You are on page 1of 7

Role of Human -defensins

in HIV Infection
like receptor 4 (Biragyn et al., 2002a). Since (1) the
A. Weinberg1*, M.E. Quiones-Mateu2, constitutively expressed hBD-1 and inducible hBD-2 and hBD-
M.M. Lederman3 3 are present in normal human oral epithelium and cells
1Department of Biological Sciences, School of Dental Medicine, Case (NHOECs) (Krisanaprakornkit et al., 1998, 2000; Weinberg et al.,
Western Reserve University (CWRU), 10900 Euclid Ave., Cleveland, OH 1998; Dunsche et al., 2002), (2) they are important mediators of
44106, USA; 2Lerner Research Institute, Cleveland Clinic Foundation, innate mucosal defense against microbial infection (Weinberg
Center for AIDS Research, CWRU; and 3 University Hospitals of et al., 1998), (3) these peptides may be involved in
Cleveland, OH; *corresponding author, aaron.weinberg@case.edu. immunomodulation of the adaptive immune system (Biragyn
Adv Dent Res 19:42-48, April, 2006 et al., 2002a,b), and (4) -defensins can impair adenoviral
infections (Gropp et al., 1999), we explored the possible role of
these molecules in defense against HIV infection. This review

M
echanisms of resistance to HIV-1 infection in the describes novel information related to the antiretroviral
human oral cavity are incompletely understood. activity of mucosal beta-defensins.
While salivary components have been implicated in
protection, there is growing evidence that human
beta-defensins (hBDs), originating in oral epithelial cells, may
Background
be playing an important role in the prevention of HIV HIV-1 and the oral cavity
infection. New antiviral, chemotactic, and immunosurveillance
properties are being attributed to hBDs, which are small At the outset of the AIDS epidemic, there was concern that
cationic antimicrobial innate response molecules expressed in HIV could be transmitted from the oral secretions of HIV
mucosal epithelium. Inducible hBDs are always expressed in carriers during kissing, dental treatment, biting, or through
normal oral epithelium, a property not shared by other aerosolization. It became clear, however, that oral transmission
mucosal barriers. Data reviewed in this paper demonstrate of HIV is actually rare (Rogers et al., 1990; Gooch et al., 1993;
that: (1) HIV-1 X4 and R5 phenotypes induce hBD-2 and -3 Moore et al., 1993). Interestingly, the frequency with which
mRNA in normal human oral epithelial cells; (2) hBD-2 and -3 infectious HIV can be found in the saliva of HIV-1-infected
inhibit HIV-1 infection by both viral strains, with greater patients is low, approximately 1%, although infectious virus
activity against X4 viruses; and (3) this inhibition is due to a can generally be isolated from the blood of untreated infected
direct interaction with virions and through modulation of the persons (Barr et al., 1992; Moore et al., 1993; Coppenhaver et al.,
CXCR4 co-receptor. These properties may be exploited as 1994). This finding is not due to a lack of virus in the oral
strategies for mucosal protection against HIV-1 transmission. cavity; HIV-1 RNA, proviral DNA, and infected cells are
detected in oropharyngeal tissues and salivary secretions of
infected persons (Goto et al., 1991; Baron et al., 1999). The
Introduction clinical importance of this dissociation is suggested by the
Epidemiological evidence suggests that exogenous infection difficulty with which HIV is transmitted through the oral
with HIV-1 through the oral cavity is quite rare, yet the mucosa (Herz et al., 2002). The question, therefore, is, why?
mechanisms for this protection are not understood. Moreover, This is particularly important, since > 90% of HIV cases
while persons chronically infected with HIV may be infected worldwide have been transmitted via mucosal surfaces,
with viruses that utilize CCR5 or CXCR4 co-receptors for primarily across genital mucosal surfaces and across lower
cellular entry, in acute infection, CCR5-tropic viruses almost gastrointestinal tract mucosa (Smith et al., 1999).
always predominate. The mechanisms underlying this Several explanations have been suggested to explain the
restriction are also not completely understood. While adaptive paucity of oral HIV-1 infection: (i) a thick multilayered mucosal
cellular immune responses are critical regulators of HIV surface (first line of defense against microbial invasion), (ii)
replication once infection has occurred, and there is some low salivary HIV-1 titers (Ho et al., 1985), and (iii) endogenous
evidence that persons at high risk for HIV infection who antiviral factors present in oral secretions (Fultz, 1986; Shugars,
remain uninfected may have low-level cellular responses to 1999). During the last 15 years, multiple studies have been
HIV-1, there is increasing interest in the importance of innate conducted to identify the source(s) and identity of HIV-
immune factors emanating from mucosal tissues and fluids. inhibitory activity in the saliva of healthy and infected
Recent findings in the field of innate immunity are individuals (Fultz, 1986; McNeely et al., 1995; Wahl et al., 1997;
shedding new light on the importance of host defense Baqui et al., 1999; Becquart et al., 1999; Shugars, 1999; Pillay et
antimicrobial peptides. With the recent discoveries of human al., 2001). Many endogenous inhibitors of HIV-1 in saliva have
-defensins (hBDs) in mucosal epithelium, a new line of been proposed (e.g., amylase, lactoferrin, proline-rich peptides,
investigation is emerging to test the hypothesis that these salivary mucins, thrombospondin; and secretory leukocyte
peptides, integral members of the innate host immune protease inhibitor [SLPI], reviewed in Shugars and Wahl, 1998).
response, function to protect the host against microbial
pathogenesis at the mucosal barrier. While their antibacterial
and antifungal properties are well-established (Weinberg et al., Key Words
1998; Zasloff, 2002), new findings point to hBDs acting like Human beta-defensin, epithelial cell, innate immunity, HIV.
chemokines in 'cross-talking' with the adaptive immune
Presented at the Fifth World Workshop on Oral Health and Disease
system, and possibly orchestrating immunosurveillance in AIDS, Phuket, Thailand, July 6-9, 2004, sponsored by Prince of
through maturation of dendritic cells. They recruit immature Songkla University, Thailand, the International Association for Dental
dendritic cells and T-cells by interacting with the chemokine Research, the World Health Organization, the NIDCR/National
receptor CCR6 (Yang et al., 1999) and, in mice, have recently Institutes of Health, USA, and the University of California-San
been shown to activate immature dendritic cells through Toll- Francisco Oral AIDS Center.

