ArticlePDF AvailableLiterature Review

Sex Drives Dimorphic Immune Responses to Viral Infections

Authors:

Abstract and Figures

New attention to sexual dimorphism in normal mam-malian physiology and disease has uncovered a previ-ously unappreciated breadth of mechanisms by which females and males differentially exhibit quantitative phenotypes. Thus, in addition to the established mod-ifying effects of hormones, which prenatally and postpubertally pattern cells and tissues in a sexually dimorphic fashion, sex differences are caused by extra-gonadal and dosage effects of genes encoded on sex chromosomes. Sex differences in immune responses, especially during autoimmunity, have been studied pre-dominantly within the context of sex hormone effects. More recently, immune response genes have been local-ized to sex chromosomes themselves or found to be reg-ulated by sex chromosome genes. Thus, understanding how sex impacts immunity requires the elucidation of complex interactions among sex hormones, sex chromo-somes, and immune response genes. In this Brief Re-view, we discuss current knowledge and new insights into these intricate relationships in the context of viral infections.
Content may be subject to copyright.
of March 15, 2017.
This information is current as
Viral Infections
Sex Drives Dimorphic Immune Responses to
Soumitra Ghosh and Robyn S. Klein
http://www.jimmunol.org/content/198/5/1782
doi: 10.4049/jimmunol.1601166
2017; 198:1782-1790; ;J Immunol
References http://www.jimmunol.org/content/198/5/1782.full#ref-list-1
, 55 of which you can access for free at: cites 158 articlesThis article
Subscriptions http://jimmunol.org/subscriptions is online at: The Journal of ImmunologyInformation about subscribing to
Permissions http://www.aai.org/ji/copyright.html
Submit copyright permission requests at:
Email Alerts http://jimmunol.org/cgi/alerts/etoc
Receive free email-alerts when new articles cite this article. Sign up at:
Print ISSN: 0022-1767 Online ISSN: 1550-6606.
Immunologists, Inc. All rights reserved.
Copyright © 2017 by The American Association of
9650 Rockville Pike, Bethesda, MD 20814-3994.
The American Association of Immunologists, Inc.,
is published twice each month byThe Journal of Immunology
by guest on March 15, 2017http://www.jimmunol.org/Downloaded from by guest on March 15, 2017http://www.jimmunol.org/Downloaded from
Sex Drives Dimorphic Immune Responses to
Viral Infections
Soumitra Ghosh* and Robyn S. Klein*
,,
New attention to sexual dimorphism in normal mam-
malian physiology and disease has uncovered a previ-
ously unappreciated breadth of mechanisms by which
females and males differentially exhibit quantitative
phenotypes. Thus, in addition to the established mod-
ifying effects of hormones, which prenatally and post-
pubertally pattern cells and tissues in a sexually
dimorphic fashion, sex differences are caused by extra-
gonadal and dosage effects of genes encoded on sex chro-
mosomes. Sex differences in immune responses,
especially during autoimmunity, have been studied pre-
dominantly within the context of sex hormone effects.
More recently, immune response genes have been local-
ized to sex chromosomes themselves or found to be reg-
ulated by sex chromosome genes. Thus, understanding
how sex impacts immunity requires the elucidation of
complex interactions among sex hormones, sex chromo-
somes, and immune response genes. In this Brief Re-
view, we discuss current knowledge and new insights
into these intricate relationships in the context of viral
infections. The Journal of Immunology, 2017, 198:
1782–1790.
Age, sex, and immune state of the host are considered
salient biological factors that determine the extent
and strength of pathogen clearance during infectious
diseases (1–3). With regard to viral infections, epidemiolog-
ical studies revealed that males have a higher mortality com-
pared with females, who reportedly display stronger antiviral
cellular and humoral immune responses (4). Although
stronger immune responses may provide better protection
against certain pathogens, in some chronic viral infections it
can lead to aberrant antigenic responses with immunopa-
thology (5, 6). Net sex differences or the degree of sexual
dimorphism in biological responses derive from genetic dif-
ferences in chromosome complement and induce their effects
via acute activational or prenatal organizational/epigenetic
effects of gonadal sex hormones (estrogen, progesterone, and
androgens), extragonadal effects of sex chromosome–encoded
genes, and compensatory mechanisms, such as reduction in
gene-dosage differences through X-chromosome inactivation.
These processes underlie sex differences in most tissue re-
sponses during normal and disease states, including immu-
nological responses to infectious diseases (Fig. 1).
Differences in susceptibility and response to viral pathogens
observed in males and females have primarily been attributed
to activational effects of sex hormones and dosages of genes on
the X and Y chromosomes (4). The onset of puberty is as-
sociated with the numbers and functions of circulating
granulocytes and monocytes, which are decreased, but acti-
vated, in females as a result of rising levels of progesterone in
the setting of ovulation or pregnancy (7, 8). In addition,
myeloid cells and lymphocytes express receptors for estrogen,
progesterone, and androgens, which orchestrate transcrip-
tional pathways and ligand-dependent or ligand-independent
signaling cascades that influence innate and adaptive immune
responses to viruses (9–12). With regard to gene dosage on sex
chromosomes, X-linked genes, such as IL-13,IL-4,IL-10,
XIST,TLR7,FOXP3, and sex-determining region Y box 9
(SOX9), on the X chromosome and sex-determining region Y
(SRY; testis-determining factor) and SOX9 on Y chromosome
may underlie sexually dimorphic responses that contribute to
stronger innate, cellular, and humoral immune responses and
susceptibility to autoimmune diseases in females compared
with males (13–15). Studies in humans and animal models
indicate sexually dimorphic mechanisms that contribute to
virologic control during infections with HIV-1, vesicular
stomatitis virus, hantavirus (Seoul virus), influenza virus
(H1N1), hepatitis C virus, Theiler’s murine encephalomy-
elitis virus, HSV-1 and coxsackievirus B3 (CVB3) (5, 16–20).
In this review, we introduce the various mechanisms that
impose sexual dimorphism in immune function in males and
females. We then discuss the impact of sexually dimorphic
immune responses on pathogenesis during viral infections.
Organizational effects of sex hormones on immune function
The initiation of sexual dimorphism occurs through early
embryonic development due to effects of genes on sex chro-
mosomes (21, 22). The SRY gene on the Y chromosome
*Department of Internal Medicine, Washington University School of Medicine,
St. Lo uis , MO 63110;
Department of Pathology and Immunology, Washington Uni-
versity School of Medicine, St. Louis, MO 63110; and
Department of Neuroscience,
Washington University School of Medicine, St. Louis, MO 63110
ORCID: 0000-0001-6785-6362 (S.G.).
Received for publication July 5, 2016. Accepted for publication October 24, 2016.
This work was supported by National Institutes of Health Grants R01 NS052632 and
U19 AI083019 and a grant from the National Multiple Sclerosis Society (all to R.S.K.).
Address correspondence and reprint requests to Dr. Robyn S. Klein, Department of
Medicine, Washington University School of Medicine, 660 South Euclid Avenue,
St. Louis, MO 63110. E-mail address: rklein@dom.wustl.edu
Abbreviations used in this article: AR, androgen receptor; CVB3, coxsackievirus B3; E2,
17b-estradiol; E3, estriol; ER, estrogen receptor; FCG, four-core genotype; MCMV,
murine CMV; pDC, plasmacytoid dendritic cell; RSPO, R-spondin; SLE, systemic
lupus erythematosus; SOX9, sex-determining region Y box 9; SRY, sex-determining
region Y; Treg, regulatory T cell.
Copyright Ó2017 by The American Association of Immunologists, Inc. 0022-1767/17/$30.00
www.jimmunol.org/cgi/doi/10.4049/jimmunol.1601166
Journal of
e
Immunology
Brief Reviews
by guest on March 15, 2017http://www.jimmunol.org/Downloaded from
induces male supporting cell precursors to differentiate into
testosterone-expressing Sertoli cells, leading to masculiniza-
tion of all tissues within the developing embryo (23–25).
During embryonic development, the increased expression of
multiple genes maintains the XX sex phenotype and inhibits
the expression of genes, such as SOX9, a transcription factor
that favors the XY male phenotype (15). In males, expression
of SOX9 protein downstream of Sry is crucial in inducing
male sex phenotype. SOX9 can independently induce male-
to-female sex reversal in male embryos through deletion from
chromosome 17 (23, 26, 27). Gonadal R-spondin (RSPO)1 is
a secreted protein that is highly expressed in female blastocysts
and interacts with Wnt4, which promotes ovarian develop-
ment and estrogen production via the Wnt4/b-catenin sig-
naling pathway (28, 29). Loss-of-function mutational studies
in RSPO1 and RSPO
2/2
XX mice confirmed this observa-
tion (27–30). FOXL2, a forkhead transcriptional regulator,
also contributes to ovarian differentiation and maintenance
during embryonic development in females via suppression of
testis-specific or male sex–determining genes, such as SRY
target gene SOX9 and fibroblast growth factor 9, the latter of
which represses Wnt4 (31–34). Although SOX9 deletion is
embryonically lethal, homozygous conditional inactivation
and mutational studies of SOX9 confirmed that SOX9 in-
duces suppression of Wnt4, Foxl2, and b-catenin to prevent
feminization of the male embryo (15). In addition, SOX9
increases the expression of genes required for male phenotype
development in mammals, including steroidogenic factor1,
doublesex and mab-3 related transcription factor 1, and GATA4-
binding protein (23, 31, 35, 36).
Studies in rodents show that sexual dimorphism in immune
function occurs through embryonic development and is
maintained postnatally via the actions of gonadal hormones,
such as estrogen and testosterone (37–39). Prenatal castration
of male mice results in postpubertal thymic involution and
aberrant T cell subset differentiation (40). Females exposed to
higher concentrations of androgen prenatally as a result of
congenital adrenal hyperplasia exhibit less modeling of be-
havior shown to them by other females, suggesting that
gender-related behavior change is due to prenatal hormonal
exposure (41). Surprisingly, immune response to heterologous
MLR (MLR-A) is unaffected by perinatal masculinization in
both sexes and is strongest in unmanipulated females (42, 43).
Consistent with this, loss of feminization at prenatal stages
leads to decreased T cell/B cell ratios compared with normally
feminized mice, whose ratios mirror those observed in male
animals (44). Several studies also report the role of gonadotropin-
releasing hormone, which maintains early levels of gonadal hor-
mones in both sexes, in the prenatal patterning of the immune
system. Postnatal gonadotropin-releasing hormone antagonism,
which inhibits expression of gonadal hormones, results in lower
numbers of circulating CD8
+
T and B cells in male rodents and
primates (45). Similar studies in female rats showed reduced
numbers of CD4
+
T cells and reduced immune responses of
thymocytes and splenocytes to T cell–mediated Ag (46, 47). The
effects of estrogen and testosterone on B cell development and
differentiation also directly influence the production of IgG (48,
49). Prenatal secretion of testosterone limits the ability of males to
produce Igs compared with females, who maintain much higher
default plasma anti-DNA Ig levels compared with males (2, 50).
Additional studies in seagull chicks reveal a reduced T cell and
plasma Ig–mediated immunity in prenatal testosterone-treated
chicks compared with control chicks (51). Activational effects
of hormones at puberty further enhance sex differences in Ab
production that are present at birth (see below), leading to per-
sistently higher humoral immune response in females throughout
their lifetime.
Activational effects of hormones on immune function
Postpubertal expression of gonadal hormones leads to acute
and reversible effects in adulthood that maintain physical and
behavioral sex differences (52). Estrogen and progesterone in
females and testosterone in males are the prime gonadal
hormones secreted during the activational period. The effect
of hormonal secretion during this period is not limited to the
reproductive system; it extends to multiple tissues, including
those of the immune system (53). The androgen testosterone
is synthesized in gonadal and adrenal tissues of males and
females but is predominantly converted to estrogens via aroma-
tization in the latter (54). Endogenous estrogens produced in
female mammals include estrone, 17b-estradiol (E2), and
estriol (E3). E2 is the predominant form in females and is
produced by theca and granulosa cells of the ovaries in pre-
menopausal women. The level of hormones secreted in post-
pubertal female mammals varies in a cyclic fashion to facilitate
ovulation and subsequent pregnancy.
Estrogen receptors (ERs) exist in two forms, ERaand ERb,
which bind estrone, E2, and E3 ligands to mediate gene expres-
sion. B and T lymphocytes, mast cells, macrophages, dendritic
cells, and NK cells predominantly express ERa(55). Hema-
topoietic progenitor cells express ERaand ERb(56). Based
FIGURE 1. Mechanisms of tissue sexual dimorphism that underlie sex dif-
ferences in immune responses. Sex hormones, such as estrogens and androgens,
contribute to organizational and activational effects via gene effects that include
promoter activation and chromatin remodeling (epigenetic modifications). Sex
chromosome complement exerts its effect in promoting sexual dimorphism
independent of sex hormones. X-dosage compensation and escape from X-in-
activation influence differential gene expression of innate immune molecules. Y
chromosome contributions include Y gene–associated polymorphisms. Studies
evaluating sexual dimorphism in immune responses focus on the interdepen-
dence of these factors, as well as their independent contributions.
