ArticlePDF Available

Immune activation and inflammation in lactating women on combination antiretroviral therapy: role of gut dysfunction and gut microbiota imbalance

Frontiers
Frontiers in Immunology
Authors:

Abstract and Figures

Introduction Combination antiretroviral therapy (cART) effectively controls HIV; however, chronic low-level viremia and gut microbiota dysbiosis remain significant drivers of gut and systemic inflammation. In this study, we explored the relationship between gut microbiota composition, intestinal inflammation, microbial translocation, and systemic inflammation in women on cART in Sub-Saharan Africa. Methods We conducted a study in HIV-infected and HIV-uninfected lactating women followed up at 6 weeks and 6 months postpartum in Harare, Zimbabwe. We used 16S ribosomal Ribonucleic Acid (rRNA) sequencing and MesoScale Discovery V-Plex assays to examine the gut microbiome and to quantify plasma inflammatory biomarkers, respectively. In addition, we measured fecal calprotectin, plasma lipopolysaccharide-binding protein (LBP), and soluble cluster of differentiation 14 (sCD14) by enzyme-linked immunosorbent assay to assess gut inflammation, microbial translocation, and monocyte/macrophage activation. Results A group of 77 lactating women were studied, of which 35% were HIV-infected. Fecal calprotectin levels were similar by HIV status at both follow-up time points. In the HIV-infected group at 6 weeks postpartum, fecal calprotectin was elevated: median (interquartile range) [158.1 µg/g (75.3–230.2)] in women who had CD4+ T-lymphocyte counts <350 cells/µL compared with those with ≥350 cells/µL [21.1 µg/g (0–58.4)], p = 0.032. Plasma sCD14 levels were significantly higher in the HIV-infected group at both 6 weeks and 6 months postpartum, p < 0.001. Plasma LBP levels were similar, but higher levels were observed in HIV-infected women with elevated fecal calprotectin. We found significant correlations between fecal calprotectin, LBP, and sCD14 with proinflammatory cytokines. Gut microbial alpha diversity was not affected by HIV status and was not affected by use of antibiotic prophylaxis. HIV significantly affected microbial beta diversity, and significant differences in microbial composition were noted. The genera Slackia and Collinsella were relatively more abundant in the HIV-infected group, whereas a lower relative abundance of Clostriduim sensu_stricto_1 was observed. Our study also found correlations between gut microbial taxa abundance and systemic inflammatory biomarkers. Discussion and conclusion HIV-infected lactating women had increased immune activation and increased microbial translocation associated with increased gut inflammation. We identified correlations between the gut inflammation and microbial composition, microbial translocation, and systemic inflammation. The interplay of these parameters might affect the health of this vulnerable population.
This content is subject to copyright.
Immune activation and
inammation in lactating women
on combination antiretroviral
therapy: role of gut dysfunction
and gut microbiota imbalance
Privilege Tendai Munjoma
1
*, Panashe Chandiwana
1
,
Jacqueline Wyss
2,3
, Arthur John Mazhandu
1
,
Sebastian Bruno Ulrich Jordi
2,3
, Rutendo Gutsire
1
,
Leolin Katsidzira
4
, Bahtiyar Yilmaz
2,3
, Benjamin Misselwitz
2,3
and Kerina Duri
1
1
Immunology Unit, Department of Laboratory Diagnostic and Investigative Sciences, University of
Zimbabwe Faculty of Medicine and Health Sciences (UZ-FMHS), Harare, Zimbabwe,
2
Department of
Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern,
Bern, Switzerland,
3
Department for Biomedical Research, Maurice Müller Laboratories, University of
Bern, Bern, Switzerland,
4
Department of Internal Medicine, University of Zimbabwe Faculty of
Medicine and Health Sciences (UZ-FMHS), Harare, Zimbabwe
Introduction: Combination antiretroviral therapy (cART) effectively controls HIV;
however, chronic low-level viremia and gut microbiota dysbiosis remain signicant
drivers of gut and systemic inammation. In this study, we explored the relationship
between gut microbiota composition, intestinal inammation, microbial
translocation, and systemic inammation in women on cART in Sub-Saharan Africa.
Methods: We conducted a study in HIV-infected and HIV-uninfected lactating
women followed up at 6 weeks and 6 months postpartum in Harare, Zimbabwe. We
used 16S ribosomal Ribonucleic Acid (rRNA) sequencing and MesoScale Discovery
V-Plex assays to examine the gut microbiome and to quantify plasma inammatory
biomarkers, respectively. In addition, we measured fecal calprotectin, plasma
lipopolysaccharide-binding protein (LBP), and soluble cluster of differentiation 14
(sCD14) by enzyme-linked immunosorbent assay to assess gut inammation,
microbial translocation, and monocyte/macrophage activation.
Results: A group of 77 lactating women were studied, of which 35% were HIV-
infected. Fecal calprotectin levels were similar by HIV status at both follow-up
time points. In the HIV-infected group at 6 weeks postpartum, fecal calprotectin
was elevated: median (interquartile range) [158.1 µg/g (75.3230.2)] in women
who had CD4+ T-lymphocyte counts <350 cells/µL compared with those with
350 cells/µL [21.1 µg/g (058.4)], p = 0.032. Plasma sCD14 levels were
signicantly higher in the HIV-infected group at both 6 weeks and 6 months
postpartum, p < 0.001. Plasma LBP levels were similar, but higher levels were
observed in HIV-infected women with elevated fecal calprotectin. We found
signicant correlations between fecal calprotectin, LBP, and sCD14 with
proinammatory cytokines. Gut microbial alpha diversity was not affected by
Frontiers in Immunology frontiersin.org01
OPEN ACCESS
EDITED BY
Giulia Carla Marchetti,
University of Milan, Italy
REVIEWED BY
Camilla Tincati,
University of Milan, Italy
Eugenio Nelson Cavallari,
Sapienza University of Rome, Italy
*CORRESPONDENCE
Privilege Tendai Munjoma
privilegemunjoma@gmail.com;
ptmunjoma@medsch.uz.ac.zw
These authors have contributed
equally to this work and share
last authorship
RECEIVED 21 August 2023
ACCEPTED 17 October 2023
PUBLISHED 16 November 2023
CITATION
Munjoma PT, Chandiwana P, Wyss J,
Mazhandu AJ, Jordi SBU, Gutsire R,
Katsidzira L, Yilmaz B, Misselwitz B and
Duri K (2023) Immune activation and
inammation in lactating women on
combination antiretroviral therapy:
role of gut dysfunction and gut
microbiota imbalance.
Front. Immunol. 14:1280262.
doi: 10.3389/fimmu.2023.1280262
COPYRIGHT
© 2023 Munjoma, Chandiwana, Wyss,
Mazhandu,Jordi,Gutsire,Katsidzira,Yilmaz,
Misselwitz and Duri. This is an open-access
article distributed under the terms of the
Creative Commons Attribution License
(CC BY). The use, distribution or
reproduction in other forums is permitted,
provided the original author(s) and the
copyright owner(s) are credited and that
the original publication in this journal is
cited, in accordance with accepted
academic practice. No use, distribution or
reproduction is permitted which does not
comply with these terms.
TYPE Original Research
PUBLISHED 16 November 2023
DOI 10.3389/fimmu.2023.1280262
HIV status and was not affected by use of antibiotic prophylaxis. HIV signicantly
affected microbial beta diversity, and signicant differences in microbial
composition were noted. The genera Slackia and Collinsella were relatively
more abundant in the HIV-infected group, whereas a lower relative abundance
of Clostriduim sensu_stricto_1 was observed. Our study also found correlations
between gut microbial taxa abundance and systemic inammatory biomarkers.
Discussion and conclusion: HIV-infected lactating women had increased
immune activation and increased microbial translocation associated with
increased gut inammation. We identied correlations between the gut
inammation and microbial composition, microbial translocation, and systemic
inammation. The interplay of these parameters might affect the health of this
vulnerable population.
KEYWORDS
fecal calprotectin, microbial translocation, systemic inammation, HIV, gut microbiota,
lactating women, resource limited setting
Introduction
People living with HIV in Sub-Saharan Africa (SSA) constitute
about 54% of the worlds HIV-infected population (1). The
introduction of combination antiretroviral therapy (cART) has
reduced the burden of HIV in SSA with a signicant reduction in
morbidity and mortality. However, chronic persistent low-level
viremia due to residual HIV remains a signicant contributor to
microbial translocation, chronic monocyte activation, and
inammation in this population (24). Understanding the
relationship between these biological systems contributes to the
global effort of interventions toward the mitigation of HIV-
associated morbidities.
The acute phase of HIV infection involves depletion of the CD4+
T-cell population, causing major damage to gut-associated lymphoid
tissue, which is not fully restored by cART (5,6). Upon gut
inammation, neutrophils serve as a reliable defense mechanism.
Gut inammation causes increased gut permeability and release of
calprotectin from neutrophils. The fecal calprotectin levels are thus a
useful marker of gut inammation and an indirect marker of intestinal
permeability (7). Faecal calprotectin levels have been reported to be
higherintheHIV-infectedpopulationevenifoncARTcomparedwith
that in HIV-uninfected peers (8,9). Despite these studies, the inuence
of gut inammation on microbial translocation and systemic
inammation is still insufciently understood.
Inammation of the gut epithelial lining causes translocation of
microbial antigens into circulation driving HIV disease progression
through monocyte activation and inammation (10). The monocyte/
macrophage bound cluster of differentiation 14 (CD14) is a co-
receptor for lipopolysaccharide (LPS) and causes the secretion of
soluble CD14 (sCD14) (11) on exposure to bacterial toxins. Thus,
both LPS-binding protein (LBP) and sCD14 are considered
biomarkers of endotoxemia and intestinal permeability, which also
alters the gut microbiota (12). Increased systemic inammation with
accompanied gut permeability has been observed in cART-treated
women (13). However, long-term exposure to cART has been shown
to decrease biomarkers of gut permeability, microbial translocation,
and vascular injury in adults with chronic HIV infection (6,14,15).
A healthy gut microbiota is usually dominated by commensal
microorganisms that continually face perturbations such as HIV-
induced gut damage and antibiotics. Dysbiosis as a result of HIV
infection is generally characterized by a decrease in alpha diversity
(16,17) with a low abundance of Bacteroides and an increased
abundance of Prevotella (18). However, further evidence is needed
to further understand the impact of HIV-associated gut microbiota
dysbiosis on the production of proinammatory cytokines and the
consequent systemic inammation (19). Microbial antigens
translocated into circulation causes immune activation with
higher levels being associated with increased T-cell activation in
cART-treated individuals (15). In early chronic HIV infection,
circulating LPS has been shown to be a predictor of HIV disease
progression independent of HIV viremia and CD4+ T-lymphocyte
count (20). Evidence of the role of HIV-induced gut microbiota
dysbiosis in microbial translocation and inammation has been
conicting due to possible confounders in HIV management such
as antibiotic prophylaxis and cART.
To gain an understanding of the intricate relationships between
the processes, we hypothesized that microbial dysbiosis and
inammation of the gut due to HIV infection cause microbial
translocation and, ultimately, systemic inammation. Our study
aims to provide insights into the gut microbiota diversity and
abundance in HIV-infected and HIV-uninfected lactating women
at 6 weeks and 6 months postpartum. Biomarkers of systemic
inammation and their association with gut microbiota
abundance, gut inammation, and microbial translocation
were investigated.
Munjoma et al. 10.3389/fimmu.2023.1280262
Frontiers in Immunology frontiersin.org02
Materials and methods
Study design
This investigation was performed as a prospective longitudinal
study nested in the University of Zimbabwe Birth Cohort Study
(UZBCS). The UZBCS has been previously described in detail (21).
In brief, lactating women were longitudinally followed up at 6 weeks
and 6 months postpartum as part of a longitudinal follow-up to 2
years after birth.
Study participants
The study followed up women enrolled in the UZBCS who were
HIV-infected and HIV-uninfected and receiving postnatal care
services. They were monitored at 6 weeks and 6 months after
giving birth at four primary healthcare clinics located in areas with
low socio-economic status in Harare, Zimbabwe (Budiriro,
Glenview, Kuwadzana, and Rujeko clinics).
Inclusion and exclusion criteria
We recruited pregnant women beyond the 20th week of pregnancy
seeking antenatal care services. All participants gave written informed
consent to participate and had been tested for HIV. Women who failed
to adhere to the study procedures due to any health disorders such as
mental issues were not included in this sub-study. For this particular
study, only women who enrolled in the UZBCS in 2019 and had stool
samples available were included. These women were then followed up
at 6 weeks and 6 months after giving birth.
Data collection, sample collection,
and storage
Data were collected using paper-based approved questionnaires
and entered into a Research Electronic Data Capture (REDCap)
databasea secure, web-based software platform designed to support
data capture for research studies (22). A physical examination
including anthropometric assessments was carried out by trained
and qualied nurses. A total of 4 mL of whole venous blood samples,
collected using ethylenediamine-tetraacetic acid as an anticoagulant,
were obtained from each participant. The blood collection procedure
was carried out by trained and qualied nurses, ensuring adherence
to proper protocols. The collected samples were promptly processed
within a maximum time frame of 6 h from the time of collection.
