytes occurs in the gut-associated lymphoid tissue. This is accompanied by rupturing of the epithelial barrier, which triggers alterations in the intestinal lumen as well as the composition of the microbiota (at least at the bacterial level) [18]. This dysbiosis favors the passage of microorganisms and their components to the lamina propria and, therefore, to the circulation, which is known as bacterial translocation (BT), leading to subsequent intestinal and systemic inflammation [19]. (A) Gut homeostasis. (B) Gut dysbiosis after HIV infection. Modified from Pérez-Matute P et al. [22].
Figure 1. Under normal physiological conditions, microorganisms in the intestinal lumen interact with intestinal cells in a state of symbiosis; however, when HIV infection occurs, depletion of CD4+ T lymphocytes occurs in the gut-associated lymphoid tissue. This is accompanied by rupturing of the epithelial barrier, which triggers alterations in the intestinal lumen as well as the composition of the microbiota (at least at the bacterial level) [18]. This dysbiosis favors the passage of microorganisms and their components to the lamina propria and, therefore, to the circulation, which is known as bacterial translocation (BT), leading to subsequent intestinal and systemic inflammation [19]. (A) Gut homeostasis. (B) Gut dysbiosis after HIV infection. Modified from Pérez-Matute P et al. [22].
Ijerph 18 06837 g001
Figure 2. Flow chart of the study selection concerning the effects of prebiotics, probiotics, and symbiotics in HIV-infected people.
Figure 2. Flow chart of the study selection concerning the effects of prebiotics, probiotics, and symbiotics in HIV-infected people.
Ijerph 18 06837 g002
Table 1. Clinical trials concerning prebiotics registered in ClinicalTrials.gov.
Table 1. Clinical trials concerning prebiotics registered in ClinicalTrials.gov.
NCT NumberCountryReferenceStatusInterventionNumber of ParticipantsAge of Participants (Years)ObjectiveMain Outcomes
NCT01838915SpainSerrano Villar et al., 2017 [30]CompletedscGOS/lcFOS + glutamine
6 weeks
4518–70To assess the effects in naive and ART-treated HIV-infected patients on:
  • safety
  • GM composition
  • bacterial translocation
  • immune activation
  • endothelial dysfunction.
  • Safe.
  • Modification of GM composition in naive patients only.
  • Decreased activation of CD4+ T lymphocytes in naive and ART-treated patients.
NCT04058392CanadaNot yet recruitingCamu camu (2 capsules/day)
12 weeks
22
(enrolled)
≥18To evaluate the effects in ART-treated HIV-infected patients on:
  • tolerance
  • GM composition
  • inflammation
  • changes of gut barrier.
ART, antiretroviral treatment; HIV, human immunodeficiency virus); GM, gut microbiota; lcFOS, long-chain fructo-oligosaccharide; scGOS, short-chain galacto-oligosaccharides.
Table 2. Clinical trials concerning probiotics registered in ClinicalTrials.gov.
Table 2. Clinical trials concerning probiotics registered in ClinicalTrials.gov.
NCT NumberCountryReferenceStatusInterventionNumber of ParticipantsAge of Participants (Years)ObjectiveMain Outcomes
NCT02164344ItalyD’Etorre et al., 2015 [31]UnknownS. salivarius spp. thermophilus 2.04 × 1014 CFU, B. breve, B. infantis, B. longum 9.3 × 1013 CFU, L. acidophilus 2 × 1012 CFU, L. plantarum 2.2 × 108 CFU, L. casei 2.2 × 108 CFU, L. delbrueckii ssp. Bulgaricus 3 × 108 CFU and S. faecium 3 × 107 CFU
1 daily dose
48 weeks
3018–75To determine the effects in ART-treated HIV-infected patients on:
  • microbial translocation-induced inflammation
  • immune function and activation.
  • Safe.
  • Decreased LBP levels.
  • Increased CD4+ T lymphocytes.
  • Decreased activation of markers CD38 and HLA-DR presented on CD4+ T lymphocytes.
NCT01439841Norway and SwedenStiksrud et al., 2015 [32]CompletedBiola (L. rhamnosous GG 108 CFU, L. acidophilus La-5 107 CFU and Bifidobacterium animalis subsp. lactis Bb-12 108 CFU)
1 daily dose
8 weeks
32≥18To investigate the effects in ART-treated HIV-infected patients on:
  • safety and tolerability
  • GM composition
  • immune activation
  • microbial translocation
  • disease progression.