42
Moreover, these agents are found in seminal fluid and vaginal molecular characteristics, and purported modes of activity are
secretions, routinely harvested from sites that are very reviewed in Weinberg et al. (1998) and Zasloff (2002). The
susceptible to infection (Shugars, 1999). Importantly, there is a defensin peptides are a superfamily of peptide antibiotics with
paucity of information regarding the contributions of innate a characteristic -sheet structure stabilized by two or three
immune factors emanating from the oral mucosal epithelium intramolecular disulfide bonds. They are strongly cationic by
itself. virtue of their numerous arginine and lysine residues. Their
amphipathic and cationic characteristics are important for
Transmission of HIV-1 binding to anionic microbial surfaces, such as LPS, through
A significant genetic bottleneck is apparent during transmission displacement of divalent, lipopolysaccharide (LPS)-associated
by any route of HIV-1 infection. The selective factors imposing cell-surface cations, followed by membrane insertion through
the bottleneck are diverse and likely include: (i) host factors the 'self-promoted uptake' pathway (Hancock, 1997) and
such as innate immune response, (ii) density of target cells generation of stable pores (for review, see Weinberg et al.,
and/or their co-receptors at the site of infection, (iii) number of 1998).
transmitted virions, and (iv) the structure of transmitted viral There is a paucity of information regarding defensin
quasi-species (swarms of mutants). Environmental differences antiviral effects. What has been published suggests that
(pH, target cells, mucosal composition) at the site of exposure defensins can inactivate a host of different viruses. Epithelial-
may affect the efficiency of transmission of the infecting isolates cell-derived beta-defensins have been shown to inhibit
(Overbaugh et al., 1999; Blauvelt et al., 2000). Although there adenoviral infections (Gropp et al., 1999). Alpha-defensins,
may be an element of chance in the expansion of a particular found in azurophilic granules of human neutrophils, inhibit
HIV-1 clone, phenotypic selection does occur in nearly every adenoviral infection in vitro (Bastian and Schafer, 2001),
HIV-1 infectioni.e., selection of R5 isolates occurs in nearly inactivate cytomegalovirus, vesicular stomatitis virus,
every HIV-1 infection. The heterogeneous HIV-1 envelope influenza virus, and herpes simplex virus 1 and 2, but not two
glycoproteins have been identified as responsible for two major non-enveloped viruses, echovirus type 11 and reovirus type 3
viral bio-phenotypes: (i) macrophage-tropic non-syncytium- (Daher et al., 1986). Alpha-defensins have also been shown to
inducing/CCR5-tropic (NSI/R5) HIV-1 isolates, and (ii) T-cell- inhibit HIV replication in vitro (Nakashima et al., 1993; Zhang et
line tropic isolates, forming cell syncytia during active al., 2002). Theta-defensins, originally isolated from the rhesus
replication in tumor T-cell lines, which utilize the CXCR4 co- monkey, Macacca mulatta, and not expressed in humans (Tang
receptor for entry (SI/X4) (Alkhatib et al., 1996; Deng et al., 1996; et al., 1999), were recently shown to prevent infection by T- and
Dragic et al., 1996; Feng et al., 1996). SI/X4 HIV-1 isolates often M-tropic strains of HIV-1 (Cole et al., 2002). The mode(s) of
dominate the quasi-species late in disease, and yet the NSI/R5 viral inactivation in all of the cited studies were not
variant is typically transmitted to a newly infected person determined, and therefore leave a definite void in our
regardless of the route of transmission (reviewed in Fenyo et al., understanding of this potentially important biological activity.
2000). Preferential transmission of NSI/R5 over SI/X4 HIV-1
isolates is contradictory to increased replication of SI/X4 HIV- -defensins and the oral cavity
1 over NSI/R5 isolates in culture (Tersmette et al., 1988; The recent discoveries that -defensins originate in
Bjorndal et al., 1997). Thus, selectivity of transmission is likely mammalian mucosal epithelium, including human (Diamond
not simply a reflection of replicative fitness, but may also be et al., 1991; Schonwetter et al., 1995; Zhao et al., 1996; Harder et
observed in the transmission of different NSI/R5 HIV-1 al., 1997, 2001; McCray and Bentley, 1997; Boe et al., 1999;
isolates in the human population (Blackard et al., 2001). O'Neil et al., 1999; Haynes et al., 2000; Garcia et al., 2001), has
Although in vivo findings suggest that NSI/R5 HIV-1 isolates led to the hypothesis that these antimicrobial peptides function
may out-compete the SI/X4 variants at the site of primary to protect the host against microbial pathogenesis at these
infection, one report suggests that the NSI/R5 isolates critical confrontational sites. We have extended this hypothesis
predominate only after a temporary expansion of SI/X4 HIV-1 to encompass the oral epithelium as well (Krisanaprakornkit et
isolates is quenched by an activated immune response al., 1998, 2000; Weinberg et al., 1998; Dale et al., 2001). This
(Cornelissen et al., 1995). However, this observation is difficult tissue, and cells derived from it, constitutively express hBD-1
to reconcile with the finding that humans who are and can be induced to express hBD-2 and -3.
homozygous for a 32-base-pair deletion in the CCR5 open- The first evidence of -defensins in a mammalian oral
reading frame, and who lack CCR5 on any cell surface, are cavity was described by Schonwetter et al. (1995). The study
typically resistant to HIV-1 infection (Dean et al., 1996; O'Brien identified a -defensin in the upper surface of the bovine
and Moore, 2000). To date, the universal factors (i.e., those tongue, which was markedly increased in the epithelium
found in almost every human host) involved in the selection surrounding naturally occurring tongue lesions, areas of both
of NSI/R5 HIV-1 isolates during transmission and acute and chronic inflammation. This agent was shown to be
asymptomatic disease are not well-defined. Langerhans cells an effective antibacterial and antifungal agent. Since then, we
(LC) are found embedded in mucosa (i.e., vaginal and oral and others have described the presence of -defensins in the
mucosa) and may be the first cell targets for primary human oral cavity (Weinberg et al., 1998; Bonass et al., 1999;
heterosexual transmission (Soto-Ramirez et al., 1996; Blauvelt Mathews et al., 1999; Sahasrabudhe et al., 2000; Dale et al., 2001;
et al., 2000). Thus, LC may play a role in NSI/R5 HIV-1 Dunsche et al., 2002). In gingival tissue, mRNA for both hBD-1
selection, since CCR5 is better expressed in situ and in the and -2 was localized in suprabasal stratified epithelium, and
absence of external stimuli (Blauvelt et al., 2000). For example, the peptides were detected in upper epithelial layers,
a recent report describes increased replication of an NSI/R5 consistent with the formation of the stratified epithelial barrier
(HIV-1Bal) over an SI/X4 (HIV-1III-B) isolate in LC embedded (Dale et al., 2001). hBD-1 and -2 were not detected in the
in skin-derived explants, even though the opposite is true in junctional epithelium (JE) that serves as the attachment to the
PBMC cultures or other permissive cell lines (Soto-Ramirez et tooth surface. In contrast, -defensins and LL-37the only
al., 1996; Blauvelt et al., 2000). However, this is not altogether other cationic antimicrobial peptide known to be expressed in
clear, since another report indicates that CXCR4 is functionally most mucosal epithelium, and belonging to the cathelicidin
expressed on the surfaces of freshly isolated and unstimulated family of antimicrobial peptides (reviewed in Weinberg et al.,
LC (Tchou et al., 2001). 1998)are detected only in the polymorphonuclear
neutrophils (PMNs) that migrate through the JE (Dale et al.,
Defensin peptides and their antiviral activity 2001), a localization that persists during inflammation, when
The human defensin antimicrobial peptide family, its the JE and surrounding tissue are highly infiltrated with