Plasma was isolated and stored at 80°C until enzyme-linked
immunosorbent assays (ELISAs) and MesoScale Discovery (MSD)
V-plex assays were performed. About 50 g of feces was collected in
sterile containers, aliquoted, and stored at 80°C until fecal
calprotectin and DNA extraction assays were done.
HIV RNA load and CD4+
T-lymphocyte counts
Results of HIV RNA load and CD4+ T-lymphocyte counts
measured during the third trimester of pregnancy were obtained
from the UZBCS REDCap database. The assaying methods for these
HIV disease progression markers were previously described (21).
All HIV-infected women in this study were taking cART at both
follow-up time points, and used a formulation of efavirenz,
lamivudine, and tenofovir disoproxil fumarate (Tenolam-E),
following the World Health Organization guidelines (23).
Systemic inammation and
immune biomarkers
The MSD multi-spot V-plex assays (Rockville, Maryland, USA)
were used to quantify proinammatory and vascular injury immune
markers in 60 µL of plasma. The assays were carried out following the
manufacturers instructions and as previously described (24).
Originally, 48 biomarkers were quantiedinplasma,and,forthis
study, the proinammatory and vascular injury V-plex panels were of
interest. The proinammatory V-plex panel included interferon-
gamma (IFN-g), interleukin-1-beta (IL-1b), interkeukin-2 (IL-2),
interleukin-4 (IL-4), interleukin-6 (IL-6), interleukin-8 (IL-8),
interleukin-10 (IL-10), interleukin-12p70 (IL-12p70), interleukin-13
(IL-13), and tumor necrosis factor (TNF), and the vascular injury V-
plex panel included serum amyloid A (SAA), C-reactive protein (CRP),
vascular cell adhesion molecule 1 (VCAM-1), and intercellular
AdhesionMolecule1(ICAM-1).ThebiomarkerSAAwasexcluded
from our analyses due to calibration failure in one of the assays.
Biomarker of gut inammation
Fecal calprotectin was quantied from stool using a Buhlmann
fecal calprotectin sandwich ELISA assay (EK-CAL2-WEX,
Schönenbuch, Switzerland) based on the manufacturers
instructions. The Buhlmann Calex Cap (B-CALEX-C200,
Schönenbuch, Switzerland) was utilized to prepare stool extracts
following the manufacturers instructions. The extracts were then
diluted at a ratio of 1:5 with an appropriate incubation buffer before
proceeding with the assay. All assays were conducted in duplicate,
and ELISA plates were read at 450 nm using Gen 5 software
(BioTek, Winooski, VT, USA). Fecal calprotectin concentrations
were determined using a standard curve and categorized based on
the manufacturers clinical cutoffs: normal (<80 µg/g), borderline/
grey zone (80160 µg/g), and elevated (>160 µg/g).
Munjoma et al. 10.3389/fimmu.2023.1280262
Frontiers in Immunology frontiersin.org03
Biomarkers of microbial translocation and
monocyte activation
Plasma LBP and sCD14 levels were quantied using ELISA
assays (Hycult Biotech, Wayne, USA) according to the
manufacturers instructions. Absorbance was read at 450 nm, and
sample concentrations were determined from a standard curve. All
assays were performed in duplicate, and ELISA plates were read
using Gen 5 software (BioTek, Winooski, VT, USA).
Stool DNA Extraction and 16S
rRNA sequencing
Fecal samples were collected into sterile 50-mL sample cups,
aliquoted into 2-mL tubes, and stored at 80°C prior to assays. Fecal
DNA extraction was carried out from about 250 mg of stool sample
using the QIAamp PowerFecal Pro DNA kit (Qiagen, Dusseldorf,
Germany) as previously described (25). Total bacterial DNA was
eluted with 70 mL of elution buffer and then stored at 20°C prior to
PCR amplication. The eluted DNA was amplied using PCR,
targeting the V5 and V6 regions of the 16S rRNA gene. Previously
described bacteria-specic primers (forward: 5-
CCATCTCATCCCTGCGTGTCTCCGACTCAGC-barcode-
ATTAGATACCCYGGTAGTCC-3and reverse: 5-CCTCTCTAT
GGGCAGTCGGTGATA CGAGCTGACGACARCCATG-3) were
utilized (26). PCR conditions consisted of an initial denaturation at
94°C for 5 min, followed by 35 cycles of denaturation at 94°C for
1 min, annealing at 46°C for 20 s, elongation at 72°C for 30 s, and a
nal elongation at 72°C for 7 min.
The PCR amplicons were run on 1% agarose gel electrophoresis
at 100 volts for 1 h, with an expected product length of
approximately 350 base pairs. The amplicons were puried using
the QIAQuick Gel Extraction Kit (Qiagen, Dusseldorf, Germany).
The concentration of amplicons was determined using a Qubit
dsDNA HS Assay Kit on the Qubit 3.0 Fluorometer (ThermoFisher
Scientic) and then set to 26 pM for sequencing library preparation.
Sequencing was carried out on the Ion PGMSystem
(ThermoFisher Scientic) using an Ion PGMSequencing kit
and chip, following a previously described method (27).
Data analysis
Sociodemographic and participant
characteristics
Data analysis was conducted using R software version 4.2.2
(http://www.r-project.org/). Continous variables were tested for
normality using the ShapiroWilk test, and data were
summarized using median and interquartile range (IQR) or using
mean ± standard deviation (SD) where appropriate. Continous data
between groups were compared using the MannWhitney U-test,
KruskalWallis test, or Student T-test depending on the
distribution of the data. Categorical data were reported as
proportions and associations determined by Fishers exact test or
Chi-squared test where appropriate.
Microbial translocation, monocyte
activation, gut inammation, and systemic
inammatory bıomarkers
Continous data were tested for normalcy using the Shapiro
Wilk test and data reported as median (IQR) or mean ± SD where
appropriate, depending on data distribution. Concentrations of
proinammatory and vascular injury immune markers below the
assay detection limit were assigned the concentration of the lowest
calibrator as previously described (24). A q-value was calculated to
correct for multiple testing using the Bonferroni test, and q < 0.05
was considered signicant. Correction was done for the 48
biomarkers originally tested during the assays. Spearman rho (r)
correlation coefcient was used to determine associations between
biomarkers of HIV disease progression, gut inammation,
microbial translocation, and systemic inammation.
Computational analysis of 16S rRNA
microbial data
The Fastq sequencing les generated from Ion Torrent PGM
System were processed using the Quantitative Insights into Microbial
Ecology 2 (QIIME2) version 2021.11.0 pipeline (https://qiime2.org/), as
previously described (26,28,29). Amplicon sequence variants were
assigned with a 97% sequence identity threshold, using the default
optionsinQIIME2aswellastheq2-feature-classier plugin and a
Naïve Bayes classier. Taxonomic weights were assembled using the
SILVA database (https://www.arb-silva.de/).
The feature table and mapping le were used to generate a
phyloseq object in R (version 4.2.2) package phyloseq (30). Only
samples with more than 2,000 high-quality reads were further
analyzed. Diversity within communities was determined using
alpha diversity indices (Simpson and Shannon index), and inter-
community diversity was determined using beta diversity [Bray
Curtis dissimilarity using principal coordinate analysis (PCoA)] (31).
MannWhitney U-tests and Adonis (PERMANOVA) tests for alpha
diversity and beta diversity were performed to test for signicance of
any group differences, respectively. Taxonomy proling was
performed using microbiome Multivariable Association with Linear
Models (MaAsLin2) package (https://huttenhower.sph.harvard.edu)
to determine associations of the gut microbiota with categorical and
continuous variables (32). A q-value was calculated to correct for
multiple testing using the BenjaminiHochberg (BH) false discovery
rate (FDR) correction as a default step in MaAsLin2. A q-value <0.05
was considered signicant, and microbiota plots were generated using
the package phyloseq and GraphPad prism version 9.0.0 (GraphPad,
San Diego, CA).
Munjoma et al. 10.3389/fimmu.2023.1280262
Frontiers in Immunology frontiersin.org04
Results
Ofthe97womenenrolledsince2019,77lactatingwomenwere
successfully followed up at both 6 weeks and 6 months postpartum.
Maternal socio-demographics, concurrent medications, and clinical
characteristics at 6 weeks postpartum are shown in Table 1.TheHIV-
infected women [median age, 32 years (IQR, 2935)] were older than
the HIV-uninfected counterparts [median age, 26 years (IQR, 2130)].
At 6 weeks postpartum, all HIV-infected women were on cART,
and 15 (55.6%) were taking cotrimoxazole prophylaxis. In all these
women, cART was started pre-conception or during pregnancy. HIV-
infected women were more likely to be on antibiotics during the study
period, p < 0.0001. In addition, HIV-infected women had a
signicantly higher mid-upperarm circumference (MUAC)
(27.0 cm; IQR, 25.728.0) compared with their HIV-uninfected peers
(25.4 cm; IQR, 24.027.3), p = 0.031. Interestingly, HIV-infected
women had a higher body mass index (BMI) compared with their
uninfected counterparts (p =0.020),asindicatedinTable 1.
Socio-demographics and clinical characteristics at 6 months
postpartum are shown in Table 2. HIV-infected women were more
likely to be in employed and to have a higher median monthly family
income (Table 2). There were no associations between HIV infection
status and toilet facilities, drinking water sources, and water
treatment. Similar to the 6-week postpartum ndings, HIV-infected
women were more likely to be on antibiotics (see Table 1). In the
HIV-infected group 55.6% and 57.1% reported taking cotrimoxazole
antibiotics at 6 weeks and 6 months postpartum, respectively.
Biomarkers of gut inammation and gut
microbial translocation
Biomarkers of intestinal inammation (fecal calprotectin),
microbial translocation, and immune activation (plasma LBP and
sCD14) were quantied to assess the association between HIV
infection with gut inammation and microbial gut translocation.
Overall, fecal calprotectin levels did not signicantly differ by HIV
infection status and antibiotic use at 6 weeks and 6 months
postpartum. However, at 6 weeks, higher fecal calprotectin levels
(158.1 µg/g; IQR, 75.3230.2) were observed in HIV-infected
women with third-trimester CD4+ T-lymphocyte counts <350
cells/µL compared with those with 350 cells/µL (21.1 µg/g; IQR,
058.4), p = 0.032. In a sub-analysis of the HIV infected at 6 weeks
postpartum, we investigated the effects of cART and HIV viremia.
We stratied cART duration into early (<2 years) and long-term
(2 years) (33) and HIV RNA load into low level viremia (<200
copies/mL) and viremic (200 copies/mL) groups (4). Fecal
calprotectin levels were non-signicantly higher in unsuppressed
(HIV RNA > 1,000 copies/mL) versus suppressed (1,000 copies/
mL) in viremic versus low level viremia groups (p = 0.610 and p =
0.614, respectively). Levels were non-signicantly higher (116.7 µg/
g; IQR, 43.3230) in the early ART group compared with that in the
long-term ART group (61.3 µg/g; IQR, 16.3165.3).
HIV-infected participants had signicantly higher plasma sCD14
levels (29.7 ng/mL; IQR, 22.435.5) at 6 weeks postpartum when
TABLE 1 Socio-demographic and clinical data of the study participants
at 6 weeks postpartum (n =77), stratied by HIV status.
HIV-
infected
(n = 27)
HIV-
uninfected
(n = 50)
p-
value
Social demographics
Age (years)
[Median (IQR)] 32 (2935) 26 (2130) 3.95e-05
Breastfeeding type
Exclusive
Mixed
25 (92.6%)
2 (7.4%)
42 (84%)
8 (16%)
0.479
Postpartum alcohol use
Yes
No
1 (3.7%)
26 (96.3%)
1 (2%)
49 (98%)
1.000
Household meals
per day
[Median (IQR)] 3 (2-3) 3 (2-3) 0.569
Average stool frequency
Once daily
Greater or equal to
twice daily
Once every 2 days
21 (77.8%)
4 (14.8%)
2 (7.4%)
37 (75.5%)
9 (18.4%)
3 (6.1%)
Missing=1
0.883
Concurrent medications
Antibiotics use
Yes
No
15 (55.6%)
12 (44.4%)
2 (4%)
48 (96%)
4.48e-07
Anti-acid use
Yes
No
0 (0%)
27 (100%)
3 (6%)
47 (94%)
0.547
cART use
Yes
No
27 (100%)
0 (0%)
NA _
Clinical data
MUAC (cm)
[Median (IQR)] 27.0 (25.728.0) 25.4 (24.027.3) 0.031
BMI (kg/m
2
)
[Median (IQR)] 24.2 (22.125.6) 22.1 (19.623.9) 0.020
Mode of delivery
Spontaneous
Caesarean section
24 (88.9)
3 (11.1%)
50 (100%)
0 (0%)
0.039
cART duration
(months)
[Median (IQR); min-
max]
40.8 (13.586.1);
1.81455.4
NA _
cART duration group
Early ART (<2 years)
Long-term ART
(2 years)
8 (29.6%)
19 (70.4%)
NA _
Third trimester CD4
count (cells/µL)
[Median (IQR);
min-max]
355 (253449);
176635
NA _
Third trimester HIV
RNA suppression
Suppressed (1,000 23 (85.2%) NA _
(Continued)
Munjoma et al. 10.3389/fimmu.2023.1280262
Frontiers in Immunology frontiersin.org05
compared with their uninfected counterparts (18.9 ng/mL; IQR,
15.921.2), p < 0.0001 (Figure 1A). Similar results were found at 6
months postpartum where increased sCD14 levels were found in
HIV-infected (33.2 ng/mL; IQR, 25.036.2) compared with that in
HIV-uninfected women (22.7 ng/mL; IQR, 19.027.0), p = 0.0006.
