Download as pdf or txt
Download as pdf or txt
You are on page 1of 23

Frontiers in Neuroendocrinology 35 (2014) 347–369

Contents lists available at ScienceDirect

Frontiers in Neuroendocrinology
journal homepage: www.elsevier.com/locate/yfrne

Review

Gender differences in autoimmune disease


S.T. Ngo a,b,1, F.J. Steyn a,1, P.A. McCombe b,c,⇑
a
School of Biomedical Sciences, University of Queensland, St Lucia, Queensland, Australia
b
University of Queensland Centre for Clinical Research, University of Queensland, Herston, Queensland, Australia
c
Department of Neurology, Royal Brisbane & Women’s Hospital, Herston, Queensland, Australia

a r t i c l e i n f o a b s t r a c t

Article history: Autoimmune diseases are a range of diseases in which the immune response to self-antigens results in
Available online 2 May 2014 damage or dysfunction of tissues. Autoimmune diseases can be systemic or can affect specific organs
or body systems. For most autoimmune diseases there is a clear sex difference in prevalence, whereby
Keywords: females are generally more frequently affected than males. In this review, we consider gender differences
Autoimmunity in systemic and organ-specific autoimmune diseases, and we summarize human data that outlines the
Gender prevalence of common autoimmune diseases specific to adult males and females in countries commonly
Sex differences
surveyed. We discuss possible mechanisms for sex specific differences including gender differences in
Immune system
immune response and organ vulnerability, reproductive capacity including pregnancy, sex hormones,
genetic predisposition, parental inheritance, and epigenetics. Evidence demonstrates that gender has a
significant influence on the development of autoimmune disease. Thus, considerations of gender should
be at the forefront of all studies that attempt to define mechanisms that underpin autoimmune disease.
Ó 2014 Published by Elsevier Inc. Open access under CC BY-NC-ND license.

1. Introduction 1.1. Autoimmunity and autoimmune disease – key considerations for


sexual dimorphism
Autoimmune disease, first recognized by Ian Mackay and
Macfarlane Burnet, is characterized by the failure of an organism Autoimmune diseases comprise a range of diseases in which the
to tolerate its own cells and tissues, resulting in an aberrant immune response to self-antigens results in damage or dysfunction
immune response by lymphocytes and/or antibodies (Mackay of tissues (Mackay and Burnet, 1963). Criteria have been developed
and Burnet, 1963). This leads to pathological changes and dysfunc- that allow a disease to be classified as autoimmune (Rose and
tion of the tissue that is the target of the self-directed immune Bona, 1993). These criteria include the identification of a target
response. Autoimmune diseases can be systemic or can affect antigen, the presence of antibodies and/or T cells in the target
specific organs or body systems including the endocrine, gastro- organ, and the transfer of disease to animals by cells or antibodies
intestinal and liver, rheumatological, and neurological systems. In (Rose and Bona, 1993; Witebsky et al., 1957). The immune system
this review, we discuss systemic and organ-specific autoimmune is comprised of multiple cell types of a single lineage. Collectively,
diseases. In general there is a greater prevalence of autoimmune these cells contribute to acquired and innate immunity. Interac-
disease in females than males, although there are exceptions. To tions between acquired and innate immune function may aggra-
demonstrate sex differences in autoimmune disease, we summa- vate autoimmune responses. A strong inflammatory component,
rize human data to provide an overview of the prevalence in males with infiltration of tissue with macrophages and lymphocytes, is
and females of common autoimmune diseases in countries associated with many organ-specific autoimmune diseases, such
commonly surveyed. We then discuss possible mechanisms for as in rheumatoid arthritis (RA) and multiple sclerosis (MS). Non-
sex specific differences. inflammatory conditions like myasthenia gravis (MG) are anti-
body-mediated, although there may be some minor lymphocyte
infiltration. Systemic lupus erythematosus (SLE) is another impor-
tant antibody-mediated disease.
The origin of autoimmune disease is a disruption of immune
⇑ Corresponding author at: University of Queensland Centre for Clinical Research, tolerance of self-antigens. This is thought to occur in genetically
University of Queensland, Herston, Queensland, Australia. Fax: +61 7 38327447. susceptible individuals after exposure to environmental triggers
E-mail address: pamela.mccombe@uq.edu.au (P.A. McCombe). (Mackay, 2001). Autoimmune diseases are strongly associated with
1
Authors contributed equally.

http://dx.doi.org/10.1016/j.yfrne.2014.04.004
0091-3022/Ó 2014 Published by Elsevier Inc. Open access under CC BY-NC-ND license.
348 S.T. Ngo et al. / Frontiers in Neuroendocrinology 35 (2014) 347–369