  • Modification of GM composition.
  • Decreased D-dimer and IL-6 levels.
NCT00517803CanadaCompletedProbiotic yogurt (175 g)
1 daily yogurt
16 weeks
2418–85To compare the effects of several levels of fortified probiotic yogurt vs. placebo in HIV-infected patients with cancer on:
  • inflammation
  • immune function.
NCT02448238USACompletedVSL#3 (S. thermophilus, B. breve, B. longum, B. infantis, L. plantarum, L. acidophilus, L. paracasei and L. bulgaricus (9 × 1011 CFU))
1 daily dose
12 weeks
23 (females only)≥18To assess the effects in naive HIV-infected Malian women on:
  • safety, tolerability, and level of stress
  • inflammation
  • bacterial translocation
  • immune function.
NCT02764684DenmarkCompletedLactobacillus rhamnosus
2 daily doses
8 weeks
45≥18To evaluate the effects in naive HIV-infected patients on:
  • GM composition
  • microbial translocation
  • lipid metabolism
  • cardiovascular risk factors
  • systemic and gut inflammation.
NCT02441231CanadaUnknownVisbiome
2 daily doses (9 × 1014 CFU/day)
48 weeks
36 (males only)≥19To investigate the effects in ART-treated HIV-infected men on:
  • microbial translocation
  • immune cells function and activation.
NCT00536848TanzaniaHummelen et al., 2010 [26]UnknownLactobacillus rhamnosus GR-1 and L. reuteri RC-14 (2 × 109 CFU)
1 daily dose
24 weeks
65 (females only)18–45To determine the effects in naive HIV-infected women with vaginosis on:
  • diarrhea
  • immune system
  • bacterial vaginosis.
No improvements in bacterial vaginosis.
NCT01908049SpainVillar-García et al., 2015 [25]UnknownS. boulardii
3 daily doses
12 weeks
44≥18To assess the effects in ART-treated HIV-infected patients on:
  • Bacterial translocation
  • GM composition
  • immune function
  • viral load.
Decreased IL-6 and LBP levels.
NCT02706717USACompletedVisbiome
1 daily dose
24 weeks
93≥18To evaluate the effects in ART-treated HIV-infected patients on:
  • Inflammation
NCT02276326ItalyCeccarelli et al., 2017 [33]RecruitingVSL#3 (S. thermophilus, B. breve, B. longum, B. infantis, L. plantarum, L. acidophilus, L. paracasei, and L. bulgaricus (9 × 1011 CFU))
4 daily doses
16 weeks
20≥18To determine the effects in ART-treated HIV-infected patients on neurocognitive profile.
  • Modification of GM composition.
  • Improved neurocognitive profile.
NCT02640625NorwayCompletedLactobacillus rhamnosus GG, L. acidophilus, L. bulgaricus, B. animalis subsp. lactis, and S. thermophilus
1 daily dose
10 weeks
60 (males only)25–65To assess the effects in ART-treated HIV-infected men on:
  • safety
  • biological effects
  • To investigate differences between IR and INR in:
  • microbial composition
  • mucosal barrier function.
NCT01184456USACompletedGanedenBC30 (Bacillus coagulans GBI-30 andPTA-6086 (2 × 1012 CFU))
1 daily dose
12 weeks
24≥18To evaluate the effects in ART-treated HIV-infected patients on bacterial translocation.
NCT04297501ChinaCompletedBifidobacterium (3 × 1012) and Lactobacillus (1012)
1 daily dose
12 months
5018–65To investigate the effects in ART-treated HIV-infected patients on:
  • microbial composition and diversity
  • immune recovery and activation
  • gut damage
  • microbial translocation
  • inflammation.
NCT04297488ChinaNot yet recruitingBifidobacterium (3 × 1012) and Lactobacillus (1012)
1 daily dose
6 months
2018–65To explore the effects in INR HIV-infected patients on:
  • microbial composition and diversity
  • immune recovery and activation
  • gut damage
  • microbial translocation
  • inflammation.
NCT04175223FranceNot yet recruitingVivomixx
2 daily doses
6 months
50≥18To evaluate the effects in ART-treated HIV-infected patients with neurocognitive disorders on:
  • immune activation
  • cognitive performance.