IntheHIV-infectedgroupat6weekspostpartum,sCD14levels
were similar when compared by cART duration and HIV viremia
groups (p = 0.449 and p = 0.921 respectively). In all women, at 6 weeks
postpartum, those taking antibiotics had signicantly higher plasma
sCD14 levels (24.4 ng/mL; IQR, 22.135.3) (Figure 1B)comparedwith
those not taking antibiotics (20.1 ng/mL; IQR, 16.524.7). Plasma
sCD14 levels did not differ by antibiotic use in the subgroups of HIV-
infected and HIV-uninfected women (Figure 1C). At 6 months
postpartum, only HIV-infected women were on regular antibiotics,
and no signicant difference in median sCD14 levels was noted on
comparison by antibiotic usage. Furthermore, plasma sCD14 levels at 6
weeksand6monthspostpartumdidnotdifferbetweenwomenwith
elevated and normal fecal calprotectin levels.
Plasma LBP levels were similar between the HIV subgroups at 6
weeks or 6 months postpartum. In the HIV-infected group at 6 weeks
postpartum, LBP levels were similar when compared by cART duration
and HIV viremia groups (p = 0.632 and p = 0.453, respectively). At the
same time point, most HIV-infected women (88.2%) were on
cotrimoxazole prophylaxis, and there was no signicant difference in
plasma LBP levels by antibiotic use. At 6 weeks postpartum,
signicantly lower LBP levels were noted in women with normal
fecal calprotectin levels compared with those with elevated levels [18.2
ng/mL (IQR, 14.726.5) versus 26.4 ng/ml (IQR, 19.931.8), p = 0.022]
(Figure 2A). A similar trend was observed in the HIV-infected
subgroupatthesametimepoint(Figure 2B).
Correlation between biomarkers of gut
inammation, microbial translocation, and
systemic inammation
To determine whether biomarkers of gut inammation (fecal
calprotectin) and microbial translocation (sCD14 and LBP) are
associated with the systemic immune environment, a correlation
matrix of these biomarkers and 13 proinammatory cytokines and
chemokines was computed (Figure 3). These correlations were
calculated after stratication by HIV status to minimize confounding
by known effects of HIV on the cytokine and chemokine environment.
TABLE 1 Continued
HIV-
infected
(n = 27)
HIV-
uninfected
(n = 50)
p-
value
copies/mL)
Unsuppressed (>1,000
copies/mL)
4 (14.8%)
Third trimester HIV
Viremia
Low-level (<200 copies/
mL)
Viremic (200 copies/
mL)
23 (85.2%)
4 (14.8%)
NA _
BMI, body mass index; MUAC, mid-upperarm circumference; IQR, interquartile range;
HIV, human immunodeciency virus; CD4, cluster of differentiation 4; cART, combination
anti-retroviral therapy; RNA, ribonucleic acid. Statistical analysis: Group comparisons were
done using MannWhitney U-test or Fishers exact test where appropriate. P-values in bold
font are statistically signicant at p < 0.05.
TABLE 2 Socio-demographic, water, hygiene and sanitation and
concurrent medications at 6 months postpartum (n = 77), stratied by
HIV status.
HIV-
infected
(n = 28)
HIV-
uninfected
(n = 49)
p-
value
Social demographics
Employment status
Employed
Unemployed
14 (50%)
14 (50%)
11 (22.4%)
38 (77.6%)
0.044
Household size
[Median (IQR)] 5 (45) 4 (35) 0.074
Family monthly income
(USD)
[Median (IQR)] 1,150 (800
1,450)
650 (4801,285) 0.017
Water, hygiene and sanitation
Toilet facility in household
Flush (outside/inside)
Blair
27 (96.4%)
1 (3.6%)
47 (95.9%)
2 (4.1%)
0.155
Households sharing toilet
[Median (IQR)] 1 (14) 3 (24) 0.106
Main drinking water source
Borehole
Piped water into dwelling
Protected well
18 (64.3%)
1 (3.6%)
9 (32.1%)
22 (44.9%)
5 (10.2%)
22 (44.9%)
0.293
Drinking water treatment
No
Yes
21 (75%)
7 (25%)
42 (85.7%)
7 (14.3%)
0.357
Current sewer burst/overspill
No
Yes
25 (89.3%)
3 (10.7%)
41 (85.4%)
7 (14.6%)
Missing = 1
0.736
Current diarrhea
(participant/household
member)
No
Yes
26 (92.9%)
2 (7.1%)
42 (89.4%)
5 (10.6%)
Missing = 2
0.705
Average stool frequency
Once daily
Greater or equal to twice daily
Once every 2 days
23 (82.1%)
4 (14.3%)
1 (3.6%)
34 (69.4%)
14 (28.6%)
1 (2%)
0.357
Concurrent medications
Antibiotics use
Yes
No
16 (57.1%)
12 (42.9%)
0 (0%)
49 (100%)
2.23e-09
Anti-acid use
Yes
No
0 (0%)
27 (100%)
Missing = 1
0 (0%)
49 (100%)
_
IQR, interquartile range; HIV, human immunodeciency virus. Statistical analysis: Group
comparisons were done using the MannWhi tney U-test or Fi shers exact test where
appropriate. P-values in bold font are statistically signicant at p < 0.05.
Munjoma et al. 10.3389/fimmu.2023.1280262
Frontiers in Immunology frontiersin.org06
In the HIV-uninfected women, at 6 weeks postpartum, fecal
calprotectin and LBP correlated positively with IL-10 (r= 0.38,
p = 0.0376) and IL-13 (r= 0.47, p = 0.0082), respectively (Figure 3A).
In the same group at 6 months postpartum, LBP correlated positively
with IL-6 (r= 0.40, p = 0.024) and fecal calprotectin (r= 0.31,
p = 0.032), whereas fecal calprotectin correlated positively with IL-2
(r= 0.43, p = 0.018) and IFN-g(r= 0.42, p = 0.022). The biomarker
sCD14 correlated positively with IL-2 (r= 0.36, p = 0.046) in this
subgroup at 6 months postpartum (Figure 3C).
In the HIV-infected group, pregnancy third-trimester CD4+ T-
lymphocyte count (En CD4) and pregnancy third-trimester HIV
RNA load (En VL) were included in the correlation analysis at 6
weeks postpartum (Figure 3B). We assumed that the time since the
third trimester of pregnancy may have caused insignicant changes
in CD4+ T-lymphocyte counts and HIV RNA load. Interestingly,
third-trimester CD4+ T-lymphocyte counts negatively correlated
with LBP (r=0.47, p = 0.019), IL-6 (r=0.43, p = 0.033) and
CRP (r=0.42, p = 0.035). Third-trimester HIV RNA levels
(En VL) positively correlated with eight proinammatory
cytokines and chemokines, p < 0.05 (Figure 3B). In the HIV-
infected group at 6 weeks postpartum, fecal calprotectin
correlated positively with LBP (r= 0.44, p = 0.027), IL-2
(r= 0.42, p = 0.037), and IFN-g(r= 0.49, p = 0.012), whereas
LBP correlated positively with IL-2 (r= 0.56, p = 0.002), IFN-g
(r= 0.48, p = 0.010), and IL-6 (r= 0.43, p = 0.026).
At 6 weeks postpartum, sCD14 levels positively correlated
with IL-8 (r= 0.42, p = 0.029), TNF (r= 0.39, p = 0.044), and
IL-6 (r= 0.42, p = 0.029) in the HIV-infected group (Figure 3B). At
6 months postpartum, fecal calprotectin positively correlated
with IL-12p70 (r= 0.41, p= 0.044) in the HIV-infected
group (Figure 3D), whereas LBP levels positively correlated with
IL-2 (r= 0.50, p = 0.010), IL-1b(r= 0.42, p = 0.037), IL-6 (r= 0.68,
p = 0.0002), TNF (r= 0.50, p = 0.011), and IFN-g(r= 0.55,
p = 0.004). In the same group, plasma sCD14 levels positively
correlated with CRP (r= 0.52, p = 0.006) (Figure 3D).
Comparison of biomarkers of
systemic inammation and vascular
injury according to HIV infection
status and follow-up time point
To explore the impact of HIV infection on the cytokine/
chemokine environment, we conducted a comparative analysis of
median biomarker levels between HIV-infected and HIV-
uninfected women at 6 weeks and 6 months postpartum
(Supplementary Figure 1). We did not observe any statistically
signicant differences in the biomarker levels between the HIV-
infected and HIV-uninfected women even after applying multiple
testing correction to account for potential false positives.
BCA
FIGURE 1
Plasma sCD14 levels. Plasma sCD14 levels in all participants at 6 weeks postpartum, according to HIV infection status (A), antibiotic use (B), and
stratied for HIV status (C). Statistics: MannWhitney U-test.
BA
FIGURE 2
Plasma LBP levels by fecal calprotectin group. Plasma LBP levels in all participants (A) and HIV-infected women (B) at 6 weeks postpartum stratied
for fecal calprotectin group. Statistics: MannWhitney U-test.
Munjoma et al. 10.3389/fimmu.2023.1280262
Frontiers in Immunology frontiersin.org07
Gut microbiota assessment at 6 weeks and
6 months postpartum
For gut microbiota analysis, samples from 73 of the 77
participants at 6 weeks and 65 of the 77 participants at 6 months
postpartum could be used. There were four participants with
missing stool samples at 6 weeks and ve participants with
missing stool samples at 6 months. Seven samples were dropped
from analysis due to low sequence read numbers. Overall, alpha
diversity measures (Shannon and Simpson index) did not differ
signicantly between the two follow-up time points or by HIV
status (Figure 4A). However, there was a signicant difference in
beta diversity in all participants between the two time points
(p = 0.001). Beta diversity also differed signicantly by HIV status
at 6 weeks (Figure 4B) and 6 months (Figure 4C) postpartum.
We conducted a comparative analysis of the gut microbiota in the
HIV-infected group at 6 weeks postpartum, considering third-
trimester HIV RNA suppression (cutoff of 1,000 copies/mL) and
third-trimester CD4+ T-lymphocyte status (cutoff of 350 cells/µL).
There were no signicant differences in the alpha diversity (assessed
by Shannon and Simpson indices) and beta diversity according to
HIV RNA suppression and CD4+ T-lymphocyte status. In addition,
there were no signicant differences in either alpha or beta diversity
stratied by antibiotic use in the HIV-infected group at 6 weeks and 6
months postpartum (data not shown). All HIV-infected participants
in our study were receiving cART; therefore, the effects of HIV and
cART on the gut microbiota could not be separated.
Our study examined the mean relative abundance of the top 10
phyla of the gut microbiota in all women (Figure 5). The
predominant phyla identied were Firmicutes (86.4%),
Actinobacteriota (7.6%), Bacteroidota (4.9%), Proteobacteria (0.8%),
Spirochaetota (0.08%), Verrucomicrobiota (0.06%), and
Desulfobacterota (0.04%). In addition to that, the mean relative
abundance of the top 10 genera was also determined. The
predominant genera were Clostriduim_sensu_stricto_1 (16.6%),
Romboutsia (16.1%), Agathobacter (5.3%), Faecalibacterium (4.9%),
Prevotella (4.4%), Sarcina (4.4%), and Blautia (3.8%). Notably, in the
HIV-infected group, a lower relative abundance of the genera
B
CD
A
FIGURE 3
Correlation matrix analysis of plasma biomarkers in HIV-infected and HIV-uninfected women at 6 weeks and 6 months postpartum. HIV-uninfected
participants at 6 weeks postpartum (A), HIV-infected participants at 6 weeks postpartum (B), HIV-uninfected participants at 6 months postpartum
(C), and HIV-infected participants at 6 months postpartum (D). Correlations marked by X were either not present or non-signicant at p < 0.05.
Munjoma et al. 10.3389/fimmu.2023.1280262
Frontiers in Immunology frontiersin.org08
FIGURE 5
Overall relative abundance for the top 10 phyla of the gut microbiota. The top 10 phyla by order of decreasing relative abundance at both 6 weeks
and 6 months postpartum are indicated.
BC
A
FIGURE 4
Microbiota characteristics at 6 weeks and 6 months postpartum according to HIV status. Alpha diversity (Shannon and Simpson index) comparison
by HIV status at both 6 weeks and 6 months postpartum (A), PCoA for beta diversity (BrayCurtis) comparison by HIV infection status at 6 weeks
postpartum (B), and PCoA for beta diversity (Bray-Curtis) comparison by HIV status at 6 months postpartum (C).
Munjoma et al. 10.3389/fimmu.2023.1280262
Frontiers in Immunology frontiersin.org09
Akkermansia and Collinsella was noted at 6 months postpartum
compared with that at 6 weeks postpartum. However, in the same
group, a higher relative abundance of the genera Lachnospira,
Prevotella,Bacteroides, and UCG.005 from the Oscillospiraceae
family was noted at 6 months compared with that at 6 weeks
postpartum. These ndings highlight dynamic shifts in the relative
abundance of specic taxa in the postpartum period.