Adv Dent Res 19:42-48, April, 2006 Role of Human -defensins in HIV Infection 43
system. Moreover, the fact that hBDs are
also expressed in less-differentiated
basally oriented epithelial cells indicates
that either differentiation is not essential
for hBD expression, as previously
suggested (Dale et al., 2001), or that hBD-
expressing cells, other than epithelial cells,
are infiltrating the oral mucosae. With
recent findings that hBD-1 and hBD-2 are
also expressed systemically, in human
monocytes, macrophages, and dendritic
cells (Duits et al., 2002), it is tempting to
speculate that they are the source, in part,
for hBD expression in the basal lamina.

Human Beta-defensins and HIV-1


As described above, the mechanisms
underlying mucosal transmission of HIV-1
are incompletely understood, as are the
mechanisms of resistance to HIV-1 infection
in the oral cavity. Infectious virus is rarely
isolated from saliva in HIV-1 infection, and
while salivary inhibitors of HIV-1i.e.,
mucins, amylase, SLPI, proline-rich
Fig. 1 - Immunohistochemical demonstration of human -defensin 1 (hBD-1) and hBD-2 in normal peptides, etc.have been identified, these
gingival epithelium. Gingival samples of periodontally healthy subjects were processed with polyclonal molecules are also found abundantly in
antibodies to hBD-1 (a,b) and hBD-2 (c,d) according to a standard immunohistochemical protocol. The vaginal and seminal secretions, sites where
tissue show intense expression of hBD-2 (c,d) and less intense expression of hBD-1 (a,b) in the granular, infection is more common. Finally, there is
spinous, and basal cell layers of gingival epithelia. The encircled areas in (a) and (c) are amplified in (b)
and (d), respectively, and show positive hBD expression in basal layers. Scale bar: 100 m. (Legend and
diminished infectivity of HIV-1 within oral
Fig. appear with permission from Dr. L.J. Jin [Lu et al., 2004]). tissue when compared with other
susceptible mucosae. For these reasons, we
proposed to test the hypothesis that
mucosal hBDs have anti-HIV-1 activity. Our
PMNs. Therefore, the undifferentiated JE contains exogenously findings are detailed in Quiones-Mateu et al. (2003) and are
expressed -defensins and LL-37, and the stratified epithelium reviewed herein.
contains endogenously expressed -defensins. Calprotectin, an
anionic epithelial cell and PMN-associated antimicrobial HIV-1 induces hBD-2 and -3 but not hBD-1 mRNA
peptide, is constitutively expressed in normal oral in normal human oral epithelial cells (NHOECs)
keratinocytes (Ross and Herzberg, 2001). These findings show NHOEC monolayers were challenged with HIV-1 strains
that defensins and other microbicidal peptides are localized in representing both viral bio-phenotypes, i.e., X4 and R5. Both
specific sites in the gingiva, are synthesized in different cell phenotypes were found to induce hBD-2 and -3 mRNA up to
types, and are likely to serve different roles in various regions 78-fold above baseline (see Quiones-Mateu et al., 2003). In
of the periodontium. addition, while HIV-1 can infect epithelial cells from other
A notable difference between oral and most other epithelia mucosal surfaces (Yahi et al., 1992; Han et al., 2000; Fotopoulos
is the expression of hBD-2 and -3. These defensins are et al., 2002), we could not detect infection of NHOECs by
expressed only in the presence of infection or inflammation in HIV-1 by viral reverse-transcriptase activity in culture
most tissues, including skin, trachea, and gut epithelium supernatant and after real-time PCR analysis to detect
(O'Neil et al., 1999). However, they are expressed in normal proviral DNA. These results differ from those of Liu et al.
uninflamed gingival tissue (Dale et al., 2001; Dunsche et al., (2003), who recently reported low-level replication of the
2002). Present evidence suggests that this baseline level of virus after high-dose viral challenge of NHOECs. The
hBD-2 and -3 could be due to the exposure of the tissue to discrepancy between their findings and ours may be due to
specific commensal bacteria. Our ongoing work in this area has differences in experimental conditions. We omitted polybrene
identified a novel strategy of Fusobacterium nucleatum, whereby from our infection assays, since this detergent is not
this ubiquitous Gram-negative organism of the human oral physiologic. They also infected cells using a viral inoculum
cavity stimulates hBD-2 and -3 expression in normal human eight times higher than ours. Finally, Liu et al. (2003) found
oral epithelial cells (NHOECs)a result that confers protection HIV-1 co-receptor expression on two-week-cultured
to the cells from P. gingivalis invasion (Weinberg et al., NHOECs, while our flow cytometric analyses did not reveal
unpublished observations; manuscript in review). CD4, CCR5, CXCR4, or galactosylceramide expression on
Most recently, Lu et al. (2004) demonstrated the three- to four-day-cultured NHOECs.
expression of hBD-1 and -2 not just in the granular and
spinous regions of normal gingival epithelia, but also in the hBD-2 and -3 inhibit HIV-1 replication without being
basal cell layers (Fig. 1). This is significant, since beta- cytotoxic to immunocompetent cells
defensins have been shown to possess immunomodulatory Pre-incubation of HIV-1 (X4 and R5 strains) with respective
activitysuch as chemoattraction of immature dendritic cells hBDs (recombinant forms generated in the Weinberg
(iDCs), monocytes, and T-cells (Yang et al., 1999; Wu et al., laboratory; see Quiones-Mateu et al. [2003] for details) at
2003)and maturation of iDCs (Biragyn et al., 2002a). concentrations ranging from 5 g/mL to 40 g/mL, followed
Expression of inducible hBDs throughout the oral epithelial by exposure of GHOST X4/R5 cells (osteosarcoma cells co-
mucosa supports the notion that these agents are also transfected with the HIV-2 long-terminal repeat driving
involved in 'cross-talk' with cells of the adaptive immune expression of the green fluorescent protein {hGFP}),