human leukocyte antigen (HLA) genes through mechanisms that Data highlight the very high (9:1) gender bias toward females
could involve genetic variability in the immune response and for systemic autoimmune diseases including SLE (Chiche et al.,
genetic variability in the ability to present antigens (Gough and 2012; Eaton et al., 2007; Feldman et al., 2013; Li et al., 2012;
Simmonds, 2007). Modern genetic studies of autoimmune diseases Ramagopalan et al., 2013) and Sjögren’s syndrome (SS) (Eaton
such as MS, type I diabetes (T1D) and inflammatory bowel disease et al., 2007; Misra et al., 2003; Nannini et al., 2013; Ramagopalan
(IBD) have shown that many other genes, each with small effect, et al., 2013; Seror et al., 2013; Tsuboi et al., 2013; Xiang and Dai,
also contribute to genetic susceptibility to autoimmunity 2009), occurring independent of country of assessment. By con-
(Australia and New Zealand Multiple Sclerosis Genetics, 2009; trast, some neurological diseases (including MG (Guptill et al.,
Barrett et al., 2009; Khor et al., 2011). At times autoimmune dis- 2011; Guy-Coichard et al., 2008; Huang et al., 2013; Pedersen
eases run in families (Bias et al., 1986; McCombe et al., 1990). This et al., 2013; Ramagopalan et al., 2013; Singhal et al., 2008;
was once thought to indicate an autosomal dominant inheritance, Suzuki et al., 2011) and Guillain–Barré syndrome (GBS)
but it is now thought to be due to the interaction of common genes (Alshekhlee et al., 2008; Chen et al., 2014; Dhadke et al., 2013;
(Clayton, 2009). However, what is often overlooked is that a very Eaton et al., 2007; Mitsui et al., 2013)) had a near equal prevalence
significant component of the genetic contribution to autoimmunity in males and females, although recent data suggest a greater prev-
is gender, with the presence or absence of a Y chromosome influ- alence of GBS in males in Asian countries including Japan (Mitsui
encing the risk for autoimmune disease. et al., 2013), China (Chen et al., 2014) and India (Dhadke et al.,
Environmental factors also predispose to autoimmune disease. 2013). In our recent study of GBS in Australia, there was a male
This is well illustrated by twin studies, where the risk of develop- preponderance (Blum et al., 2013). However, in MS, the most com-
ing autoimmune disease cannot be solely explained by genetics. mon neurological autoimmune disease, females accounted for two
For example, in MS there is greater concordance in monozygotic thirds of all cases (Bentzen et al., 2010; Cheng et al., 2009; El Adssi
than dizygotic twins, indicating that genetic factors are important et al., 2012; Hadjimichael et al., 2008; McCombe, 2003; Osoegawa
(Willer et al., 2003). However, the concordance in monozygotic et al., 2009; Pandit and Kundapur, 2014; Ramagopalan et al., 2013).
twins is incomplete, suggesting a role for environmental factors. Again, this occurred mostly independent of country of assessment.
In a study from Finland, it was shown that the concordance of The prevalence of endocrine related autoimmune diseases pre-
MS in dizygotic twins has increased in two decades, with this rapid dominantly favored females (including Grave’s Disease (GD)
increase again suggesting a significant role for environmental fac- (Carle et al., 2011; Gaujoux et al., 2006; Guo et al., 2013;
tors (Kuusisto et al., 2008). Possible environmental factors that Phitayakorn et al., 2013) and Hashimoto’s thyroiditis (HT)
predispose to autoimmunity include exposure to infectious agents (Ramagopalan et al., 2013; Zhang et al., 2012)). This was not the
and to dietary components (Lamb et al., 2008; Pender and Greer, case for T1D (Anderson et al., 2012; Bhadada et al., 2011; Eaton
2007), exposure to chemicals, xenobiotics or toxins (Rieger and et al., 2007; Kawasaki and Eguchi, 2004; Menke et al., 2013), where
Gershwin, 2007), stress (Rieger and Gershwin, 2007), or reduced prevalence was equal in males and females. This varied somewhat
exposure to factors that protect from the development of autoim- between countries, and prevalence slightly favored males in the
mune diseases (e.g. sunlight during childhood and adulthood) USA (Menke et al., 2013) and Denmark (Eaton et al., 2007), but
(Fraser et al., 2003; Ponsonby et al., 2005). Different exposure or females in Japan (Kawasaki and Eguchi, 2004) and Australia
response to these environmental factors could contribute to gender (Speight et al., 2012). Prevalence for rheumatological autoimmune
differences in autoimmunity. diseases varied greatly between countries, however trends
remained the same. For ankylosing spondylitis (AS), prevalence
favored males in the USA (Reveille, 2011), England (Ramagopalan
2. Sexual dimorphism in autoimmune diseases – prevalence et al., 2013), China (Xiang and Dai, 2009), India (Aggarwal and
and consequence Malaviya, 2009), and Australia (Oldroyd et al., 2009). For RA prev-
alence greatly favored females, with reports ranging from 66% to
2.1. Effect of gender on the prevalence of autoimmune diseases 86% dominance (Biver et al., 2009; Li et al., 2012; Myasoedova
et al., 2011; Pedersen et al., 2011; Ramagopalan et al., 2013;
For most autoimmune diseases there are clear sex differences in Yamanaka et al., 2013). Of the gastrointestinal diseases that we
prevalence whereby females are generally more frequently studied, PBC showed a strong female dominance (values ranging
affected than males (Eaton et al., 2007). This gender bias can be from 80% to 100%), regardless of country (Amarapurkar and Patel,
modest, as is seen in MS (Bentzen et al., 2010; Cheng et al., 2007; Eaton et al., 2007; Harada et al., 2013; McNally et al.,
2009; El Adssi et al., 2012; Hadjimichael et al., 2008; McCombe, 2011; Shen et al., 2009; Sood et al., 2004). While celiac disease
2003; Osoegawa et al., 2009; Pandit and Kundapur, 2014; was generally female dominant (60% to 80% incidence), an excep-
Ramagopalan et al., 2013), or strong, as is seen in primary biliary tion was the recent observations by Nijhawan et al. (2013) that
cirrhosis (PBC) (Amarapurkar and Patel, 2007; Eaton et al., 2007; found a 60% incidence for adult males (Nijhawan et al., 2013). A
Harada et al., 2013; McNally et al., 2011; Shen et al., 2009; Sood similar rate of prevalence in male children was reported
et al., 2004). Importantly, prevalence between males and females (Bhattacharya et al., 2013). Incidence of ulcerative colitis (UC)
may be changing. For example, the predominance of MS in females and Crohn’s disease (CD) in males and females did not vary greatly
is increasing; initial early reports suggest equal prevalence in by country, and generally did not favor females or males
males and females (Buckley, 1954), by the 1980’s prevalence was (Gathungu et al., 2012; Jain et al., 2012; Jess et al., 2007; Nerich
approximately 2:1 (female:male) (Confavreux et al., 1980), and et al., 2006; Ng et al., 2013; Ramagopalan et al., 2013; Zhao
recent reports suggest prevalence in some instances of up to 3:1 et al., 2013). Of interest, Asakura et al. (2009) showed a deviation
(female:male) (Ramagopalan et al., 2013). Such a rapid change is in this trend, with 70% CD subjects in Japan being male (Asakura
likely to be due to environmental factors. To provide a more et al., 2009). The studies detailed above do not consider severity
detailed assessment of the current status of gender bias in autoim- of symptoms.
mune diseases, we summarize adult data from recent studies,
focusing predominantly (when possible) on the United States 2.2. Effect of gender on the severity of autoimmune diseases
(USA), England, France, Denmark, Japan, China, India, and Australia.
Measures are further categorized based on systemic, endocrine, As well as prevalence varying with gender, the severity of
gastrointestinal, neurological or rheumatological diseases (Fig. 1). autoimmune diseases, meaning the severity of symptoms and the
S.T. Ngo et al. / Frontiers in Neuroendocrinology 35 (2014) 347–369 349

Fig. 1. Prevalence of autoimmune disease in USA, England, France, Denmark, Japan, China, India, and Australia. There is a very high gender bias toward females for systemic
diseases including systemic lupus erythematosus (Chiche et al., 2012; Eaton et al., 2007; Feldman et al., 2013; Li et al., 2012; Ramagopalan et al., 2013) and Sjögren’s
syndrome (Eaton et al., 2007; Misra et al., 2003; Nannini et al., 2013; Ramagopalan et al., 2013; Seror et al., 2013; Tsuboi et al., 2013; Xiang and Dai, 2009). Neurological
diseases including myasthenia gravis (Guptill et al., 2011; Guy-Coichard et al., 2008; Huang et al., 2013; Pedersen et al., 2013; Ramagopalan et al., 2013; Singhal et al., 2008;
Suzuki et al., 2011) and Guillain–Barré syndrome (Alshekhlee et al., 2008; Chen et al., 2014; Dhadke et al., 2013; Eaton et al., 2007; Mitsui et al., 2013) have near parity
distribution between males and females, although recent data suggests a greater prevalence of Guillain–Barré syndrome in males in Japan (Mitsui et al., 2013), China (Chen
et al., 2014) and India (Dhadke et al., 2013). In Australia, there is a male preponderance in Guillain–Barré syndrome (Blum et al., 2013). For multiple sclerosis, females account
for two thirds of all cases (Bentzen et al., 2010; Cheng et al., 2009; El Adssi et al., 2012; Hadjimichael et al., 2008; McCombe, 2003; Osoegawa et al., 2009; Pandit and
Kundapur, 2014; Ramagopalan et al., 2013). The prevalence of endocrine related diseases predominantly favors females (including Grave’s Disease (Carle et al., 2011; Gaujoux
et al., 2006; Guo et al., 2013; Phitayakorn et al., 2013) and Hashimoto’s thyroiditis (Ramagopalan et al., 2013; Zhang et al., 2012)). Prevalence is near parity for type 1 diabetes
(Anderson et al., 2012; Bhadada et al., 2011; Eaton et al., 2007; Kawasaki and Eguchi, 2004; Menke et al., 2013), although prevalence slightly favors males in the USA (Menke
et al., 2013) and Denmark (Eaton et al., 2007), or females in Japan (Kawasaki and Eguchi, 2004) and Australia (Speight et al., 2012). Prevalence for rheumatological diseases
varies between countries, however trends remained the same. For ankylosing spondylitis, prevalence favors males in the USA (Reveille, 2011), England (Ramagopalan et al.,
2013), China (Xiang and Dai, 2009), India (Aggarwal and Malaviya, 2009), and Australia (Oldroyd et al., 2009). For rheumatoid arthritis, prevalence greatly favors females
(Biver et al., 2009; Li et al., 2012; Myasoedova et al., 2011; Pedersen et al., 2011; Ramagopalan et al., 2013; Yamanaka et al., 2013). Primary biliary cirrhosis shows a strong
female dominance (Amarapurkar and Patel, 2007; Eaton et al., 2007; Harada et al., 2013; McNally et al., 2011; Shen et al., 2009; Sood et al., 2004). Celiac disease is generally
female dominant, although prevalence seems to favor males in India (Nijhawan et al., 2013). Incidence of ulcerative colitis (UC) and Crohn’s disease (CD) in males and females
does not vary greatly by country, and generally does not favor females or males (Gathungu et al., 2012; Jain et al., 2012; Jess et al., 2007; Nerich et al., 2006; Ng et al., 2013;
Ramagopalan et al., 2013; Zhao et al., 2013), although a deviation in this trend is seen in Japan (Asakura et al., 2009).

