Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Infection (2014) 42:951–959

DOI 10.1007/s15010-014-0666-5

REVIEW

Intestinal barrier dysfunction in HIV infection: pathophysiology,


clinical implications and potential therapies
S. F. Assimakopoulos • D. Dimitropoulou •

M. Marangos • C. A. Gogos

Received: 21 April 2014 / Accepted: 11 July 2014 / Published online: 29 July 2014
Ó Springer-Verlag Berlin Heidelberg 2014

Abstract local and systemic inflammatory response. This chronic


Introduction Current pathogenetic aspects on HIV immune activation is associated with HIV disease pro-
infection highlight the importance of a chronic immune gression, suboptimal response to HAART and development
activation ultimately leading to T lymphocyte homeostasis of non-AIDS comorbidities.
disruption and immune deregulation associated with dis- Conclusions We have reached a point where the effective
ease manifestations and progression. It is widely accepted control of HIV viremia by HAART should be combined
that this continuous immune activation in HIV infection is with emerging pharmacological approaches aiming at the
principally driven by the phenomenon of pathological restoration of the intestinal barrier, targeting its diverse
microbial translocation (MT). levels of structure and function. Elimination of the MT
Methods Review of the literature on the role of intestinal phenomenon would mitigate its effect on immune
barrier dysfunction in HIV infection, with emphasis on the homeostasis, which might improve the prognosis of the
implicated pathophysiological mechanisms, clinical impli- HIV-infected patient in terms of morbidity and mortality.
cations and potentially effective therapeutic interventions.
Findings MT in HIV infection is promoted by a multi- Keywords HIV  AIDS  Intestinal barrier  Intestinal
factorial disruption of all major levels comprising the permeability  Microbial translocation  Bacterial
intestinal barrier defense. Specifically, HIV infection dis- translocation  Endotoxemia
rupts the integrity of the intestinal biological (quantitative
and qualitative alterations of gut microecology, overgrowth
of pathogenic bacteria), immune (depletion of CD4(?) T Introduction
cells, especially Th17 cells, increased CD4? FoxP3?
Tregs, decreased mucosal macrophages phagocytic capac- The current pathogenetic aspects on HIV infection high-
ity, development of intestinal proinflammatory milieu) and light the importance of a chronic immune activation ulti-
mechanical barrier (enterocytes’ apoptosis, disruption of mately leading to T lymphocyte homeostasis disruption
tight junctions). Intestinal barrier dysfunction allows the and immune deregulation associated with disease mani-
passage of microbes and immunostimulatory bioproducts festations and progression [1–3]. It is currently widely
from the gut lumen first in the lamina propria and thereafter accepted that chronic immune activation in HIV infection
in the systemic circulation, thus continuously promoting a is to a great extent dependent on the pathological microbial
translocation (MT) of intestinal microbes or their products
S. F. Assimakopoulos and D. Dimitropoulou have contributed equally
from the gastrointestinal tract to portal and systemic cir-
to this review. culation [3, 4]. This pathological MT has been repeatedly
confirmed in experimental and clinical HIV infection, takes
S. F. Assimakopoulos (&)  D. Dimitropoulou  place even from the early phase of acute infection and
M. Marangos  C. A. Gogos
throughout the course of chronic disease, and has been
Department of Internal Medicine, Division of Infectious
Diseases, University Hospital of Patras, 26504 Patras, Greece associated with HIV disease progression, response to
e-mail: sassim@upatras.gr treatment and non-AIDS comorbidities [5–7].

123
952 S. F. Assimakopoulos et al.

The intestinal barrier: a brief overview even from the early stages of HIV infection. This is related
to the fact that most of the CD4? T cells of the intestine
The intestinal epithelium has a complex role as it allows are CCR5? activated memory CD4? T cells which are
absorption and excretion of substances necessary for body preferred cellular targets for HIV [14, 15]. Depletion of
homeostasis and at the same time acts as a barrier to var- intestinal CD4? CCR5? T cells continues throughout the
ious pathogens and their products. The gut barrier function chronic phase of the disease to a greater extent in the
consists of three major lines of defense [8]: (a) the bio- intestinal tissue as compared to blood and lymph nodes
logical barrier, which is made up of normal intestinal flora [16].
responsible for colonization resistance; (b) the immune The Th17 cells, which are subpopulations of CD4? T
barrier, composed of locally acting factors such as secre- cells producing IL-17, exert pivotal role in the prevention
tory IgA, intra-mucosal lymphocytes, Payer’s nodules, of MT by stimulating epithelial cell proliferation and
mesenteric lymph nodes and the systemic host defense antibacterial defensin expression and by recruiting neu-
represented mainly by the reticuloendothelial system; and trophils to the gut-associated lymphoid tissue to clear
(c) the mechanical barrier, consisting of the closed-lining pathogens and their bioproducts [17, 18]. In HIV infection,
intestinal epithelial cells and the capillary endothelial cells. the early and massive loss of Th17 cells from the intestinal
The epithelial and endothelial cells come into the closest mucosal epithelium has been proposed as an important
possible contact in the most apical part of the lateral cell promoter of MT [19]. Previous studies have additionally
membranes (‘‘kissing points’’) by specific structures named shown that not only the loss of Th17 cells, but also the
‘‘tight junctions’’ (TJs), which interconnect the cells and increased presence of CD4? FoxP3? Tregs might con-
restrict the passage of ions, molecules and cells through the tribute to the breakdown of the intestinal barrier integrity
paracellular space [8, 9]. [20]. The selective depletion of Th17 cells from the
intestinal mucosa in HIV infection could be explained
either by a direct cytotoxic effect of HIV in this cell pop-
The intestinal barrier in HIV infection ulation or by the inhibitory effect of proinflammatory
cytokines and in particular of IFN-c on Th17 cells devel-
HIV and the intestinal biological barrier opment, which is exerted with a concurrent promoting
action on Treg differentiation, thus altering the intestinal
The specific composition of the gut microbiome exerts immune cell balance [20].
many immunologic, metabolic and structural functions that Additionally, a recent study demonstrated the important
are essential to maintain gut barrier integrity [10]. Previous role of mucosal macrophages in the maintenance of gut
studies on HIV-infected patients have demonstrated the epithelial barrier integrity. It was found that the intestinal
presence of quantitative and qualitative alterations of their mucosa of untreated HIV-infected patients was signifi-
gut microbiota [11, 12]. Bifidobacteria and lactobacilli, cantly infiltrated by macrophages secreting proinflamma-
which have been shown to have a positive impact on tory cytokines that may promote epithelial injury [21].
mucosal immune function and barrier integrity, were found Moreover, the phagocytic activity of the accumulated
at significantly lower levels in HIV-infected patients as intestinal mucosa macrophages was significantly impaired
compared to healthy subjects. On the other hand, oppor- contributing to the development of the MT phenomenon
tunistic pathogens such as Pseudomonas aeruginosa and [21].
Candida albicans, were detected at significantly increased
numbers in the vast majority of HIV-infected patients. HIV and the intestinal mechanical barrier
Most importantly, this impairment of the normal bowel
flora was present even from the early stages of HIV Histologically, HIV infection, especially in its chronic
infection and was associated with elevated levels of phase, induces morphologic changes in the intestinal
intestinal inflammatory parameters detected in stools, such mucosa on routine light microscopy mainly represented by
as fecal calprotectin, indicating a possible correlation of villous atrophy [22, 23]. In addition, there is currently
disrupted gut microecology with intestinal inflammation, evidence of important cellular (increased epithelial apop-
mucosal damage and mucosal immune activation [11, 13]. tosis) and paracellular (altered tight junction expression)
alterations that might contribute to the barrier defect of the
HIV and the intestinal immune barrier intestinal epithelium associated with HIV infection [7, 22].
Increased epithelial apoptosis seems to be an early
The intestine which is the largest single immunological intestinal alteration in HIV infection as it has been detected
organ of the body consists one of the initial targets of HIV, in patients with acute HIV infection [22]. Increased
resulting in the disruption of the intestinal immune barrier expression of genes related to immune activation and