The Journal of Immunology 1783
by guest on March 15, 2017http://www.jimmunol.org/Downloaded from
on studies in ERa-andERb-deficient mice, ERaappears to
be the key regulator in the differentiation of hematopoietic
progenitor cells (57). Females produce higher levels of estro-
gen and regulate ER activation on their immune cells, which
helps them to exert a stronger humoral and cellular immune
response (37, 58).
ERs play vital roles in several signaling pathways, acting as a
signal transduction molecule in calcium regulation across cell
membranes and inducing activation of G coupled– and other
surface receptors, such as receptors that drive expression of
insulin growth factor 1 and activation of ERK/MAPK, pro-
tein kinase C, PI3K, and cAMP signaling (59, 60). ERais
also required for proper dendritic cell differentiation and
CD40-mediated cytokine production (61). Although ERs can im-
pact signaling pathways in ligand-dependent and -independent
manners (62), signaling in immune cells is ligand-dependent,
whereas signaling through coactivator-associated arginine methyl-
transferase 1 is ligand independent. E2 receptor [specifically
ERa46 (63)] mediates anti-inflammatory signaling in mono-
cytes and macrophages through suppression of CXCL8 (63,
64). ERs activate STAT signaling pathways during T and B cell
proliferation, maintenance, and activation. Under inflammatory
conditions, ER activation also induces NO synthase and IFN-g
expression in T cells. ER ligands also mediate phosphorylation,
nuclear localization, and transcriptional activation of STAT1,
STAT3, and STAT5 in B cells and circulating monocytes (65,
66). E2 additionally regulates STAT activity by increasing
expression of cytokine-inhibitor proteins, such as suppressor
of cytokine signaling 1 and 5, in T cells and macrophages
(67–70). In macrophages, ER signaling mediates inhibitory
responses toward proinflammatory genes regulated by NF-kB,
such as IL-6. In contrast, E2 directly suppresses the expression
of CCL2 in leukocytes, leading to a reduction in migration
(71, 72).
It is well established that ERs play an important role in
promoting sexual dimorphism in the neonatal brain through
chromatin remodeling, particularly via promoter methylation
and acetylation. This process is mediated by ERaactivation
and dimerization, followed by nuclear translocation, DNA
binding through estrogen-response elements, and recruitment
of receptor coactivators and corepressors, such as nuclear re-
ceptor corepressors, leading to epigenetic modifications and
regulation of downstream transcriptional factors (73–75).
Current studies on epigenetics also indicate that methylation
of the ERapromoter contributes to sex differences in the
brain, as well as maintains sexual dimorphism throughout life
by creating methyl marks on the DNA of the individual (76).
However, direct evidence for ER- or androgen receptor (AR)-
mediated epigenetic modifications that contribute to immune
cell differentiation and function has not been found.
In addition to endogenous ER ligands, ligands that effect ER
signaling are found in environmental sources, including food
(e.g., phytoestrogens) and pharmaceuticals (e.g., tamoxifen,
toremifene and raloxifene, which are selective ER modulators).
Selective ER modulators have been used as therapeutics in
multiple sclerosis, ovarian cancer, breast cancer, and Ebola
virus infection to suppress the activity of Th1 cells and induce
Th2 cytokine expression (77–81). Further studies are required
to understand the impact of environmental and exogenous
estrogens, either independently or in combination with other
factors, on immune function during exposure to pathogens.
Progesterone also mediates stimulatory and suppressive roles
in immune responses. Progesterone receptors are primarily
expressed by T and NK cells, but recent studies detected them
on dendritic and mesenchymal stem cells, where they suppress
Th1 cytokine secretion and increase Th2 cytokine secretion
(82, 83). Suppression of T cell cytotoxicity, as well as regu-
latory T cell (Treg) proliferation, is also mediated by pro-
gesterone (84). Progesterone also inhibits the activity of NK
cells via downregulation of IFN-gsecretion (85). In macro-
phages, progesterone suppresses NO levels and inhibits FcgR
expression and microparticle release, thereby dampening the
initial immune response at initial stages of infection. During
pregnancy, progesterone enhances immunomodulatory func-
tions of mesenchymal stem cells through upregulation of
PGE-2 and IL-6. This is essential in females to maintain the
fetal–maternal interface (83).
Most testosterone (98%) is irreversibly converted to an
active metabolite, dihydrotestosterone (86), which binds with
higher affinity than testosterone to ARs, which are expressed
at various levels by leukocytes (87). In innate immune cells,
such as neutrophils, AR signaling maintains cellular differ-
entiation via induction of G-CSF signaling through activation
of ERK1/2 and STAT3 (88). In wound-healing studies, AR
regulates the chemotactic ability of macrophages through
upregulation of CCL2, TNF-a, and CCR2 (89). AR signal-
ing also regulates T and B cell function and development.
CD4
+
thymocytes express lower levels of inducible AR,
whereas CD4
2
CD8
2
and CD8
+
thymocytes express the
highest levels (90). Although AR signaling promotes Th1-
mediated T cell immune response, it also acts as an antago-
nist to NF-kB and IFN type I signaling pathways (91).
Specifically, splenic CD4
+
and CD8
+
T cells express inducible
AR that binds to testosterone in males (92). Castration studies
revealed that, in the absence of testosterone, AR signaling
suppresses the activation of CD4
+
and CD8
+
T cells via
overproduction of IL-2 and IL-2R (47, 93). Moreover, the
absence of testosterone leads to dampening of Th1 differen-
tiation from naive CD4 T cells in autoimmune disease con-
ditions through suppression of IFN-gand IL-2 expression in
males (94, 95).
Sex chromosome complement and immune function
Sex chromosome complement arises from fundamental genetic
differences in XX and XY cells that are mediated by several
processes, such as X chromosome inactivation, X gene dosage,
and epigenetic modifications (96). To maintain X gene dosage
in the female blastocyst, one of the X chromosomes is inac-
tivated by formation of Barr bodies in individual cells (97).
Barr bodies are formed as a result of Xist gene expression and
histone modification, making one of the X chromosomes
inactive in every cell (98). This process, which is exclusive to
XX somatic cells, is random and leads to cells with active
maternal or paternal X and compensates X gene dosages in all
somatic cells (14). The gene products of X or Y gene also
facilitate epigenetic modifications on the DNA of an indi-
vidual. SRY interacts with Kruppel-associated box domain
transcriptional factor through the high mobility group box on
SRY to recruit histone-modifying enzymes, such as histone
deacetylase and heterochromatin protein 1, which remodels
chromatin (99, 100). Few studies evaluated sexual dimor-
phism in chromatin remodeling, irrespective of hormonal
1784 BRIEF REVIEWS: SEX DIFFERENCES IN VIRAL INFECTIONS
by guest on March 15, 2017http://www.jimmunol.org/Downloaded from
influence, and those that did focused on the CNS. Thus, H3
methylation is more prevalent in cortical regions of the brain
in males compared with females (101), and histone deacety-
lase 2 and 4 bound more prevalently to promoters of Esr1 and
Cyp19a during brain differentiation in males compared with
females. This increased interaction leads to stronger deacety-
lation and higher gene expression (102).
microRNAs are also sexually dimorphic in nature and
regulate SRY-related genes. For example, in the prenatal stage,
miR-124 is highly expressed in female-supporting cells and
suppresses SOX9 gene expression. In contrast, miR-202-5p/
3p is highly expressed in males through SOX9, which is
generated by male-supporting cells, and is necessary for male
sex determination. Data also suggest that miR-202-5p is a
direct transcriptional target of SOX9 during testis differenti-
ation (103). Interestingly, miR-124 also was shown to inhibit
STAT3 signaling, which suppresses T cell proliferation and
leads to Foxp3
+
Treg induction, including upregulation of
IL-2, IFN-g, and TNF-a(104, 105). Another study using a
murine model of multiple sclerosis found that peripheral
administration of miR-124 leads to systemic deactivation of
macrophages, reduced activation of myelin-specific T cells,
and suppression of disease progression (106). Because research
regarding the role of sex chromosome complement in many
physiologic and disease processes is still in its early stages, less
is known about their contributions to antiviral immunity.
However, studies using four-core genotype (FCG) mice, in
which the role of XX versus XY genes can be separated from
those of gonadal hormonal effects, are providing new insights
into the impact of sex chromosome complement on immune
responses, particularly during autoimmune diseases.
Female bias in disease expression of autoimmunity is well
established, with female/male ratios in systemic lupus ery-
thematosus (SLE) and multiple sclerosis approaching 4:1 and
9:1, respectively. A study of sex chromosome aneuploidy in
male subjects expressing an excess X chromosome found that
they were at a higher risk for SLE (107). Thus, the X chro-
mosome likely plays a crucial role in disease incidence inde-
pendently of hormonal effects (107, 108). The FCG mouse
model is a novel tool for examining the effect of sex chro-
mosome complement XX and XY on phenotypic sex differ-
ences induced in male and female mice without the
confounding effects of hormonal patterning. In the FCG
model, the testis-determining gene Sry is deleted from the Y
chromosome on male B6 or SJL mice (XY
2
), and a transgenic
Sry is inserted at multiple sites on an autosome (109). The
model generates four genotypes of mice; the Sry transgene is
present in XX and XY
2
mice, which develop testis in contrast
to XY
2
mice, which develop ovaries similar to XX mice (109).
Gonadectomy of FCG mice allows further examination of the
genetic contributions of sex chromosomes in adult animals
that do not express sex steroids.
Using FCG mice, XXSry mice were found to display in-
creased susceptibility for SLE and experimental autoimmune
encephalomyelitis compared with XY
2
Sry mice. Because both
XXSry and XY
2
Sry mice had testes during development, and
the only difference between the two groups is the presence of
the Y chromosome complement, this study confirmed that sex
complement alone could promote susceptibility to diseases,
irrespective of the hormonal secretions (52, 108). Further
comparison between XX and XY
2
mice found higher levels of
IL-13Ra2 and reduced levels of Th2 cytokines in spleen cells
isolated from XX mice. These results suggest that subdued
Th2 cytokine levels that result from an increase in X linked
gene IL-13Ra2 expression could be due to X chromosome
complement (108).
Sexually dimorphic immunity to viral infections
Differences in susceptibility to viral infections are likely due to
inherent differences in the immune system of females and
males. Females mount a stronger immune response to viral
infections compared with males as the result of more robust
humoral and cellular immune responses (Fig. 2). Clinical
studies of viral infections in humans are complicated by the
impact of nonbiological factors, such as exposure rates, social
behavior, habitat and diet, on viral pathogenesis in a sex-
specific fashion. However, studies in a controlled setting
suggested that levels of estrogen and testosterone differentially
alter the expression of genes involved in innate immunity,
such as those encoding TLRs and IFNs, in females and males,
thereby contributing to sexual dimorphism in viral infections.
Immune response to viral infections
The innate immune response is the first line of defense against
any viral infection. Males and females exhibit a different
pattern of response to viral infections. The innate response is
primarily mediated by three classes of pattern recognition
receptors (PRRs): TLRs, retinoic acid–inducible gene I–like
receptors, and nucleotide oligomerization domain–like re-
ceptors (110, 111). These PRRs detect viral components, such
as genomic DNA, dsRNA, ssRNA, RNA with 59-triphos-
phate ends, and viral proteins. TLRs and retinoic acid–
inducible gene I–like receptors specifically regulate the
production of type 1 IFNs and other cytokines. In contrast,
nucleotide oligomerization domain–like receptors regulate
FIGURE 2. The interplay of sex chromosomes, sex hormones, and immune
responses influence sex differences in virologic control. Females display in-
creased innate and adaptive immune responses to most viral infections
compared with males as a result of differences in the effects of sex hormones
(estrogen, progesterone, and androgen). X and Y chromosome complement
also contribute to sexually dimorphic immune responses to viruses in females
and males. The relative increase in immune responses in females may con-
tribute to differential levels of virologic control during acute infections and/or
immunopathologic effects of antiviral T cells that may lead to chronic
inflammation
The Journal of Immunology 1785
by guest on March 15, 2017http://www.jimmunol.org/Downloaded from
IL-1bthrough caspase-1 activation (110, 112, 113). Sexual
dimorphism is observed during antiviral responses mediated
by TLR and IFN pathways (114, 115). Immune cells in
females exhibit a 10-fold higher expression of TLRs com-
pared with males (116). In mammals, the number and ac-
tivity of innate immune cells, such as monocytes, macrophages,
and dendritic cells, are higher in females than in males. As a
result, responses to Ags, vaccines, and infections are also higher
in females compared with males (3, 117). The adaptive im-
mune response also exhibits many sexual differences in response
to viral infections. Depending on the stage of the infection,
females exhibit higher inflammatory Th1 and anti-inflammatory
Th2 compared with males (3). Additionally, upregulation of
anti-inflammatory genes and higher cytotoxic T cell activity are
observed in females. Some studies also showed higher numbers
of Tregs in females compared with males. Clinical investiga-
tions in humans also reported lower instances of CD3
+
,CD4
+
,
and CD4
+
:CD8
+
ratios in Th cells in males compared with
females (44, 118).