degree of disability, may also differ between males and females. more severe in girls (Gupta et al., 2007), whereas in diseases like
However, compared to prevalence, this is not easily defined. The RA, gender does not necessarily predict outcome (Courvoisier
effect of gender on severity varies among autoimmune diseases. et al., 2008). In MS, disease severity, measured as disability pro-
For example, psoriasis is more severe in males (Sakai et al., gression, appears to be greater in males (Runmarker and
2005). Men develop autoimmune hepatitis at a younger age and Andersen, 1993; Weinshenker et al., 1991). A recent study demon-
have high relapse rates, but also have better prognosis than women strates greater disease severity in male SLE patients; male patients
(Al-Chalabi et al., 2008). By contrast, the clinical course in CD is were more likely to suffer renal and cardiovascular co-morbidities
350 S.T. Ngo et al. / Frontiers in Neuroendocrinology 35 (2014) 347–369

while female patients were more likely to suffer from urinary tract
infections, hypothyroidism, depression, esophageal reflux, asthma,
and fibromyalgia (Crosslin and Wiginton, 2011).

2.3. Effect of gender on the pathological features of autoimmune


disease

Gender differences are reported in the T cell and antibody


responses that occur in autoimmune disease. In MS for instance,
females tend to secrete higher levels of IFNc in response to prote-
olipid protein (PLP) peptides when compared to control females
and males with MS (Pelfrey et al., 2002), and increased T-cell reac-
tivity occurs at a higher frequency (Greer et al., 2004). On the other
hand, a small study in SLE patients report that the IgM and IgG
reactivity to dsDNA Ro and La follows no gender bias (Ferreira
et al., 2005). In diabetes, females have higher levels of antibodies
to glutamic acid decarboxylase (Lindholm et al., 2004).
There are no systematic studies of the effects of gender on the
histopathological features of autoimmune disease, but it has been Fig. 2. Factors underlying sexual dimorphism in autoimmune disease. While the
suggested that autoimmune disease in males is young onset, with underlying basis for sexual dimorphism in autoimmune disease is yet to be
determined, a number of factors may contribute to gender differences in autoim-
acute inflammation and autoantibodies, while in females autoim-
mune disease. Females show increased immune reactivity, differences in the
mune disease is either acute onset and non-inflammatory and/or number or responsiveness of cells that constitute the immune response, and
associated with chronic pathology (Fairweather et al., 2008). Much differential resistance to target organ damage, and this may influence propensity to
remains to be studied in this area. autoimmune disease. Hormonal changes during pubertal maturation, pregnancy,
and menopause may alter susceptibility to autoimmunity, and it is generally
accepted that pregnancy is associated with improved symptoms in autoimmune
2.4. Effect of gender on survival in autoimmune disease disease. The protective effects that are exerted by female (estrogen, progesterone,
and prolactin) and male (androgens) hormones may also explain gender differences
in specific autoimmune diseases. Differential exposure to environmental factors
Autoimmune diseases are generally not considered to be fatal, (including sunlight) can influence the prevalence and risk of developing an
but in many diseases, reduced life expectancy can be demon- autoimmune disease or the severity of disease. Genetic factors that might influence
strated, compared to the general population. The measure of this the gender specific development of autoimmune disease may be related to
susceptibility genes, chromosomal differences, or epigenetics. When genetic factors
is the standarized mortality rate (SMR), which compares the
are combined with environmental factors, this can influence parental inheritance of
observed mortality in a group with that expected in that popula- autoimmune disease. Epigenetic changes that arise in autoimmune disease can be
tion. This has been well studied in MS where survival duration is related to the sex of the parent, or can result from external factors. Genetic
greater for women than men. However, when survival is consid- imprinting, and in particular, miRNA imprinting, could also contribute to the sexual
ered and adjusted relative to general mortality rates, females with dimorphism seen in autoimmune disease.

MS have poorer survival outcomes (Grytten Torkildsen et al.,


2008). In T1D, relative mortality was found to be higher for Finnish
prevalence, susceptibility and severity of autoimmune disease
women than for men (Asao et al., 2003). Increased mortality in a
(Fig. 2). These factors are discussed below.
large multicentre international cohort of SLE patients have been
reported to be associated with female gender (Bernatsky et al.,
2006). Similar observations were documented in a Spanish popula- 3.1. Differences between female and male immune systems
tion (Ruiz et al., 2014). For PBC, a higher number of deaths have
been observed in English/Welsh women (Hamlyn and Sherlock, The gender differences in autoimmunity could be due to differ-
1974). While data suggests that gender can influence the mortality ences between male and female immune systems (McCombe and
of autoimmune disease, it is important to note that co-morbidities Greer, 2013a). Such differences are found in most animal species;
may significantly alter the course of disease. Moreover, survival males have immune suppression when compared to females and
can be influenced by factors other than severity of disease, such this is linked to male sexual activity (McKean and Nunney,
as quality of care, age of onset, and time to diagnosis. 2005). Females show increased immune reactivity (Hewagama
et al., 2009), and this greater immunocompetence may translate
to greater resilience to infectious and some non-infectious dis-
3. Factors underlying sexual dimorphism in autoimmune eases. However, it is possible that this greater immune reactivity
diseases makes women more prone to developing autoimmune disease
(Zandman-Goddard et al., 2007).
Above we have described the observable effects of gender on
the incidence and severity of autoimmune disease. Now we will 3.1.1. T lymphocytes, B lymphocytes and natural killer cells
consider the possible underlying mechanisms that could lead to T lymphocytes, B lymphocytes and natural killer (NK) cells com-
these clinical effects. We limit our considerations to human obser- prise the lymphocytes of the immune system. T lymphocyte
vations and refer to mouse data when human data is restricted. We secreted cytokines including IL-2, IL-4, IL-10, IFNc and TNFa
have previously provided an extensive review of data pertaining to underlie cell-mediated adaptive immunity. B lymphocytes produce
mouse models of autoimmune disease (McCombe et al., 2009). The IgG and IgM antibodies against foreign antigens to mount antibody
underlying basis for sexual dimorphism in autoimmune disease is driven adaptive immune responses. NK cells are effectors of cell-
yet to be determined. However, significant research has investi- mediated, innate immunity.
gated the impact of differences between males and females in While there are age-associated changes in human lymphocyte
the immune response, organ vulnerability, reproductive function, subtype numbers (Yan et al., 2010), the overall number of lympho-
microchimerism, sex hormones, and the environment on the cytes in males and females is the same. However, males have a
S.T. Ngo et al. / Frontiers in Neuroendocrinology 35 (2014) 347–369 351