123
HIV and intestinal barrier 953

decreased expression of genes related to epithelial repair phenomenon of pathological MT in HIV infection has been
have been associated, as a contributing underlying mech- additionally confirmed by the use of diverse other markers
anism, with increased intestinal apoptosis through a cas- of MT, such as the plasma levels of soluble CD14 (sCD14),
pase-3 dependent pathway [24]. A variety of endotoxin core antibodies measured by the EndoCAb assay
immunological triggers, such as increased infiltration of and bacterial 16s rDNA, all of which were found elevated in
intestinal mucosa by perforin-expressing cytotoxic T lym- HIV-infected patients [35–37]. The extent of damage to the
phocytes in acute HIV infection and their loss in later epithelial barrier and the magnitude of MT, measured as the
stages, increased production of proinflammatory cytokines amount of LPS within the underlying mucosa or the extent
and activation of Fas on enterocytes and Fas-ligand on of translocated microbial products in draining and remote
lamina propria lymphocytes, have been associated with lymph node tissues, are positively correlated [34, 38].
increased intestinal epithelial apoptosis in HIV infection HIV infection is characterized by persistent immune
[24–27]. A recent study demonstrated also that the HIV-1 activation even in phases of viral load decline such as after
transactivator factor (Tat) induces enterocyte apoptosis the acute phase of infection or during administration of
through an oxidative stress-mediated mechanism [28]. suppressive anti-retroviral therapy [39, 40]. Persistent
Regarding the intestinal paracellular barrier, the first immune activation is evidenced by the presence of activated
observations on the detrimental effects of HIV infection on phenotypes of macrophages, cytokinemia (TNF-a, IL-6),
enterocytes’ TJs come from in vitro experiments. Specifi- increased acute phase proteins (C- reactive protein) and
cally, in colonic epithelial cell culture models, incubation increased elements of the coagulation cascade (d-dimmers,
with the HIV-1 or its envelope glycoprotein (gp) 120 resulted tissue factor). Regarding the adaptive immune system,
in decreased transepithelial electrical resistance associated previous studies have demonstrated increased turnover and
with downregulation of the key TJ molecular components exhaustion of T cells, low thymic output, increased turnover
occludin and claudins -1, -2 and -4 [29, 30]. A potential of B cells and hyperimmunoglobulinemia [41–43].
mechanism might be the increased production of TNF-alpha, The phenomena of pathological MT and persistent
IL-6, IL-8 and other inflammatory cytokines from epithelial immune activation in HIV infection are closely interrelated
cells when exposed to the HIV envelope glycoprotein, which according to the results of previous studies. Specifically, it
subsequently exert a detrimental effect on TJs’ structure and has been demonstrated that a significant correlation exists
function [29, 31]. In addition, the T84 enteric cell line when between the magnitude of endotoxemia, used as a marker
exposed to HIV-1 displayed disruption of the ZO-1, which is of MT, and diverse indices of innate and adaptive immu-
a macromolecule located underneath the TJs in the ‘‘tight nity, such as plasma IFN-a and T cell activation levels [33,
junctional plaque’’ serving a key role in bringing several 35–37, 44]. There are two main theories of how MT can
components of the tight junctional plaque with TJs and the lead to immune activation. The first one supports the view
cytoskeleton [29]. It is hypothesized that gp120, after bind- that after the disruption of the intestinal barrier integrity,
ing to the HIV co-receptor Bob/GPR15, which is abundant at intestinal bacteria and endotoxins translocate in mesenteric
the basal surface of small intestinal epithelium, induces lymph nodes as well as in portal circulation, thereafter
microtubule loss via a calcium-dependent signaling pathway entering into the systemic circulation and thus invading
[32]. The causative interrelation of HIV with intestinal epi- normally sterile extraintestinal tissues [45]. This process
thelial cells TJs’ disruption was proven by restoration of the causes systemic infections and promotes a systemic
TJs structure and function with administration of gp120 inflammatory response syndrome. The second theory sup-
neutralizing antibodies [29]. ports that, after the disruption of gut barrier integrity,
bacteria and endotoxins cross the mucosal barrier and
activate an intestinal inflammatory response, even when
Intestinal barrier dysfunction, immune activation these translocating factors are trapped within the gut wall
and clinical implications for HIV infection or intestinal lymph nodes and do not reach the systemic
circulation [46]. According to later view, the gut becomes a
Pathogenetic link of intestinal barrier dysfunction proinflammatory organ and gut-derived inflammatory fac-
and immune activation tors, carried mainly in the mesenteric lymph, induce a
systemic inflammatory response with detrimental effects in
A number of previous studies have demonstrated that HIV multiple organs’ structure and function [47].
infection is associated with pathological MT due to intes-
tinal barrier integrity disruption. Specifically, it has been Clinical implications
shown that HIV-infected patients present significantly
increased levels of circulating lipopolysaccharide (LPS) In HIV infection, the continuous immune activation driven
compared to uninfected controls [3, 33, 34]. The by the phenomenon of pathological MT exerts a pivotal