The gut microbiota was compared to determine taxa
signicantly associated with HIV infection status at both 6 weeks
and 6 months postpartum. At 6 weeks postpartum, HIV infection
was signicantly associated with lower abundance of genus
Clostriduim_sensu_stricto_1 and family Clostridiaceae from the
Firmicutes phylum (Figure 6A). At the same time point, HIV
infection was signicantly associated with a higher abundance of
taxa from the Actinobacteriota phylum including the genera Slackia
and Collinsella (Figure 6A). At 6 months postpartum, only taxa
from the Firmicutes phylum signicantly differed by HIV infection
status (Figure 6B). The genus Clostriduim_sensu_stricto_1 was
signicantly abundant in the HIV-uninfected group at both 6
weeks and 6 months postpartum.
We investigated longitudinal changes in the gut microbiota from
pregnancy to 6 months postpartum. The pregnancy gut microbiota
from our study population has been previously described in detail (34).
In brief, species richness was lower (Shannon, p = 0.0092, and Simpson,
p = 0.012) in the HIV-infected women compared with that in the
uninfected peers. Beta diversity assessed using BrayCurtis dissimilarity
index showed signicant differences in diversity between HIV-infected
and HIV-uninfected pregnant women. Alpha diversity did not differ by
CD4+ T-lymphocyte group and viral load suppression using similar
cutoffs used in our study. Infection with HIV was associated with
reduced abundance of Clostridium,Bacteroides,Bidobacterium,and
Faecalibacterium with an observed increase in Actinomyces.
In HIV-uninfected women, there were no differences in
microbial evenness between pregnancy and the two follow-up
time points (Supplementary Figure 2A). However, a signicance
was noted in microbial richness (p = 0.017) when comparing the
three time points. In the HIV-infected group, both evenness and
richness indices showed an increase from pregnancy to 6 months
postpartum although the difference was non-signicant for
evenness (Supplementary Figure 2B). Beta diversity comparison
showed a signicant difference (p = 0.001) when compared by time
point in HIV-uninfected (Supplementary Figure 2C) and HIV-
infected groups, p = 0.001 (Supplementary Figure 2D).
Furthermore, the effects of cART duration and HIV viremia on
the gut microbiota in the HIV-infected group were investigated at 6
weeks postpartum. No differences were observed for both Alpha
and Beta diversity when compared between early versus long-term
cART groups and low-level viremia versus viremic groups.
Association of inammatory and
vascular injury biomarkers with
the gut microbiota abundance
We determined the association of the gut microbiota with
inammatory and vascular injury biomarkers after stratication by
HIV infection status using the MaAsLin2 package in R. Only
associations remaining signicant (q < 0.05) after BH FDR
correction were reported (Table 3). To test for microbial taxa
associations with proinammatory and vascular injury biomarkers,
microbial features that appeared in 25% of the total number of
samples assayed. In the HIV-infected group at 6 months postpartum,
the genera Catenibacterium and Haemophilus positively associated
with levels of IL-2 and IL-6 respectively (q = 0.021 and q =
0.017, respectively).
In the HIV-uninfected group at 6 months, the Actinobacteriota
phylum positively associated with LBP levels (q = 0.018), whereas the
Firmicutes phylum negatively associated with IL-2 levels (q = 0.041).
The family Coriobacteriaceae positively associated with IL-2 levels
(q = 0.029), and the genus Clostridia_UCG.014 negatively associated
with IFN-glevels (Table 3). In the same group, the genus Actinomyces
positively associated with fecal calprotectin levels (q = 0.032).
B
A
FIGURE 6
Association of bacterial taxa with HIV infection status. (A) Comparisons at 6 weeks postpartum and (B) 6 months postpartum are shown. *p < 0.05
and ***p < 0.001.
Munjoma et al. 10.3389/fimmu.2023.1280262
Frontiers in Immunology frontiersin.org10
Discussion
Our study addresses associations of gut inammation, microbial
composition, and systemic inammation in HIV-infected lactating
women on cART in SSA where data have been scarce. None of the
women in our study suffered from AIDS or HIV-related symptoms,
and the levels of systemic inammation were low. Therefore, no
strong HIV-related alterations were observed; however, we would like
to emphasize the following key results.
1. In our study population, similar levels of gut inammation
(fecal calprotectin levels), microbial translocation (LBP
levels), and important descriptors of systemic
inammation (TNF, IL-6, IL-8, and CRP) were found in
HIV-infected and HIV-uninfected individuals.
2. HIV infection can affect intestinal inammation in more
immune-compromised individuals, because fecal
calprotectin levels correlated with third trimester CD4+
T-lymphocyte counts.
3. HIV-infected participants showed increased levels monocyte
activation (increased sCD14 levels), despite taking cART
and prophylactic antibiotics.
4. There is a relationship between intestinal inammation,
microbial translocation, and systemic inammation
because fecal calprotectin, LBP, and sCD14 correlated
with biomarkers of systemic inammation.
5. Stratied by HIV status, the gut microbiota differed in beta
diversity but not alpha diversity.
6. Several gut microbial taxa were signicantly associated with
HIV status and systemic inammation.
TABLE 3 Gut microbial taxa and systemic inammation.
Feature Metadata Coefcient N N not 0% q-value
HIV-infected (6 weeks postpartum)
p:Proteobacteria.c:Gammaproteobacteria.o:Burkholderiales.f:Oxalobacteraceae CRP 0.6080931 25 4 0.023359
p:Firmicutes.c:Clostridia.o:Lachnospirales.f:Lachnospiraceae.g:Eubacterium_eligens_group ICAM-1 0.43383237 25 3 0.000235
HIV-uninfected (6 weeks postpartum)
p:Firmicutes.c:Bacilli.o:Lactobacillales.f:Leuconostocaceae.g:Weissella IL-4 0.9326621 48 9 6.04e-08
HIV-infected (6 months postpartum)
p:Firmicutes.c:Bacilli.o:Erysipelotrichales.f:Erysipelatoclostridiaceae.g:Catenibacterium IL-2 1.32753279 23 9 0.021138
p:Proteobacteria.c:Gammaproteobacteria.o:Pasteurellales.f:Pasteurellaceae.g:Haemophilus IL-6 1.58409509 23 6 0.017096
p:Firmicutes.c:Clostridia.o:Lachnospirales.f:Lachnospiraceae.g:Eubacterium_eligens_group IL-2 0.55998378 23 4 0.001936
p:Firmicutes.c:Clostridia.o:Lachnospirales.f:Lachnospiraceae.g:Eubacterium_eligens_group IL-10 0.60010197 23 4 0.000557
p:Firmicutes.c:Clostridia.o:Lachnospirales.f:Lachnospiraceae.g:Anaerostignum IL-4 0.45052327 23 3 0.017532
p:Firmicutes.c:Clostridia.o:Lachnospirales.f:Lachnospiraceae.g:Butyrivibrio IL-6 0.76872521 23 3 4.03e-05
p:Firmicutes.c:Clostridia.o:Lachnospirales.f:Lachnospiraceae.g:Butyrivibrio TNF 0.70911458 23 3 0.001431
HIV-uninfected (6 months postpartum)
p:Actinobacteriota LBP 0.83076488 42 42 0.018545
p:Firmicutes IL-2 0.1308627 42 42 0.041327
p:Actinobacteriota.c:Coriobacteriia.o:Coriobacteriales.f:Coriobacteriaceae IL-2 0.91371356 42 41 0.029222
p:Firmicutes.c:Clostridia.o:Clostridia_UCG.014.f:Clostridia_UCG.014.g:Clostridia_UCG.014 IFN-g1.5113457 42 36 0.033319
p:Actinobacteriota.c:Actinobacteria.o:Actinomycetales.f:Actinomycetaceae.g:Actinomyces FC 0.97433139 42 32 0.03262
p:Firmicutes.c:Bacilli.o:Erysipelotrichales.f:Erysipelatoclostridiaceae ICAM-1 2.02529785 42 27 0.033465
p:Firmicutes.c:Bacilli.o:Erysipelotrichales.f:Erysipelotrichaceae.g:Solobacterium ICAM-1 1.74957426 42 12 0.033465
p:Actinobacteriota.c:Coriobacteriia.o:Coriobacteriales.f:Eggerthellaceae.g:Eggerthella FC 1.21725587 42 9 0.03262
p:Actinobacteriota.c:Coriobacteriia.o:Coriobacteriales.f:Eggerthellaceae.g:Eggerthella IL-2 1.3634006 42 9 0.0208
p:Firmicutes.c:Bacilli.o:Lactobacillales.f:Lactobacillaceae.g:Lactobacillus IL-8 1.04516168 42 9 0.041506
p:Fusobacteriota.c:Fusobacteriia.o:Fusobacteriales.f:Fusobacteriaceae FC 1.30098884 42 6 0.001424
Association between gut microbial taxa and microbial translocation, and systemic and gut inammation biomarkers. N = total number of samples used in model; N not 0% = number of samples
in which microbial feature is not 0%; FC, fecal calprotectin.
Munjoma et al. 10.3389/fimmu.2023.1280262
Frontiers in Immunology frontiersin.org11
Effect of HIV infection on
biomarkers of gut inammation
and microbial translocation
In our population of asymptomatic HIV-infected and HIV-
uninfected lactating women, intestinal inammation (as assessed by
fecal calprotectin levels) was not affected by HIV status. Subgroups
with elevated fecal calprotectin levels included women with third-
trimester CD4+ T-lymphocyte counts <350 cells/µL consistent with
ndings from previous studies (35,36). Other studies in HIV-
infected individuals described similar (37,38) or elevated fecal
calprotectin levels (8,9), most likely depending on the degree of gut
mucosal damage and immune dysfunction despite taking cART
(39). In our study, above normal median fecal calprotectin levels
were observed in HIV-infected cART-experienced individuals, in
agreement with studies in cART-naïve Italians (40,41).
In line with an overall well-preserved intestinal barrier with
similar translocation of LPS and microbial antigens into the
systemic circulation, we found no effects of HIV status on plasma
LBP levels in our population. This contradicts previous studies
showing elevated plasma LBP levels in HIV-infected adults from
Europe and Africa (3,10). However, our ndings resembled results
from studies in Ugandan, American, and Chinese adults (8,4244).
Moreover, a study in Swedish HIV-infected adults revealed a
decrease in LBP upon the commencement of cotrimoxazole
prophylaxis (45). However, plasma LBP correlated positively with
fecal calprotectin levels in some of our analyses, conrming that gut
inammation can impact the translocation of microbes and/or LPS
in HIV-infected individuals on cART.
HIV-infected individuals in our study showed activation of
innate immunity, indicated by persistently high plasma sCD14
levels, a biomarker for innate immune activation including
monocytes and macrophages (46), and an independent predictor
for mortality in HIV-infected individuals on cART (47,48). Elevated
sCD14 levels were also found in HIV-infected European and African
children as well as African and Chinese adults (2,3,8,13,36,43,44,
46,49,50). We did not determine the source of monocyte activation
in our population but our results are consistent with HIV-induced
microbial translocation potentially driving monocyte/macrophage
activation as demonstrated in another study (47).
In our study, plasma sCD14 levels did not differ signicantly by
antibiotic use and correlated neither with fecal calprotectin nor with
plasma LBP levels. These ndings are inconsistent with another
study in cART-naïve Ugandan adults with recent HIV infection,
which reported signicant correlations between LBP and sCD14
levels (51). Differences between both study populations and/or
cotrimoxazole prophylaxis in our study might explain this
discrepancy; however, this warrants further investigations.
Association of systemic inammation
biomarkers with HIV infection, microbial
translocation, and gut inammation
In line with an overall preserved immune function in our study
population, we found overall similar plasma levels of important
systemic inammatory biomarkers (TNF, IL-6, IL-8, and CRP) in
HIV-infected and HIV-uninfected women, consistent with previous
work (2,17,52). Our results thus likely reect protection from
systemic inammation due to cART treatment, as observed in other
studies (14,53). The situation was shown to be different in cART-
naïve and more immune-suppressed cART-experienced individuals
who showed increased levels of inammatory biomarkers (49,
5357).
However, our detailed analyses revealed some effects of HIV
infection on the intestinal immune system: Fecal calprotectin
positively correlated with proinammatory cytokines in the HIV-
infected group, consistent with ndings from another study (8).
Furthermore, in the HIV-infected group, LBP, a biomarker of
microbial translocation, positively correlated with IL-2, IL-6, 1L-
1b, IFN-g, and CRP. Moreover, as shown in some (58,59) but not
all previous studies (60), CD4+ T-lymphocyte counts inversely
correlated with LBP, CRP, and IL-6. These ndings indicate some
effects of gut inammation and microbial translocation on systemic
inammation mainly in the more immune-compromised
individuals in our study.
Furthermore, some cytokines seem to be sensitive to low-level
HIV viremia; we observed a strong correlation of third-trimester
HIV RNA levels with IL-4, IL-10, IL-6, IL-13, IL-1b, IL-13, IL-
12p70, and TNF. Similar ndings were also observed in suppressed
and unsuppressed HIV-infected French and African participants
(51,61). Some effects of HIV infection on the immune system are
likely mediated by macrophage/monocyte activation and
inammation (62). In our and a previous Spanish study (4), the
monocyte/macrophage activation marker sCD14 correlated with
IL-2, IL-6, TNF, and CRP.