DNA virus members of the Herpesviridae family
Like many DNA viruses, herpesviruses replicate within host
cell nuclei, with their genome persisting as an episome for the
life of the host. In this study we will discuss sexually dimorphic
responses to herpesvirus infections that have been traced to
activational effects of gonadal hormones.
HSV-1 and HSV-2. Infection with HSVs, DNA viruses that
cause oral and genital herpes, and rare encephalitis in im-
munocompetent individuals can lead to devastating disease in
neonates and immunocompromised patients via extensive
dissemination to visceral organs and the CNS (119). In males,
the T cell–suppressive effects of androgens appear to protect
against inflammatory-mediated demyelination infected with
HSV-1 (120). Studies in mice showed that E2 treatment
increases the chances of survival and decreases vaginal pathol-
ogy and inflammation in HSV-1–infected females (121).
Consistent with this, females infected with HSV generate
higher levels of HSV-specific IgG and IgM compared with
males (122). Similarly, females infected with HSV-2 are
protected against neurologic damage and viral reactivation
via virus-specific CD8
+
T cell activation (123, 124). Studies
in ovariectomized mouse models indicated that progesterone
treatment increases the susceptibility of females to genital HSV-2
infection, whereas estrogen treatment helps to clear the infection
rapidly. Interestingly, combined treatment with estrogen and pro-
gesterone in ovariectomized mice resulted in increased infection
spread that was accompanied by persistent inflammation and
neutrophil infiltration (125, 126). Although studies do not
directly indicate the organizational effect, the effect of estrogen
and progesterone in ovariectomized animals suggests an effect of
sex hormones in HSV-2 infection.
CMV and murine CMV. CMV, a member of the Herpesviridae
family with a large genome, causes systemic viral infections
in immunocompetent individuals that may be devastating
and life-threatening in the immunocompromised (127).
Knowledge of sexual dimorphism in CMV infection stems
from experiments in murine CMV (MCMV), which is
genetically similar to CMV and has been used to study the
pathogenesis of CMV in mouse models. In MCMV-infected
female mice, IFN-a/bproduction by splenic plasmacytoid
dendritic cells (pDCs) controls viral replication and is required
to prevent viral reactivation (128). Studies in MyD88
2/2
mice
infected with MCMV showed suppression of TLR9 signaling in
neutrophils of female mice, which wasassociatedwithincreased
viral replication (129). Additionally, CD4
+
T cell–mediated
responses, including expression of TNF-a, IL-12, IL-6, and
IFN-g, which are required for clearance of CMV, are also
mediated primarily by TLR9 signaling, which is decreased in
females (130, 131).
RNA viruses
RNA viruses may replicate in either the nucleus or the cytoplasm
and, except for HIV-1, are generally cleared by host adaptive
immune responses. In this article we outline sexually dimorphic
immune responses to clinically relevant RNA viruses.
Hantavirus. Hantavirus is a negative-sense RNA virus of the
Bunyaviridae family that predominantly infects rodents.
Airborne transmission of virus occurs in humans by exposure
to rodent urine, feces, and saliva and leads to hantavirus
pulmonary syndrome, a severe respiratory disease that may be
fatal (132). Male rats infected with the Seoul virus strain
exhibit higher viral burdens in target organs and shed virus
for a longer duration than do similarly infected female
animals (16, 133, 134). Accordingly, antiviral and proinflammatory
factors, such as Tlr7,MyD88,Ifn-b,TNF-a,andCcl5, are more
highly expressed in female rodents (16, 135). Consistent with
this, acute infection with Puumala virus strain in humans is
associated with higher concentrations of IL-9 and GM-CSF
in females compared with males (136, 137).
CVB3. Coxsackieviruses (genus Enterovirus) belong to the
Picornaviridae family of positive-sense ssRNA viruses. CVB3
infection leads to myocarditis during the acute and chronic
phases, which affects more males than females, with double
the mortality in infected individuals under the age of 40
(138). Cardiomyocytes are directly infected by CVB3 during
the acute phase, which is followed by a chronic phase with a
prolonged T cell–mediated immune response and persistence of
CVB3 within the heart (139). Coxsackievirus AR is required for
CVB3 entry into cardiomyocytes (140). Chronic-phase CVB3
myocarditis is an autoimmune disease that requires Th17 cells
and whose differentiation and expression of IL-17 are suppressed
by estrogen, making females less susceptible (138, 141). In
contrast, lack of estrogen and the presence of testosterone
induce Th17 cell differentiation in CVB3-infected males,
enhancing autoimmune-mediated cardiac damage. In females,
Th2- and Treg-mediated immune responses, which are
increased through ERasignaling in T cells and macrophages
in heart tissues, suppress CVB3-mediated immunopathology
while clearing infection (139, 142). A recent study also
indicates that sex chromosome complement plays a significant
role in survival from CVB3 infection. Survival of CVB3-infected
B6-ChrY consomic male mice was exclusively dependent on the
Y
CVB3
loci on chromosome Y and independent of prenatal or
adult testosterone (24). In summary, the immune make-up of
the males and the presence of male sex steroid testosterone
promote the spread of CVB3, leading to myocarditis. Sex
complement, as well as the activational effect of hormones,
contributes to CVB3 pathogenesis.
Influenza. The incidences of H5N1 avian influenza and H1N1
and H2N2 pandemic influenza, RNA viruses that cause severe,
inflammatory-induced respiratory diseases, all demonstrate
1786 BRIEF REVIEWS: SEX DIFFERENCES IN VIRAL INFECTIONS
by guest on March 15, 2017http://www.jimmunol.org/Downloaded from
significant sexual dimorphism, affecting more females than
males (143, 144). Reports on H1N1 pandemic influenza
indicated higher mortality in females of reproductive age
(20–49 y), implicating roles for female gonadal hormones,
especially during pregnancy (145). In murine studies,
H1N1 infection in female mice prolongs the diestrus cycle,
leading to lower serum levels of E2 (5, 145, 146). This
reduction in E2 significantly increases the expression of
inflammatory cytokines and chemokines, including TNF-a,
IFN-g, IL-6, and CCL2, the latter of which promoted the
influx of mononuclear cells into virally infected lungs and
exacerbated immune responses (5, 147). Under chronic
infection, low levels of E2 bind to ER-aand modulate
NF-kB transcriptional activity, further contributing to augmented
inflammatory responses and immunopathology (114, 145).
In contrast, lower levels of estrogen and higher levels of
androgen in influenza-infected males were linked to immuno-
suppression and reduced T cell counts, which may limit
immunopathology in males (148). Clearly, more studies
emphasizing the interplay among sex, hormones, and genes
are required to further understand the effect of sexual
dimorphism on influenza pathogenesis.
HIV-1. Untreated HIV-1 infection leads to AIDS, with near-
complete loss of CD4
+
T cells. Meta-analysis data show 41%
less HIV RNA in women compared with men following
primary infection, which gradually increase to a higher viral
load set point compared with males after chronic infection
(149–151). In females, HIV-1 promotes type 1 IFN production
by pDCs via TLR7 signaling, which itself is increased
downstream of E3 signaling (115, 152). These effects lead to
strong, initial cellular responses that limit HIV-1 replication
(153). However, continuous production of type 1 IFN by
pDCs in females leads to chronic T cell activation with CCR5
expression, providing more targets for HIV-1 (154). In HIV-1–
infected women, pDCs produce more IFN-athrough TLR7
ligand activation. This results in a secondary activation of
CD8
+
T cells (115). Follow-up studies showed that, even with
the same viral load, pDCs ob taine d fr om f emale s showed
higher CD8
+
T cell activation compared with men (115).
Additionally, females mount a stronger B and T cell activation
following HIV-1 infection as a result of the higher baseline
count of CD4
+
T and CD8
+
T cells compared with men
(115, 151). The concentration of estrogen-binding SHBG is
increased in HIV-infected males. Although the mechanism
is not well understood, suppressing SHBG levels decreases
the severity of HIV infection in male patients (155, 156).
Another recent study in SCID mice showed that ERs ESR1
and ESR2 and X chromosome complement in females are
necessary for IFN-aand TNF-aproduction under TLR7
activation in pDCs (157). This study confirmed that
estrogen, female sex hormones, and X chromosome dosage
confer stronger innate and adaptive immune responses in
females compared with males in HIV infection. Altogether,
these studies suggest that estradiol treatment could facilitate a
stronger immune response to HIV infection.
Although sexual dimorphism is observed in HIV patho-
genesis, studies are needed to better understand the underlying
mechanisms that contribute to sex-based immune cell regu-
lation in HIV-1–infected patients. These will assist in the
design of experiments and the accuracy of clinical trials in
HIV-1–infected populations.
Vaccine for viral infections
A sexually dimorphic response to viral vaccines is observed in
males and females. Viral vaccines against hepatitis A, hepatitis
B, and HSV-2 all exhibit a stronger side effect in young females
following immunization. Higher IgA and IgG Ab responses, as
well as higher transcriptional activation of genes important in
immune cell signaling, such as IFN and TLR7, are also ob-
served in females compared with males (143, 158). These
studies clearly demonstrate that the development of vaccines
against viral infection should carefully consider the effect of
differential immune responses in males and females.
Conclusions
This review highlights early and recent findings regarding the
impact of sex on immune cell numbers and function, with a
focus on sexually dimorphic phenotypes during viral infec-
tions. Although still an emerging field, it is clear that mech-
anisms by which mammals achieve and maintain sex
differences can directly and indirectly influence host–path-
ogen interactions at the cellular and molecular levels. These
findings support the notion that there are, in fact, “two
normals” with regard to manifestations of infectious diseases,
with sex-specific responses during acute virologic control and
in immunopathologic manifestations of viral infections.
Accordingly, the development of therapeutics to treat vari-
ousphasesofviralinfectionswill require continued study
and comparisons of viral pathogenesis in female and male
animals and humans.
Disclosures
The authors have no financial conflicts of interest.
References
1. Marriott, I., and Y. M. Huet-Hudson. 2006. Sexual dimorphism in innate im-
mune responses to infectious organisms. Immunol. Res. 34: 177–192.
2. Butterworth, M., B. McClellan, and M. Allansmith. 1967. Influence of sex in
immunoglobulin levels. Nature 214: 1224–1225.
3. Klein, S. L., and K. L. Flanagan. 2016. Sex differences in immune responses. Nat.
Rev. Immunol. 16: 626–638.
4. Klein, S. L. 2012. Sex influences immune responses to viruses, and efficacy of
prophylaxis and treatments for viral diseases. BioEssays 34: 1050–1059.
5. Larcombe, A. N., R. E. Foong, E. M. Bozanich, L. J. Berry, L. W. Garratt,
R. C. Gualano, J. E. Jones, L. F. Dousha, G. R. Zosky, and P. D. Sly. 2011. Sexual
dimorphism in lung function responses to acute influenza A infection. Influenza
Other Respi. Viruses 5: 334–342.
6. Molina, V., and Y. Shoenfeld. 2005. Infection, vaccines and other environmental
triggers of autoimmunity. Autoimmunity 38: 235–245.
7. Lamason, R., P. Zhao, R. Rawat, A. Davis, J. C. Hall, J. J. Chae, R. Agarwal,
P. Cohen, A. Rosen, E. P. Hoffman, and K. Nagaraju. 2006. Sexual dimorphism
in immune response genes as a function of puberty. BMC Immunol. 7: 2.
8. Luppi, P., C. Haluszczak, D. Betters, C. A. Richard, M. Trucco, and J. A. DeLoia.
2002. Monocytes are progressively activated in the circulation of pregnant women.
J. Leukoc. Biol. 72: 874–884.
9. Kaushic, C., K. L. Roth, V. Anipindi, and F. Xiu. 2011. Increased prevalence of
sexually transmitted viral infections in women: the role of female sex hormones in
regulating susceptibility and immune responses. J. Reprod. Immunol. 88: 204–209.
10. Oertelt-Prigione, S. 2012. The influence of sex and gender on the immune re-
sponse. Autoimmun. Rev. 11: A479–A485.
11. Klein, S. L., I. Marriott, and E. N. Fish. 2015. Sex-based differences in immune
function and responses to vaccination. Trans. R. Soc. Trop. Med. Hyg. 109: 9–15.
12. Fischer, J., N. Jung, N. Robinson, and C. Lehmann. 2015. Sex differences in
immune responses to infectious diseases. Infection 43: 399–403.
13. Wang, J., C. M. Syrett, M. C. Kramer, A. Basu, M. L. Atchison, and
M. C. Anguera. 2016. Unusual maintenance of X chromosome inactivation pre-
disposes female lymphocytes for increased expression from the inactive X. Proc.
Natl. Acad. Sci. USA 113: E2029–E2038.
14. Dixon-McDougall, T., and C. Brown. 2016. The making of a Barr body: the
mosaic of factors that eXIST on the mammalian inactive X chromosome. Biochem.
Cell Biol. 94: 56–70.
15. Barrionuevo, F., S. Bagheri-Fam, J. Klattig, R. Kist, M. M. Taketo, C. Englert,
and G. Scherer. 2006. Homozygous inactivation of Sox9 causes complete XY sex
reversal in mice. Biol. Reprod. 74: 195–201.