123
954 S. F. Assimakopoulos et al.

pathophysiological role in disease progression [48]. In activation [7, 49]. In addition, MT and its associated
particular, MT promotes the proliferation of activated T chronic immune activation in HIV infection seem to exert
cells which in turn provide new targets for HIV infection, an important role in diverse non-AIDS comorbidities.
leading to further impairment of the immune function HIV infection is a major cause of acceleration of hep-
through T and B cell dysfunction with subsequent poor atitis C virus-related liver disease, cirrhosis and death [50,
control of pathogens and further priming of immune 51]. The most widely proposed mechanisms implicated in

123
HIV and intestinal barrier 955

b Fig. 1 Overview of intestinal barrier dysfunction in HIV infection Hodgkin’s lymphoma [58, 59]. Among contributing
and interconnection with chronic immune activation: HIV infection mechanisms, chronic immune activation seems to exert an
induces a multifactorial disruption of intestinal barrier integrity by
negatively affecting all of its major levels of defense. a Gut important role. It has been previously shown that markers
microecology is altered allowing overgrowth of pathogenic bacteria of immune activation, such as IL-6, IL-10, CRP, sCD23,
with consequent increase of luminal endotoxin load. b The intestinal sCD27 and sCD30, are elevated several years preceding the
immune barrier is significantly impaired through depletion of CD4(?) diagnosis of AIDS-associated non-Hodgkin lymphoma
T cells, especially Th17 cells. Immune dysregulation is also
evidenced by increased CD4? FoxP3? Tregs and increased infiltra- [60]. Considering the pivotal pathophysiological role of
tion of lamina propria by mucosal macrophages, which however MT in chronic immune activation, a recent study demon-
present decreased phagocytic capacity. c The integrity of the strated that elevated serum markers of MT, such as LPS
intestinal mechanical barrier, which comprises the intestinal epithelial and sCD14, are significantly associated with increased risk
cells and their close interconnections (tight junctions––TJs), is
disrupted; HIV alters the expression of enterocytes’ TJs thus opening for non-Hodgkin lymphoma development, supporting an
the paracellular route and additionally promotes the apoptotic death of etiologic role for MT in lymphomagenesis among HIV-
intestinal epithelial cells. Pathogenic microbes and LPS from the infected individuals [61].
intestinal lumen pass through the disrupted mechanical barrier to the Chronic immune activation seems to be a key mecha-
lamina propria, where they cannot be cleared by the dysfunctional
immune system. Intestinal macrophages are activated to produce nism in the pathogenesis of HIV-associated neurocognitive
proinflammatory cytokines and reactive oxygen species. This inflam- disorders as well [62]. Activated monocytes can traffic the
matory and oxidative environment further promotes apoptotic HIV virus into the central nervous system and then infect/
epithelial cell death and disrupts the structural and functional activate other macrophages or other cell types [62].
integrity of enterocytes’ TJs, while further aggravating the defects
of the local immunological defense by altering the intestinal immune Infected macrophages represent the major cell type sup-
cell balance through inhibition of Th17 cells development and porting productive HIV infection in the brain. Also,
promotion of Treg differentiation. More microbes and LPS can now infected or activated macrophages can release numerous
escape the intestinal lumen and gain access to the systemic neurotoxic factors which have been implicated in the
circulation, where they induce a systemic inflammatory response
associated with HIV clinical progression, suboptimal CD4(?)T cell pathogenesis of HIV-associated neurocognitive impairment
recovery on ART and with development of non-AIDS comorbidities such as HIV proteins, cytokines and chemokines [63]. The
from diverse systems gut-derived bacterial products (LPS) can trigger monocyte
activation via CD14 and TLR4-mediated signaling [64]. A
recent study demonstrated that plasma LPS and sCD14
the acceleration of liver injury in HIV/HCV co-infection levels predict the development of HIV-associated dementia
are increased liver expression of profibrogenic cytokines, independently of viral load, suggesting the important role
increased hepatic oxidative stress and hepatocellular of MT in the pathogenesis of HIV-associated dementia
apoptosis. Pathological MT and immune activation in the [65].
setting of HIV infection seem to exert a pivotal role in the
induction of the above-stated alterations [52–54]. Gut-
derived translocating bacteria and/or their products might Interventions for prevention of HIV-associated
promote liver fibrosis either by directly interacting with intestinal barrier dysfunction and MT
Kupffer cells and hepatic stellate cells or indirectly through
induction of systemic immune activation and promotion of HAART
hepatocellular apoptosis [52–55].
Also, according to previous clinical studies, MT and its Numerous previous studies have shown that HAART,
associated persistent immune activation and proinflamma- despite the effective clearance of plasma HIV-1 RNA to
tory state have been associated with dyslipidemia, insulin undetectable levels, only partly decreases plasma levels of
resistance and cardiovascular complications in HIV-infec- diverse markers of MT and immune activation [3, 23, 66–
ted patients [56]. Another very recent study examined 68]. According to previous studies, the potential beneficial
plasma LPS and sCD14 levels in untreated and treated HIV effect of HAART on the intestinal barrier function might
patients and correlated these with metabolic indices [57]. In be based on restoration of enterocyte TJ proteins’ expres-
the whole group of patients, plasma LPS levels were neg- sion, thus increasing the transepithelial electrical resistance
atively correlated with large particle HDL levels and posi- of the intestinal mucosa, and on intestinal mucosal CD4?
tively correlated with triglyceride levels, while sCD14 was T cell reconstitution, especially when HAART is instituted
negatively correlated with HDL cholesterol levels. These early [23, 67]. With regard to other parameters of intestinal
findings imply that MT possibly contributes to the increased mucosal immunity such as CD8? function, NK cell
cardiovascular disease risk observed during HIV infection. reconstitution, immunoglobulin production and lymphatic
Patients with HIV infection are at markedly increased tissue architecture, overall HAART seems to support par-
risk for the development of multiple subtypes of non- tial mucosal immune recovery [67].