lower number of T lymphocytes (Bouman et al., 2004), and post- including the brain (Silverman et al., 2000; St John and Abraham,
menopausal women have decreased numbers of B lymphocytes 2013) and take part in the host innate immune defence by releas-
and helper T cells (Th cells) (Giglio et al., 1994; Yang et al., ing chemoattractants that recruit eosinophils (Wardlaw et al.,
2000). B lymphocyte counts do not differ between genders, how- 1986) and monocytes (Perry et al., 1993). Dendritic cells, which
ever, women produce higher levels of circulating antibodies than are the antigen-presenting cells of the immune system, are gener-
men (Rowley and Mackay, 1969), which could underlie their pro- ally present on tissues that are exposed to the environment
pensity to produce higher levels of autoantibodies in autoimmune (Banchereau and Steinman, 1998; Silverman et al., 2000), but they
diseases (Whitacre et al., 1999). Indeed, females have increased have also recently been suggested to be resident in brain (albeit
levels of IgM when compared to their male counterparts mouse brain) (Bulloch et al., 2008).
(Butterworth et al., 1967; Lichtman et al., 1967). While data on A number of in vivo studies provide insight into the roles of
NK cells is limited, when compared to men, fertile females have microglia, astrocytes, mast cells, and dendritic cells in the neuro-
decreased NK cell cytotoxic activity, but women with premature immune response in general. Interestingly, such studies have also
menopause have higher numbers of NK cells (Giglio et al., 1994). generated observations that might be of relevance when consider-
Females produce stronger humoral and cellular immune ing gender differences in autoimmune disease. For example: estro-
responses to antigen than males (Ansar Ahmed et al., 1985). Stud- gen inhibits microglia and astrocyte activation in the striatum of a
ies in humans suggest that females produce higher levels of mouse model of Parkinson’s Disease (Tripanichkul et al., 2006);
CD4 + T cells in response to immunization (Ho et al., 1995). More- astrocytes in female mice may be more resistant to oxidative
over, the antibody response of females surpasses that of males stress-induced toxicity (Giordano et al., 2013), estrogen increases
after vaccination for influenza, hepatitis B, rubella and tetanus the number and activation of mast cells in the brain (Boes and
(Cook, 2008). In some instances, adverse effects in response to Levy, 2012), and female mice have increased recruitment of den-
immunization can be observed in females in the absence of a sig- dritic cells to the brain after stroke (Manwani et al., 2013).
nificant humoral response (Shohat et al., 2000). In the context of autoimmunity, studies in mice suggest that
Gender differences are also seen in the outcome of immunity to estrogen may play a key role in the activation of microglia and
bacterial, parasitic, and viral infections. With bacterial infections, astrocytes, and the generation of tolerogenic dendritic cells. Myelin
decreased production of anti-lipoarabinomannan IgM is observed basic protein-primed T cells from female mice and castrated male
in males with Mycobacterium tuberculosis infections (Demkow mice induce microglial expression of proinflammatory cytokines
et al., 2007). In secondary syphilis, females are more resistant as (Dasgupta et al., 2005), expression of the RNA binding protein
CD4 + and CD8 + T lymphocytes are both elevated (Ho et al., HuR in spinal cord astrocytes provides greater attenuation of dis-
1995). The prevalence, severity or susceptibility of parasitic infec- ease in female experimental autoimmune encephalomyelitis
tions, Schistosoma mansoni, Schistosoma haematobiainfections (Degu (EAE) mice (and this is reversed following ovariectomy) (Wheeler
et al., 2002), and Leishmania donovani (Goble and Konopka, 1973) is et al., 2012), an estrogen receptor alpha ligand reduces inflamma-
greater in males than females. The decreased prevalence and tion and protects against axonal loss and reactive astrogliosis in
severity of Schistosoma infections in females may be attributed to EAE mice (Spence et al., 2011), and estriol leads to the generation
their ability to secrete greater amounts of anti-inflammatory cyto- of splenic dendritic cells that when transferred to mice prior to
kines and differing antibody isotypes (Demkow et al., 2007), and active induction of EAE leads to protection from the development
the decreased susceptibility in females to Leishmania may be due of EAE (Papenfuss et al., 2011).
to increased secretion of granulocyte macrophage colony-stimulat- Data pertaining to human autoimmune disease is currently
ing factor (Lezama-Davila et al., 2007). In the case of viruses, stud- limited to astrocytes and dendritic cells. Astrocytes in the rim
ies illustrate higher prevalence of herpes simplex virus type 2 in and center of MS lesions in male and female patients have recently
females (Glynn et al., 2008; Howard et al., 2003; Rabenau et al., been found to upregulate different gender steroid-specific
2002), and this may be due to higher levels of progesterone in enzymes, suggesting that these responses might underlie gender-
females since progesterone contraceptives hastens herpes simplex specific differences in steroid synthesis and signaling in the brains
virus type 1 (Stringer et al., 2007) (the role of progesterone in auto- of MS patients, and thus, the gender bias in MS prevalence and sus-
immunity will be discussed below). Increased levels of lympho- ceptibility (Luchetti et al., 2014). While dendritic cell numbers are
cytes and neutrophils in young females (Pembrey et al., 2008) reported to be lower in MS (de Andres et al., 2009), primary SS
may underlie their increased susceptibility to hepatitis C virus (Vogelsang et al., 2010), and SLE (Henriques et al., 2012), whether
(European Paediatric Hepatitis, 2005; Zanetti et al., 1998), although there are gender differences in the expression or function of den-
chronic adult hepatitis C virus infection results in increased fibrosis dritic cells in these diseases remains unknown. Thus, future
in males (Poynard et al., 1997). research that aims to determine whether gender differences in
autoimmune disease can be attributed to differential expression
3.1.2. Microglia, astrocytes, mast cells, and dendritic cells and function of microglia, astrocytes, mast cells, and dendritic cells
The immune cells of that are resident in tissues could poten- will generate significant interest.
tially show gender differences that could influence autoimmune
disease. Little is known about gender differences in these cells. 3.2. Sexual dimorphism in target organ vulnerability to damage
To illustrate this issue we will use examples from the CNS. Microg-
lia and astrocytes account for the majority of the brain cell popula- Autoimmune disease results when an autoimmune attack on
tion, and there is no indication of any gender difference with target organs leads to dysfunction. Autoimmunity can occur with-
respect to the spatial distribution of these cell types in the male out clinical disease being evident. This means that there are two
and female brain (Stark et al., 2007). While microglia represent issues involved in the development of autoimmune disease: the
the primary immune cell of the central nervous system, astrocytes presence of autoimmunity and the ability of the target organ to
are also considered to be immunocompetent, as they possess the resist the autoimmune attack. For example, in SLE, the presence
ability to synthesize immune molecules in response to insult/ of autoantibodies does not always lead to end organ damage
injury (Farina et al., 2007; Gimsa et al., 2013; Jensen et al., 2013; (Lewis et al., 2013). The role of the target organ in autoimmunity
Kaur et al., 2010; Pivneva, 2008; Wirenfeldt et al., 2011). In the has been reviewed elsewhere (Hill et al., 2007). To consider gender
context of immunity, mast cells and dendritic cells are also of par- differences in the target organ that could lead to gender differences
ticular importance. Mast cells are resident in a multitude of tissues in the incidence and severity of autoimmune disease, we need to
352 S.T. Ngo et al. / Frontiers in Neuroendocrinology 35 (2014) 347–369