Microbial translocation might also be relevant in the HIV-
uninfected group, where we found a positive association of taxa
from the Actinobacteriota phylum with LBP and fecal calprotectin,
as well as the genus Actinomyces with fecal calprotectin in line with
existing knowledge (63).
The gut microbiota and its association with
microbial translocation, gut, and systemic
inammation biomarkers
HIV infection can lead to a decrease in gut microbiota diversity,
potentially resulting in the loss of benecial bacteria and the
proliferation of harmful ones (64). In contrast, the gut microbiota
richness and evenness in our study were similar between the HIV-
infected and HIV-uninfected groups in line with previous studies in
cART-treated American and Spanish adults as well as South African
and Italian children (37,50,6568). In contrast, in other studies,
reduced (16,17,64,69) or increased (41) alpha diversity was
observed in HIV-infected cART-naïve individuals. Furthermore,
in our study, microbiota richness or evenness was not inuenced by
immune status, in agreement with some (34,44,70,71) but not all
previous studies in HIV-infected individuals (72). Most likely,
cART-mediated viral suppression can restore or preserve immune
status sufciently that gross effects of HIV on the gut microbiota are
no longer detectable.
Munjoma et al. 10.3389/fimmu.2023.1280262
Frontiers in Immunology frontiersin.org12
We observed a decrease in microbial richness in the HIV-
uninfected group from pregnancy to 6 weeks postpartum, possibly
related to effects of birth and lactation. We also observed increasing
richness in the HIV-infected group from pregnancy to 6 months
after birth. The increase could have been an effect of better maternal
compliance with cART in pregnancy and lactation to prevent HIV
mother-to-child transmission, which would also drive a partial
restoration of the gut-associated lymphoid tissue. However, in
other studies, cART use caused substantial alterations to the
composition of the gut microbiota (41) and a decrease in alpha
diversity with time (73,74).
We show signicant inter-community differences (beta
diversity) of the gut microbiota between HIV-uninfected and
HIV-infected lactating women on cART. Similar ndings were
observed in cART-treated American, Spanish, and Chinese adults
as well as South African and Italian cART-treated children (37,44,
50,64,66,67,75). However, no signicant variability in gut
microbiota composition due to HIV was found in other studies
(17,68), possibly related to varying degrees of immune dysfunction
or co-infection in the study populations.
At phylum level, we found high abundance of Firmicutes,
Actinobacteriota, and Proteobacteria in all participants as
previously reported in Asian and American cART-treated adults
(44,68,76). Increased relative abundances of taxa from the
Actinobacteriota phylum were observed in the HIV-infected
group at 6 weeks postpartum in line with a Japanese study in
cART-treated adults (76).
We found a higher relative abundance of the order
Coriobacteriales and the genera Collinsella and Slackia in the
HIV-infected group, consistent with previous ndings in
ZimbabweanchildrenandcART-experienced Japanese
participants (75,76). The genus Collinsella has been previously
linked to detrimental outcomes such as obesity, non-alcoholic
steatohepatitis, and dyslipidemia (77). The lower relative
abundance of genus Collinsella in the HIV-infected group noted
by Zhao et al. was inconsistent with our results but concurred with
other studies in American and Chinese adults (44,68).
At genus level, we observed lower relative abundance of genus
Clostridium sensu_stricto_1 in the HIV-infected group at 6 weeks
postpartum, consistent with previous studies in African and
Chinese adults (44,59).Clostridiumspecies are important
obligate anaerobes in the human gut with a signicant role in
fermentation and metabolism of carbohydrates and amino acids. In
another Chinese study, Clostridium sensu_stricto_1 positively
correlated with CD4+ and CD8+ T-lymphocytes counts,
suggesting this taxon as a potential marker of improved immune
status in HIV-infected participants (70).
In our study, the relative abundance of the genera Romboutsia
and Clostridium sensu_stricto_1 was lower in the HIV-infected
group at 6 months postpartum in line with a study in South
African children (37). The signicantly lower relative abundances
of Romboutsia and Clostridium sensu_stricto_1 in the HIV-infected
women may signify compromised metabolism and imbalanced
intestinal homeostasis compared with the HIV-uninfected peers.
In line with the benecial role of Clostridium species, we found a
negative association of the genus Clostridia_UCG.014 with IFN-gin
the HIV-uninfected at 6 months postpartum, supporting a role in
attenuating inammation within the human gut.
More than 50% of the HIV-infected participants in our study
were taking cotrimoxazole prophylaxis following the local and
WHO guidelines (78). In our study, cotrimoxazole prophylaxis
had no signicant effects on general descriptors of the gut
microbiota consistent with ndings from a study in HIV-
uninfected Ugandan adults and children (79,80). These ndings
were conrmed with antibiotics other than cotrimoxazole in HIV-
infected and HIV-uninfected individuals (69). However, in a study
in African children, signicant differences were noted in seven
species of the gut microbiota with lower abundances in the
cotrimoxazole treated group (80).
Assessing the relationship between HIV-associated microbial
dysbiosis and systemic inammation, we identied signicant
correlations between the relative abundance of certain taxa and
plasma inammatory biomarkers, warranting further investigations.
Our ndings were inconsistent with studies in Australian cART-naïve
adults and Italian cART-experienced children where correlation of
gut microbiota with systemic cytokines and microbial translocation
markers was not found (17,50).
Strengths and limitations of the study
The strengths of our study lie in its comprehensive approach,
encompassing longitudinal assessments of multiple intestinal and
systemic inammatory biomarkers, as well as the gut microbiota in
HIV-infected and HIV-uninfected controls from the same
community. However, there are limitations that should be
acknowledged. First, because of the inclusion criteria of our study,
we were unable to disentangle the effects of cART and HIV infection,
as all HIV-infected women were receiving cART. Furthermore, the
relatively small sample size hampers the generalizability of our
conclusions, and it is possible that certain associations between
microbial taxa and inammatory biomarkers may have been
overlooked. We are therefore also underpowered to draw
meaningful conclusions from cART duration and HIV viremia
group comparisons. Moreover, our study exclusively comprised
lactating female participants, with no age-matched non-lactating
controls that may restrict the generalizability of our ndings to
other populations. In addition, we did not directly assess microbial
translocation, and measurements of 16S rDNA levels, as previously
done (15) and could be used in follow-up studies. Finally, non-
bacterial infections, such as protozoa, were not tested in our study.
Conclusion
In our study population of HIV-infected lactating women on
cART without AIDS and HIV-related symptoms, many important
parameters of the immune system were not affected by HIV.
However, we identied effects of HIV on gut inammation,
microbial composition, and translocation as well as some
cytokines, in line with a role of intestinal pathology contributing
to systemic inammation in HIV.
Munjoma et al. 10.3389/fimmu.2023.1280262
Frontiers in Immunology frontiersin.org13
Data availability statement
The data presented in this manuscript are tabulated in the main
paper and Supplementary Materials. All sequencing data generated
in the preparation of this manuscript, les and entire details of
used samples used has been deposited in https://doi.org/10.6084/
m9.gshare.24455392.v1.
Ethics statement
The studies involving humans were approved by The Joint
Parirenyatwa Hospitals and University of Zimbabwe Research
Ethics Committee (JREC), JREC/114/20 and the Medical Research
Council of Zimbabwe (MRCZ), MRCZ/A/2663. The studies were
conducted in accordance with the local legislation and institutional
requirements. The participants provided their written informed
consent to participate in this study.
Author contributions
PM: Conceptualization, Formal Analysis, Investigation,
Methodology, Writing original draft. PC: Investigation,
Methodology, Writing review & editing. JW: Formal Analysis,
Writing review & editing. AM: Methodology, Writing review &
editing. SJ: Formal Analysis, Writing review & editing. RG:
Writing review & editing. LK: Conceptualization, Writing
review & editing. BY: Funding acquisition, Methodology,
Supervision, Writing review & editing. BM: Conceptualization,
Funding acquisition, Investigation, Supervision, Writing review &
editing. KD: Conceptualization, Investigation, Supervision, Writing
review & editing.
Funding
The author(s) declare nancial support was received for the
research, authorship, and/or publication of this article. This work
was supported nancially by the Botnar Foundation, Department of
Visceral Surgery and Medicine, Inselspital, Bern University,
Switzerland and the Welcome Trust. BM was supported by the Swiss
National Science Foundation (SNF), Grant Number: 320030_197815
and BY is supported by SNF Ambizione Grant: PZ00P3_185880.
Conict of interest
The authors declare that the research was conducted in the
absence of any commercial or nancial relationships that could be
construed as a potential conict of interest.
Publishers note
All claims expressed in this article are solely those of the authors
and do not necessarily represent those of their afliated
organizations, or those of the publisher, the editors and the
reviewers. Any product that may be evaluated in this article, or
claim that may be made by its manufacturer, is not guaranteed or
endorsed by the publisher.
Supplementary material
The Supplementary Material for this article can be found online at:
https://www.frontiersin.org/articles/10.3389/mmu.2023.1280262/
full#supplementary-material
SUPPLEMENTARY FIGURE 1
Plasma proinammatory and vascular injury biomarkers in HIV infected and
HIV uninfected women. Comparison of plasma proinammatory and vascular
injury biomarkers in HIV-infected vs. HIV uninfected women at 6 weeks
postpartum and 6 months postpartum. Signicance after Bonferroni
correction is indicated as non-signicant (ns), q<0.05 (*), q< 0.01 (**) or q<
0.001 (***). CRP, c-reactive protein; ICAM, intercellular adhesion molecule;
IL, Interleukin; TNF, tumour necrosis factor; INF-g, interferon gamma; VCAM,
vascular cell adhesion molecule.
SUPPLEMENTARY FIGURE 2
Comparison of pregnancy, 6 weeks and 6 months postpartum gut
microbiota. Alpha diversity comparison in HIV-uninfected women (A) and
HIV-infected women (B). Beta diversity comparison in HIV-uninfected
women (C) and HIV-infected women (D).
References
1. Parker E, Judge MA, Macete E, Nhampossa T, Dorward J, Langa DC, et al. HIV
infection in Eastern and Southern Africa: Highest burden, largest challenges, greatest
potential. South Afr J HIV Med (2021) 22(1):1237. doi: 10.4102/sajhivmed.v22i1.1237
2. Dirajlal-Fargo S, Strah M, Ailstock K, Sattar A, Karungi C, Nazzinda R, et al.
Persistent immune activation and altered gut integrity over time in a longitudinal study
of Ugandan youth with perinatally acquired HIV. Front Immunol (2023) 14:1165964.
doi: 10.3389/mmu.2023.1165964
3. Kroeze S, Wit FW, Rossouw TM, Steel HC, Kityo CM, Siwale M, et al. Plasma
biomarkers of human immunodeciency virus-related systemic inammation and
immune activation in sub-saharan Africa before and during suppressive
antiretroviral therapy. J Infect Dis (2019) 220(6):102933. doi: 10.1093/infdis/jiz252
4. Reus S, Portilla J, Sanchez-PayaJ, Giner L, Frances R, Such J, et al. Low-level HIV
viremia is associated with microbial translocation and inammation. J Acquir Immune
Dec Syndr 1999 (2013) 62(2):12934. doi: 10.1097/QAI.0b013e3182745ab0
5. Vidya Vijayan KK, Karthigeyan KP, Tripathi SP, Hanna LE. Pathophysiology of
CD4+ T-cell depletion in HIV-1 and HIV-2 infections. Front Immunol (2017) 8:580.