123
956 S. F. Assimakopoulos et al.

Beyond HAART interventions Interventions aiming at the translocating microbial and/


or endotoxin burden pre- or post-translocation
Beyond HAART, a number of other preventive treatment
strategies have a reasonable basis for use in HIV infection i. Elimination of the gut luminal bacteria and/or their
aiming at elimination of MT, either by strengthening the bioproducts before their translocation.
above-described diverse levels of gut barrier integrity or by
Elimination of the intestinal bacterial and/or endotoxin load
eliminating the burden of translocated bioproducts by act-
could be achieved by oral administration of non-absorbed
ing before or after their escape from the gut lumen.
antibiotics such as rifaximin [80]. However, the need for
long-term intervention in HIV infection carries the risk for
Interventions aiming at enhancing the intestinal barrier
antibiotic-resistant bacteria selection. Another option,
integrity
simple in its conception and relatively safe, is elimination of
luminal endotoxin load by oral lactulose administration.
i. Restoration of the intestinal biological barrier.
Lactulose exerts endotoxin-binding capacity, thus neutral-
Maintenance of the gut microecology could exert beneficial izing before translocation this important pathophysiological
effects on preservation of enterocyte homeostasis and pre- trigger for chronic immune activation in HIV infection. A
vention of mucosal immune activation. A number of clini- disadvantage of this second option is the diarrheal effect of
cal studies have shown that supplementation with probiotics lactulose that could limit its clinical application, especially
prevents intestinal hyperpermeability in diverse pathologi- in HIV-infected patients with associated chronic diarrhea.
cal conditions [69–72]. In HIV-1 infection probiotics were
ii. Clearance of the systemically translocated bacterial
shown to exert beneficial effects in children who become
bioproducts.
infected before the development of a normal gut flora and
are at risk for chronic immune activation and growth The bactericidal/permeability-increasing protein is a
abnormalities, while HIV-1-infected adults consuming naturally occurring endotoxin-binding protein produced in
probiotic-enriched yogurt presented a significant increase of neutrophils, eosinophils and some epithelial cells, which
CD4 cells compared to patients not consuming yogurt [73]. binds endotoxin, neutralizing the activity and inhibiting
cytokine production by mononuclear cells. Animal studies
ii. Restoration of the intestinal immune barrier.
with models of systemic endotoxemia have shown that this
Disruption of T cell homeostasis in the intestinal compound reduces the endotoxin-induced mortality to a
immune system is a cardinal feature of HIV infection. death rate comparable to non-endotoxemic experimental
Given that IL-7 is an essential cytokine for T cell devel- animals. The rationale for its potential use in HIV-infected
opment and homeostasis, a recent clinical study examined patients is further strengthened by its demonstrated defi-
the effect of recombinant IL-7 administration on the gut- ciency in this patient population [81].
residing naı̈ve and memory CD4(?) T cells in HIV-
infected patients on HAART with inadequate CD4? T cell
recovery with promising results [74]. Conclusions
iii. Restoration of the intestinal mechanical barrier.
HIV infection disrupts the integrity of the intestinal bio-
Considerable experimental and clinical evidence sug- logical, immune and mechanical barrier, promoting the
gests that enteral immunonutrition using glutamine, argi- phenomenon of microbial translocation which is a crucial
nine and omega-3 fatty acids exerts beneficial effects on determinant of chronic immune activation (Fig. 1). To our
preservation of gut barrier integrity against diverse injuri- opinion, we have reached a point where the effective
ous insult [75, 76]. The enhancement of intestinal perme- control of HIV viremia by HAART should be combined
ability by glutamine may be explained by its antiapoptotic with emerging pharmacological approaches aiming at the
effect on the intestinal mucosa [77]. A recent study dem- restoration of the intestinal barrier targeting its diverse
onstrated that nutritional supplementation with glutamine levels of structure and function. Elimination of the MT
in HIV-infected patients improved intestinal barrier func- phenomenon would mitigate its effect on immune
tion [78]. In addition, considering the fact that oxidative homeostasis, which might improve the prognosis of the
stress has been implicated in intestinal injury in HIV HIV-infected patient in terms of morbidity and mortality.
infection, previous studies demonstrated that administra-
tion of the antioxidants N-acetyl-cysteine or monosodium
luminol prevent intestinal barrier disruption in HIV infec- Conflict of interest On behalf of all authors, the corresponding
tion [28, 79]. author states that there is no conflict of interest.