The Journal of Immunology 1787
by guest on March 15, 2017http://www.jimmunol.org/Downloaded from
16. Hannah, M. F., V. B. Bajic, and S. L. Klein. 2008. Sex differences in the recog-
nition of and innate antiviral responses to Seoul virus in Norway rats. Brain Behav.
Immun. 22: 503–516.
17. Geurs, T. L., E. B. Hill, D. M. Lippold, and A. R. French. 2012. Sex differences in
murine susceptibility to systemic viral infections. J. Autoimmun. 38: J245–J253.
18. Hagen, S., and M. Altfeld. 2016. The X awakens: multifactorial ramifications of
sex-specific differences in HIV-1 infection. J Virus Erad 2: 78–81.
19. Fish, E. N. 2008. The X-files in immunity: sex-based differences predispose im-
mune responses. Nat. Rev. Immunol. 8: 737–744.
20. Peretz, J., A. Pekosz, A. P. Lane, and S. L. Klein. 2016. Estrogenic compounds
reduce influenza A virus replication in primary human nasal epithelial cells derived
from female, but not male, donors. Am. J. Physiol. Lung Cell. Mol. Physiol. 310:
L415–L425.
21. Bermejo-Alvarez, P., D. Rizos, P. Lonergan, and A. Gutierrez-Adan. 2011.
Transcriptional sexual dimorphism during preimplantation embryo development
and its consequences for developmental competence and adult health and disease.
Reproduction 141: 563–570.
22. Lowe, R., C. Gemma, V. K. Rakyan, and M. L. Holland. 2015. Sexually di-
morphic gene expression emerges with embryonic genome activation and is dy-
namic throughout development. BMC Genomics 16: 295.
23. Sekido, R., and R. Lovell-Badge. 2008. Sex determination involves synergistic
action of SRY and SF1 on a specific Sox9 enhancer. Nature 453: 930–934.
24. Case, L. K., L. Toussaint, M. Moussawi, B. Roberts, N. Saligrama, L. Brossay,
S. A. Huber, and C. Teuscher. 2012. Chromosome y regulates survival following
murine coxsackievirus b3 infection. G3 2: 115–121.
25. Makiyan, Z. 2016. Studies of gonadal sex differentiation. Organogenesis 12: 42–51.
26. Durando, M., L. Cocito, H. A. Rodrı
´guez, J. Varayoud, J. G. Ramos,
E. H. Luque, and M. Mun
˜oz-de-Toro. 2013. Neonatal expression of amh, sox9
and sf-1 mRNA in Caiman latirostris and effects of in ovo exposure to endocrine
disrupting chemicals. Gen. Comp. Endocrinol. 191: 31–38.
27. Lavery,R.,A.A.Chassot,E.Pauper,E.P.Gregoire,M.Klopfenstein,D.G.deRooij,
M. Mark, A. Schedl, N. B. Ghyselinck, and M. C. Chaboissier. 2012. Testicular
differentiation occurs in absence of R-spondin1 and Sox9 in mouse sex reversals. PLoS
Genet. 8: e1003170.
28. Tomizuka, K., K. Horikoshi, R. Kitada, Y. Sugawara, Y. Iba, A. Kojima,
A. Yoshitome, K. Yamawaki, M. Amagai, A. Inoue, et al. 2008. R-spondin1 plays
an essential role in ovarian development through positively regulating Wnt-4
signaling. Hum. Mol. Genet. 17: 1278–1291.
29. Chassot, A. A., F. Ranc, E. P. Gregoire, H. L. Roepers-Gajadien, M. M. Taketo,
G. Camerino, D. G. de Rooij, A. Schedl, and M. C. Chaboissier. 2008. Activation
of beta-catenin signaling by Rspo1 controls differentiation of the mammalian
ovary. Hum. Mol. Genet. 17: 1264–1277.
30. Smith, C. A., C. M. Shoemaker, K. N. Roeszler, J. Queen, D. Crews, and
A. H. Sinclair. 2008. Cloning and expression of R-Spondin1 in different verte-
brates suggests a conserved role in ovarian development. BMC Dev. Biol. 8: 72.
31. Kim, Y., A. Kobayashi, R. Sekido, L. DiNapoli, J. Brennan, M. C. Chaboissier,
F. Poulat, R. R. Behringer, R. Lovell-Badge, and B. Capel. 2006. Fgf9 and Wnt4
act as antagonistic signals to regulate mammalian sex determination. PLoS Biol. 4:
e187.
32. Schmidt, D., C. E. Ovitt, K. Anlag, S. Fehsenfeld, L. Gredsted, A. C. Treier, and
M. Treier. 2004. The murine winged-helix transcription factor Foxl2 is required for
granulosa cell differentiation and ovary maintenance. Development 131: 933–942.
33. Ottolenghi, C., S. Omari, J. E. Garcia-Ortiz, M. Uda, L. Crisponi, A. Forabosco,
G. Pilia, and D. Schlessinger. 2005. Foxl2 is required for commitment to ovary
differentiation. Hum. Mol. Genet. 14: 2053–2062.
34. Uhlenhaut, N. H., S. Jakob, K. Anlag, T. Eisenberger, R. Sekido, J. Kress,
A. C. Treier, C. Klugmann, C. Klasen, N. I. Holter, et al. 2009. Somatic sex
reprogramming of adult ovaries to testes by FOXL2 ablation. Cell 139: 1130–1142.
35. Kashimada, K., T. Svingen, C. W. Feng, E. Pelosi, S. Bagheri-Fam, V. R. Harley,
D. Schlessinger, J. Bowles, and P. Koopman. 2011. Antagonistic regulation of
Cyp26b1 by transcription factors SOX9/SF1 and FOXL2 during gonadal devel-
opment in mice. FASEB J. 25: 3561–3569.
36. Matson, C. K., M. W. Murphy, A. L. Sarver, M. D. Griswold, V. J. Bardwell, and
D. Zarkower. 2011. DMRT1 prevents female reprogramming in the postnatal
mammalian testis. Nature 476: 101–104.
37. Kovats, S. 2015. Estrogen receptors regulate innate immune cells and signaling
pathways. Cell. Immunol. 294: 63–69.
38. Tejera-Alhambra, M., B. Alonso, R. Teijeiro, R. Ramos-Medina, C. Aristimun
˜o,
L. Valor, C. de Andre
´s, and S. Sa
´nchez-Ramo
´n. 2012. Perforin expression by CD4+
regulatory T cells increases at multiple sclerosis relapse: sex differences. Int. J. Mol. Sci.
13: 6698–6710.
39. McCarthy, M. M., and A. P. Arnold. 2011. Reframing sexual differentiation of the
brain. Nat. Neurosci. 14: 677–683.
40. Azad, N., N. LaPaglia, L. Agrawal, J. Steiner, S. Uddin, D. W. Williams,
A. M. Lawrence, and N. V. Emanuele. 1998. The role of gonadectomy and tes-
tosterone replacement on thymic luteinizing hormone-releasing hormone pro-
duction. J. Endocrinol. 158: 229–235.
41. Hines, M., V. Pasterski, D. Spencer, S. Neufeld, P. Patalay, P. C. Hindmarsh,
I. A. Hughes, and C. L. Acerini. 2016. Prenatal androgen exposure alters girls’
responses to information indicating gender-appropriate behaviour. Philos. Trans.
R. Soc. Lond. B Biol. Sci. 371: 20150125.
42. Konstadoulakis, M. M., K. N. Syrigos, C. N. Baxevanis, E. I. Syrigou,
M. Papamichail, P. Peveretos, M. Anapliotou, and B. C. Golematis. 1995. Effect
of testosterone administration, pre- and postnatally, on the immune system of rats.
Horm. Metab. Res. 27: 275–278.
43. Weinstein, Y., S. Ran, and S. Segal. 1984. Sex-associated differences in the regulation of
immune responses controlled by the MHC of the mouse. J. Immunol. 132: 656–661.
44. Amadori, A., R. Zamarchi, G. De Silvestro, G. Forza, G. Cavatton, G. A. Danieli,
M. Clementi, and L. Chieco-Bianchi. 1995. Genetic control of the CD4/CD8
T-cell ratio in humans. Nat. Med. 1: 1279–1283.
45. Gould, K. G., M. A. Akinbami, and D. R. Mann. 1998. Effect of neonatal
treatment with a gonadotropin releasing hormone antagonist on developmental
changes in circulating lymphocyte subsets: a longitudinal study in male rhesus
monkeys. Dev. Comp. Immunol. 22: 457–467.
46. Mann, D. R., and H. M. Fraser. 1996. The neonatal period: a critical interval in
male primate development. J. Endocrinol. 149: 191–197.
47. Page, S. T., S. R. Plymate, W. J. Bremner, A. M. Matsumoto, D. L. Hess,
D. W. Lin, J. K. Amory, P. S. Nelson, and J. D. Wu. 2006. Effect of medical
castration on CD4+ CD25+ T cells, CD8+ T cell IFN-gamma expression, and NK
cells: a physiological role for testosterone and/or its metabolites. Am. J. Physiol.
Endocrinol. Metab. 290: E856–E863.
48. Paavonen, T., L. C. Andersson, and H. Adlercreutz. 1981. Sex hormone regulation
of in vitro immune response. Estradiol enhances human B cell maturation via
inhibition of suppressor T cells in pokeweed mitogen-stimulated cultures. J. Exp.
Med. 154: 1935–1945.
49. Sthoeger, Z. M., N. Chiorazzi, and R. G. Lahita. 1988. Regulation of the immune
response by sex hormones. I. In vitro effects of estradiol and testosterone on
pokeweed mitogen-induced human B cell differentiation. J. Immunol. 141: 91–98.
50. Ainbender, E., R. B. Weisinger, M. Hevizy, and H. L. Hodes. 1968. Difference in
the immunoglobulin class of polioantibody in the serum of men and women. J.
Immunol. 101: 92–98.
51. Muller, W., T. G. Groothuis, A. Kasprzik, C. Dijkstra, R. V. Alatalo, and
H. Siitari. 2005. Prenatal androgen exposure modulates cellular and humoral
immune function of black-headed gull chicks. Proc. Biol. Sci. 272: 1971–1977.
52. Arnold, A. P. 2009. The organizational-activational hypothesis as the foundation
for a unified theory of sexual differentiation of all mammalian tissues. Horm.
Behav. 55: 570–578.
53. Gaumond, I., P. Arsenault, and S. Marchand. 2002. The role of sex hormones on
formalin-induced nociceptive responses. Brain Res. 958: 139–145.
54. Lai, J. J., K. P. Lai, W. Zeng, K. H. Chuang, S. Altuwaijri, and C. Chang. 2012.
Androgen receptor influences on body defense system via modulation of innate and
adaptive immune systems: lessons from conditional AR knockout mice. Am. J.
Pathol. 181: 1504–1512.
55. Nilsson, S., S. Ma
¨kela
¨, E. Treuter, M. Tujague, J. Thomsen, G. Andersson,
E. Enmark, K. Pettersson, M. Warner, and J. A. Gustafsson. 2001. Mechanisms of
estrogen action. Physiol. Rev. 81: 1535–1565.
56. Phiel, K. L., R. A. Henderson, S. J. Adelman, and M. M. Elloso. 2005. Differential
estrogen receptor gene expression in human peripheral blood mononuclear cell
populations. Immunol. Lett. 97: 107–113.
57. Nakada, D., H. Oguro, B. P. Levi, N. Ryan, A. Kitano, Y. Saitoh, M. Takeichi,
G. R. Wendt, and S. J. Morrison. 2014. Oestrogen increases haematopoietic stem-
cell self-renewal in females and during pregnancy. Nature 505: 555–558.
58. Bird, M. D., J. Karavitis, and E. J. Kovacs. 2008. Sex differences and estrogen
modulation of the cellular immune response afterinjury. Cell. Immunol. 252: 57–67.
59. Zhou, T., Z. Yang, Y. Chen, Y. Chen, Z. Huang, B. You, Y. Peng, and J. Chen.
2016. Estrogen accelerates cutaneous wound healing by promoting proliferation of
epidermal keratinocytes via Erk/Akt signaling pathway. Cell. Physiol. Biochem. 38:
959–968.
60. Xie, H., C. Hua, L. Sun, X. Zhao, H. Fan, H. Dou, L. Sun, and Y. Hou. 2011.
17b-estradiol induces CD40 expression in dendritic cells via MAPK signaling
pathways in a minichromosome maintenance protein 6-dependent manner. Ar-
thritis Rheum. 63: 2425–2435.
61. Douin-Echinard, V., S. Laffont, C. Seillet, L. Delpy, A. Krust, P. Chambon,
P. Gourdy, J. F. Arnal, and J. C. Gue
´ry. 2008. Estrogen receptor alpha, but not
beta, is required for optimal dendritic cell differentiation and [corrected] CD40-
induced cytokine production. [Published erratum appears in 2008 J. Immunol.
180: 7047.] J. Immunol. 180: 3661–3669.
62. Carascossa, S., P. Dudek, B. Cenni, P. A. Briand, and D. Picard. 2010. CARM1
mediates the ligand-independent and tamoxifen-resistant activation of the estrogen
receptor alpha by cAMP. Genes Dev. 24: 708–719.