44 Weinberg et al. Adv Dent Res 19:42-48, April, 2006


demonstrated that hBD-2 and hBD-3 were
preferentially able to inhibit X4 virus from
infecting the GHOST cells, but not hBD-1
(Fig. 2B). Moreover, reduced GFP
fluorescence coincided with reduced viral
reverse-transcriptase activity (Fig. 2C).
Moreover, conditions that mimic the oral
mucosal interface, low salt and no serum
(Mandel, 1972), were found to elicit the
best in vitro protection by the hBDs
(Quiones-Mateu et al., 2003). Interestingly,
the concentration range of the hBDs that
were used fell well within the reported
concentrations of hBD-2 in normal oral
epithelium (Sawaki et al., 2002). A
thaizolyl-blue-based colorimetric assay
(MTT method) (Pauwels et al., 1988)
revealed no cytotoxicity against any of the
immunocompetent cells that were used in
the study, even at the highest hBD
concentrations that demonstrated anti-
HIV-1 activity.
hBD-2 and -3 down-modulate CXCR4,
but not CCR5
Flow cytometric analyses of sham-treated
vs. hBD-treated immunocompetent cells
(PBMCs) revealed a significant selective Fig. 2 - Anti-HIV-1 activity of hBDs. HIV-1 strains (X4 HXB2 and R5 93US142) were incubated in 10 mM
phosphate buffer with increasing concentrations of hBDs and used to infect GHOST X4/R5 cells. (a) Qualitative
reduction of the CXCR4 receptor in cells determination of HIV-1 infection, measured by GFP fluorescence, in the absence (-C) and presence (+C) of
pre-treated with hBD-2 or hBD-3, but not virus pre-incubated in 10 mM PB. (b) Anti-HIV-1 activity of hBDs in GHOST X4/R5 cells examined by
hBD-1. Similar results were obtained with fluorescence microscopy. (c) Anti-HIV-1 activity of hBDs measured by reverse-transcriptase (RT) activity in cell-
CEM cells (T-cell lines) expressing CXCR4 free culture supernatant, relative to the positive control (i.e., HIV-1 infection in the absence of hBD).
and CCR5. Confocal microscopy results Reproduced with permission from Quiones-Mateu et al. (2003).
suggested that hBD-2 and -3 bind to cell-
surface CXCR4 and induce internalization
of the bound complex (Fig. 3). In the case of hBD-3, this down-
regulation was maintained for at least 24 hrs. Interestingly,
hBD modulation of CXCR4 occurred even in the presence of
high salt concentrations, suggesting that the hBD activity could
be occurring on cells deeper in the mucosal epithelium. Finally,
immunoelectron microscopy confirmed hBD localization to the
cell membrane (Fig. 4).
hBD-2 and -3 interact directly with HIV-1
After noticing that multiple washings of HIV-1 virions that had
been pre-exposed to hBD2 or -3 did not reverse the anti-
retroviral effects of the hBDs, we conducted immunoelectron
microscopy and saw direct localization of the beta-defensins to
the virion particles (Fig. 4). Electrostatic interactions with
virion-specific membrane structures, such as gp120, are
plausible. Polyanionic compounds exert their anti-HIV-1
activity by binding to the positively charged sites in the V3
loop of gp120 (Schols et al., 1990; Witvrouw et al., 1994). Like
other polycationic peptides which block infection with X4 HIV-
1 isolates (e.g., T22, T134, and ALX40-4C) (De Clercq, 2002), the
direct anti-retroviral effects of beta-defensins might be
predicted to be very different, perhaps interacting with other
viral surface domains. Theta-defensins have been shown to
inhibit HIV-1 replication in vitro by binding to envelope sugar
residues (Wang et al., 2003). Whether hBD2 and -3 exhibit
Fig. 3 - Confocal microscopy analysis of hBD-2 and -3 on CXCR4 and CCR5
lectin-like properties remains to be seen.
surface expression. CEM X4/R5 cells were stained with anti-CXCR4 PE, anti-CCR5
Significance PE, anti-hBD-2, and anti-hBD-3 monoclonal antibodies in the presence and
absence of 20 g/mL of hBD-2 and -3. Pre-incubation with hBD-2 (a2) or hBD-3
More than any other single development, the advances in (b2) specifically blocked the detection of CXCR4 but not CCR5 on the cell surface.
HIV/AIDS therapeutics have caused a decrease in both AIDS Detection of hBD-2 (c,e) or -3 (d,f) with live, fixed, or live cells post-perfusion.
incidence and death in the US and Europe. Despite these Following incubation with hBD-2 or -3, polyclonal antibodies against the hBDs
gratifying advances, the worldwide incidence of HIV failed to detect them on the surfaces of live cells (c2, d2). 1% paraformaldehyde
fixation, followed by hBD-2 or -3 incubation and antibody labeling,
infection is rising, and strategies to prevent the acquisition of demonstrated hBDs bound to the cell membrane (e1, f1). To visualize hBD-2 and
HIV infection are urgently needed (Little et al., 2002). The -3 internalization, we incubated live CEM cells with hBD-2 or -3, followed by cell
discovery, in the mid-1990s, of the major HIV-1 co-receptors permeabilization (FACS/PERM) and staining with anti-hBD antibodies (e2, f2).
(Cocchi et al., 1995) has led to rapid development of novel Reproduced with permission from Quiones-Mateu et al. (2003).