123
HIV and intestinal barrier 957

References 17. Kim CJ, McKinnon LR, Kovacs C, Kandel G, Huibner S, Chege
D, Shahabi K, Benko E, Loutfy M, Ostrowski M, Kaul R.
1. Miedema F, Hazenberg MD, Tesselaar K, van Baarle D, de Boer Mucosal Th17 cell function is altered during HIV infection and is
RJ, Borghans JA. Immune activation and collateral damage in an independent predictor of systemic immune activation.
AIDS pathogenesis. Front Immunol. 2013;4:298. J Immunol. 2013;191:2164–73.
2. Ipp H, Zemlin AE, Erasmus RT, Glashoff RH. Role of inflam- 18. Bixler SL, Mattapallil JJ. Loss and dysregulation of Th17 cells
mation in HIV-1 disease progression and prognosis. Crit Rev Clin during HIV infection. Clin Dev Immunol. 2013;2013:852418.
Lab Sci. 2014;51:98–111. 19. Dandekar S, George MD, Baumler AJ. Th17 cells, HIV and the
3. Brenchley JM, Price DA, Schacker TW, Asher TE, Silvestri G, gut mucosal barrier. Curr Opin HIV AIDS. 2010;5:173–8.
Rao S, Kazzaz Z, Bornstein E, Lambotte O, Altmann D, Blazar 20. Kanwar B, Favre D, McCune JM. Th17 and regulatory T cells:
BR, Rodriguez B, Teixeira-Johnson L, Landay A, Martin JN, implications for AIDS pathogenesis. Curr Opin HIV AIDS.
Hecht FM, Picker LJ, Lederman MM, Deeks SG, Douek DC. 2010;5:151–7.
Microbial translocation is a cause of systemic immune activation 21. Allers K, Fehr M, Conrad K, Epple HJ, Schurmann D, Geelhaar-
in chronic HIV infection. Nat Med. 2006;12:1365–71. Karsch A, Schinnerling K, Moos V, Schneider T. Macrophages
4. Funderburg N, Luciano AA, Jiang W, Rodriguez B, Sieg SF, accumulate in the gut mucosa of untreated HIV-infected patients.
Lederman MM. Toll-like receptor ligands induce human T cell J Infect Dis. 2014;209:739–48.
activation and death, a model for HIV pathogenesis. PLoS ONE. 22. Epple HJ, Allers K, Troger H, Kuhl A, Erben U, Fromm M, Zeitz
2008;3:e1915. M, Loddenkemper C, Schulzke JD, Schneider T. Acute HIV
5. Pilakka-Kanthikeel S, Arheart K, Selvaraj A, Swaminathan S, infection induces mucosal infiltration with CD4? and CD8? T
Pahwa S. Immune activation is associated with increased gut cells, epithelial apoptosis, and a mucosal barrier defect. Gastro-
microbial translocation in treatment naive HIV infected children enterology. 2010;139:1289–300.
in a resource limited setting. J Acquir Immune Defic Syndr. 23. Epple HJ, Schneider T, Troeger H, Kunkel D, Allers K, Moos V,
2014;66:16–24. Amasheh M, Loddenkemper C, Fromm M, Zeitz M, Schulzke JD.
6. Tincati C, Bellistri GM, Ancona G, Merlini E, d’Arminio Mon- Impairment of the intestinal barrier is evident in untreated but
forte A, Marchetti G. Role of in vitro stimulation with lipopoly- absent in suppressively treated HIV-infected patients. Gut.
saccharide on T-cell activation in HIV-infected antiretroviral- 2009;58:220–7.
treated patients. Clin Dev Immunol. 2012;2012:935425. 24. Sankaran S, George MD, Reay E, Guadalupe M, Flamm J,
7. Marchetti G, Tincati C, Silvestri G. Microbial translocation in the Prindiville T, Dandekar S. Rapid onset of intestinal epithelial
pathogenesis of HIV infection and AIDS. Clin Microbiol Rev. barrier dysfunction in primary human immunodeficiency virus
2013;26:2–18. infection is driven by an imbalance between immune response
8. Assimakopoulos SF, Scopa CD, Vagianos CE. Pathophysiology and mucosal repair and regeneration. J Virol. 2008;82:538–45.
of increased intestinal permeability in obstructive jaundice. 25. Du Plessis J, Vanheel H, Janssen CE, Roos L, Slavik T, Stivaktas
World J Gastroenterol. 2007;13:6458–64. PI, Nieuwoudt M, van Wyk SG, Vieira W, Pretorius E, Beukes
9. Assimakopoulos SF, Papageorgiou I, Charonis A. Enterocytes’ M, Farre R, Tack J, Laleman W, Fevery J, Nevens F, Roskams T,
tight junctions: from molecules to diseases. World J Gastrointest Van der Merwe SW. Activated intestinal macrophages in patients
Pathophysiol. 2011;2:123–37. with cirrhosis release NO and IL-6 that may disrupt intestinal
10. Paiardini M, Frank I, Pandrea I, Apetrei C, Silvestri G. Mucosal barrier function. J Hepatol. 2013;58:1125–32.
immune dysfunction in AIDS pathogenesis. AIDS Rev. 26. Epple HJ, Zeitz M. HIV infection and the intestinal mucosal
2008;10:36–46. barrier. Ann N Y Acad Sci. 2012;1258:19–24.
11. Gori A, Tincati C, Rizzardini G, Torti C, Quirino T, Haarman M, 27. Sankaran S, Guadalupe M, Reay E, George MD, Flamm J,
Ben Amor K, Van Schaik J, Vriesema A, Knol J, Marchetti G, Prindiville T, Dandekar S. Gut mucosal T cell responses and gene
Welling G, Clerici M. Early impairment of gut function and gut expression correlate with protection against disease in long-term
flora supporting a role for alteration of gastrointestinal mucosa in HIV-1-infected nonprogressors. Proc Natl Acad Sci USA.
human immunodeficiency virus pathogenesis. J Clin Microbiol. 2005;102:9860–5.
2008;46:757–8. 28. Buccigrossi V, Laudiero G, Nicastro E, Miele E, Esposito F,
12. McKenna P, Hoffmann C, Minkah N, Aye PP, Lackner A, Liu Z, Guarino A. The HIV-1 transactivator factor (Tat) induces en-
Lozupone CA, Hamady M, Knight R, Bushman FD. The maca- terocyte apoptosis through a redox-mediated mechanism. PLoS
que gut microbiome in health, lentiviral infection, and chronic ONE. 2011;6:e29436.
enterocolitis. PLoS Pathog. 2008;4:e20. 29. Nazli A, Chan O, Dobson-Belaire WN, Ouellet M, Tremblay MJ,
13. Ton H, Brandsnes, Dale S, Holtlund J, Skuibina E, Schjonsby H, Gray-Owen SD, Arsenault AL, Kaushic C. Exposure to HIV-1
Johne B. Improved assay for fecal calprotectin. Clin Chim Acta. directly impairs mucosal epithelial barrier integrity allowing
2000;292:41–54. microbial translocation. PLoS Pathog. 2010;6:e1000852.
14. Mattapallil JJ, Douek DC, Hill B, Nishimura Y, Martin M, Roederer 30. Schmitz H, Rokos K, Florian P, Gitter AH, Fromm M, Scholz P,
M. Massive infection and loss of memory CD4? T cells in multiple Ullrich R, Zeitz M, Pauli G, Schulzke JD. Supernatants of HIV-
tissues during acute SIV infection. Nature. 2005;434:1093–7. infected immune cells affect the barrier function of human HT-
15. Mehandru S, Poles MA, Tenner-Racz K, Horowitz A, Hurley A, 29/B6 intestinal epithelial cells. Aids. 2002;16:983–91.
Hogan C, Boden D, Racz P, Markowitz M. Primary HIV-1 31. Canani RB, Cirillo P, Mallardo G, Buccigrossi V, Secondo A,
infection is associated with preferential depletion of CD4? T Annunziato L, Bruzzese E, Albano F, Selvaggi F, Guarino A.
lymphocytes from effector sites in the gastrointestinal tract. J Exp Effects of HIV-1 Tat protein on ion secretion and on cell pro-
Med. 2004;200:761–70. liferation in human intestinal epithelial cells. Gastroenterology.
16. Gordon SN, Cervasi B, Odorizzi P, Silverman R, Aberra F, 2003;124:368–76.
Ginsberg G, Estes JD, Paiardini M, Frank I, Silvestri G. Dis- 32. Clayton F, Kotler DP, Kuwada SK, Morgan T, Stepan C, Kuang
ruption of intestinal CD4? T cell homeostasis is a key marker of J, Le J, Fantini J. Gp120-induced Bob/GPR15 activation: a pos-
systemic CD4? T cell activation in HIV-infected individuals. sible cause of human immunodeficiency virus enteropathy. Am J
J Immunol. 2010;185:5169–79. Pathol. 2001;159:1933–9.