doi: 10.3389/mmu.2017.00580
6. Vassallo M, MercieP, Cottalorda J, Ticchioni M, Dellamonica P. The role of
lipopolysaccharide as a marker of immune activation in HIV-1 infected patients: a
systematic literature review. Virol J (2012) 9:174. doi: 10.1186/1743-422X-9-174
7. Seethaler B, Basrai M, Neyrinck AM, Nazare JA, Walter J, Delzenne NM, et al.
Biomarkers for assessment of intestinal permeability in clinical practice. Am J Physiol
Gastrointest Liver Physiol (2021) 321(1):G117. doi: 10.1152/ajpgi.00113.2021
8. Eckard AR, Hughes HY, Hagood NL, ORiordan MA, Labbato D, Kosco JC, et al.
Fecal calprotectin is elevated in HIV and related to systemic inammation. J Acquir
Immune Dec Syndr 1999 (2021) 86(2):2319. doi: 10.1097/QAI.0000000000002538
9. Littleeld KM, Schneider JM, Neff CP, Soesanto V, Siebert JC, Nusbacher NM,
et al. Elevated inammatory fecal immune factors in men who have sex with men with
Munjoma et al. 10.3389/fimmu.2023.1280262
Frontiers in Immunology frontiersin.org14
HIV associate with microbiome composition and gut barrier function. Front Immunol
(2022) 13:1072720. doi: 10.3389/mmu.2022.1072720
10. Younas M, Psomas C, Reynes C, Cezar R, Kundura L, Portales P, et al. Microbial
translocation is linked to a specic immune activation prole in HIV-1-infected adults
with suppressed viremia. Front Immunol (2019) 10:2185. doi: 10.3389/mmu.2019.02185
11. De Voeght A, Maes N, Moutschen M. sCD14 is not a bona-de biomarker of
microbial translocation in HIV-1-infected Africans living in Belgium. AIDS Lond Engl
(2016) 30(6):9214. doi: 10.1097/QAD.0000000000000996
12. Ouyang J, Yan J, Zhou X, Isnard S, Harypursat V, Cui H, et al. Relevance of
biomarkers indicating gut damage and microbial translocation in people living with
HIV. Front Immunol (2023) 14:1173956. doi: 10.3389/mmu.2023.1173956
13. Siedner MJ, Zanni M, Tracy RP, Kwon DS, Tsai AC, Kakuhire B, et al. Increased
systemic inammation and gut permeability among women with treated HIV infection
in rural Uganda. J Infect Dis (2018) 218(6):9226. doi: 10.1093/infdis/jiy244
14. Temu TM, Zifodya JS, Polyak SJ, Wagoner J, Wanjalla CN, Masyuko S, et al.
Antiretroviral therapy reduces but does not normalize immune and vascular
inammatory markers in adults with chronic HIV infection in Kenya. AIDS Lond
Engl (2021) 35(1):4551. doi: 10.1097/QAD.0000000000002729
15. Jiang W, Lederman MM, Hunt P, Sieg SF, Haley K, Rodriguez B, et al. Plasma
levels of bacterial DNA correlate with immune activation and the magnitude of
immune restoration in persons with antiretroviral-treated HIV infection. J Infect Dis
(2009) 199(8):117785. doi: 10.1086/597476
16. Dubourg G, Lagier JC, Hüe S, Surenaud M, Bachar D, Robert C, et al. Gut
microbiota associated with HIV infection is signicantly enriched in bacteria tolerant to
oxygen. BMJ Open Gastroenterol (2016) 3(1):e000080. doi: 10.1136/bmjgast-2016-
000080
17. Mak G, Zaunders JJ, Bailey M, Seddiki N, Rogers G, Leong L, et al. Preservation
of gastrointestinal mucosal barrier function and microbiome in patients with controlled
HIV infection. Front Immunol (2021) 12:688886. doi: 10.3389/mmu.2021.688886
18. Pinacchio C, Scagnolari C, Iebba V, Santinelli L, Innocenti GP, Frasca F, et al.
High abundance of genus Prevotella is associated with dysregulation of IFN-I and T cell
response in HIV-1-infected patients. AIDS Lond Engl (2020) 34(10):146773. doi:
10.1097/QAD.0000000000002574
19. Al Bander Z, Nitert MD, Mousa A, Naderpoor N. The gut microbiota and
inammation: an overview. Int J Environ Res Public Health (2020) 17(20):7618. doi:
10.3390/ijerph17207618
20. Marchetti G, Cozzi-Lepri A, Merlini E, Bellistrì GM, Castagna A, Galli M, et al.
Microbial translocation predicts disease progression of HIV-infected antiretroviral-
naive patients with high CD4+ cell count. AIDS (2011) 25(11):1385. doi: 10.1097/
QAD.0b013e3283471d10
21. Duri K, Gumbo FZ, Munjoma PT, Chandiwana P, Mhandire K, Ziruma A, et al.
The University of Zimbabwe College of Health Sciences (UZ-CHS) BIRTH COHORT
study: rationale, design and methods. BMC Infect Dis (2020) 20(1):725. doi: 10.1186/
s12879-020-05432-6
22. Harris PA, Taylor R, Minor BL, Elliott V, Fernandez M, ONeal L, et al. The
REDCap consortium: Building an international community of software platform
partners. J BioMed Inform (2019) 95:103208. doi: 10.1016/j.jbi.2019.103208
23. Update of recommendations on rst- and second-line antiretroviral regimens.
Available at: https://www.who.int/publications-detail-redirect/WHO-CDS-HIV-19.15.
24. Chandiwana P, Munjoma PT, Mazhandu AJ, Mazengera LR, Misselwitz B, Jordi
SBU, et al. Antenatal and postpartum immunological markers levels in women with
HIV infection and malnutrition in a low resource setting: A pilot study. Eur J Inamm
(2022) 20:1721727X221139261. doi: 10.1177/1721727X221139261
25. Yilmaz B, Juillerat P, Øyås O, Ramon C, Bravo FD, Franc Y, et al. Microbial
network disturbances in relapsing refractory Crohns disease. Nat Med (2019) 25
(2):32336. doi: 10.1038/s41591-018-0308-z
26. Yilmaz B, Spalinger MR, Biedermann L, Franc Y, Fournier N, Rossel JB, et al.
The presence of genetic risk variants within PTPN2 and PTPN22 is associated with
intestinal microbiota alterations in Swiss IBD cohort patients. PloS One (2018) 13(7):
e0199664. doi: 10.1371/journal.pone.0199664
27. Whiteley AS, Jenkins S, Waite I, Kresoje N, Payne H, Mullan B, et al. Microbial 16S
rRNA Ion Tag and community metagenome sequencing using the Ion Torrent (PGM)
Platform. J Microbiol Methods (2012) 91(1):808. doi: 10.1016/j.mimet.2012.07.008
28. Bolyen E, Rideout JR, Dillon MR, Bokulich NA, Abnet CC, Al-Ghalith GA, et al.
Reproducible, interactive, scalable and extensible microbiome data science using
QIIME 2. Nat Biotechnol (2019) 37(8):8527. doi: 10.1038/s41587-019-0209-9
29. Caporaso JG, Kuczynski J, Stombaugh J, Bittinger K, Bushman FD, Costello EK,
et al. QIIME allows analysis of high-throughput community sequencing data. Nat
Methods (2010) 7(5):3356. doi: 10.1038/nmeth.f.303
30. McMurdie PJ, Holmes S. phyloseq: an R package for reproducible interactive
analysis and graphics of microbiome census data. PloS One (2013) 8(4):e61217. doi:
10.1371/journal.pone.0061217
31. Callahan BJ, Sankaran K, Fukuyama JA, McMurdie PJ, Holmes SP. Bioconductor
Workow for Microbiome Data Analysis: from raw reads to community analyses.
F1000Research (2016) 5:1492. doi: 10.12688/f1000research.8986.2
32. Mallick H, Rahnavard A, McIver LJ, Ma S, Zhang Y, Nguyen LH, et al.
Multivariable association disco very in population-scale meta-omics studies. PloS
Comput Biol (2021) 17(11):e1009442. doi: 10.1371/journal.pcbi.1009442
33. Wojcik-Cichy K, Piekarska A, Jabłonowska E. Intestinal barrier impairment and
immune activation in HIV-infected advanced late presenters are not dependent on
CD4 recovery. Arch Immunol Ther Exp (Warsz) (2018) 66(4):3217. doi: 10.1007/
s00005-018-0508-8
34. Chandiwana P, Munjoma PT, Mazhandu AJ, Li J, Baertschi I, Wyss J, et al.
Antenatal gut microbiome proles and effect on pregnancy outcome in HIV infected
and HIV uninfected women in a resource limited setting. BMC Microbiol (2023) 23
(1):4. doi: 10.1186/s12866-022-02747-z
35. Hestvik E, Olafsdottir E, Tylleskar T, Aksnes L, Kaddu-Mulindwa D, Ndeezi G,
et al. Faecal calprotectin in HIV-infected, HAART-naïve Ugandan children. J Pediatr
Gastroenterol Nutr (2012) 54(6):78590. doi: 10.1097/MPG.0b013e318241a683
36. Ruiz-Briseño MDR, De Arcos-Jimenez JC, Ratkovich-Gonzalez S, Sanchez-
Reyes K, Gonzalez-Hern andez LA, Andrade-Villanueva JF, et al. Association of
intestinal and systemic inammatory biomarkers with immune reconstitution in
HIV+ patients on ART. JInammation Lond Engl (2020 ) 17:32. doi: 10.1186/
s12950-020-00262-4
37. Goosen C, Proost S, Baumgartner J, Mallick K, Tito RY, Barnabas SL, et al.
Associations of HIV and iron status with gut microbiota composition, gut
inammation and gut integrity in South African school-age children: a two-way
factorial case-control study. J Hum Nutr Diet Off J Br Diet Assoc (2023) 36(3):819
32. doi: 10.1111/jhn.13171
38. Mantegazza C, Maconi G, Giacomet V, Furfaro F, Mameli C, Bezzio C, et al. Gut
and mesenteric lymph node involvement in pediatric patients infected with human
immunodeciency virus. HIVAIDS Auckl NZ (2014) 6:6974. doi: 10.2147/HIV.S60157
39. Tincati C, Merlini E, Braidotti P, Ancona G, Savi F, Tosi D, et al. Impaired gut
junctional complexes feature late-treated individuals with suboptimal CD4+ T-cell
recovery upon virologically suppressive combination antiretroviral therapy. AIDS Lond
Engl (2016) 30(7):9911003. doi: 10.1097/QAD.0000000000001015
40. Gori A, Tincati C, Rizzardini G, Torti C, Quirino T, Haarman M, et al. Early
impairment of gut function and gut ora supporting a role for alteration of
gastrointestinal mucosa in human immunodeciency virus pathogenesis. JClin
Microbiol (2008) 46(2):7578. doi: 10.1128/JCM.01729-07
41. Ancona G, Merlini E, Tincati C, Barassi A, Calcagno A, Augello M, et al. Long-
term suppressive cART is not sufcient to restore intestinal permeability and gut
microbiota compositional changes. Front Immunol (2021) 12:639291. doi: 10.3389/
mmu.2021.639291
42. Kyosiimire-Lugemwa J, Anywaine Z, Abaasa A, Levin J, Gombe B, Musinguzi K,
et al. Effect of stopping cotrimoxazole preventive therapy on microbial translocation
and inammatory markers among human immunodeciency virus-infected Ugandan
adults on antiretroviral therapy: the COSTOP trial immunology substudy. J Infect Dis
(2020) 222(3):38190. doi: 10.1093/infdis/jiz494
43. Neff CP, Krueger O, Xiong K, Arif S, Nusbacher N, Schneider JM, et al. Fecal
microbiota composition drives immune activation in HIV-infected individuals.
EBioMedicine (2018) 30:192202. doi: 10.1016/j.ebiom.2018.03.024
44. Zhao H, Feng A, Luo D, Wu H, Zhang G, Zhang L, et al. Altered gut microbiota
is associated with different immunologic responses to antiretroviral therapy in HIV-
infected men who have sex with men. J Med Virol (2023) 95(3):e28674. doi: 10.1002/
jmv.28674
45. Vesterbacka J, Barqasho B, Häggblom A, Nowak P. Effects of co-trimoxazole on
microbial translocation in HIV-1-infected patients initiating antiretroviral therapy.
AIDS Res Hum Retroviruses (2015) 31(8):8306. doi: 10.1089/aid.2014.0366
46. Knudsen AD, Bouazzi R, Afzal S, Gelpi M, Beneld T, gh J, et al. Monocyte
count and soluble markers of monocyte activation in people living with HIV and
uninfected controls. BMC Infect Dis (2022) 22(1):451. doi: 10.1186/s12879-022-07450-y
47. Hunt PW, Sinclair E, Rodriguez B, Shive C, Clagett B, Funderburg N, et al. Gut
epithelial barrier dysfunction and innate immune activation predict mortality in treated
HIV infection. J Infect Dis (2014) 210(8):122838. doi: 10.1093/infdis/jiu238
48. Sandler NG, Wand H, Roque A, Law M, Nason MC, Nixon DE, et al. Plasma
levels of soluble CD14 independently predict mortality in HIV infection. J Infect Dis
(2011) 203(6):78090. doi: 10.1093/infdis/jiq118
49. Temu TM, Polyak SJ, Zifodya JS, Wanjalla CN, Koethe JR, Masyuko S, et al.
Endothelial dysfunction is related to monocyte activation in antiretroviral-treated
people with HIV and HIV-negative adults in Kenya. Open Forum Infect Dis (2020) 7
(10):ofaa425. doi: 10.1093/od/ofaa425
50. Tincati C, Ficara M, Ferrari F, Augello M, Dotta L, Tagliabue C, et al. Gut-
dependent inammation and alterations of the intestinal microbiota in individuals with
perinatal HIV exposure and different HIV serostatus. AIDS Lond Engl (2022) 36
(14):191725. doi: 10.1097/QAD.0000000000003324
51. Redd AD, Dabitao D, Bream JH, Charvat B, Laeyendecker O, Kiwanuka N, et al.
Microbial translocation, the innate cytokine response, and HIV-1 disease progression in
Africa. Proc Natl Acad Sci USA (2009) 106(16):671823. doi: 10.1073/pn as.0901983106
52. Watanabe M, Jergovic M, Davidson L, LaFleur BJ, Castaneda Y, Martinez C,
et al. Inammatory and immune markers in HIV-infected older adults on long-term
antiretroviral therapy: Persistent elevation of sCD14 and of proinammatory effector
memory T cells. Aging Cell (2022) 21(9):e13681. doi: 10.1111/acel.13681
53. Espı
ndola MS, Lima LJG, Soares LS, Cacemiro MC, Zambuzi FA, de Souza
Gomes M, et al. Dysregulated immune activation in second-line HAART HIV+
Patients is similar to that of untreated patients. PloS One (2015) 10(12):e0145261.