examine gender differences in the ability of different target organs an example of how factors other than hormones could influence
to tolerate damage. The processes that lead to an ability to with- the effect of puberty on disease.
stand damage is sometimes referred to as cytoprotection; or neu-
roprotection or cardioprotection when referring to the ability of
3.3.2. Pregnancy
the nervous system or the heart, respectively, to withstand dam-
Pregnancy involves physiological changes in the mother,
age. A full discussion of this topic is beyond the scope of this
including elevation of cardiac output, increased basal metabolic
review, but the processes involved are fundamental and include
rate, increased lipid levels, and weight gain (Forsum and Lof,
susceptibility to apoptosis, autophagy (Lista et al., 2011), mito-
2007; Granger, 2002; Lain and Catalano, 2007; Tan and Tan,
chondrial function (Martel et al., 2012), and biochemical pathways
2013). In pregnancy there are changes in the levels of hormones
that promote survival such as the nrf2 pathway (Chen and Kunsch,
such as estriol, progesterone, prolactin, early pregnancy factor
2004; Lee et al., 2005).
(EPF), alpha-fetoprotein (Brunton and Russell, 2010) and leptin
There are some studies that indicate that there are gender dif-
(Molvarec et al., 2011), and elevated levels of growth factors such
ferences in the ability to protect cells from damage. In general it
as insulin-like growth factor (IGF) (Lauszus et al., 2003). After
is thought that females have greater resistance, because of the
delivery, there is a rapid decline in the levels of pregnancy hor-
cytoprotective properties of estrogen (see below). Much of the
mones and maternal physiology rapidly returns to normal,
work in this area has been done in the nervous system, although
although during lactation the levels of prolactin and oxytocin are
estrogen also has protective properties on endothelium (Si et al.,
elevated (Uvnas-Moberg et al., 1990). It is generally thought that
2001). It is generally accepted that females tend to have improved
the changes in maternal physiology are mediated through the
outcome after traumatic brain injury (Groswasser et al., 1998), and
effects of the pregnancy hormones. In pregnancy, the mother plays
although this has been debated (Bayir et al., 2004; Farace and
host to the fetus, which contains foreign antigens. Immune
Alves, 2000; Wagner et al., 2000), this is thought to occur in
changes in pregnancy tend to suppress the maternal immune
response to the effects of estrogen (Roof and Hall, 2000). While less
system, which is thought to be important in preventing rejection
is known about gender differences in the processes and pathways
of the fetus (Munoz-Suano et al., 2011; Veenstra van
involved in neuroprotection, there is a study suggesting that there
Nieuwenhoven et al., 2003; Yip et al., 2006).
are gender differences in autophagy (Lista et al., 2011). Additional
research is needed to determine in more detail whether gender
differences that target organ resistance are responsible for gender 3.3.2.1. Immune responses in pregnancy. Regulatory T cells (Tregs)
differences in autoimmune disease. mediate active immune tolerance to prevent any maternal lym-
phocyte-mediated damage to the fetus (Aluvihare et al., 2004;
Saito et al., 2005). A shift in Th1/Th2 activity in pregnancy favors
3.3. Autoimmune disease and reproductive function
increased Th2-related and reduced Th1-related activities. This
results in an improvement in Th1-mediated diseases and an exac-
Reproductive load may significantly alter susceptibility to auto-
erbation of Th2-mediated diseases (Doria et al., 2002, 2004;
immunity, particularly in females. Careful consideration relative to
Ostensen et al., 2005). Indeed, the remission of the Th1 diseases
pubertal maturation (thus the onset of mature reproductive age),
RA and AS during pregnancy is coupled with an inhibition of
and life events that alter reproductive function (including
Th1-related activities via an upregulation in cytokine inhibitors
pregnancy and menopause) should be made. For pregnancy, these
(Ostensen et al., 2005). These findings are further corroborated
considerations extend to that of the fetus. We now survey the data
by a Th1 phenotype in abortion, and a Th2 phenotype during nor-
relating to reproductive issues and autoimmune disease.
mal pregnancy (Raghupathy, 2001). While it is accepted that the
Th1/Th2 paradigm is a marker for pregnancy success, it has been
3.3.1. Puberty superseded by the Treg/Th17 paradigm. In 2004, Tregs were found
The incidence of autoimmune disease varies relative to the to be elevated during normal pregnancy (Somerset et al., 2004).
onset of reproductive function, although with the exception of epi- Elevated levels of Tregs correlate well with decreased activity of
demiological studies, few studies differentiate between pre-puber- Th1-related autoimmune diseases (Forger et al., 2008). The ratio
tal and post-pubertal incidence, specifically in relation to gender of Treg and IL-17 appears to be of pivotal importance during preg-
bias. This is surprising, given the impact of puberty on gender bias nancy as an imbalance in the Treg/IL-17 proportion is linked to
in autoimmune diseases. For example, while pre-pubertal onset of complications during pregnancy, including recurrent pregnancy
MS is rare, with only 3–5% of cases reported in individuals under loss and pre-eclampsia (Darmochwal-Kolarz et al., 2012; Saito
18 years of age (Chitnis et al., 2009; Duquette et al., 1987; Ghezzi et al., 2011). It is likely that steroid hormones drive these immuno-
et al., 1997), gender bias within these cases of MS is absent logical changes in pregnancy; improvements in psoriasis (Th1-
(reviewed in (Chitnis, 2013)). Following the onset of puberty, how- dependent disease) is correlated with higher levels of estrogen
ever, incidence changes rapidly and pubertal girls are found to be (Murase et al., 2005), and the increase in Tregs may be driven by
at greater risk of developing MS than pre-pubertal girls estradiol since low levels of Tregs are positively correlated with
(Ramagopalan et al., 2010). Moreover, earlier onset of puberty in low levels of estradiol in patients with missed abortion (Cao
girls is also associated with increased risk of developing MS et al., 2014). The steroid-associated immunological changes seen
(Ramagopalan et al., 2009b). However, in SLE, the male to female in pregnancy may very well contribute to sex differences in auto-
ratio of 1:9 may be as low as 1:2 prior to puberty (reviewed in immune disease, as discussed below.
(Tedeschi et al., 2013)). By contrast, the incidence of UC in adoles- Another pregnancy-associated change that may underlie gender
cents is equivalent in males and females, but by early-adulthood differences in autoimmune disease is a small protein of the heat
the ratio of male to female patients is 4:1 (Bove, 2013). These shock family, chaperonin 10 (cpn 10) (heat shock protein 10, hsp
reports could suggest that exposure to hormonal changes during 10). Cpn 10 is released into blood during early pregnancy
pubertal development could underlie the gender bias in autoim- (Morton et al., 1977). In line with its immunomodulatory capacity,
mune diseases. However, environmental effects may also play a a 12 week course of cpn 10 treatment improves disease measures
role. For example, relocation from a low-risk geographic to high- in patients with RA (Vanags et al., 2006). Moreover, decreased T
risk geographic area during puberty significantly increased MS risk lymphocyte-associated secretion of TNFa and interleukin 1b
(Dean and Elian, 1997). Although this was not gender specific, it is occurs after 12 weeks of cpn 10 in psoriasis (Vanags et al., 2006).
S.T. Ngo et al. / Frontiers in Neuroendocrinology 35 (2014) 347–369 353