63. Murphy, A. J., P. M. Guyre, C. R. Wira, and P. A. Pioli. 2009. Estradiol regulates
expression of estrogen receptor ERalpha46 in human macrophages. PLoS One 4:
e5539.
64. Lasarte, S., D. Elsner, T. Sanchez-Elsner, A. Fernandez-Pineda, L. A. Lo
´pez-
Ferna
´ndez, A. L. Corbı
´, M. A. Mun
˜oz-Fernandez, and M. Relloso. 2013. Estradiol
downregulates NF-kb translocation by Ikbkg transcriptional repression in den-
dritic cells. Genes Immun. 14: 462–469.
65. Ho, H. H., and L. B. Ivashkiv. 2006. Role of STAT3 in type I interferon re-
sponses. Negative regulation of STAT1-dependent inflammatory gene activation.
J. Biol. Chem. 281: 14111–14118.
66. Kuchipudi, S. V. 2015. The complex role of STAT3 in viral infections. J.
Immunol. Res. 2015: 272359. Available at: http://dx.doi.org/10.1155/2015/
272359.
67. Alexander, W. S., R. Starr, J. E. Fenner, C. L. Scott, E. Handman, N. S. Sprigg,
J. E. Corbin, A. L. Cornish, R. Darwiche, C. M. Owczarek, et al. 1999. SOCS1 is
a critical inhibitor of interferon gamma signaling and prevents the potentially fatal
neonatal actions of this cytokine. Cell 98: 597–608.
68. Kinjyo, I., T. Hanada, K. Inagaki-Ohara, H. Mori, D. Aki, M. Ohishi,
H. Yoshida, M. Kubo, and A. Yoshimura. 2002. SOCS1/JAB is a negative regu-
lator of LPS-induced macrophage activation. Immunity 17: 583–591.
69. Cornish, A. L., G. M. Davey, D. Metcalf, J. F. Purton, J. E. Corbin,
C. J. Greenhalgh, R. Darwiche, L. Wu, N. A. Nicola, D. I. Godfrey, et al. 2003.
Suppressor of cytokine signaling-1 has IFN-gamma-independent actions in T cell
homeostasis. J. Immunol. 170: 878–886.
1788 BRIEF REVIEWS: SEX DIFFERENCES IN VIRAL INFECTIONS
by guest on March 15, 2017http://www.jimmunol.org/Downloaded from
70. Brender, C., R. Columbus, D. Metcalf, E. Handman, R. Starr, N. Huntington,
D. Tarlinton, N. Ødum, S. E. Nicholson, N. A. Nicola, et al. 2004. SOCS5 is
expressed in primary B and T lymphoid cells but is dispensable for lymphocyte
production and function. Mol. Cell. Biol. 24: 6094–6103.
71. Pan, T., L. Zhong, S. Wu, Y. Cao, Q. Yang, Z. Cai, X. Cai, W. Zhao, N. Ma,
W. Zhang, et al. 2016. 17b-Oestradiol enhances the expansion and activation of
myeloid-derived suppressor cells via signal transducer and activator of tran-
scription (STAT)-3 signaling in human pregnancy. Clin. Exp. Immunol. 185:
86–97.
72. Kawana, K., Y. Kawana, and D. J. Schust. 2005. Female steroid hormones use
signal transducers and activators of transcription protein-mediated pathways to
modulate the expression of T-bet in epithelial cells: a mechanism for local immune
regulation in the human reproductive tract. Mol. Endocrinol. 19: 2047–2059.
73. Schwarz, J. M., B. M. Nugent, and M. M. McCarthy. 2010. Developmental and
hormone-induced epigenetic changes to estrogen and progesterone receptor genes
in brain are dynamic across the life span. Endocrinology 151: 4871–4881.
74. Pedram, A., M. Razandi, M. Lewis, S. Hammes, and E. R. Levin. 2014.
Membrane-localized estrogen receptor ais required for normal organ development
and function. Dev. Cell 29: 482–490.
75. Zhang, X., K. Tanaka, J. Yan, J. Li, D. Peng, Y. Jiang, Z. Yang, M. C. Barton,
H. Wen, and X. Shi. 2013. Regulation of estrogen receptor aby histone
methyltransferase SMYD2-mediated protein methylation. Proc. Natl. Acad. Sci.
USA 110: 17284–17289.
76. Edelmann, M. N., and A. P. Auger. 2011. Epigenetic impact of simulated ma-
ternal grooming on estrogen receptor alpha within the developing amygdala. Brain
Behav. Immun. 25: 1299–1304.
77. Banchs, H. L., V. Gonza
´lez, I. Gonza
´lez Cancel, C. Quintana, R. Caldero
´n, and
P. I. Altieri. 2005. Heart transplantation in females: the experience in Puerto Rico.
Bol. Asoc. Med. P. R. 97: 248–256.
78. Nalbandian, G., V. Paharkova-Vatchkova, A. Mao, S. Nale, and S. Kovats. 2005.
The selective estrogen receptor modulators, tamoxifen and raloxifene, impair
dendritic cell differentiation and activation. J. Immunol. 175: 2666–2675.
79. Marino, M., P. Galluzzo, and P. Ascenzi. 2006. Estrogen signaling multiple
pathways to impact gene transcription. Curr. Genomics 7: 497–508.
80. Johansen, L. M., J. M. Brannan, S. E. Delos, C. J. Shoemaker, A. Stossel, C. Lear,
B. G. Hoffstrom, L. E. Dewald, K. L. Schornberg, C. Scully, et al. 2013. FDA-
approved selective estrogen receptor modulators inhibit Ebola virus infection. Sci.
Transl. Med. 5: 190ra79.
81. Wisdom, A. J., Y. Cao, N. Itoh, R. D. Spence, and R. R. Voskuhl. 2013. Estrogen
receptor-bligand treatment after disease onset is neuroprotective in the multiple
sclerosis model. J. Neurosci. Res. 91: 901–908.
82. Butts, C. L., S. A. Shukair, K. M. Duncan, E. Bowers, C. Horn, E. Belyavskaya,
L. Tonelli, and E. M. Sternberg. 2007. Progesterone inhibits mature rat dendritic
cells in a receptor-mediated fashion. Int. Immunol. 19: 287–296.
83. Zhao, X., L. Liu, D. Liu, H. Fan, Y. Wang, Y. Hu, and Y. Hou. 2012. Proges-
terone enhances immunoregulatory activity of human mesenchymal stem cells via
PGE2 and IL-6. Am. J. Reprod. Immunol. 68: 290–300.
84. Enninga, E. A., S. G. Holtan, D. J. Creedon, R. S. Dronca, W. K. Nevala,
S. Ognjanovic, and S. N. Markovic. 2014. Immunomodulatory effects of sex
hormones: requirements for pregnancy and relevance in melanoma. Mayo Clin.
Proc. 89: 520–535.
85. Arruvito, L., S. Giulianelli, A. C. Flores, N. Paladino, M. Barboza, C. Lanari, and
L. Fainboim. 2008. NK cells expressing a progesterone receptor are susceptible to
progesterone-induced apoptosis. J. Immunol. 180: 5746–5753.
86. Llewelyn, D. E., G. F. Read, and S. G. Hillier. 1975. Proceedings: novel procedure
for the simultaneous determination of testosterone and 5alpha-dihydrotestosterone
concentrations in unpurified plasma extracts by radioimmunoassay. J. Endocrinol.
67: 7P–8P.
87. Penning, T. M., D. R. Bauman, Y. Jin, and T. L. Rizner. 2007. Identification of
the molecular switch that regulates access of 5alpha-DHT to the androgen re-
ceptor. Mol. Cell. Endocrinol. 265–266: 77–82.
88. Chuang, K. H., S. Altuwaijri, G. Li, J. J. Lai, C. Y. Chu, K. P. Lai, H. Y. Lin,
J. W. Hsu, P. Keng, M. C. Wu, and C. Chang. 2009. Neutropenia with impaired
host defense against microbial infection in mice lacking androgen receptor. J. Exp.
Med. 206: 1181–1199.
89. Lai, J. J., K. P. Lai, K. H. Chuang, P. Chang, I. C. Yu, W. J. Lin, and C. Chang.
2009. Monocyte/macrophage androgen receptor suppresses cutaneous wound
healing in mice by enhancing local TNF-alpha expression. J. Clin. Invest. 119:
3739–3751.
90. Olsen, N. J., and W. J. Kovacs. 2001. Effects of androgens on T and B lymphocyte
development. Immunol. Res. 23: 281–288.
91. Kissick, H. T., M. G. Sanda, L. K. Dunn, K. L. Pellegrini, S. T. On, J. K. Noel,
and M. S. Arredouani. 2014. Androgens alter T-cell immunity by inhibiting T-
helper 1 differentiation. Proc. Natl. Acad. Sci. USA 111: 9887–9892.
92. Benten, W. P., A. Becker, H. P. Schmitt-Wrede, and F. Wunderlich. 2002. De-
velopmental regulation of intracellular and surface androgen receptors in T cells.
Steroids 67: 925–931.
93. Lanini, S., A. Nanni Costa, P. A. Grossi, F. Procaccio, A. Ricci,
M. R. Capobianchi, N. A. Terrault, and G. Ippolito. 2016. Liver transplant re-
cipients and prioritization of anti-HCV therapy: an Italian cohort analysis. Liver
Int. 36: 410–417.
94. Bao, M., Y. Yang, H. S. Jun, and J. W. Yoon. 2002. Molecular mechanisms for
gender differences in susceptibility to T cell-mediated autoimmune diabetes in
nonobese diabetic mice. J. Immunol. 168: 5369–5375.
95. Bebo, B. F., Jr., J. C. Schuster, A. A. Vandenbark, and H. Offner. 1999. An-
drogens alter the cytokine profile and reduce encephalitogenicity of myelin-reactive
T cells. J. Immunol. 162: 35–40.
96. Arnold, A. P., X. Chen, and Y. Itoh. 2012. What a difference an X or Y makes: sex
chromosomes, gene dose, and epigenetics in sexual differentiation. Handb. Exp.
Pharmacol. 214: 67–88.
97. Ercan, S. 2015. Mechanisms of x chromosome dosage compensation. J Genomics 3:
1–19.
98. Simon, M. D., S. F. Pinter, R. Fang, K. Sarma, M. Rutenberg-Schoenberg,
S. K. Bowman, B. A. Kesner, V. K. Maier, R. E. Kingston, and J. T. Lee. 2013.
High-resolution Xist binding maps reveal two-step spreading during X-
chromosome inactivation. Nature 504: 465–469.
99. Prokop, J. W., F. J. Rauscher III, H. Peng, Y. Liu, F. C. Araujo, I. Watanabe,
F. M. Reis, and A. Milsted. 2014. MAS promoter regulation: a role for Sry and
tyrosine nitration of the KRAB domain of ZNF274 as a feedback mechanism.
Clin. Sci. 126: 727–738.
100. Oh, H. J., Y. Li, and Y. F. Lau. 2005. Sry associates with the heterochromatin
protein 1 complex by interacting with a KRAB domain protein. Biol. Reprod. 72:
407–415.
101. Tsai, H. W., P. A. Grant, and E. F. Rissman. 2009. Sex differences in histone
modifications in the neonatal mouse brain. Epigenetics 4: 47–53.
102. Matsuda, K. I., H. Mori, B. M. Nugent, D. W. Pfaff, M. M. McCarthy, and
M. Kawata. 2011. Histone deacetylation during brain development is essential for
permanent masculinization of sexual behavior. Endocrinology 152: 2760–2767.
103. Wainwright, E. N., J. S. Jorgensen, Y. Kim, V. Truong, S. Bagheri-Fam,
T. Davidson, T. Svingen, S. L. Fernandez-Valverde, K. S. McClelland, R. J. Taft,
et al. 2013. SOX9 regulates microRNA miR-202-5p/3p expression during mouse
testis differentiation. Biol. Reprod. 89: 34.
104. Wei, J., F. Wang, L. Y. Kong, S. Xu, T. Doucette, S. D. Ferguson, Y. Yang,
K. McEnery, K. Jethwa, O. Gjyshi, et al. 2013. miR-124 inhibits STAT3 signaling
to enhance T cell-mediated immune clearance of glioma. Cancer Res. 73: 3913–
3926.
105. Jiang, S., C. Li, G. McRae, E. Lykken, J. Sevilla, S. Q. Liu, Y. Wan, and Q. J. Li.
2014. MeCP2 reinforces STAT3 signaling and the generation of effector CD4+
T cells by promoting miR-124-mediated suppression of SOCS5. Sci. Signal. 7: ra25.
106. Ponomarev, E. D., T. Veremeyko, N. Barteneva, A. M. Krichevsky, and
H. L. Weiner. 2011. MicroRNA-124 promotes microglia quiescence and sup-
presses EAE by deactivating macrophages via the C/EBP-a-PU.1 pathway. Nat.
Med. 17: 64–70.
107. Dillon, S. P., B. T. Kurien, S. Li, G. R. Bruner, K. M. Kaufman, J. B. Harley,
P. M. Gaffney, D. J. Wallace, M. H. Weisman, and R. H. Scofield. 2012. Sex
chromosome aneuploidies among men with systemic lupus erythematosus. J.
Autoimmun. 38: J129–J134.