NCT0409943ItalyRecruitingVivomixx
4 daily doses
6 months
To assess the effects in ART-treated HIV-infected patients with HPV genital infection on:
  • anal HPV clearance
  • anal dysplasia.
ART, antiretroviral treatment; CFU, colony forming unit; GM, gut microbiota; HIV, human immunodeficiency virus; HPV, human papilloma virus IL-6, interleukin-6; IR, immunological responder; INR, immunologic non-responder.
Table 3. Clinical trials concerning symbiotics registered in ClinicalTrials.gov.
Table 3. Clinical trials concerning symbiotics registered in ClinicalTrials.gov.
NCT NumberCountryReferenceStatusInterventionNumber of ParticipantsAge of Participants (Years)ObjectiveMain Outcomes
NCT03009032SpainSerrano-Villar et al., 2019 [34]CompletedPMT25341
1 daily dose
48 weeks
7718–80To investigate the effects of the intervention in naive HIV-infected patients who initiate ART on:
  • safety
  • immunological recovery
  • inflammatory markers
  • GM composition.
  • No effects on microbiota composition
  • No effects on CD4+ T lymphocytes.
  • No effects on markers of inflammation and bacterial translocation.
NCT00688311USASchunter et al., 2012 [35]CompletedSynbiotic 2000 (4 probiotic bacteria (1010 CFU/bacteria) + 4 dietary fibers (2.5 g/fiber))
1 daily dose
4 weeks
34 (only females)≥18To determine the effects in ART-treated HIV-infected women on:
  • intestinal function
  • immune system overactivation
  • immune function.
  • Safe.
  • Modification of GM composition.
NCT03568812IndonesiaRecruitingRillus [L. plantarum 8.55 mg, S. thermophilus 8.55 mg and B. bifidum 2.55 mg (109 CFU) + FOS (480 mg)]
1 daily dose
12 weeks
8018–55To assess the effects INR HIV-infected patients on:
  • gut mucosal integrity and immunity
  • bacterial translocation
  • gut inflammation.
NCT03542786SpainRecruitingI3.1 (L. plantarum CECT7484, L. plantarum CECT7485 and P. acidilactici) + ProSheed
1 daily dose
48 weeks
40≥18To evaluate the effects in ART-treated HIV-infected patients on inflammaging (premature aging).
NCT02180035BrazilSantos et al., 2017 [36]CompletedLactobacillus paracasei, Lactobacillus rhamnosus, Lactobacillus acidophilus and Bifidobacterium lactis + FOS
2 daily doses
24 weeks
290≥19To investigate the effects in ART-treated HIV-infected patients on lipid metabolism.Reduced diarrhea, nausea and/or vomiting, constipation, and dyspepsia.
ART, antiretroviral treatment; CF, colony forming unit; GM, gut microbiota; HIV, human immunodeficiency virus; INR, immunological non-responder.
Table 4. Bacteria whose relative abundance is modified by the interventions described in this review and their functions in health and disease.
Table 4. Bacteria whose relative abundance is modified by the interventions described in this review and their functions in health and disease.
PhylumClassOrderFamilyGenusSpeciesFunction(s)
It INCREASEFirmicutesClostridiaClostridialesRuminococcaceaeFaecalibacterium One of the main butyrate producers in the gut. Plays a crucial role in gut physiology and host wellbeing [38].
Ruminiclostridium Significantly enriched in healthy donors compared with people with metabolic issues [39].
LachnospiraceaeBlautia Contributes to alleviation of inflammatory diseases and metabolic diseases and has antibacterial activity against specific microorganisms [40].
Anaritides Unknown.
Dorea Capable of degrading polysaccharides, oligosaccharides and sugars [41].
Fusicatenibacter Butyrate production [42].
EubacteriaceaeEubacterium Butyrate production. Plays a critical role in energy homeostasis, colonic motility, immunomodulation, and suppression of inflammation in the gut. Bile acid and cholesterol transformations [43].
ErysipelotrichiaErysipelotrichalesErysipelotrichaceaeCatenibacterium Polysaccharide degradation [44].
NegativicutesSelenomonadalesVeillonellaceaeMegamonas Production of the SCFAs acetate, propionate, and butyrate production, which are beneficial for health [45].