Thus, elevated levels of cpn 10 may contribute to the beneficial As detailed above, current data indicates that the differential
effects of pregnancy on autoimmune disease. effects imparted by pregnancy in autoimmune diseases are very
specific to autoimmune disease type. While it has been suggested
3.3.2.2. Autoimmune disease and pregnancy. The effects of preg- that some autoimmune diseases are not improved by pregnancy
nancy on autoimmune disease differ for inflammatory autoimmune (Adams Waldorf and Nelson, 2008), this is generally only the case
diseases, which tend to remit, and antibody mediated autoimmune for SLE, which is an antibody mediated disease. We note that preg-
diseases, where the effects are more variable. For instance, the nancy is documented to exert protective effects in AS, CD, MS, and
symptoms of MG vary considerably from woman to woman and RA, which are inflammatory diseases. In particular, there is over-
from pregnancy to pregnancy in the same woman (Hantai et al., whelming evidence to demonstrate that pregnancy is associated
2004), and pregnancy has been reported to worsen, improve, or with improved symptoms in MS and RA.
provide no change in MG symptoms (Batocchi et al., 1999). Like-
wise, pregnancy with psoriasis can lead to similar variations in dis-
3.3.2.3. Pregnancy, microchimerism and autoimmunity. Microchim-
ease response (Murase et al., 2005). In AS, pregnancy may not
erism, the trafficking of cells from the fetus to the mother and vice
aggravate disease (Lui et al., 2011), but it has been reported to cause
versa, occurs during pregnancy (Lo et al., 1996). It is well known
remission (Ostensen and Ostensen, 1998). For CD, the activity of
that, as a normal phenomenon, fetal cells persist in the maternal
disease was significantly lower during pregnancy as determined
circulation years after pregnancy (fetal microchimerism) (Bianchi
by the Harvey-Bradshaw index (Agret et al., 2005). In MS
et al., 1996), and that maternal cells persist in immunodeficient
(Borchers et al., 2010; Confavreux et al., 1998; McCombe and
and immunocompetent offspring into adult life (maternal
Greer, 2013b), and RA (Hench, 1983; Nelson and Ostensen, 1997;
microchimerism) (Evans et al., 1999; Maloney et al., 1999). Micro-
Ostensen and Villiger, 2007) however, it is widely accepted that
chimerism has been considered to be a possible trigger for autoim-
pregnancy is associated with the remission of symptoms, although
munity and has also been put forward as a protective mechanism
there can be a flare of disease post-partum. In contrast to MS and
(Fig. 3).
RA, SLE an antibody mediated disease, is reported to worsen during
If microchimerism were indeed linked to the development of
pregnancy (Ruiz-Irastorza et al., 1996; Stojan and Baer, 2012), and
autoimmune disease, then chimeric cells may persist as inactive
this can vary in the degree of severity (Lateef and Petri, 2012).
dendritic cells and lymphocytes (McNallan et al., 2007) until an
Pregnancy can also trigger the onset of autoimmune disease.
environmental factor, a shift in cytokine proportion, an infection,
Fulminant autoimmune diabetes can present during pregnancy
or a hormonal trigger activates these cells, thereby causing an allo-
or in the post-partum period (Inagaki et al., 2002), and other dor-
graft reaction, and subsequently, autoimmune disease in either the
mant autoimmune diseases, like thyroiditis, can become clinically
mother or the offspring (Nelson, 1999, 2002). The evidence for a
evident post-partum (Muller et al., 2001; Premawardhana et al.,
role for fetal microchimerism in the development of autoimmune
2004; Thompson and Farid, 1985). With evidence indicating a role
disease is conflicting. Fetal microchimeric cells have been found
of pregnancy in autoimmune disease, it must be noted however
in the blood and thyroid of women with autoimmune thyroid dis-
that heterogeneous study designs, variable presentation of disease,
ease (Lepez et al., 2012; Renne et al., 2004). Lepez and colleagues
and patient ethnicity can render the interpretation of pregnancy-
have also demonstrated that these fetal cells in women with thy-
associated data difficult.
roid disease were cytotoxic T lymphocytes, and hence, may have
Autoimmune disease during pregnancy can also result in fetal
been capable of initiating a graft-vs-host reaction to elicit autoim-
manifestations of disease. This is seen in diseases including MG
mune thyroid disease (Lepez et al., 2011). In line with a role for
(Eymard et al., 1991; Gveric-Ahmetasevic et al., 2008), SLE (Lee,
fetal micochimerism, women with increased odds of developing
2009; Yildirim et al., 2013), thyrotoxicosis (Batra, 2013; Radetti
autoimmune thyroid disease were noted to have had a previous
et al., 2002), autoimmune thrombocytopenic purpura (Sukenik-
pregnancy (Friedrich et al., 2008), but Bulow-Pederson and col-
Halevy et al., 2008; van der Lugt et al., 2013) and cutaneous lupus
leagues found no association between pregnancy and autoimmune
(Johnson, 2013). When placental transmission of autoantibodies
occurs, a temporary autoimmune disease can occur in the neonate.
In more severe cases, passively acquired autoimmunity can result
in congenital heart block due to transplacental transfer of anti-Ro
and anti-La from mothers with SLE (Brucato et al., 2001;
Wahren-Herlenius and Sonesson, 2006), or, from transfer of acetyl-
choline receptors antibodies, in arthrogryposis multiplex congenita
(Donaldson et al., 1981) or pulmonary hypoplasia (Newsom-Davis,
2007). Thus, given fetal complications associated with pregnant
mothers with autoimmune disease, due care is required for the
management of disease during the gestational period.
Autoimmune disease can also appear under conditions of
assisted pregnancy and ovarian hyperstimulation. Increased MS
activity and relapses of MS can occur after assisted reproductive
technology (ART) (Correale et al., 2012; Hellwig et al., 2008;
Laplaud et al., 2006), and this may be due to stress, immunological
changes in response to hormone therapy, or termination of
immune therapy upon the instigation of ART. ART pregnancies
have also been associated with exacerbation of SLE (Ben-Chetrit Fig. 3. Microchimerism and autoimmunity. Microchimerism is the trafficking of
and Ben-Chetrit, 1994; Casoli et al., 1997). Furthermore, complica- cells from the fetus to the mother and vice versa. Fetal microchimerism has been
tions can arise in autoimmune patients undergoing ART. Women proposed to have a beneficial role in helping a parous woman to recover from injury
with thyroid antibodies before the first cycle of ART have increased through the provision of additional mutipotential stem cells that could be used in
repair of damage. By contrast, maternal microchimerism may be harmful since
risk of miscarriage (Poppe et al., 2003), and a trend toward worse maternal cells are a possible source of graft versus host response, as is seen in
pregnancy and live-birth rate is becoming evident in SLE mothers systemic sclerosis. Since microchimerism occurs during pregnancy, microchimer-
undergoing ART (Costa and Colia, 2008). ism may contribute to gender differences in autoimmune disease.
354 S.T. Ngo et al. / Frontiers in Neuroendocrinology 35 (2014) 347–369