108. Smith-Bouvier, D. L., A. A. Divekar, M. Sasidhar, S. Du, S. K. Tiwari-Woodruff,
J. K. King, A. P. Arnold, R. R. Singh, and R. R. Voskuhl. 2008. A role for sex
chromosome complement in the female bias in autoimmune disease. J. Exp. Med.
205: 1099–1108.
109. Itoh, Y., R. Mackie, K. Kampf, S. Domadia, J. D. Brown, R. O’Neill, and
A. P. Arnold. 2015. Four core genotypes mouse model: localization of the Sry
transgene and bioassay for testicular hormone levels. BMC Res. Notes 8: 69.
110. Medzhitov, R. 2007. Recognition of microorganisms and activation of the im-
mune response. Nature 449: 819–826.
111. Kanneganti, T. D., M. Lamkanfi, and G. Nu
´n
˜ez. 2007. Intracellular NOD-like
receptors in host defense and disease. Immunity 27: 549–559.
112. Akira, S., S. Uematsu, and O. Takeuchi. 2006. Pathogen recognition and innate
immunity. Cell 124: 783–801.
113. Cao, X. 2016. Self-regulation and cross-regulation of pattern-recognition receptor
signalling in health and disease. Nat. Rev. Immunol. 16: 35–50.
114. Robinson, D. P., M. E. Lorenzo, W. Jian, and S. L. Klein. 2011. Elevated 17b-
estradiol protects females from influenza A virus pathogenesis by suppressing in-
flammatory responses. PLoS Pathog. 7: e1002149.
115. Meier, A., J. J. Chang, E. S. Chan, R. B. Pollard, H. K. Sidhu, S. Kulkarni,
T. F. Wen, R. J. Lindsay, L. Orellana, D. Mildvan, et al. 2009. Sex differences in
the Toll-like receptor-mediated response of plasmacytoid dendritic cells to HIV-1.
Nat. Med. 15: 955–959.
116. Scotland, R. S., M. J. Stables, S. Madalli, P. Watson, and D. W. Gilroy. 2011. Sex
differences in resident immune cell phenotype underlie more efficient acute in-
flammatory responses in female mice. Blood 118: 5918–5927.
117. vom Steeg, L. G., and S. L. Klein. 2016. SeXX matters in infectious disease
pathogenesis. PLoS Pathog. 12: e1005374.
118. Wikby, A., I. A. Ma
˚nsson, B. Johansson, J. Strindhall, and S. E. Nilsson. 2008.
The immune risk profile is associated with age and gender: findings from three
Swedish population studies of individuals 20-100 years of age. Biogerontology 9:
299–308.
119. Kollias, C. M., R. B. Huneke, B. Wigdahl,and S. R. Jennings. 2015. Animal models
of herpes simplex virus immunity and pathogenesis. J. Neurovirol. 21: 8–23.
120. Yirrell, D. L., W. A. Blyth, and T. J. Hill. 1987. The influence of androgens on
paralysis in mice following intravenous inoculation of herpes simplex virus. J. Gen.
Virol. 68: 2461–2464.
121. MacDonald, E. M., A. Savoy, A. Gillgrass, S. Fernandez, M. Smieja,
K. L. Rosenthal, A. A. Ashkar, and C. Kaushic. 2007. Susceptibility of human
female primary genital epithelial cells to herpes simplex virus, type-2 and the effect
of TLR3 ligand and sex hormones on infection. Biol. Reprod. 77: 1049–1059.
122. Han, X., P. Lundberg, B. Tanamachi, H. Openshaw, J. Longmate, and E. Cantin.
2001. Gender influences herpes simplex virus type 1 infection in normal and
gamma interferon-mutant mice. J. Virol. 75: 3048–3052.
123. Mueller, S. N., and L. K. Mackay. 2016. Tissue-resident memory T cells: local
specialists in immune defence. Nat. Rev. Immunol. 16: 79–89.
124. van Lint, A., M. Ayers, A. G. Brooks, R. M. Coles, W. R. Heath, and
F. R. Carbone. 2004. Herpes simplex virus-specific CD8+ T cells can clear
The Journal of Immunology 1789
by guest on March 15, 2017http://www.jimmunol.org/Downloaded from
established lytic infections from skin and nerves and can partially limit the early
spread of virus after cutaneous inoculation. J. Immunol. 172: 392–397.
125. Gillgrass, A. E., S. A. Fernandez, K. L. Rosenthal, and C. Kaushic. 2005. Estradiol
regulates susceptibility following primary exposure to genital herpes simplex virus
type 2, while progesterone induces inflammation. J. Virol. 79: 3107–3116.
126. Ragupathy, V., W. Xue, J. Tan, K. Devadas, Y. Gao, and I. Hewlett. 2016.
Progesterone augments cell susceptibility to HIV-1 and HIV-1/HSV-2 co-infec-
tions. J. Mol. Endocrinol. 57: 185–199.
127. Vancı
´kova
´, Z., and P. Dvora
´k. 2001. Cytomegalovirus infection in immuno-
competent and immunocompromised individuals–a review. Curr. Drug Targets
Immune Endocr. Metabol. Disord. 1: 179–187.
128. Chang, W. L., P. A. Barry, R. Szubin, D. Wang, and N. Baumgarth. 2009.
Human cytomegalovirus suppresses type I interferon secretion by plasmacytoid
dendritic cells through its interleukin 10 homolog. Virology 390: 330–337.
129. Delale, T., A. Paquin, C. Asselin-Paturel, M. Dalod, G. Brizard, E. E. Bates,
P. Kastner, S. Chan, S. Akira, A. Vicari, et al. 2005. MyD88-dependent and
-independent murine cytomegalovirus sensing for IFN-alpha release and initiation
of immune responses in vivo. J. Immunol. 175: 6723–6732.
130. Traub, S., O. Demaria, L. Chasson, F. Serra, B. Desnues, and L. Alexopoulou.
2012. Sex bias in susceptibility to MCMV infection: implication of TLR9. PLoS
One 7: e45171.
131. Lester, S. N., and K. Li. 2014. Toll-like receptors in antiviral innate immunity. J.
Mol. Biol. 426: 1246–1264.
132. Jonsson, C. B., L. T. Figueiredo, and O. Vapalahti. 2010. A global perspective on
hantavirus ecology, epidemiology, and disease. Clin. Microbiol. Rev. 23: 412–441.
133. Klein, S. L., B. H. Bird, and G. E. Glass. 2001. Sex differences in immune re-
sponses and viral shedding following Seoul virus infection in Norway rats. Am. J.
Trop. Med. Hyg. 65: 57–63.
134. Easterbrook, J. D., and S. L. Klein. 2008. Corticosteroids modulate Seoul virus
infection, regulatory T-cell responses and matrix metalloprotease 9 expression in
male, but not female, Norway rats. J. Gen. Virol. 89: 2723–2730.
135. Klein, S. L., A. Cernetich, S. Hilmer, E. P. Hoffman, A. L. Scott, and G. E. Glass.
2004. Differential expression of immunoregulatory genes in male and female
Norway rats following infection with Seoul virus. J. Med. Virol. 74: 180–190.
136. Vapalahti, O., J. Mustonen, A. Lundkvist, H. Henttonen, A. Plyusnin, and
A. Vaheri. 2003. Hantavirus infections in Europe. Lancet Infect. Dis. 3: 653–661.
137. Ahlm, C., M. Linderholm, P. Juto, B. Stegmayr, and B. Settergren. 1994. Prev-
alence of serum IgG antibodies to Puumala virus (haemorrhagic fever with renal
syndrome) in northern Sweden. Epidemiol. Infect. 113: 129–136.
138. Robinson, D. P., S. A. Huber, M. Moussawi, B. Roberts, C. Teuscher,
R. Watkins, A. P. Arnold, and S. L. Klein. 2011. Sex chromosome complement
contributes to sex differences in coxsackievirus B3 but not influenza A virus
pathogenesis. Biol. Sex Differ. 2: 8.
139. Huber, S. A., and B. Pfaeffle. 1994. Differential Th1 and Th2 cell responses in
male and female BALB/c mice infected with coxsackievirus group B type 3. J.
Virol. 68: 5126–5132.
140. Kallewaard, N. L., L. Zhang, J. W. Chen, M. Guttenberg, M. D. Sanchez, and
J. M. Bergelson. 2009. Tissue-specific deletion of the coxsackievirus and adeno-
virus receptor protects mice from virus-induced pancreatitis and myocarditis. Cell
Host Microbe 6: 91–98.
141. Li, Z., Y. Yue, and S. Xiong. 2013. Distinct Th17 inductions contribute to the
gender bias in CVB3-induced myocarditis. Cardiovasc. Pathol. 22: 373–382.
142. Frisancho-Kiss, S., J. F. Nyland, S. E. Davis, J. A. Frisancho, M. A. Barrett,
N. R. Rose, and D. Fairweather. 2006. Sex differences in coxsackievirus B3-
induced myocarditis: IL-12Rbeta1 signaling and IFN-gamma increase inflamma-
tion in males independent from STAT4. Brain Res. 1126: 139–147.
143. Klein, S. L., and A. Pekosz. 2014. Sex-based biology and the rational design of
influenza vaccination strategies. J. Infect. Dis. 209(Suppl. 3): S114–S119.
144. Klein, S. L., C. Passaretti, M. Anker, P. Olukoya, and A. Pekosz. 2010. The
impact of sex, gender and pregnancy on 2009 H1N1 disease. Biol. Sex Differ. 1: 5.
145. Klein, S. L., A. Hodgson, and D. P. Robinson. 2012. Mechanisms of sex dis-
parities in influenza pathogenesis. J. Leukoc. Biol. 92: 67–73.
146. Torcia, M. G., L. Nencioni, A. M. Clemente, L. Civitelli, I. Celestino,
D. Limongi, G. Fadigati, E. Perissi, F. Cozzolino, E. Garaci, and A. T. Palamara.
2012. Sex differences in the response to viral infections: TLR8 and TLR9 ligand
stimulation induce higher IL10 production in males. PLoS One 7: e39853.
147. Gilliver, S. C. 2010. Sex steroids as inflammatory regulators. J. Steroid Biochem.
Mol. Biol. 120: 105–115.
148. Hoffmann, J., A. Otte, S. Thiele, H. Lotter, Y. Shu, and G. Gabriel. 2015. Sex
differences in H7N9 influenza A virus pathogenesis. Vaccine 33: 6949–6954.
149. Sterling, T. R., D. Vlahov, J. Astemborski, D. R. Hoover, J. B. Margolick, and
T. C. Quinn. 2001. Initial plasma HIV-1 RNA levels and progression to AIDS in
women and men. N. Engl. J. Med. 344: 720–725.
150. Sterling, T. R., C. M. Lyles, D. Vlahov, J. Astemborski, J. B. Margolick, and
T. C. Quinn. 1999. Sex differences in longitudinal human immunodeficiency virus
type 1 RNA levels among seroconverters. J. Infect. Dis. 180: 666–672.
151. Ziegler, S., and M. Altfeld. 2016. Sex differences in HIV-1-mediated immuno-
pathology. Curr. Opin. HIV AIDS 11: 209–215.
152. Sterling, T. R., T. Pisell-Noland, J. L. Perez, J. Astemborski, J. R. McGriff,
L. Nutting, D. R. Hoover, D. Vlahov, and R. C. Bollinger. 2005. Sex-based
differences in T lymphocyte responses in HIV-1-seropositive individuals. J. In-
fect. Dis. 191: 881–885.
153. Li, G., M. Cheng, J. Nunoya, L. Cheng, H. Guo, H. Yu, Y. J. Liu, L. Su, and
L. Zhang. 2014. Plasmacytoid dendritic cells suppress HIV-1 replication but
contribute to HIV-1 induced immunopathogenesis in humanized mice. PLoS
Pathog. 10: e1004291.
154. Tsao, L. C., H. Guo, J. Jeffrey, J. A. Hoxie, and L. Su. 2016. CCR5 interaction
with HIV-1 Env contributes to Env-induced depletion of CD4 T cells in vitro and
in vivo. Retrovirology 13: 22.
155. Monroe, A. K., A. S. Dobs, X. Xu, F. J. Palella, L. A. Kingsley, M. D. Witt, and
T. T. Brown. 2011. Sex hormones, insulin resistance, and diabetes mellitus among
men with or at risk for HIV infection. J. Acquir. Immune Defic. Syndr. 58: 173–
180.
156. Martin, M. E., C. Benassayag, C. Amiel, P. Canton, and E. A. Nunez. 1992.
Alterations in the concentrations and binding properties of sex steroid binding
protein and corticosteroid-binding globulin in HIV+patients. J. Endocrinol. Invest.
15: 597–603.
157. Laffont, S., N. Rouquie
´, P. Azar, C. Seillet, J. Plumas, C. Aspord, and J. C. Gue
´ry.
2014. X-Chromosome complement and estrogen receptor signaling independently
contribute to the enhanced TLR7-mediated IFN-aproduction of plasmacytoid
dendritic cells from women. J. Immunol. 193: 5444–5452.