123
958 S. F. Assimakopoulos et al.

33. Papasavvas E, Pistilli M, Reynolds G, Bucki R, Azzoni L, 50. Hsu DC, Sereti I, Ananworanich J. Serious Non-AIDS events:
Chehimi J, Janmey PA, DiNubile MJ, Ondercin J, Kostman JR, immunopathogenesis and interventional strategies. AIDS Res
Mounzer KC, Montaner LJ. Delayed loss of control of plasma Ther. 2013;10:29.
lipopolysaccharide levels after therapy interruption in chronically 51. Sagnelli C, Uberti-Foppa C, Pasquale G, De Pascalis S, Coppola
HIV-1-infected patients. AIDS. 2009;23:369–75. N, Albarello L, Doglioni C, Lazzarin A, Sagnelli E. Factors
34. Vassallo M, Mercie P, Cottalorda J, Ticchioni M, Dellamonica P. influencing liver fibrosis and necroinflammation in HIV/HCV
The role of lipopolysaccharide as a marker of immune activation coinfection and HCV monoinfection. Infection. 2013;41:959–67.
in HIV-1 infected patients: a systematic literature review. Virol J. 52. Bica I, McGovern B, Dhar R, Stone D, McGowan K, Scheib R,
2012;9:174. Snydman DR. Increasing mortality due to end-stage liver disease
35. Sandler NG, Wand H, Roque A, Law M, Nason MC, Nixon DE, in patients with human immunodeficiency virus infection. Clin
Pedersen C, Ruxrungtham K, Lewin SR, Emery S, Neaton JD, Infect Dis. 2001;32:492–7.
Brenchley JM, Deeks SG, Sereti I, Douek DC. Plasma levels of 53. Graham CS, Baden LR, Yu E, Mrus JM, Carnie J, Heeren T,
soluble CD14 independently predict mortality in HIV infection. Koziel MJ. Influence of human immunodeficiency virus infection
J Infect Dis. 2011;203:780–90. on the course of hepatitis C virus infection: a meta-analysis. Clin
36. Bukh AR, Melchjorsen J, Offersen R, Jensen JM, Toft L, Stovring Infect Dis. 2001;33:562–9.
H, Ostergaard L, Tolstrup M, Sogaard OS. Endotoxemia is asso- 54. Rosenthal E, Salmon-Ceron D, Lewden C, Bouteloup V, Pialoux
ciated with altered innate and adaptive immune responses in G, Bonnet F, Karmochkine M, May T, Francois M, Burty C,
untreated HIV-1 infected individuals. PLoS ONE. 2011;6:e21275. Jougla E, Costagliola D, Morlat P, Chene G, Cacoub P. Liver-
37. Jiang W, Lederman MM, Hunt P, Sieg SF, Haley K, Rodriguez B, related deaths in HIV-infected patients between 1995 and 2005 in
Landay A, Martin J, Sinclair E, Asher AI, Deeks SG, Douek DC, the French GERMIVIC Joint Study Group Network (Mortavic
Brenchley JM. Plasma levels of bacterial DNA correlate with 2005 study in collaboration with the Mortalite 2005 survey,
immune activation and the magnitude of immune restoration in ANRS EN19). HIV Med. 2009;10:282–9.
persons with antiretroviral-treated HIV infection. J Infect Dis. 55. Puoti M, Prestini K, Putzolu V, Zanini B, Baiguera C, Antonini
2009;199:1177–85. MG, Pagani P, Airoldi M, Carosi G. HIV/HCV co-infection:
38. Hofer U, Schlaepfer E, Baenziger S, Nischang M, Regenass S, natural history. J Biol Regul Homeost Agent. 2003;17:144–6.
Schwendener R, Kempf W, Nadal D, Speck RF. Inadequate 56. Pedersen KK, Pedersen M, Troseid M, Gaardbo JC, Lund TT,
clearance of translocated bacterial products in HIV-infected Thomsen C, Gerstoft J, Kvale D, Nielsen SD. Microbial trans-
humanized mice. PLoS Pathog. 2010;6:e1000867. location in HIV infection is associated with dyslipidemia, insulin
39. Deeks SG, Kitchen CM, Liu L, Guo H, Gascon R, Narvaez AB, resistance, and risk of myocardial infarction. J Acquir Immune
Hunt P, Martin JN, Kahn JO, Levy J, McGrath MS, Hecht FM. Defic Syndr. 2013;64:425–33.
Immune activation set point during early HIV infection predicts 57. Timmons T, Shen C, Aldrovandi G, Rollie A, Gupta SK, Stein
subsequent CD4? T-cell changes independent of viral load. JH, Dube MP. Microbial translocation and metabolic and body