Adv Dent Res 19:42-48, April, 2006 Role of Human -defensins in HIV Infection 45
Acknowledgments
Work at the CCF (M.E.Q-M) was supported by grant 5-K01-
HL67610-03. Work at CWRU was supported by CFAR A136219
and A1 51649 (M.M.L.). Work at the CWRU School of Dental
Medicine was supported by R01 DE12589, R01 DE13992, and
R01 DE015510 (A.W.).

References
Alkhatib G, Combadiere C, Broder CC, Feng Y, Kennedy PE, Murphy
PM, et al. (1996). CC CKR5: a RANTES, MIP-1alpha, MIP-1beta
receptor as a fusion cofactor for macrophage-tropic HIV-1. Science
272:1955-1958.
Baqui AA, Meiller TF, Falkler WA Jr (1999). Enhanced secretory
leukocyte protease inhibitor in human immunodeficiency virus
type 1-infected patients. Clin Diagn Lab Immunol 6:808-811.
Fig. 4 - Immunoelectron microscopy analysis showing the interaction of hBD-2
Baron S, Poast J, Cloyd MW (1999). Why is HIV rarely transmitted by
and -3 with HIV-1 and with T-cell membrane. X4 HIV-1 HXB2 strain and MT4 oral secretions? Saliva can disrupt orally shed, infected leukocytes.
cells (T-cell line) were incubated with hBD-2 or -3 (20 g/mL), 37C, 1 hr. Arch Intern Med 159:303-310.
Polyclonal anti-hBD-2 or -3 antibodies were added, followed by the addition Barr CE, Miller LK, Lopez MR, Croxson TS, Schwartz SA, Denman H,
of secondary IgG conjugated with 10-nm gold particles. Arrows indicate hBD- et al. (1992). Recovery of infectious HIV-1 from whole saliva. J Am
2 and -3 localization to virions and the cell membrane. Reproduced with Dent Assoc 123:36-37, 39-48.
permission from Quiones-Mateu et al. (2003).
Bastian A, Schafer H (2001). Human alpha-defensin 1 (HNP-1) inhibits
adenoviral infection in vitro. Reg Pept 101:157-161.
Becquart P, Gresenguet G, Hocini H, Kazatchkine MD, Belec L (1999).
antiretroviral strategies that target binding of HIV to these co- Secretory leukocyte protease inhibitor in colostrum and breast
receptors. Moreover, observations of intrinsic resistance to the milk is not a major determinant of the protection of early postnatal
acquisition of HIV infection have demonstrated that failure of transmission of HIV. AIDS 13:2599-2602.
CCR5 expression is associated with high-level resistance to Biragyn A, Ruffini PA, Leifer CA, Klyushnenkova E, Shakhov A,
HIV infection (Dean et al., 1996; Liu et al., 1996). Nonetheless, Chertov O, et al.(2002a). Toll-like receptor 4-dependent activation
studies in several cohorts of high-risk seronegative of dendritic cells by beta-defensin 2. Science 298:1025-1029.
individuals have clearly demonstrated that the 32-base-pair Biragyn A, Belyakov IM, Chow YH, Dimitrov DS, Berzofsky JA, Kwak
mutation in the CCR5 open-reading frame, which results in LW (2002b). DNA vaccines encoding human immunodeficiency
the failure of surface expression of this chemokine receptor, is virus-1 glycoprotein 120 fusions with proinflammatory
found at best in only a minority of high-risk HIV-seronegative chemoattractants induce systemic and mucosal immune responses.
individuals (Kokkotou et al., 1998; Kaul et al., 2000; Salkowitz Blood 100:1153-1159.
et al., 2001). Thus, it is likely that other mechanisms Bjorndal A, Deng H, Jansson M, Fiore JR, Colognesi C, Karlsson A, et
determine risk for the acquisition of HIV infection in these al. (1997). Coreceptor usage of primary human immunodeficiency
groups. We propose that studies of the oral cavity may virus type 1 isolates varies according to biological phenotype. J
provide insights into intrinsic mechanisms of resistance to Virol 71:7478-7487.
HIV infection. New drugs in development target different Blackard JT, Renjifo B, Fawzi W, Hertzmark E, Msamanga G,
steps in the viral life cycle (Jacobson et al., 2000), gp 120- Mwakagile D, et al. (2001). HIV-1 LTR subtype and perinatal
binding to the chemokine receptors CCR5 or CXCR4 (Cocchi transmission. Virology 287:261-265.
et al., 1995), and the fusion of the viral and cellular Blauvelt A, Glushakova S, Margolis LB (2000). HIV-infected human
membranes (Kilby et al., 1998). The fact that the oral cavity is Langerhans cells transmit infection to human lymphoid tissue ex
naturally resistant to HIV-1 infection prompts us to ask how vivo. AIDS 14:647-651.
and why? While salivary components have been identified Boe R, Silvola J, Yang J, Moens U, McCray PB Jr, Stenfors LE, et al.
and are continuing to be studied as potential antiretroviral (1999). Human beta-defensin-1 mRNA is transcribed in tympanic
agents, we believe that there is much to be learned, and membrane and adjacent auditory canal epithelium. Infect Immun
potentially tapped, from the oral mucosal epithelium. There 67:4843-4846.
is new, exciting information emanating from leading Bonass WA, High AS, Owen PJ, Devine DA (1999). Expression of beta-
laboratories that defensin molecules may be a new frontier defensin genes by human salivary glands. Oral Microbiol Immunol
for studying the host's defense against HIV-1 (Yang et al., 14:371-374.
1999; Biragyn et al., 2002a,b; Zhang et al., 2002). Our Cocchi F, DeVico AL, Garzino-Demo A, Arya SK, Gallo RC, Lusso P
preliminary studies suggest that an intriguing dynamic is (1995). Identification of RANTES, MIP-1 alpha, and MIP-1 beta as
occurring between the human oral epithelial cell and HIV-1 the major HIV-suppressive factors produced by CD8+ T cells [see
virions, possibly leading to mucosal protection through - comments]. Science 270:1811-1815. Comment in: Science 274:1393-
defensin expression. We hypothesize that 'turning on' 1395.
inducible beta-defensins, locally, could result in less viral Cole AM, Hong T, Boo LM, Nguyen T, Zhao C, Bristol G, et al. (2002).
transmission through the mucosal barrier. Moreover, other Retrocyclin: a primate peptide that protects cells from infection by
epithelial-cell-derived antimicrobial agents may be induced T- and M-tropic strains of HIV-1. Proc Natl Acad Sci USA 99:1813-
along with the hBDs, upon HIV-1 challenge, and could then 1818.
act collectively in protecting the oral cavity from HIV-1 Coppenhaver DH, Sriyuktasuth-Woo P, Baron S, Barr CE, Qureshi MN
infection. This may include such agents as SLPI, whose (1994). Correlation of nonspecific antiviral activity with the ability
mechanism for selectively inhibiting R5 virus was recently to isolate infectious HIV-1 from saliva. N Engl J Med 330:1314-1315.
elucidated (Ma et al., 2004). Results presented herein are Cornelissen M, Mulder-Kampinga G, Veenstra J, Zorgdrager F, Kuiken
intended to shed light on the properties of the oral mucosa C, Hartman S, et al. (1995). Syncytium-inducing (SI) phenotype
that could contribute to its relative resistance to HIV-1, which suppression at seroconversion after intramuscular inoculation of a
could then be translated into promoting similar protection in non-syncytium-inducing/SI phenotypically mixed human
more vulnerable mucosal barriers. immunodeficiency virus population. J Virol 69:1810-1818.