thyroid disease, and thus argue that there is no association et al., 2005; Sanchez-Guerrero et al., 2001; Urowitz et al., 2006).
between fetal microchimerism and autoimmune disease (Bulow In RA, postmenopausal women report higher physical disability
Pedersen et al., 2006). scores than premenopausal women (Kuiper et al., 2001). Meno-
The disease that has been most studied with respect to a caus- pause has also been associated with the onset of autoimmune dis-
ative role for fetal microchimerism is systemic sclerosis (sclero- ease. For example, autoimmune hepatitis occurs most frequently in
derma) given that chronic graft-vs-host disease after bone the late teenage years in males, but occurs in women in the late
marrow transplantation results in clinical presentations that are teens and after menopause (Werner et al., 2008). By contrast, men-
similar to systemic sclerosis (Artlett et al., 1998; Gratwohl et al., opause is not associated with high occurrence of SLE (Mok, 2008),
1977). Much like the studies for thyroid disease however, those and SLE after menopause is generally less severe (Boddaert et al.,
that have investigated microchimerism as an underlying cause 2004). Early menopause however has shown to be associated with
for systemic sclerosis have presented variable results (Ichikawa an increased risk of developing SLE and RA (Costenbader et al.,
et al., 2001; Murata et al., 1999). Maternal microchimerism may 2007; Pikwer et al., 2012a,b). There are a number of immunological
present as tissue-specific maternal microchimerism in neonates, changes associated with menopause including increased produc-
as has been suggested to be the case in neonatal lupus congenital tion and response to pro-inflammatory cytokines, decreased secre-
heart block (Stevens et al., 2003). The rate of maternal microchim- tion of anti-inflammatory cytokines, decreased lymphocyte levels,
erism is also higher in twins with MS (Willer et al., 2006). By con- and decreased activity of NK cells (Gameiro and Romao, 2010;
trast, maternal microchimerism appears to play no role in PBC Gameiro et al., 2010). These changes, coupled with altered endo-
(Nomura et al., 2004). crine function, may underlie the incidence and risk of autoimmune
Fetal microchimerism has also been proposed to have a disease that is associated with menopausal transition.
beneficial role in helping a parous woman to recover from injury.
Microchimeric cells have been demonstrated at sites of tissue 3.4. Role of hormones in the sexual dimorphism in autoimmunity
inflammation during pregnancy, forming blood vessels. After deliv-
ery, microchimeric cells have been found in the maternal liver after The strong gender bias in autoimmune disease susceptibility
liver injury, and many years after pregnancy, microchimeric fetal could be due to the differential effects of gender-specific
cells have been found at sites of injury where these cells adopt a hormones. Indeed, a considerable amount of evidence now demon-
variety of phenotypes (indicating their multipotent capacity). This strates not only the role of gender-specific hormones in contribut-
transfer of fetal stem cells could be helpful and provide additional ing to disease, but also the differential roles of these hormones
mutipotential stem cells that could be used in repair of damage relative to altered reproductive states (Fig. 4). For example, disease
(Guillot et al., 2007; Johnson and Bianchi, 2004; Khosrotehrani flares for SLE are influenced by the age of onset of menarche, the
and Bianchi, 2005; Khosrotehrani et al., 2004; Lo et al., 1996). This use of oral contraceptives, the age of onset of menopause, and
is speculative, but further investigation could be rewarding. hormonal treatments in response to menopause (Costenbader
et al., 2007). These changes in reproductive function are all charac-
3.3.2.4. Long-term effects of pregnancy on autoimmunity. Another terized by significant changes in the hormonal milieu. The effects
consideration in autoimmune disease is the long-term effect of of various sex hormones including estrogens, progesterone, andro-
pregnancy. There is little information in this area, except in the gens, and prolactin are considered below. When possible, we will
case of MS. Overall, women with MS who have been pregnant have discuss evidence of contribution to immunity, contribution to
a better long-term outcome than those who have not been preg- autoimmune disease, and outcomes following treatments targeting
nant (D’Hooghe et al., 2010). It seems that the effects are cumula- these hormones.
tive since multiparous women seem to have a better outcome than
women with fewer, or who have no pregnancies (Runmarker and 3.4.1. Estrogens
Andersen, 1995; Verdru et al., 1994). Having been pregnant does Acting through estrogen receptor subtypes (ERs) alpha (ERa)
not increase the risk of secondary progression of disease (Koch and beta (ERb), estrogen mediates transcription activity of intracel-
et al., 2009). Women who have children with different partners lular machinery to produce genomic effects, whereas the rapid,
do not have greater risk of disability as might be expected to occur non-genomic actions of estrogen are triggered through activation
if the immune response to paternal genes was harmful (Basso et al., of membrane-associated ERs (mER). This results in changes in
2004). Of interest, recent findings suggested that pregnancy fre- intracellular activity that occur independent of transcription
quency lowers mortality in T1D (Sjöberg et al., 2013). This awaits pathways and protein synthesis. Endogenous estrogens include
further validation, as it is a difficult topic to evaluate. There may estrogen (E1), estradiol (E2) and estriol (E3, produced only in preg-
be reverse causation at work, such that women with less severe nancy). These compounds differentially act on intracellular ERs
disease could be more likely to decide to have children. However that are present in all cells of the immune system (Deroo and
there are some plausible biological mechanisms for long-term ben- Korach, 2006), including T and B lymphocyte subsets, and periph-
eficial effects of pregnancy, including fetal microchimerism which eral NK cells (Pernis, 2007; Pierdominici et al., 2010).
could provide a reservoir of stem cells that could aid in repair (as It has become clear that estrogens impact immunity by modu-
discussed above), and epigenetic changes (as discussed below). lating lymphocyte development and function (Grimaldi et al.,
2002; Smithson et al., 1998). Indeed, estrogens may facilitate
3.3.3. Menopause critical protective effects that negate detrimental changes follow-
Menopause is a natural occurrence that marks the end of cycli- ing inflammatory responses. For example, a decrease in CD4+/
cal changes associated with reproductive function in women. It is CD8 + T-cell-subset ratios in cultures of human lymphocytes is
characterized by lower levels of estrogens and progesterone, peak- seen following estrogen treatment (Athreya et al., 1993). Similarly,
ing of FSH levels, follicular depletion, and irreversible cessation of estrogen enhances immunoglobulin secretion from these cells
menses. There are conflicting reports about the effects of meno- (Kanda and Tamaki, 1999). Estrogen may also directly affect B
pause on MS that has developed earlier in life (Holmqvist et al., lymphocyte subsets. In animal studies, estrogens increase bone
2006) (Smith and Studd, 1992; Wundes et al., 2011). In SLE, marrow progenitor B cells by protecting them from apoptosis
however, menopausal transition has been reported to decrease (Grimaldi et al., 2002; Medina et al., 2000), and by increasing the
the frequency of flares, but to cause more damage in the organs survival of B cells (Grimaldi et al., 2002). In humans, estrogens
in which individual postmenopausal flares occur (Fernandez induce polyclonal activation of B cells. Specifically, estrogen
S.T. Ngo et al. / Frontiers in Neuroendocrinology 35 (2014) 347–369 355

Fig. 4. The role of hormones in the sexual dimorphism in autoimmunity. The strong gender bias in autoimmune disease susceptibility could be due to the differential effects
of gender-specific hormones in contributing to disease, and the differential roles of these hormones relative to altered reproductive states. Estrogen, which is seen in high
levels during pregnancy, impacts immunity by modulating lymphocyte development and function, and promoting cytoprotection. These actions of estrogen could lead to
improved cell-mediated disease or worsened antibody-mediated disease. Progesterones and androgens exert anti-inflammatory and immunosuppressive effects respectively,
and this is generally beneficial in autoimmune disease. Prolactin, which is high during pregnancy, induces pro-inflammatory effects, and this tends to worsen autoimmune
disease.