Blood. 2004;104:942–7. composition measures in treated and untreated HIV infection.
40. Brenchley JM, Douek DC. The mucosal barrier and immune acti- AIDS Res Hum Retroviruses. 2014;30:272–7.
vation in HIV pathogenesis. Curr Opin HIV AIDS. 2008;3:356–61. 58. Epeldegui M, Widney DP, Martinez-Maza O. Pathogenesis of
41. Lane HC, Masur H, Edgar LC, Whalen G, Rook AH, Fauci AS. AIDS lymphoma: role of oncogenic viruses and B cell activation-
Abnormalities of B-cell activation and immunoregulation in associated molecular lesions. Curr Opin Oncol. 2006;18:444–8.
patients with the acquired immunodeficiency syndrome. N Engl J 59. Ehren K, Hertenstein C, Kummerle T, Vehreschild JJ, Fischer J,
Med. 1983;309:453–8. Gillor D, Wyen C, Lehmann C, Cornely OA, Jung N, Gravemann
42. Hellerstein M, Hanley MB, Cesar D, Siler S, Papageorgopoulos S, Platten M, Wasmuth JC, Rockstroh JK, Boesecke C,
C, Wieder E, Schmidt D, Hoh R, Neese R, Macallan D, Deeks S, Schwarze-Zander C, Fatkenheuer G. Causes of death in HIV-
McCune JM. Directly measured kinetics of circulating T lym- infected patients from the Cologne–Bonn cohort. Infection.
phocytes in normal and HIV-1-infected humans. Nat Med. 2014;42:135–40.
1999;5:83–9. 60. Breen EC, Hussain SK, Magpantay L, Jacobson LP, Detels R,
43. Valdez H, Lederman MM. Cytokines and cytokine therapies in Rabkin CS, Kaslow RA, Variakojis D, Bream JH, Rinaldo CR,
HIV infection. AIDS Clin Rev 1997-1998;187–228. Ambinder RF, Martinez-Maza O. B-cell stimulatory cytokines and
44. Nixon DE, Landay AL. Biomarkers of immune dysfunction in markers of immune activation are elevated several years prior to the
HIV. Curr Opin HIV AIDS. 2010;5:498–503. diagnosis of systemic AIDS-associated non-Hodgkin B-cell lym-
45. Deitch EA. The role of intestinal barrier failure and bacterial phoma. Cancer Epidemiol Biomarkers Prev. 2011;20:1303–14.
translocation in the development of systemic infection and mul- 61. Marks MA, Rabkin CS, Engels EA, Busch E, Kopp W, Rager H,
tiple organ failure. Arch Surg. 1990;125:403–4. Goedert JJ, Chaturvedi AK. Markers of microbial translocation
46. Senthil M, Brown M, Xu DZ, Lu Q, Feketeova E, Deitch EA. and risk of AIDS-related lymphoma. AIDS. 2013;27:469–74.
Gut-lymph hypothesis of systemic inflammatory response syn- 62. Zhou L, Saksena NK. HIV associated neurocognitive disorders.
drome/multiple-organ dysfunction syndrome: validating studies Infect Dis Rep. 2013;5:e8.
in a porcine model. J Trauma. 2006;60:958–65 (discussion 65–7). 63. Genis P, Jett M, Bernton EW, Boyle T, Gelbard HA, Dzenko K,
47. Deitch EA. Gut lymph and lymphatics: a source of factors leading Keane RW, Resnick L, Mizrachi Y, Volsky DJ, et al. Cytokines
to organ injury and dysfunction. Ann N Y Acad Sci. and arachidonic metabolites produced during human immuno-
2010;1207:E103–11. deficiency virus (HIV)-infected macrophage–astroglia interac-
48. Douek DC, Roederer M, Koup RA. Emerging concepts in the tions: implications for the neuropathogenesis of HIV disease.
immunopathogenesis of AIDS. Annu Rev Med. 2009;60:471–84. J Exp Med. 1992;176:1703–18.
49. Hunt PW, Brenchley J, Sinclair E, McCune JM, Roland M, Page- 64. Kim D, Kim JY. Anti-CD14 antibody reduces LPS responsive-
Shafer K, Hsue P, Emu B, Krone M, Lampiris H, Douek D, ness via TLR4 internalization in human monocytes. Mol Immu-
Martin JN, Deeks SG. Relationship between T cell activation and nol. 2014;57:210–5.
CD4? T cell count in HIV-seropositive individuals with unde- 65. Ancuta P, Kamat A, Kunstman KJ, Kim EY, Autissier P, Wurcel
tectable plasma HIV RNA levels in the absence of therapy. A, Zaman T, Stone D, Mefford M, Morgello S, Singer EJ, Wo-
J Infect Dis. 2008;197:126–33. linsky SM, Gabuzda D. Microbial translocation is associated with