46 Weinberg et al. Adv Dent Res 19:42-48, April, 2006


Daher KA, Selsted ME, Lehrer RI (1986). Direct inactivation of viruses antibiotic from human skin [letter]. Nature 387:861.
by human granulocyte defensins. J Virol 60:1068-1074. Harder J, Bartels J, Christophers E, Schroder JM (2001). Isolation and
Dale BA, Kimball JR, Krisanaprakornkit S, Roberts F, Robinovitch M, characterization of human beta-defensin-3, a novel human
O'Neal R, et al. (2001). Localized antimicrobial peptide expression inducible peptide antibiotic. J Biol Chem 276:5707-5713.
in human gingiva. J Periodontal Res 36:285-294. Haynes RJ, McElveen JE, Dua HS, Tighe PJ, Liversidge J (2000).
De Clercq E (2002). New developments in anti-HIV chemotherapy. Expression of human beta-defensins in intraocular tissues. Invest
Biochim Biophys Acta 1587:258-275. Ophthalmol Vis Sci 41:3026-3031.
Dean M, Carrington M, Winkler C, Huttley GA, Smith MW, Allikmets Herz AM, Robertson MN, Lynch JB, Schmidt A, Rabin M, Sherbert C, et
R, et al. (1996). Genetic restriction of HIV-1 infection and al. (2002). Viral dynamics of early HIV infection in neonatal
progression to AIDS by a deletion allele of the CKR5 structural macaques after oral exposure to HIV-2287: an animal model with
gene. Hemophilia Growth and Development Study, Multicenter implications for maternal-neonatal HIV transmission. J Med
AIDS Cohort Study, Multicenter Hemophilia Cohort Study, San Primatol 31:29-39.
Francisco City Cohort, ALIVE Study [see comments] [erratum Ho DD, Byington RE, Schooley RT, Flynn T, Rota TR, Hirsch MS (1985).
appears in Science 274:1069, 1996]. Science 273:1856-1862. Comment Infrequency of isolation of HTLV-III virus from saliva in AIDS
in: Science 273:1979-1798. [letter]. N Engl J Med 313:1606.
Deng H, Liu R, Ellmeier W, Choe S, Unutmaz D, Burkhart M, et al. Jacobson JM, Lowy I, Fletcher CV, O'Neill TJ, Tran DN, Ketas TJ, et al.
(1996). Identification of a major co-receptor for primary isolates of (2000). Single-dose safety, pharmacology, and antiviral activity of
HIV-1 [see comments]. Nature 381:661-666. Comment in: Nature the human immunodeficiency virus (HIV) type 1 entry inhibitor
381:647-648. PRO 542 in HIV-infected adults. J Infect Dis 182:326-329.
Diamond G, Zasloff M, Eck H, Brasseur M, Maloy WL, Bevins CL Kaul R, Plummer FA, Kimani J, Dong T, Kiama P, Rostron T, et al.
(1991). Tracheal antimicrobial peptide, a cysteine-rich peptide from (2000). HIV-1-specific mucosal CD8+ lymphocyte responses in the
mammalian tracheal mucosa: peptide isolation and cloning of a cervix of HIV-1-resistant prostitutes in Nairobi. J Immunol
cDNA. Proc Natl Acad Sci USA 88:3952-3956. 164:1602-1611.
Dragic T, Litwin V, Allaway GP, Martin SR, Huang Y, Nagashima KA, Kilby JM, Hopkins S, Venetta TM, DiMassimo B, Cloud GA, Lee JY, et
et al. (1996). HIV-1 entry into CD4+ cells is mediated by the al. (1998). Potent suppression of HIV-1 replication in humans by T-
chemokine receptor CC-CKR-5 [see comments]. Nature 381:667- 20, a peptide inhibitor of gp41-mediated virus entry [see
673. Comment in: Nature 381:647-648. comments]. Nat Med 4:1302-1307. Comment in: Nat Med 4:1232-
Duits LA, Ravensbergen B, Rademaker M, Hiemstra PS, Nibbering PH 1233.
(2002). Expression of beta-defensin 1 and 2 mRNA by human mono- Kokkotou E, Philippon V, Gueye-Ndiaye A, Mboup S, Wang WK, Essex
cytes, macrophages and dendritic cells. Immunology 106:517-525. M, et al. (1998). Role of the CCR5 delta 32 allele in resistance to
Dunsche A, Acil Y, Dommisch H, Siebert R, Schroder JM, Jepsen S HIV-1 infection in west Africa. J Hum Virol 1:469-474.
(2002). The novel human beta-defensin-3 is widely expressed in Krisanaprakornkit S, Weinberg A, Perez CN, Dale BA (1998).
oral tissues. Eur J Oral Sci 110:121-124. Expression of the peptide antibiotic human beta-defensin 1 in
Feng Y, Broder CC, Kennedy PE, Berger EA (1996). HIV-1 entry cultured gingival epithelial cells and gingival tissue. Infect Immun
cofactor: functional cDNA cloning of a seven-transmembrane, G 66:4222-4228.