158. Lindsey, N. P., B. A. Schroeder, E. R. Miller, M. M. Braun, A. F. Hinckley,
N. Marano, B. A. Slade, E. D. Barnett, G. W. Brunette, K. Horan, et al. 2008.
Adverse event reports following yellow fever vaccination. Vaccine 26: 6077–6082.
1790 BRIEF REVIEWS: SEX DIFFERENCES IN VIRAL INFECTIONS
by guest on March 15, 2017http://www.jimmunol.org/Downloaded from
... It has been well documented that sex is a key factor in shaping the immune responses, contributing to differences in the pathogenesis of infectious diseases, between male and female patients. 33,34 Moreover, metabolic homeostasis is differentially regulated between men and women, and there are sex-specific effects on lipid and cholesterol metabolism. Briefly, sexual differences exist in ApoH deletion-regulated lipid metabolic dysbiosis, HBsAg secretion, and the development of liver injury in chronic HBV infection. ...
Article
Full-text available
Apolipoprotein H (APOH) is involved in lipid metabolism and functions as an acute-phase protein during hepatitis B virus (HBV) infection. Herein, we explored whether APOH acts on the development of fatty liver upon chronic HBV infection. Serum APOH level was significantly lower in cirrhosis patients than in healthy controls or patients with chronic infection. It showed sex bias, with elevated levels in female patients with chronic infection. Also, serum APOH levels were negatively correlated with HBV surface antigen (HBsAg) but positively correlated with albumin and triglyceride levels. In In vitro HBV infection model, HBV upregulated APOH expression in a non-temporal manner, and HBsAg levels were elevated by silencing APOH. RNA sequencing (RNA-seq) demonstrated bidirectional expression of APOH, which impacted the immunoregulation upon infection or the metabolic regulation in HepG2.2.15 cells. Then, ApoH -/- mice with persistent HBV replication displayed steatohepatitis and gut microbiota dysbiosis with synergistic sex differences. Our study deciphers the roles of APOH in chronic liver diseases.
... We also found that women had faster declines in NP RNA between Day 0 and Day 3 compared with men, which persisted in analyses adjusting for serostatus, prior symptom duration, and NP RNA level at Day 0. This is a novel finding in the outpatient setting, but is consistent with reports of hospitalized patients with COVID-19 that found male sex to be associated with prolonged duration of SARS-CoV-2 RNA shedding [33][34][35][36] as well as an population surveillance study in Italy, which reported higher proportions of women achieving early viral clearance [37]. These data are consistent with a large body of literature that suggest females are able to clear pathogens faster than males, potentially due to greater innate and adaptive immune responses to infections, including SARS-CoV-2 [38][39][40][41][42]. Whether these differences in SARS-CoV-2 clearance rates contribute to sex-based differences in COVID-19 outcomes that have been observed, including increased risk for COVID-19 hospitalization and mortality among men, is not known [43,44]. ...
Article
Full-text available
Background Identifying characteristics associated with SARS-CoV-2 RNA shedding may be useful to understand viral compartmentalization, disease pathogenesis, and risks for viral transmission. Methods Participants enrolled August 2020 to February 2021 in ACTIV-2/A5401, a placebo-controlled platform trial evaluating investigational therapies for mild-to-moderate COVID-19, and underwent quantitative SARS-CoV-2 RNA testing on nasopharyngeal and anterior nasal swabs, oral wash/saliva, and plasma at entry (day 0, pre-treatment) and days 3, 7, 14 and 28. Concordance of RNA levels (copies/mL) across compartments and predictors of nasopharyngeal RNA levels were assessed at entry (n = 537). Predictors of changes over time were evaluated among placebo recipients (n = 265) with censored linear regression models. Results Nasopharyngeal and anterior nasal RNA levels at study entry were highly correlated (r = 0.84); higher levels of both were associated with greater detection of RNA in plasma and oral wash/saliva. Older age, White non-Hispanic race/ethnicity, lower body mass index (BMI), SARS-CoV-2 IgG seronegativity, and shorter prior symptom duration were associated with higher nasopharyngeal RNA at entry. In adjusted models, BMI and race/ethnicity associations were attenuated, but association with age remained (for every 10 years older age, mean nasopharyngeal RNA was 0.27 log10 copies/mL higher, p < 0.001). Examining longitudinal viral RNA levels, among placebo recipients, women had faster declines in nasopharyngeal RNA than men (mean change: -2.0 vs -1.3 log10 copies/mL, entry to day 3, p < 0.001). Conclusions SARS-CoV-2 RNA shedding was concordant across compartments. Age was strongly associated with viral shedding and men had slower viral clearance than women, which could explain sex differences in acute COVID-19 outcomes.
... Previously published studies reported that COVID-19 had a more severe and mortal course in men than women (22,23). It is widely accepted that sex hormones, sex chromosomes, genomic and epigenetic differences can lead to the difference in the immune responses of women and men (24,25). ...
Article
Full-text available
GİRİŞ ve AMAÇ: COVID-19 aşıları ile bağışıklanma, pandemiden çıkış stratejisi olarak tüm dünyada geniş çapta kabul edilmiştir. Aşının etkililiği ve bunu etkileyen faktörlerin tespit edilmesi, aşı başarısını arttırarak pandemi kontrolünü sağlayabilir. COVID-19 hastalarının mortalitesinde; düzensiz immunizasyon, çoklu organ yetmezliği gibi nedenler gösterilse de bunların cinsiyet ile olan ilişkisi aydınlatılmamıştır. Bu çalışmada; sağlık çalışanlarında CoronaVac aşı etkililiğini belirlemek ve etkileyen faktörleri incelemek amaçlanmıştır. YÖNTEM ve GEREÇLER: Bu retrospektif çalışmaya Eğitim ve Araştırma Hastanesi’nde çalışan 2666 sağlık personeli dahil edilmiştir. Çalışmanın bağımlı değişkeni COVID-19 gelişimidir. Bağımsız değişkenler; yaş, cinsiyet, meslek grubu (doktor/hemşire/diğer sağlık çalışanları), çalışılan klinik tipi (YBÜ/YBÜ dışı), COVID-19 kliniğinde çalışma durumu (COVID-19 kliniği/diğer klinikler), COVID-19 geçirme öyküsü (var/yok), aşıdan sonra COVID-19 tanı süresi (gün) ve aşılanma durumu (aşılı/aşısız) dur. COVID-19 tanısı alan tüm sağlık çalışanlarının klinik takibi Enfeksiyon Hastalıkları ve Klinik Mikrobiyoloji Personel Sağlığı Polikliniği’nde yapılmıştır. Ayrıca klinik sorumluları ile haftalık olarak telefonla görüşülerek veya klinik ziyaretleri yapılarak, COVID-19 gelişimi yönünden tüm sağlık çalışanları izlenmiştir. Aşılama durumu dahil tüm değişkenlerin COVID-19 gelişimi üzerindeki etkilerini analiz etmek için lojistik regresyon analizi kullanılmıştır. Aşı etkililiğine ilişkin etkileşim/etki değişiminin belirlenmesi için tabakalı analizler yapılmıştır. Veriler SPSS versiyon 20.0 paket programı kullanılarak analiz edilmiştir. Tüm istatistiksel testler için önemlilik sınırı 0.05 olarak alınmıştır. BULGULAR: Çalışmaya alınan toplam 2666 sağlık çalışanının yaş ortalaması 37,3±10,2 (n=2556) ve %55,8 (n=1488)’i kadın, %44,2 (n=1178)’si erkektir. CoronaVac aşısının sağlık çalışanlarındaki RT-PCR pozitifliği ile tanı konulan COVID-19’u önlemede etkililiği kadınlarda (%84,5 (%95GA: %73,3-91,0)), erkeklerden (%47,0 (%95GA: %1,7-71,4)) daha yüksek bulunmuştur. Meslek grupları, klinik tipi, COVID-19 kliniğinde çalışma durumu aşı etkililiğini etkilememektedir. Çok değişkenli Lojistik Regresyon analizinde doktor/hemşire olmak, YBÜ’de çalışmak, COVID-19 kliniğinde çalışmak, COVID-19 geçirmiş olmak ve aşılı olmak koruyucu faktör olarak saptanmıştır. Değişkenler arasında %96,5 ile en yüksek koruyuculuk düzeyi COVID-19 geçirme öyküsünün olmasıdır. TARTIŞMA ve SONUÇ: Cinsiyete göre COVID-19 aşı etkililiğinde farklılık, aşılama programlarının revize edilmesini gerektirebilir. Erkekler ve kadınlar için farklı prime-boost aralıkları, özel aşı platformları, farklı dozaj seviyeleri ile sonuçlanan stratejiler COVİD-19 eliminasyonu için kullanılabilir. COVID-19 aşısı olan sağlık çalışanlarının belirli aralıklarla, yaklaşık 1 yıl izlenmesi; çeşitli gruplarda aşı etkililiğindeki farklılıkların belirlenmesi; aşılama programları, sürveyans ve klinik takip gibi noktalarda önceliklendirme yapma, rapel doz aşılama, ağır hastalık geçirme riski olan çalışanları koruma, hizmet planlama ve aşı seçimi gibi pek çok konuda büyük fayda sağlayabilir. COVID-19 pandemisinin kontrol altına alınabilmesi için aşı ile immunizasyon anahtar roldedir ve aşı etkililiğini etkileyen faktörleri tespit etmek hedefe giden yolda başarıyı arttırabilir. INTRODUCTION: Immunization by vaccination has a crucial role in controlling COVID-19 pandemic. Determination of the factors affecting the effectiveness of the vaccine can increase the success rates. We aimed to investigate the effectiveness of CoronaVac and factors affecting its effectiveness in healthcare workers. METHODS: This retrospective study included healthcare personnel (n=2666) working at a training and research hospital. Logistic regression analysis was used for analyzing the effects of all variables including vaccination status on the development of COVID-19. Adjusted odds ratios calculated by logistic regression analysis were used to determine the vaccine effectiveness. Stratified analyses were performed for the determination of the interaction/effect modification regarding the vaccine effectiveness. RESULTS: Mean age of the 2666 healthcare workers included in this study was 37,3±10,2 and 55,8% (n=1488) were females and %44,2 (n=1178) were males. In this study gender and history of COVID-19 infection was found to be an effect modifier for the vaccine effectiveness by the stratified analysis. The effectiveness of the CoronaVac vaccine in preventing development of COVID-19 diagnosed by real-time polymerase chain reaction (RT-PCR) in healthcare workers was 84,5% (95%CI: 73,3-91) in women and 47% (95%CI: 1,7-71,4) in men. Being a medical doctor or a registered nurse, working in ICU or a COVID-19 clinic, a positive history of COVID-19 and COVID-19 vaccination were other protective factors against COVID-19 infection. DISCUSSION AND CONCLUSION: Determination of the factors affecting the effectiveness of the vaccine can increase the success rates. Vaccination programs may need to be modified if there is a COVID-19 history or gender-related difference in vaccine effectiveness.
... Sexual dimorphism is observed not only in the physical appearance and behaviour of the sexes, but also in the context of autoimmunity and antiviral immunity [98][99][100]. It has long been known that women are less susceptible to viral infections than men due to their ability to develop a more effective antiviral response. ...
Article
Full-text available
Antiviral type I interferons (IFN) produced in the early phase of viral infections effectively inhibit viral replication, prevent virus-mediated tissue damages and promote innate and adaptive immune responses that are all essential to the successful elimination of viruses. As professional type I IFN producing cells, plasmacytoid dendritic cells (pDC) have the ability to rapidly produce waste amounts of type I IFNs. Therefore, their low frequency, dysfunction or decreased capacity to produce type I IFNs might increase the risk of severe viral infections. In accordance with that, declined pDC numbers and delayed or inadequate type I IFN responses could be observed in patients with severe coronavirus disease (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), as compared to individuals with mild or no symptoms. Thus, besides chronic diseases, all those conditions, which negatively affect the antiviral IFN responses lengthen the list of risk factors for severe COVID-19. In the current review, we would like to briefly discuss the role and dysregulation of pDC/type I IFN axis in COVID-19, and introduce those type I IFN-dependent factors, which account for an increased risk of COVID-19 severity and thus are responsible for the different magnitude of individual immune responses to SARS-CoV-2.
... (19) The sexual dysmorphism in COVID-19 follows previous data demonstrating men and women have different immune responses to viral infections. (30,31) ...
Article
Full-text available
Background: Older adults are more vulnerable to severe infection and mortality due to COVID-19. They have atypical presentations of the disease without respiratory symptoms, making early diagnosis clinically challenging. We aimed to compare the baseline characteristics, presentation, and disease course of older (≥70 yrs & ≥90 yrs) and younger (<70 yrs) patients hospitalized with COVID-19. Methods: Retrospective review of 429 consecutive patients hospitalized at two tertiary care hospitals in Montreal, Canada, with PCR-confirmed COVID-19. Baseline characteristics, presentation, in-hospital complications, and outcomes were recorded. Desegregation by age was performed to compare older versus younger individuals. Additional subgroup analyses were performed amongst patients ≥70 stratifying by sex, living situation, and those presenting with geriatric syndromes compared to those without. Results: Patients ≥70 (n=260) presented less frequently with respiratory symptoms compared to patients <70 (n=169) (52% vs. 32%). 11% of patients ≥70 and 24% of patients ≥90 presented with a geriatric syndrome as their sole symptom compared to 3% of those <70. Older adults were more likely to develop disease complications (including delirium, acute kidney injury, and hypernatremia) and had higher in-hospital mortality (32% vs. 13%). Subgroup analyses revealed heightened vulnerability to complications in older men, those from long-term care, and those with at least one geriatric syndrome upon presentation. Conclusions: Older adults presenting to hospital with COVID-19 often have no respiratory symptoms and can present with only a geriatric syndrome. New geriatric syndromes in older individuals should trigger evaluation for COVID-19 and consideration for early initiation of multidisciplinary care to prevent complications.