BacilliLactobacillalesLactobacillaceaeLactiplantibacillusplantarumProduction of bacteriocins, antimicrobial peptides [46].
PediococcuspentosaceuAlleviation of intestinal inflammation by maintaining the intestinal integrity and modulating the immunological profiles, gut microbiome, and metabolite composition [47].
ActinobacteriaActinobacteriaCoriobacterialesCoriobacteriaceaeCollinsella Butyrate production [48].
ActinomycetalesCorynebacteriaceaeCorynebacterium Cause of significant infections [49].
BifidobacterialesBifidobacteriaceaeBifidobacterium Numerous positive health benefits [50].
ProteobacteriaDeltaproteobacteriaDesulfovibrionales Sulfate reduction [51].
It DECREASEFirmicutesClostridiaClostridialesClostridiaceaeClostridiumlituseburenseAssociation with a range of human and animal diseases [52].
histolyctum
perfringens
coccoides
PeptostreptococcaceaeClostridioidesdifficileCauses diarrhea, fulminant colitis, and death [53].
EubacteriaceaeEubacteriumrectaleContributes to colorectal cancer by promoting colitis and inflammation [54].
ErysipelotrichiaErysipelotrichalesErysipelotrichaceaeCatenibacterium Polysaccharide degradation [44].
ProteobacteriaGammaproteobacteriaEnterobacterialesEnterobacteriaceae Causes many nosocomial infections, community-acquired infection, respiratory infections, soft tissue infections, osteomyelitis, and endocarditis [55].
Bacteroidetes Significant clinical pathogens [56].
SCFA, short-chain fatty acid.
Table 5. Clinical trials concerning fecal transplantation registered in ClinicalTrials.gov.
Table 5. Clinical trials concerning fecal transplantation registered in ClinicalTrials.gov.
NCT NumberCountryReferenceStatusInterventionNumber of ParticipantsAge of Participants (Years)ObjectiveMain Outcomes
NCT02256592USACompleted300 mL of fecal suspension from a healthy donor delivered by colonoscopy and provided by OpenBiome1818–75To examine the safety and durability of the intervention
To determine the effects in ART-treated HIV-infected patients on:
  • Immune activation
  • Inflammatory biomarkers.
NCT03329560USAActive, not recruitingPRIM-DJ2727 (capsules containing lyophilized microbiota derived from 150 g of healthy donor stool)
1 daily dose
6 weeks
6(enrolled)≥18To evaluate whether the intervention is safe for ART-treated HIV-infected MSM.
NCT03008941SpainSerrano-Villar et al., 2021 [80]CompletedFT capsules provided by OpenBiome10 capsules first week + 5 capsules weekly8 weeks3018–80To assess the effects in ART-treated HIV-infected patients on:
  • bacterial metabolism
  • plasma metabolite fingerprint
  • immune function and activation
  • inflammatory markers
  • markers of enterocyte barrier function.
  • Safe.
  • Modification of microbiota composition.
  • No effects on CD4+ T lymphocytes, CD4/CD8 ratio, lymphocyte markers of immune activation, or plasma markers of inflammation and BT.
  • Decreased IFABP levels.
NCT04165200MexicoCompletedFT via frozen capsules15 capsules every 12 h for four doses 7 days prior ART start and on weeks 0, 4, 8 and 12 after ART start22≥18To monitor the effects in naive HIV-infected patients on:
  • CD4+ T lymphocytes
  • viral load during weeks 0, 4, 8, 12, and 24 after initiating ART.
ART, antiretroviral treatment; BT, bacterial translocation; FT, fecal transplantation; HIV, human immunodeficiency virus; IFABP, intestinal fatty acid-binding protein; MSM, men who have sex with men.
Table 6. Summary of the interventions described in this review and their composition.
Table 6. Summary of the interventions described in this review and their composition.