protein-coupled receptor [see comments]. Science 272:872-877. Krisanaprakornkit S, Kimball JR, Weinberg A, Darveau RP, Bainbridge
Comment in: Science 272:809-810. BW, Dale BA (2000). Inducible expression of human beta-defensin
Fenyo EM, Schuitemaker B, Asjo B, McKeating J, Sattentau Q, EC 2 by Fusobacterium nucleatum in oral epithelial cells: multiple
Concerted Action HIV Variability (2000). The history of HIV-1 signaling pathways and role of commensal bacteria in innate
biological phenotypes past, present and future [review]. In: immunity and the epithelial barrier. Infect Immun 68:2907-2915.
Human Retroviruses and AIDS 1997. Korber B, Hahn B, Foley B, Little SJ, Holte S, Routy JP, Daar ES, Markowitz M, Collier AC, et al.
Mellors JW, Leitner T, Myers G, et al., editors. Los Alamos, NM: (2002). Antiretroviral-drug resistance among patients recently
Theoretical Biology and Biophysics Group, Los Alamos National infected with HIV [see comments]. N Engl J Med 347:385-394.
Laboratory. Comment in: N Engl J Med 347: 438-439. Author reply in: N Engl J
Fotopoulos G, Harari A, Michetti P, Trono D, Pantaleo G, Kraehenbuhl Med 347:1889-1890.
JP (2002). Transepithelial transport of HIV-1 by M cells is receptor- Liu L, Heng H, Zhao C, Ganz T (1996). Human alpha- and beta-
mediated. Proc Natl Acad Sci USA 99:9410-9414. defensins evolved from a common pre-mammalian ancestor
Fultz PN (1986). Components of saliva inactivate human (abstract). J Invest Med 44:294A.
immunodeficiency virus [letter]. Lancet 2:1215. Liu X, Zha J, Chen H, Nishitani J, Camargo P, Cole SW, et al. (2003).
Garcia JR, Krause A, Schulz S, Rodriguez-Jimenez FJ, Kluver E, Human immunodeficiency virus type 1 infection and replication in
Adermann K, et al. (2001). Human beta-defensin 4: a novel normal human oral keratinocytes. J Virol 77:3470-3476.
inducible peptide with a specific salt-sensitive spectrum of Lu Q, Jin L, Darveau RP, Samaranayake LP (2004). Expression of
antimicrobial activity. FASEB J 15:1819-1821. human beta-defensins-1 and -2 peptides in unresolved chronic
Gooch B, Marianos D, Ciesielski C, Dumbaugh R, Lasch A, Jaffe H, et periodontitis. J Periodontal Res 39:221-227.
al. (1993). Lack of evidence for patient-to-patient transmission of Ma G, Greenwell-Wild T, Lei K, Jin W, Swisher J, Hardegen N, et al.
HIV in a dental practice [see comments]. J Am Dent Assoc 124:38- (2004). Secretory leukocyte protease inhibitor binds to annexin II, a
44. Comment in: J Am Dent Assoc 124:12, 14. cofactor for macrophage HIV-1 infection. J Exp Med 200:1337-1346.
Goto Y, Yeh CK, Notkins AL, Prabhakar BS (1991). Detection of Mandel ID (1972). Saliva. St. Louis: C.V. Mosby Co.
proviral sequences in saliva of patients infected with human Mathews M, Jia HP, Guthmiller JM, Losh G, Graham S, Johnson GK, et
immunodeficiency virus type 1. AIDS Res Hum Retroviruses 7:343- al. (1999). Production of beta-defensin antimicrobial peptides by
347. the oral mucosa and salivary glands. Infect Immun 67:2740-2745.
Gropp R, Frye M, Wagner TO, Bargon J (1999). Epithelial defensins McCray PB Jr, Bentley L (1997). Human airway epithelia express a
impair adenoviral infection: implication for adenovirus-mediated beta-defensin. Am J Respir Cell Mol Biol 16:343-349.
gene therapy. Hum Gene Ther 10:957-964. McNeely TB, Dealy M, Dripps DJ, Orenstein JM, Eisenberg SP, Wahl
Han Y, Ventura CL, Black KP, Cummins JE Jr, Hall SD, Jackson S (2000). SM (1995). Secretory leukocyte protease inhibitor: a human saliva
Productive human immunodeficiency virus-1 infection of protein exhibiting anti-human immunodeficiency virus 1 activity
epithelial cell lines of salivary gland origin. Oral Microbiol Immunol in vitro. J Clin Invest 96:456-464.
15:82-88. Moore BE, Flaitz CM, Coppenhaver DH, Nichols M, Kalmaz GD,
Hancock RE (1997). Peptide antibiotics. Lancet 349:418-422. Bessman JD, et al. (1993). HIV recovery from saliva before and after
Harder J, Bartels J, Christophers E, Schroder JM (1997). A peptide dental treatment: inhibitors may have critical role in viral