doi: 10.1371/journal.pone.0145261
Munjoma et al. 10.3389/fimmu.2023.1280262
Frontiers in Immunology frontiersin.org15
54. Sereti I, Krebs SJ, Phanuphak N, Fletcher JL, Slike B, Pinyakorn S, et al.
Persistent, albeit reduced, chronic inammation in persons starting antiretroviral
therapy in acute HIV infection. Clin Infect Dis Off Publ Infect Dis Soc Am (2017) 64
(2):12431. doi: 10.1093/cid/ciw683
55. Subramanya V, McKay HS, Brusca RM, Palella FJ, Kingsley LA, Witt MD, et al.
Inammatory biomarkers and subclinical carotid atherosclerosis in HIV-infected and
HIV-uninfected men in the Multicenter AIDS Cohort Study. PloS One (2019) 14(4):
e0214735. doi: 10.1371/journal.pone.0214735
56. Wallet MA, Buford TW, Joseph AM, Sankuratri M, Leeuwenburgh C, Pahor M,
et al. Increased inammation but similar physical composition and function in older-
aged, HIV-1 infected subjects. BMC Immunol (2015) 16:43. doi: 10.1186/s12865-015-
0106-z
57. Xie Y, Sun J, Wei L, Jiang H, Hu C, Yang J, et al. Altered gut microbiota correlate
with different immune responses to HAART in HIV-infected individuals. BMC
Microbiol (2021) 21(1):11. doi: 10.1186/s12866-020-02074-1
58. Kroeze S, Rossouw TM, Steel HC, Wit FW, Kityo CM, Siwale M, et al. Plasma
inammatory biomarkers predict CD4+ T-cell recovery and viral rebound in HIV-1
infected africans on suppressive antiretroviral therapy. J Infect Dis (2021) 224(4):6738.
doi: 10.1093/infdis/jiaa787
59. Parbie PK, Mizutani T, Ishizaka A, Kawana-Tachikawa A, Runtuwene LR, Seki S,
et al. Dysbiotic fecal microbiome in HIV-1 infected individuals in Ghana. Front Cell
Infect Microbiol (2021) 11:646467. doi: 10.3389/fcimb.2021.646467
60. Abad-Fernandez M, Vallejo A, Hernandez-Novoa B, Dı
az L, Gutierrez C,
Madrid N, et al. Correlation between different methods to measure microbial
translocation and its association with immune activation in long-term suppressed
HIV-1-infected individuals. J Acquir Immune Dec Syndr 1999 (2013) 64(2):14953.
doi: 10.1097/QAI.0b013e31829a2f12
61. Bastard JP, SoulieC, Fellahi S, Haïm-Boukobza S, Simon A, Katlama C, et al.
Circulating interleukin-6 levels correlate with residual HIV viraemia and markers of
immune dysfunction in treatment-controlled HIV-infected patients. Antivir Ther
(2012) 17(5):9159. doi: 10.3851/IMP2093
62. Hakansson A, Molin G. Gut microbiota and inammation. Nutrients (2011) 3
(6):63782. doi: 10.3390/nu3060637
63. Li J, Li Y, Zhou Y, Wang C, Wu B, Wan J. Actinomyces and alimentary tract
diseases: A review of its biological functions and pathology. BioMed Res Int (2018)
2018:3820215. doi: 10.1155/2018/3820215
64. Mutlu EA, Keshavarzian A, Losurdo J, Swanson G, Siewe B, Forsyth C, et al. A
compositional look at the human gastrointestinal microbiome and immune activation
parameters in HIV infected subjects. PloS Pathog (2014) 10(2):e1003829. doi: 10.1371/
journal.ppat.1003829
65. Cook RR, Fulcher JA, Tobin NH, Li F, Lee D, Javanbakht M, et al. Effects of HIV
viremia on the gastrointestinal microbiome of young MSM. AIDS Lond Engl (2019) 33
(5):793804. doi: 10.1097/QAD.0000000000002132
66. Dinh DM, Volpe GE, Duffalo C, Bhalchandra S, Tai AK, Kane AV, et al.
Intestinal microbiota, microbial translocation, and systemic inammation in chronic
HIV infection. J Infect Dis (2015) 211(1):1927. doi: 10.1093/infdis/jiu409
67. Villoslada-Blanco P, Perez-Matute P, I
ñiguez M, Recio-Fernandez E, Blanco-
Navarrete P, Metola L, et al. Integrase inhibitors partially restore bacterial translocation,
inammation and gut permeability induced by HIV infection: impact on gut
microbiota. Infect Dis Ther (2022) 11(4):154157. doi: 10.1007/s40121-022-00654-4
68. Wang Z, Usyk M, Sollecito CC, Qiu Y, Williams-Nguyen J, Hua S, et al. Altered
gut microbiota and host metabolite proles in women with human immunodeciency
virus. Clin Infect Dis Off Publ Infect Dis Soc Am (2020) 71(9):234553. doi: 10.1093/cid/
ciz1117
69. Ellis RJ, Iudicello JE, Heaton RK, Isnard S, Lin J, Routy JP, et al. Markers of gut
barrier functi on and microbial translocation associate with lower gut microbial
diversity in people with HIV. Viruses (2021) 13(10):1891. doi: 10.3390/v13101891
70. Mingjun Z, Fei M, Zhousong X, Wei X, Jian X, Yuanxue Y, et al. 16S rDNA
sequencing analyzes differences in intestinal ora of human immunodeciency virus
(HIV) patients and association with immune activation. Bioengineered (2022) 13
(2):408599. doi: 10.1080/21655979.2021.2019174
71. Abange WB, Martin C, Nanfack AJ, Yatchou LG, Nusbacher N, Nguedia CA, et al.
Alteration of the gut fecal microbiome in children living with HIV on antiretroviral therapy
in Yaounde, Cameroon. Sci Rep (2021) 11(1):7666. doi: 10.1038/s41598-021-87368-8
72. Ji Y, Zhang F, Zhang R, Shen Y, Liu L, Wang J, et al. Changes in intestinal
microbiota in HIV-1-infected subjects following cART initiation: inuence of CD4+ T
cell count. Emerg Microbes Infect (2018) 7(1):113. doi: 10.1038/s41426-018-0117-y
73. Imahashi M, Ode H, Kobayashi A, Nemoto M, Matsuda M, Hashiba C, et al.
Impact of long-term antiretroviral therapy on gut and oral microbiotas in HIV-1-
infected patients. Sci Rep (2021) 11(1):960. doi: 10.1038/s41598-020-80247-8
74. Nowak P, Troseid M, Avershina E, Barqasho B, Neogi U, Holm K, et al. Gut
microbiota diversity predicts immune status in HIV-1 infection. AIDS Lond Engl
(2015) 29(18):240918. doi: 10.1097/QAD.0000000000000869
75. Flygel TT, Sovershaeva E, Claassen-Weitz S, Hjerde E, Mwaikono KS, Odland
JØ, et al. Composition of gut microbiota of children and adolescents with perinatal
human immunodeciency virus infection taking antiretroviral therapy in Zimbabwe. J
Infect Dis (2020) 221(3):48392. doi: 10.1093/infdis/jiz473
76. Ishizaka A, Koga M, Mizutani T, Parbie PK, Prawisuda D, Yusa N, et al. Unique
gut microbiome in HIV patients on antiretroviral therapy (ART) suggests association
with chronic inammation. Microbiol Spectr (2021) 9(1):e0070821. doi: 10.1128/
Spectrum.00708-21
77. Astbury S, Atallah E, Vijay A, Aithal GP, Grove JI, Valdes AM. Lower gut
microbiome diversity and higher abundance of proinammatory genus Collinsella are
associated with biopsy-proven nonalcoholic steatohepatitis. Gut Microbes (2020) 11
(3):56980. doi: 10.1080/19490976.2019.1681861
78. Guidelines on post-exposure prophylaxis for HIV and the use of co-trimoxazole
prophylaxis for HIV-related infections among adults, adolescents and children. Available
at: https://www.who.int/publications-detail-redirect/9789241506830.
79. Monaco CL, Gootenberg DB, Zhao G, Handley SA, Ghebremichael MS, Lim ES,
et al. Altered virome and bacterial microbiome in human immunodeciency virus-
associated acquired immunodeciency syndrome. Cell Host Microbe (2016) 19(3):311
22. doi: 10.1016/j.chom.2016.02.011
80. Bourke CD, Gough EK, Pimundu G, Shonhai A, Berejena C, Terry L, et al.
Cotrimoxazole reduces systemic inammation in HIV infection by altering the gut
microbiome and immune activation. Sci Transl Med (2019) 11(486):eaav0537. doi:
10.1126/scitranslmed.aav0537
Munjoma et al. 10.3389/fimmu.2023.1280262
Frontiers in Immunology frontiersin.org16
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
The intestinal barrier has the daunting task of allowing nutrient absorption while limiting the entry of microbial products into the systemic circulation. HIV infection disrupts the intestinal barrier and increases intestinal permeability, leading to microbial product translocation. Convergent evidence has shown that gut damage and an enhanced level of microbial translocation contribute to the enhanced immune activation, the risk of non-AIDS comorbidity, and mortality in people living with HIV (PLWH). Gut biopsy procedures are invasive, and are not appropriate or feasible in large populations, even though they are the gold standard for intestinal barrier investigation. Thus, validated biomarkers that measure the degree of intestinal barrier damage and microbial translocation are needed in PLWH. Hematological biomarkers represent an objective indication of specific medical conditions and/or their severity, and should be able to be measured accurately and reproducibly via easily available and standardized blood tests. Several plasma biomarkers of intestinal damage, i.e., intestinal fatty acid-binding protein (I-FABP), zonulin, and regenerating islet-derived protein-3α (REG3α), and biomarkers of microbial translocation, such as lipopolysaccharide (LPS) and (1,3)-β-D-Glucan (BDG) have been used as markers of risk for developing non-AIDS comorbidities in cross sectional analyses and clinical trials, including those aiming at repair of gut damage. In this review, we critically discuss the value of different biomarkers for the estimation of gut permeability levels, paving the way towards developing validated diagnostic and therapeutic strategies to repair gut epithelial damage and to improve overall disease outcomes in PLWH.
Article
Full-text available
Introduction Perinatally acquired HIV infection (PHIV) occurs during a critical window of immune development. We investigated changes in systemic inflammation and immune activation in adolescents with PHIV and those without HIV (HIV-) in Uganda. Methods A prospective observational cohort study was performed in 2017-2021 in Uganda. All participants were between 10-18 years of age and without active co-infections. PHIVs were on ART with HIV-1 RNA level ≤400 copies/mL. We measured plasma and cellular markers of monocyte activation, T-cell activation (expression of CD38 and HLA-DR on CD4+ and CD8+), oxidized LDL, markers of gut integrity and fungal translocation. Groups were compared using Wilcoxon rank sum tests. Changes from baseline were examined with 97.5% confidence intervals on relative fold change. P values were adjusted for false discovery rate. Results We enrolled 101 PHIV and 96 HIV-; among these, 89 PHIV and 79 HIV- also had measurements at 96 weeks. At baseline, median (Q1, Q3) age was 13 yrs (11,15), and 52% were females. In PHIV, median CD4+ cell counts were 988 cells/µL (638, 1308), ART duration was 10 yrs (8, 11), and 85% had viral load <50 copies/mL throughout the study, 53% of participants had a regimen switch between visits, 85% of whom switched to 3TC, TDF and DTG. Over 96 weeks, while hsCRP decreased by 40% in PHIV (p=0.12), I-FABP and BDG both increased by 19 and 38% respectively (p=0.08 and ≤0.01) and did not change in HIV- (p≥0.33). At baseline, PHIVs had higher monocyte activation (sCD14) (p=0.01) and elevated frequencies of non-classical monocytes (p<0.01) compared to HIV- which remained stable over time in PHIV but increased by 34% and 80% respectively in HIV-. At both time points, PHIVs had higher T cell activation (p ≤ 0.03: CD4+/CD8+ T cells expressing HLA-DR and CD38). Only in PHIV, at both timepoints, oxidized LDL was inversely associated with activated T cells(p<0.01). Switching to dolutegravir at week 96 was significantly associated an elevated level of sCD163 (β=0.4, 95% CI=0.14,0.57, p<0.01), without changes in other markers. Conclusion Ugandan PHIV with viral suppression have some improvement in markers of inflammation over time, however T-cell activation remains elevated. Gut integrity and translocation worsened only in PHIV over time. A deeper understanding of the mechanisms causing immune activation in ART treated African PHIV is crucial.