123
HIV and intestinal barrier 959

increased monocyte activation and dementia in AIDS patients. lymphocyte restoration in chronically HIV (?) patients treated
PLoS ONE. 2008;3:e2516. with recombinant interleukin-7. In: 19th Conf Retroviruses
66. Marchetti G, Bellistri GM, Borghi E, Tincati C, Ferramosca S, La Opportun Infect, Seattle, WA.
Francesca M, Morace G, Gori A, Monforte AD. Microbial 75. Sung MK, Park MY. Nutritional modulators of ulcerative colitis:
translocation is associated with sustained failure in CD4? T-cell clinical efficacies and mechanistic view. World J Gastroenterol.
reconstitution in HIV-infected patients on long-term highly active 2013;19:994–1004.
antiretroviral therapy. AIDS. 2008;22:2035–8. 76. Zuhl MN, Lanphere KR, Kravitz L, Mermier CM, Schneider S,
67. Costiniuk CT, Angel JB. Human immunodeficiency virus and the Dokladny K, Moseley PL. Effects of oral glutamine supplemen-
gastrointestinal immune system: does highly active antiretroviral tation on exercise-induced gastrointestinal permeability and tight
therapy restore gut immunity? Mucosal Immunol. 2012;5: junction protein expression. J Appl Physiol. 1985;2014(116):
596–604. 183–91.
68. Cassol E, Malfeld S, Mahasha P, van der Merwe S, Cassol S, 77. Margaritis VG, Filos KS, Michalaki MA, Scopa CD, Spiliopou-
Seebregts C, Alfano M, Poli G, Rossouw T. Persistent microbial lou I, Nikolopoulou VN, Vagianos CE. Effect of oral glutamine
translocation and immune activation in HIV-1-infected South administration on bacterial translocation, endotoxemia, liver and
Africans receiving combination antiretroviral therapy. J Infect ileal morphology, and apoptosis in rats with obstructive jaundice.
Dis. 2010;202:723–33. World J Surg. 2005;29:1329–34.
69. Sindhu KN, Sowmyanarayanan TV, Paul A, Babji S, Ajjampur 78. Leite RD, Lima NL, Leite CA, Farhat CK, Guerrant RL, Lima
SS, Priyadarshini S, Sarkar R, Balasubramanian KA, Wanke CA, AA. Improvement of intestinal permeability with alanyl-gluta-
Ward HD, Kang G. Immune response and intestinal permeability mine in HIV patients: a randomized, double blinded, placebo-
in children with acute gastroenteritis treated with Lactobacillus controlled clinical trial. Arq Gastroenterol. 2013;50:56–63.
rhamnosus GG: a randomized, double-blind, placebo-controlled 79. Scofield VL, Yan M, Kuang X, Kim SJ, Wong PK. The drug
trial. Clin Infect Dis. 2014;58:1107–15. monosodium luminol (GVT) preserves crypt-villus epithelial
70. Yeoh N, Burton JP, Suppiah P, Reid G, Stebbings S. The role of organization and allows survival of intestinal T cells in mice
the microbiome in rheumatic diseases. Curr Rheumatol Rep. infected with the ts1 retrovirus. Immunol Lett. 2009;122:150–8.
2013;15:314. 80. Bajaj JS, Heuman DM, Sanyal AJ, Hylemon PB, Sterling RK,
71. Ohland CL, Macnaughton WK. Probiotic bacteria and intestinal Stravitz RT, Fuchs M, Ridlon JM, Daita K, Monteith P, Noble
epithelial barrier function. Am J Physiol Gastrointest Liver NA, White MB, Fisher A, Sikaroodi M, Rangwala H, Gillevet
Physiol. 2010;298:G807–19. PM. Modulation of the metabiome by rifaximin in patients with
72. Girardin M, Seidman EG. Indications for the use of probiotics in cirrhosis and minimal hepatic encephalopathy. PLoS ONE.
gastrointestinal diseases. Dig Dis. 2011;29:574–87. 2013;8:e60042.
73. Irvine SL, Hummelen R, Hekmat S, Looman CW, Habbema JD, 81. Palmer CD, Guinan EC, Levy O. Deficient expression of bac-
Reid G. Probiotic yogurt consumption is associated with an tericidal/permeability-increasing protein in immunocompromised
increase of CD4 count among people living with HIV/AIDS. hosts: translational potential of replacement therapy. Biochem
J Clin Gastroenterol. 2010;44:e201–5. Soc Trans. 2011;39:994–9.
74. Sereti I EJ, Thompson W, Fischl M, Croughs T, Beq S, Yao M,
Boulassel R, Lederman M, Routy JP (2012) Gut mucosa T

123

You might also like