InterventionDefinition/Composition
PrebioticsscGOSShort-chain galacto-oligosaccharides
lcFOSLong-chain fructo-oligosaccharides
pAOSPectin hydrolysate-derived acidic oligosaccharides
GlutamineGlutamine aminoacidic
Camu camuMyrciaria dubia
ProbioticsSupplemented fermented skimmed milkL. rhamnosus GG (108 CFU/mL), B. animalis subsp. lactis B-12 (108 CFU/mL), and L. acidophilus La-5 (107 CFU/mL)
VisbiomeS. thermophilus, B. breve, B. longum, B. infantis, L. plantarum, L. acidophilus, L. paracasei, and L. bulgaricus (9 × 1011 CFU)
Saccharomyces boulardii56.5 mg of living S. boulardii
Lactobacillus rhamnosus HN001 plus Bifidobacterium lactis Bi-07L. rhamnosus HN001 plus B. lactis Bi-07 (109 CFU/mL)
Lactobacillus rhamnosus GGL. rhamnosus GG (1010 CFU)
Packet AS. salivarius spp. thermophilus (2.04 × 1014 CFU), B. breve, B. infantis, B. longum (9.3 × 1013 CFU), L. acidophilus (2 × 1012 CFU), L. plantarum (2.2 × 108 CFU), L. casei (2.2 × 108 CFU), L. delbrueckii ssp. bulgaricus (3 × 108 CFU), and S. faecium (3 × 107 CFU)
Lactobacillus rhamnosus GR-1 and Lactobacillus reuteri RC-14L. rhamnosus GR-1 and L. reuteri RC-14
Ganeden BC30B. coagulans GBI-30, 6086 (2 × 1012 CFU)
Fortified yogurtL. rhamnosus GR-1 (1.23 × 109 CFU/mL)
BiolaL. rhamnosous GG 108 CFU, L. acidophilus La-5 107 CFU and B. animalis subsp. lactis Bb-12 108 CFU
VSL#3S. thermophilus, B. breve, B. longum, B. infantis, L. plantarum, L. acidophilus, L. paracasei and L. bulgaricus (9 × 1011 CFU)
Lactobacillus rhamnosus GG plus Lactobacillus acidophilus plus Lactobacillus bulgaricus plus Bifidobacterium animalis subsp. lactis plus Streptococcus thermophilusL. rhamnosus GG, L. acidophilus, L. bulgaricus, B. animalis subsp. lactis, and S. thermophilus
VivomixxB. breve, B. longum, B. infantis, L. acidophilus, L. plantarum, L. paracasei, L. bulgaricus, and S. thermophilus
SymbioticsAgavins plus Lactobacillus rhamnosus HN001 plus Bifidobacterium lactis Bi-0710 g of agavins from Agave tequilana plus Lactobacillus rhamnosus HN001 and Bifidobacterium lactis Bi-07 (109 CFU/mL)
Synbiotic 20004 probiotic bacteria (1010 CFU/bacteria) plus 4 dietary fibers (2.5 g/fiber)
PMT25431Unknown
LACTOFOSL. paracasei Lpc-37 SD 5275, Lactobacillus rhamnosus HN001 SD 5675, L. acidophilus N CFM SD 5221, and B. bifidum (109 CFU/strain) plus FOS
RillusL. plantarum (8.55 mg), S. thermophilus (8.55 mg), and B. bifidum 2.55 mg (109 CFU) plus FOS (480 mg)
I3.1L. plantarum CECT7484, L. plantarum CECT7485 and P. acidilactici) plus ProSheed
Lactobacillus paracasei plus Lactobacillus rhamnosus plus Lactobacillus acidophilus plus Bifidobacterium lactis plus FOSL. paracasei, Lactobacillus rhamnosus, L. acidophilus, and B. lactis plus FOS
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Villoslada-Blanco, P.; Pérez-Matute, P.; Oteo, J.A. Lights and Shadows of Microbiota Modulation and Cardiovascular Risk in HIV Patients. Int. J. Environ. Res. Public Health 2021, 18, 6837. https://doi.org/10.3390/ijerph18136837

AMA Style

Villoslada-Blanco P, Pérez-Matute P, Oteo JA. Lights and Shadows of Microbiota Modulation and Cardiovascular Risk in HIV Patients. International Journal of Environmental Research and Public Health. 2021; 18(13):6837. https://doi.org/10.3390/ijerph18136837

Chicago/Turabian Style

Villoslada-Blanco, Pablo, Patricia Pérez-Matute, and José A. Oteo. 2021. "Lights and Shadows of Microbiota Modulation and Cardiovascular Risk in HIV Patients" International Journal of Environmental Research and Public Health 18, no. 13: 6837. https://doi.org/10.3390/ijerph18136837

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