Article
Full-text available
Background: HIV and iron deficiency (ID) affect many African children. Both HIV and iron status interact with gut microbiota composition and related biomarkers. The study's aim was to determine the associations of HIV and iron status with gut microbiota composition, gut inflammation, and gut integrity in South African school-age children. Methods: In this two-way factorial case-control study, 8- to 13-year-old children were enrolled into four groups according to their HIV and iron status: (1) With HIV (HIV+) and ID (n=43); (2) HIV+ and iron-sufficient non-anaemic (n=41); (3) Without HIV (HIV-) and ID (n=44); and (4) HIV- and iron-sufficient non-anaemic (n=38). HIV+ children were virally suppressed (<50 HIV RNA copies/mL) on antiretroviral therapy (ART). Microbial composition of faecal samples (16S rRNA sequencing) and markers of gut inflammation (faecal calprotectin) and gut integrity (plasma intestinal fatty acid-binding protein (I-FABP)) were assessed. Results: Faecal calprotectin was higher in ID vs. iron-sufficient non-anaemic children (p=0.007). I-FABP did not significantly differ by HIV or iron status. ART-treated HIV (redundancy analysis (RDA) R2 =0.009, p=0.029) and age (RDA R2 = 0.013 p=0.004) explained the variance in the gut microbiota across the four groups. Probabilistic models showed that the relative abundance of the butyrate-producing genera Anaerostipes and Anaerotruncus was lower in ID vs. iron-sufficient children. Fusicatenibacter was lower in HIV+ and in ID children vs. their respective counterparts. The prevalence of the inflammation-associated genus Megamonas was 42% higher in children with both HIV and ID vs. HIV- and iron-sufficient non-anaemic counterparts. Conclusions: In our sample of 8- to 13-year-old virally suppressed HIV+ and HIV- children with or without ID, ID was associated with increased gut inflammation and changes in the relative abundance of specific microbiota. Moreover, in HIV+ children, ID had a cumulative effect that further shifted the gut microbiota to an unfavourable composition. This article is protected by copyright. All rights reserved.
Article
Full-text available
Background: Human immunodeficiency virus (HIV) severely damages the epithelial cells of the gut lining leading to an inflamed leaky gut, translocation of microbial products, and dysbiosis resulting in systemic immune activation. Also, microbiota composition and maternal gut function can be altered in pregnancy through changes in the immune system and intestinal physiology. The aim of this study was to investigate the gut microbiota in HIV-infected and HIV-uninfected pregnant women and to compare and identify the association between gut microbial composition and adverse birth outcomes. Results: A total of 94 pregnant women (35 HIV-infected and 59 HIV-uninfected controls) were recruited in Harare from 4 polyclinics serving populations with relatively poor socioeconomic status. Women were of a median age of 28 years (interquartile range, IQR: 22.3-32.0) and 55% of women were 35 weeks gestational age at enrolment (median 35.0 weeks, IQR: 32.5-37.2). Microbiota profiling in these participants showed that species richness was significantly lower in the HIV-infected pregnant women compared to their HIV-uninfected peers and significant differences in β-diversity using Bray-Curtis dissimilarity were observed. In contrast, there was no significant difference in α-diversity between immune-compromised (CD4+ < 350 cells/µL) and immune-competent HIV-infected women (CD4+ ≥ 350 cells/µL) even after stratification by viral load suppression. HIV infection was significantly associated with a reduced abundance of Clostridium, Turicibacter, Ruminococcus, Parabacteroides, Bacteroides, Bifidobacterium, Treponema, Oscillospira, and Faecalibacterium and a higher abundance of Actinomyces, and Succinivibrio. Low infant birth weight (< 2500 g) was significantly associated with high abundances of the phylum Spirochaetes, the families Spirochaeteceae, Veillonellaceae, and the genus Treponema. Conclusion: The results reported here show that the species richness and taxonomy composition of the gut microbiota is altered in HIV-infected pregnant women, possibly reflecting intestinal dysbiosis. Some of these taxa were also associated with low infant birth weight.
Article
Full-text available
Introduction People living with HIV infection (PLWH) exhibit elevated levels of gastrointestinal inflammation. Potential causes of this inflammation include HIV infection and associated immune dysfunction, sexual behaviors among men who have sex with men (MSM) and gut microbiome composition. Methods To better understand the etiology of gastrointestinal inflammation we examined levels of 28 fecal soluble immune factors (sIFs) and the fecal microbiome in well-defined cohorts of HIV seronegative MSM (MSM-SN), MSM with untreated HIV infection (MSM-HIV) and MSM with HIV on anti-retroviral treatment (MSMART). Additionally, fecal solutes from these participants were used to stimulate T-84 colonic epithelial cells to assess barrier function. Results Both MSM cohorts with HIV had elevated levels of fecal calprotectin, a clinically relevant marker of GI inflammation, and nine inflammatory fecal sIFs (GM-CSF, ICAM-1, IL-1β, IL-12/23, IL-15, IL-16, TNF-β, VCAM-1, and VEGF). Interestingly, four sIFs (GM-CSF, ICAM-1, IL-7 and IL-12/23) were significantly elevated in MSM-SN compared to seronegative male non-MSM. Conversely, IL-22 and IL-13, cytokines beneficial to gut health, were decreased in all MSM with HIV and MSM-SN respectively. Importantly, all of these sIFs significantly correlated with calprotectin, suggesting they play a role in GI inflammation. Principal coordinate analysis revealed clustering of fecal sIFs by MSM status and significant associations with microbiome composition. Additionally, fecal solutes from participants in the MSM-HIV cohort significantly decreased colonic transcellular fluid transport in vitro, compared to non-MSM-SN, and this decrease associated with overall sIF composition and increased concentrations of eight inflammatory sIFs in participants with HIV. Lastly, elevated levels of plasma, sCD14 and sCD163, directly correlated with decreased transcellular transport and microbiome composition respectively, indicating that sIFs and the gut microbiome are associated with, and potentially contribute to, bacterial translocation. Conclusion Taken together, these data demonstrate that inflammatory sIFs are elevated in MSM, regardless of HIV infection status, and are associated with the gut microbiome and intestinal barrier function.
Article
Full-text available
Objectives: Both, Human Immunodeficiency Virus (HIV) infection and malnutrition are major challenges in pregnancy and postpartum in low-resource settings and the respective cytokine levels remain poorly described. The main objectives of this study were to find immune markers that are associated with HIV infection and malnutrition in pregnant women and to determine how these would change at 14 weeks postpartum.Method: Pregnant women of at least 20 weeks gestational age were enrolled into this longitudinal observational single centre pilot study at 4 primary health clinics in high-density areas around Harare, Zimbabwe. Socio-demographic and clinical data including plasma samples were collected in pregnancy and 14 weeks postpartum (PP). Mid-upper-arm circumference (MUAC) ≤23 cm was used as an indicator for malnourishment. Fifty-six cytokines and chemokines were assayed in plasma using the Mesoscale multiplex assay. We determined cytokine/chemokine levels including markers for vascular injury in HIV-infection and malnutrition. Associations remaining significant after multiple test correction were confirmed in multivariable analyses after controlling for confounders.Results: Ninety-seven pregnant women were recruited for this study and from these, 44 were randomly selected for cytokine assaying of which 20 HIV infected, 15 malnourished, and 9 well-nourished HIV uninfected participants. HIV infection was associated with significantly higher interleukin (IL)-4 ( q < 0.05) and IL-10 ( q < 0.001) in pregnancy. Longitudinally, IL-4 ( q < 0.01) and IL-10 ( q < 0.001) significantly increased in HIV uninfected women whilst in the HIV-infected both were non-significantly decreased. IL-8 (q < 0.05) levels significantly increased in HIV-infected women from pregnancy to 14 weeks PP. Vascular Cell Adhesion Molecule 1 (VCAM-1) ( q < 0.05) and interleukin-1 receptor antagonist (IL-1RA) ( q < 0.05) were significantly lower in malnourished women in pregnancy and 14 weeks PP, respectively. Conclusions: IL-4, IL-8, IL-10, and VCAM-1 are potential biomarkers for monitoring immune functioning in HIV-infected pregnant women and malnutrition. However, studies with larger sample size are warranted to confirm these findings.
Article
Full-text available
HIV-positive patients whose viral loads are successfully controlled by active antiretroviral therapy (ART) show no clinical signs of AIDS. However, their lifespan is shorter compared with individuals with no HIV infection and they prematurely exhibit a multitude of chronic diseases typically associated with advanced age. It was hypothesized that immune system aging may correlate with, and provide useful biomarkers for, this premature loss of healthspan in HIV-positive subjects. Here, we tested whether the immune correlates of aging, including cell numbers and phenotypes, inflammatory status, and control of human cytomegalovirus (hCMV) in HIV-positive subjects on long-term successful ART (HIV+) may reveal increased "immunological age" compared with HIV-negative, age-matched cohort (HIV-) in participants between 50 and 69 years of age. Specifically, we expected that younger HIV+ subjects may immunologically resemble older individuals without HIV. We found no evidence to support this hypothesis. While T cells from HIV+ participants displayed differential expression in several differentiation and/or inhibitory/exhaustion markers in different T cell subpopulations, aging by a decade did not pronounce these changes. Similarly, while the HIV+ participants exhibited higher T cell responses and elevated inflammatory marker levels in plasma, indicative of chronic inflammation, this trait was not age-sensitive. We did find differences in immune control of hCMV, and, more importantly, a sustained elevation of sCD14 and of proinflammatory CD4 and CD8 T cell responses across age groups, pointing towards uncontrolled inflammation as a factor in reduced healthspan in successfully treated older HIV+ patients.
Article
Full-text available
Objective: HIV-exposed infected (HEI) and uninfected (HEU) children represent the two possible outcomes of maternal HIV infection. Modifications of the intestinal microbiome have been linked to clinical vulnerability in both settings, yet whether HEI and HEU differ in terms of gut impairment and peripheral inflammation/activation is unknown. Design: We performed a cross-sectional, pilot study on fecal and plasma microbiome as well as plasma markers of gut damage, microbial translocation, inflammation and immune activation in HIV-infected and uninfected children born from an HIV-infected mother. Methods: Fecal and plasma microbiome were determined by means of 16S rDNA amplification with subsequent qPCR quantification. Plasma markers were quantified via ELISA. Results: Forty-seven HEI and 33 HEU children were consecutively enrolled. The two groups displayed differences in fecal beta-diversity and relative abundance, yet similar microbiome profiles in plasma as well as comparable gut damage and microbial translocation. In contrast, monocyte activation (sCD14) and systemic inflammation (IL-6) were significantly higher in HEI than HEU. Conclusion: In the setting of perinatal HIV infection, enduring immune activation and inflammation do not appear to be linked to alterations within the gut. Given that markers of activation and inflammation are independent predictors of HIV disease progression, future studies are needed to understand the underlying mechanisms of such processes and elaborate adjuvant therapies to reduce the clinical risk in individuals with perinatal HIV infection.
Article
Full-text available
Introduction: Human immunodeficiency virus (HIV) infection can be considered a chronic disease thanks to the extended use of antiretroviral treatment (ART). In this context, low-grade chronic inflammation related to gut microbiota (GM) dysbiosis and bacterial translocation (BT) among other factors has been observed despite the use of ART. In addition, different ART regimens have demonstrated differential impacts on GM. However, the role of novel integrase strand transfer inhibitors (INSTIs) has not been investigated yet. The aim of this study was to analyse the effects of INSTIs in first-line of treatment on markers of BT, inflammation, cardiovascular risk, gut permeability and GM composition and derived short-chain fatty acids. Methods: Twenty-six non-HIV-infected volunteers and 30 HIV-infected patients (15 naïve and 15 under INSTIs regimen) were recruited. Blood samples were extracted to analyse biochemical parameters and markers of BT, inflammation, cardiovascular risk, gut permeability and bacterial metabolism. GM composition was analysed using 16S rRNA gene sequencing. Results: Our results showed that HIV infection increased BT, inflammation, cardiovascular risk and gut permeability, whereas INSTIs counteracted these effects. Regarding GM, the reduction in bacterial richness induced by HIV infection was restored by INSTIs. Beta diversity revealed that HIV-infected people were separated from the control group independently of treatment. Conclusions: Current antiretroviral regimens based on INSTIs are able to reverse the impact of HIV infection on BT, systemic inflammation, gut permeability and bacterial diversity/richness, reaching similar levels to those observed in an uninfected/control population. These results suggest a protective role of INSTIs in disease progression, subsequent immune activation and in the development of future age-related complications such as cardiovascular events.
Article
Background: The association between gut microbiota and immunologic non-response among people living with HIV (PLHIV) on antiretroviral therapy (ART) is not well documented. This study aimed to characterize gut microbiota among HIV-infected men who have sex with men (MSM) with different immunologic responses. Methods: We recruited HIV-infected MSM and HIV-uninfected MSM (healthy controls, HC) in Guangzhou, June-October 2021. HIV-infected MSM were grouped into good immunological responders (GIR) (CD4+ T cell count ≥ 350 cells/μL) and poor immunological responders (PIR) (< 350 cells/μL). Blood and stool samples were collected. Microbial translocation in serum was performed using enzyme linked immunosorbent assay (ELISA). Bacterial 16S ribosomal DNA sequencing was performed on stool samples, and microbial metabolites were obtained through gas chromatography-mass spectrometry. Results: 56 GIR, 41 PIR and 51 HC were included. Microbial translocation marker soluble cluster of differentiation 14 (sCD14) in both GIR and PIR groups was significantly higher than that in HC. Compared with PIR or HC groups, the genera of Coprococcus, Blautia, Clostridium, and SMB53 were decreased, whereas Megamonas and Megasphaera were more abundant in GIR group. Compared with GIR or PIR groups, Bifidobacterium, Collinsella, Faecalibacterium, Oscillospira, and Roseburia were more abundant, whereas Escherichia was decreased in HC group. The levels of benzenoids, imidazoles, phenylpropanoic acids, phenylpropanoids, and pyridines showed strongly significant correlations between differential genera. Conclusions: This study presented a comprehensive landscape of gut microbiota in PLHIV with different treatment outcomes. Megamonas, Coprococcus and Blautia were the major genera correlated with different immunologic responses in PLHIV. This article is protected by copyright. All rights reserved.