Chapter
The course of coronavirus disease 2019 (COVID-19) varies greatly from asymptomatic infection to severe condition resulting in death. Identification of prognostic factors is crucial to identify those patients who may progress to severe status. Common risk factors associated with severity and prognosis of COVID-19 are demographic features, clinical symptoms, comorbidities, laboratory markers, and complications. In this chapter, the authors discuss the most important factors influencing the prognosis of patients with COVID-19.
Article
Full-text available
Introduction. Sex differences in the course and outcomes of the disease were found during the COVID- 19 pandemic. Aim. To summarize the knowledge about the mechanisms underlying sex differences in COVID-19, with a focus on the role of estrogen. Materials and methods. We conducted a study using various databases until September 2022 for the keywords “estrogen” and “COVID-19”. All articles were published in English. Results. The review discusses the involvement of estrogen in the implementation of the immune response in viral infection. Individual paragraphs of the article are devoted to the effect of female sex hormones on coagulation, inflammation, and the renin-angiotensin system. Conclusion. At the end of the paper, it is concluded that there is great potential for future work deciphering hormonal effects on human physiology to explain the heterogeneity in pathogenic responses and may facilitate the development of more effective and personalized interventions.
Article
Background: Limited data exist to describe sex-based differences in the severity of cytomegalovirus (CMV) infection after solid organ transplant (SOT). We sought to identify if a difference exists in likelihood of tissue-invasive disease between male and female SOT recipients and to understand how age affects this relationship. Methods: A retrospective cohort of 180 heart, liver, and kidney recipients treated for CMV was examined. A logistic regression model was developed to assess the relationship between female sex and CMV type (noninvasive vs. invasive). A secondary regression analysis looked at the relationship of invasive CMV with a variable combining sex with age above or below 50. Results: There were 37 cases of proven or probable invasive CMV, occurring in 30% of females versus 16% of males. After adjustment for potential confounders, females with CMV infection were significantly more likely to have invasive disease (odds ratio (OR) 2.69, 95% confidence interval (CI) 1.25-5.90, p = .01). Females 50 years or older were at particular risk compared with males under 50 years (adjusted OR 4.54, 95% CI 1.33-18.83, p = .02). Conclusion: Female SOT recipients with CMV in our cohort were more likely than males to have tissue-invasive disease, with the highest risk among older females. Further prospective studies are warranted to explore underlying immunologic mechanisms.
Article
Full-text available
Sex-specific differences have been described for a variety of infectious and autoimmune diseases. In HIV-1 infection women present with significantly lower viral loads during early infection, but during chronic infection women progress faster to AIDS for the same amount of viral replication. Recent studies have shown that sex differences during HIV-1 infection might also include the size of the latent viral reservoir, which represents a major obstacle towards a cure for HIV-1. Here we review different immunological and virological aspects that can be influenced by sex hormones and sex-specific genetic factors and their contribution to viral replication, as well as the creation and maintenance of the HIV-1 reservoir.
Article
Full-text available
CD4 T cell depletion during HIV-1 infection is associated with AIDS disease progression, and the HIV-1 Env protein plays an important role in the process. Together with CXCR4, CCR5 is one of the two co-receptors that interact with Env during virus entry, but the role of CCR5 in Env-induced pathogenesis is not clearly defined. We have investigated CD4 T cell depletion mechanisms caused by the Env of a highly pathogenic CXCR4/CCR5 dual-tropic HIV-1 isolate R3A. We report here that R3A infection induced depletion of both infected and uninfected “bystander” CD4 T cells, and treatment with CCR5 antagonist TAK-779 inhibited R3A-induced bystander CD4 T cell depletion without affecting virus replication. To further define the role of Env-CCR5 interaction, we utilized an Env-mutant of R3A, termed R3A-5/6AA, which has lost CCR5 binding capability. Importantly, R3A-5/6AA replicated to the same level as wild type R3A by using CXCR4 for viral infection. We found the loss of CCR5 interaction resulted in a significant reduction of bystander CD4 T cells death during R3A-5/6AA infection, whereas stimulation of CCR5 with MIP1-β increased bystander pathogenesis induced by R3A-5/6AA. We confirmed our findings using a humanized mouse model, where we observed similarly reduced pathogenicity of the mutant R3A-5/6AA in various lymphoid organs in vivo. We provide the first evidence that shows CCR5 interaction with a dual-tropic HIV-1 Env played a significant role in Env-induced depletion of CD4 T cells.
Article
Full-text available
Significance Females have increased immune responsiveness than males, and they are more likely to develop autoimmune disorders. The mechanism underlying these observations is unclear, and hypotheses suggest an important role for the X chromosome. Here, we discover that the inactive X is predisposed to become partially reactivated in mammalian female lymphocytes, resulting in the overexpression of immunity-related genes. We also demonstrate that lymphocytes from systemic lupus erythematosus patients have different epigenetic characteristics on the inactive X that compromises transcriptional silencing. These findings are the first to our knowledge to link the unusual maintenance of X chromosome Inactivation (the female-specific mechanism for dosage compensation) in lymphocytes to the female bias observed with enhanced immunity and autoimmunity susceptibility.
Article
Full-text available
During a successful pregnancy, the maternal immune system plays a critical role in maintaining immunotolerance towards semi-allogeneic fetal antigens. Recent studies have indicated that myeloid-derived suppressor cells (MDSCs) are active players in establishing fetal-maternal tolerance; however, the underlying mechanism remains poorly understood. In this study, we observed a significant expansion of monocytic MDSCs (M-MDSCs) in the peripheral blood of pregnant women, which suppressed T cell responses in a reactive oxygen species-dependent manner and required cell-cell contact. The number of M-MDSCs positively correlated with serum estrogen and progesterone levels. Administration of 17β-estradiol, but not progesterone, enhanced both the expansion and suppressive activity of M-MDSCs through signal transducer and activator of transcription 3 (STAT3). Pretreatment with STAT3 inhibitor JSI-124 almost completely abrogated the effects of 17β-estradiol on MDSCs. Collectively, these results demonstrate that 17β-estradiol-induced STAT3 signaling plays an important role in both the expansion and activation of MDSCs during human pregnancy, which may benefit the development of novel therapeutic strategies for prevention of immune-related miscarrage. This article is protected by copyright. All rights reserved.
Article
Full-text available
Background: Previous studies have established that estrogen is capable of accelerating cutaneous wound healing through multiple mechanisms, one of which involves affecting keratinocytes biological properties, such as migration, proliferation, etc. This study aims to reveal the underlying molecular mechanisms of estrogen promoting epidermal keratinocytes proliferation. Method & Results: We found that compared with female mice with a normal estrous cycle, female mice with their ovaries removed before puberty exhibited a delayed cutaneous wound healing, thinner epidermis, and significantly fewer proliferating cell nuclear antigen (PCNA)-positive keratinocytes. Moreover, a significant increase in HaCaT proliferation was detected by a CCK8 assay when treated with 17 β-estradiol compared with those treated with control vehicle. Consistent with the results of the CCK8 assay, flow cytometry indicated a high proportion of 17 β-estradiol-treated HaCaT cells in S phase compared with vehicle-treated cells. Western blot analysis demonstrated the activation of Akt, Erk and upregulation of PCNA in HaCaT cells treated with 17 β-estradiol. Interestingly, Erk activation occurred prior to Akt activation. Upregulation of PCNA expression, elevated proliferation and high S phase fraction of HaCaT cell by 17 β-estradiol could be reversed by an Akt or Erk inhibitor. Moreover, Erk inhibition reversed 17 β-estradiol-induced Akt activation, whereas an Akt inhibitor exhibited no effect on Erk, further suggesting that Erk was on the upstream while Akt on the downstream of the signaling pathway. Conclusion: This study demonstrates that one of the critical mechanisms underlying 17 β-estradiol promoting skin wound healing is through regulation of keratinocyte proliferation via Erk/Akt signaling pathway.
Article
Full-text available
Gonadal differentiation has a determinative influence on sex development in human embryos. Disorders of sexual development (DSD) have been associated with persistent embryonal differentiation stages. Between 1998 and 2015, 139 female patients with various (DSD) underwent operations at the Scientific Center of Obstetrics, Gynaecology and Perynatology in Moscow, Russia. Clinical investigations included karyotyping, ultrasound imaging, hormonal measurement and investigations of gonadal morphology. The male characteristics in the embryo are imposed by testicular hormones. When these are absent or inactive, the fetus may be arrested at between developmental stages, or stay on indifferent stage and become phenotypically female. A systematic analysis of gonadal morphology in DSD patients and a literature review revealed some controversies and led us to formulate a new hypothesis about sex differentiation. Proliferation of the mesonephric system (tubules and corpuscles) in the gonads stimulates the masculinization of gonads to testis. Sustentacular Sertoli cells of the testes are derived from mesonephric excretory tubules, while interstitial Leydig cells are derived from the original mesenchyme of the mesonephros. According of the new hypothesis, the original mesonephric cells (tubules and corpuscles) potentially persist in the ovarian parenchyma. In female gonads, some mesonephric excretory tubules regress and lose the tubular structure, but form ovarian theca interna and externa, becoming analogous to the sustentacular Sertoli cells in the testis. The ovarian interstitial Leydig cells are derived from intertubal mesenchyme of the mesonephros, similar to what occurs in male gonads (testis). Surprisingly, the leading determinative factor in sexual differentiation of the gonads is the mesonephros, represented by the embryonic urinary system. 2016
Article
Males and females differ in their immunological responses to foreign and self-antigens and show distinctions in innate and adaptive immune responses. Certain immunological sex differences are present throughout life, whereas others are only apparent after puberty and before reproductive senescence, suggesting that both genes and hormones are involved. Furthermore, early environmental exposures influence the microbiome and have sex-dependent effects on immune function. Importantly, these sex-based immunological differences contribute to variations in the incidence of autoimmune diseases and malignancies, susceptibility to infectious diseases and responses to vaccines in males and females. Here, we discuss these differences and emphasize that sex is a biological variable that should be considered in immunological studies.
Article
In human immunodeficiency virus type 1 (HIV-1) infected women, oral or injectable progesterone containing contraceptive pills may enhance HIV-1 acquisition in vivo and the mechanism by which this occurs is not fully understood. In developing countries Herpes simplex virus type-2 (HSV-2) co-infection has been shown to be a risk for increase of HIV-1 acquisition and, if co-infected women use progesterone pills, infections may increase several fold. In the present study we used an invitro cell culture system to study progesterone effects on HIV-1 replication and to explore the molecular mechanism of progesterone effects on infected cells. In our in-vitro model, CEMss cells (lymphoblastoid cell line) were infected with either HIV-1 alone or co-infected with HSV-2. HIV-1 viral load was measured with and without sex hormone treatment. Progesterone treated cells showed an increase in HIV-1 viral load (1411.2pg/mL) compared to cells without progesterone treatment (993.1pg/mL). Increased cell death noted with HSV-2 co-infection and progesterone treated cells. Similar observations were noted in peripheral blood mononuclear cells (PBMC) cells derived from three female donors. Progesterone treated cells also showed reduced antiviral efficacy. Inflammatory cytokines and associations with bio-markers of disease progression were explored. Progesterone up regulated inflammatory cytokines and chemokines con-versely, down-regulated anti-apoptotic Bcl-2 expression. Nuclear protein analysis by electrophoretic mobility shift assay showed association of progesterone with Progesterone Response Element (PRE), which may lead to down regulation of Bcl-2. These data indicate that progesterone treatment enhances HIV-1 replication in infected cells and co-infection with HSV-2 may further fuel this process.
Article
Purpose of review: The article reviews our current knowledge regarding the role of sex and sex hormones in regulating innate immune responses to viral infections, which may account for the described sex differences in immunity to HIV-1. Recent findings: Prominent sex differences exist in various infectious and autoimmune diseases. Biological mechanisms underlying these differences include the modulation of immunological pathways by sex hormones and gene dosage effects of immunomodulatory genes encoded by the X chromosome. During HIV-1 infections, women have been shown to present with lower viral load levels in primary infection, although their progression to AIDS is faster in comparison with men when accounting for viral load levels in chronic infection. HIV-1-infected women furthermore tend to have higher levels of immune activation and interferon-stimulated gene expression in comparison with men for the same viral load, which has been associated to innate sensing of HIV-1 by Toll-like receptor 7 and the consequent interferon-α production by plasmacytoid dendritic cells. Summary: Improvement in understanding the mechanisms associated with sex differences in HIV-1-mediated immunopathology will be critical to take sex differences into consideration when designing experimental and clinical studies in HIV-1-infected populations.