Adv Dent Res 19:42-48, April, 2006 Role of Human -defensins in HIV Infection 47
inactivation. J Am Dent Assoc 124:67-74. 179(Suppl 3):S436-S440.
Nakashima H, Yamamoto N, Masuda M, Fujii N (1993). Defensins Soto-Ramirez LE, Renjifo B, McLane MF, Marlink R, O'Hara C,
inhibit HIV replication in vitro [letter]. AIDS 7:1129. Sutthent R, et al. (1996). HIV-1 Langerhans' cell tropism associated
O'Brien SJ, Moore JP (2000). The effect of genetic variation in with heterosexual transmission of HIV [see comments]. Science
chemokines and their receptors on HIV transmission and 271:1291-1293. Comment in: Science 278:786-788.
progression to AIDS. Immunolog Rev 177:99-111. Tang YQ, Yuan J, Osapay G, Osapay K, Tran D, Miller CJ, et al. (1999). A
O'Neil DA, Porter EM, Elewaut D, Anderson GM, Eckmann L, Ganz T, cyclic antimicrobial peptide produced in primate leukocytes by the
et al. (1999). Expression and regulation of the human beta- ligation of two truncated alpha-defensins [see comments]. Science
defensins hBD-1 and hBD-2 in intestinal epithelium. J Immunol 286:498-502. Comment in: Science 286:420-421.
163:6718-6724. Tchou I, Misery L, Sabido O, Dezutter-Dambuyant C, Bourlet T, Moja P,
Overbaugh J, Kreiss J, Poss M, Lewis P, Mostad S, John G, et al. (1999). et al. (2001). Functional HIV CXCR4 coreceptor on human
Studies of human immunodeficiency virus type 1 mucosal viral epithelial Langerhans cells and infection by HIV strain X4. J Leukoc
shedding and transmission in Kenya. J Infect Dis 179(Suppl Biol 70:313-321.
3):S401-S404. Tersmette M, de Goede RE, Al BJ, Winkel IN, Gruters RA, Cuypers HT,
Pauwels R, Balzarini J, Baba M, Snoeck R, Schols D, Herdewijn P, et al. et al. (1988). Differential syncytium-inducing capacity of human
(1988). Rapid and automated tetrazolium-based colorimetric assay immunodeficiency virus isolates: frequent detection of syncytium-
for the detection of anti-HIV compounds. J Virolog Meth 20:309-321. inducing isolates in patients with acquired immunodeficiency
Pillay K, Coutsoudis A, Agadzi-Naqvi AK, Kuhn L, Coovadia HM, syndrome (AIDS) and AIDS-related complex. J Virol 62:2026-2032.
Janoff EN (2001). Secretory leukocyte protease inhibitor in vaginal Wahl SM, McNeely TB, Janoff EN, Shugars D, Worley P, Tucker C, et al.
fluids and perinatal human immunodeficiency virus type 1 (1997). Secretory leukocyte protease inhibitor (SLPI) in mucosal
transmission. J Infect Dis 183:653-656. fluids inhibits HIV-I. Oral Dis 3(Suppl 1):S64-S69.
Quiones-Mateu ME, Lederman MM, Feng Z, Chakraborty B, Weber J, Wang W, Cole AM, Hong T, Waring AJ, Lehrer RI (2003). Retrocyclin,
Rangel HR, et al. (2003). Human epithelial beta-defensins 2 and 3 an antiretroviral theta-defensin, is a lectin. J Immunol 170:4708-
inhibit HIV-1 replication. AIDS 17:F39-F48. 4716.
Rogers MF, White CR, Sanders R, Schable C, Ksell TE, Wasserman RL, Weinberg A, Krisanaprakornkit S, Dale BA (1998). Epithelial
et al. (1990). Lack of transmission of human immunodeficiency antimicrobial peptides: review and significance for oral
virus from infected children to their household contacts. Pediatrics applications. Crit Rev Oral Biol Med 9:399-414.
85:210-214. Witvrouw M, Este JA, Quiones-Mateu ME, Reymen D, Andrei G,
Ross KF, Herzberg MC (2001). Calprotectin expression by gingival Snoeck R, et al. (1994). Activity of a sulfated polysaccharide
epithelial cells. Infect Immun 69:3248-3254. extracted from the red seaweed Aghardhiella tenera against human
Sahasrabudhe KS, Kimball JR, Morton TH, Weinberg A, Dale BA immunodeficiency virus and other enveloped viruses. Antivir
(2000). Expression of the antimicrobial peptide, human beta- Chem Chemother 5:297-303.
defensin 1, in duct cells of minor salivary glands and detection in Wu Z, Hoover DM, Yang D, Boulegue C, Santamaria F, Oppenheim JJ,
saliva. J Dent Res 79:1669-1674. et al. (2003). Engineering disulfide bridges to dissect antimicrobial
Salkowitz JR, Purvis SF, Meyerson H, Zimmerman P, O'Brien TR, and chemotactic activities of human beta-defensin 3. Proc Natl
Aledort L, et al. (2001). Characterization of high-risk HIV-1 Acad Sci USA 100:8880-8885.
seronegative hemophiliacs. Clin Immunol 98:200-211. Yahi N, Baghdiguian S, Moreau H, Fantini J (1992). Galactosyl
Sawaki K, Mizukawa N, Yamaai T, Yoshimoto T, Nakano M, Sugahara ceramide (or a closely related molecule) is the receptor for human
T (2002). High concentration of beta-defensin-2 in oral squamous immunodeficiency virus type 1 on human colon epithelial HT29
cell carcinoma. Anticancer Res 22:2103-2107. cells. J Virol 66:4848-4854.
Schols D, Pauwels R, Desmyter J, De Clercq E (1990). Dextran sulfate Yang D, Chertov O, Bykovskaia SN, Chen Q, Buffo MJ, Shogan J, et al.
and other polyanionic anti-HIV compounds specifically interact (1999). Beta-defensins: linking innate and adaptive immunity
with the viral gp120 glycoprotein expressed by T-cells persistently through dendritic and T cell CCR6 [see comments]. Science
infected with HIV-1. Virology 175:556-561. 286:525-528. Comment in: Science 286:420-421.
Schonwetter BS, Stolzenberg ED, Zasloff MA (1995). Epithelial Zasloff M (2002). Antimicrobial peptides of multicellular organisms.
antibiotics induced at sites of inflammation. Science 267:1645-1648. Nature 415:389-395.
Shugars DC (1999). Endogenous mucosal antiviral factors of the oral Zhang L, Yu W, He T, Yu J, Caffrey RE, Dalmasso EA, et al. (2002).
cavity. J Infect Dis 179(Suppl 3):S431-S435. Contribution of human alpha-defensin 1, 2, and 3 to the anti-HIV-1
Shugars DC, Wahl SM (1998). The role of the oral environment in HIV- activity of CD8 antiviral factor. Science 298:995-1000.
1 transmission. J Am Dent Assoc 129:851-858. Zhao C, Wang I, Lehrer RI (1996). Widespread expression of beta-
Smith PD, Li L, Meng G (1999). Mucosal events in the pathogenesis of defensin hBD-1 in human secretory glands and epithelial cells.
human immunodeficiency virus type 1 infection. J Infect Dis FEBS Lett 396:319-322.

48 Weinberg et al. Adv Dent Res 19:42-48, April, 2006

You might also like