increases IgG and IgM production of peripheral blood mononuclear evidence demonstrating a direct involvement of estrogen in the
cells. This occurs regardless of gender (Kanda and Tamaki, 1999; incidence of autoimmune disease.
Weetman et al., 1981). The role of estrogen in immunity has been The high female to male incidence rate for SLE (Fig. 1), coupled
extensively reviewed (Bouman et al., 2005). with a dramatic reduction in gender ratios prior to puberty or fol-
The potential cytoprotective role of estrogens may explain these lowing menopause (both conditions are associated with a reduc-
effects. Takao and colleagues showed that estrogen treatment tion in circulating estrogen levels) has contributed to anecdotal
dose-dependently reversed the cytotoxic effects of TNFa in T cells. evidence suggesting that estrogen mediate immunity specific to
In this study, the ER antagonist ICI-172.780 abolished the cytopro- autoimmune diseases. This has been substantiated by studies
tective effects of 17b-estradiol on T cells (Takao et al., 2005). assessing the effects of estrogens on tissue collected from patients
Numerous reports demonstrate protective actions of estrogens on suffering from autoimmune diseases. For example, lymphocytes
the EAE mouse model of MS (Bebo et al., 2001; Ito et al., 2001; collected from SLE patients are more sensitive to the actions of
Jansson et al., 1994). In this model of MS, inflammation is mediated physiological levels of E2, whereas E2 may differentially modify
by T lymphocytes that react to CNS and brain-derived autoanti- signaling pathways coupled to disease onset and severity in T cells
gens. To delineate the mechanisms through which estrogen may from SLE patients (reviewed in (Pierdominici and Ortona, 2013)).
be cytoprotective, Lélu and colleagues demonstrate that endoge- Moreover, documented associations between autoimmune dis-
nous estrogens limit the recruitment of blood-derived inflamma- eases and various facets of estrogen signaling provide reason for
tory cells (CD4 + T cells) into the CNS (Lelu et al., 2010). Thus, further assessment of potential causes. For example, in MS, ER
estrogen may protect the brain from damage following inflamma- expression in the thymus changes dramatically (Zoller and Kersh,
tory insults. This may underlie the potential cytoprotective effects 2006), whereas estrogen concentrations in synovial fluid is mark-
of estrogen during pregnancy. edly increased in RA patients (Cutolo et al., 2004). However, this
In females, circulating levels of estrogens change dramatically does not necessarily mean that altered levels of endogenous estro-
relative to reproductive function. This is important, as varying gens will increase the risk for developing SLE. For example, the
levels of exposure to estrogens greatly affects immunity. At high incidence of SLE does not change relative to pregnancy and birth
concentrations (such as in pregnancy), estrogen inhibits the Th-1 rate (Cooper et al., 2002). Moreover, while the incidence of lupus
pro-inflammatory pathways (including TNFa, IL-1b and IL-6) and flares in patients with existing SLE increase during pregnancy
stimulates Th-2 anti-inflammatory pathways (including IL-4, IL- (Walker, 2007), this is not necessarily associated with alterations
10 and TGFb). Conversely, at low levels (as seen after menopause), in circulating levels of estrogens. For example, Doria et al. (2006)
estrogen stimulates pro-inflammatory pathways (including TNFa demonstrated that estrogen levels in SLE patients may not rise dur-
and IL1-b) (Straub, 2007). ing the second and third trimesters of pregnancy, and thus disease
Relative to autoimmunity, estrogens may differentially impact flares that occur at this time cannot be directly associated with
incidence across age and reproductive capacity. Second to this, altered circulating levels of estrogens (Doria et al., 2006).
the actions of estrogens on inflammatory and immune responses Accordingly, alterations in physiology not coupled to endogenous
may change relative to the presence of autoimmune diseases. For estrogen may underlie autoimmune disease associations. To this
example, T lymphocytes from patients with SLE, but not from extent, the potential role of estrogen in the development of
healthy controls, are activated via ERa and ERb-specific agonists autoimmune disease may be better identified when considering
(Rider et al., 2006). While evidence confirms the role of estrogens exogenous estrogens, estrogen-like compounds, or modifiers of
in immunity, suggesting that they may play an important role in estrogen action.
autoimmunity, the mechanisms through which estrogens may Immune responses may be compromised by naturally occurring
directly or indirectly mediate autoimmune responses remain exogenous estrogens (phyto/myco-estrogens), or by synthetic
unknown. Elucidation of this will require more substantive estrogen-like compounds (xenoestrogens) (reviewed in
356 S.T. Ngo et al. / Frontiers in Neuroendocrinology 35 (2014) 347–369

(Chighizola and Meroni, 2012; Pierdominici and Ortona, 2013; women with RA for 6 months did not significantly improve symp-
Vandenberg et al., 2012)). These compounds are widely found in toms (Hazes et al., 1989).
products used in the production of food and around the home, As with the use of oral contraceptives, conflicting reports exist
including detergents, plastics, pesticides, and industrial chemicals. regarding the value and risk of hormone replacement therapy
While less potent than endogenous estrogens, these compounds (HRT) in women to treat autoimmune diseases. For example, HRT
can accumulate in adipose tissue, where they may exert potent does not appear to impact the ongoing course of disease in MS
effects on immunity (Chighizola and Meroni, 2012). Existing evi- (Holmqvist et al., 2006), or the risk of developing RA (Walitt
dence showing an involvement in phyto-, myco- or xenoestrogens et al., 2008). HRT is thought to benefit postmenopausal women
in autoimmune disease rely predominantly on epidemiological evi- with inflammatory bowel disease (Kane and Reddy, 2008), increase
dence, or exposure studies conducted on animal models. Thus, it bone mineral density in women with PBC (Ormarsdottir et al.,
remains unknown to what extent these compounds contribute to 2004), and improve incidence of isolated pulmonary hypertension
autoimmune disease in the real world. Moreover, epidemiological in post-menopausal women with systemic sclerosis (Beretta et al.,
studies, and resulting assessment of the effects of exogenous estro- 2006). Conversely, in SLE, HRT may increase the risk of thrombosis
gens on immune function may not necessarily accurately predict (Fernandez et al., 2007; Sanchez-Guerrero et al., 2007). Current
direct estrogenic effects on autoimmune disease incidence. clinical trials assessing the potential roles of estrogens in prevent-
Perhaps one of the most revealing cases of associations between ing or minimizing disease impact for other autoimmune diseases
autoimmune disease and estrogen replacement therapy comes (for example ongoing efforts to assess the impact of estriol in
from the use of Diethylstilbestrol (DES). Relapsing Remitting Multiple Sclerosis, ClinicalTrials.gov Identi-
DES is a synthetic estrogen, historically used to reduce risk of fier: NCT00451204) will shed light on the value of estogen-medi-
complications to pregnancy, to assist with postpartum lactation, ated effects on autoimmunity.
and for the treatment of other postmenopausal symptoms. Not
only did DES not improve pregnancy outcomes (Bamigboye and 3.4.2. Progesterone
Morris, 2003), it contributed to a number of complications. For Progesterone belongs to the class of steroid hormones known as
example, as a transplacental carcinogen, the exposure to DES con- progestogens, named for their function in gestation (pro-gesta-
tributed to a significant rise in the development of rare vaginal tion). Progesterone is produced predominantly in the gonads and
tumors (Herbst et al., 1971). Animal studies showed that DES has adrenal gland, and in pregnancy in the placenta. While critically
a profound effect on immune responses (Luster et al., 1978), involved in pregnancy, detectable levels of progesterone are in cir-
confirming fears that DES exposure may have contribute to auto- culation in females throughout the menstrual cycle and in males in
immune diseases in humans. However, while confirming that levels similar to that seen in females during the follicular phase of
females exposed to DES developed a number of autoimmune disor- the menstrual cycle. The actions of progesterone in reproduction
ders, the incidence was relatively low (Noller et al., 1988). This was are varied. In addition to directly regulating reproductive function
verified in more recent observations (Strohsnitter et al., 2010), (including oocyte maturation, endometrial differentiation, embry-
where the relative rise in incidence in RA in DES exposed females onic implantation, placental growth etc.), progesterone may also
below 45 years of age was offset by a much lower incidence in alter the actions of other steroid hormones at this time (Steyn
older females. Thus, while animal studies demonstrate a significant et al., 2008). Accordingly, interaction of progesterone with target
risk for autoimmunity following DES exposure, data from humans tissues may occur via indirect mechanisms.
provide little support for an association between the development Progesterone acts via its cytosolic receptor, the progesterone
of an autoimmune disease and exposure to DES. This important receptor (PR). This receptor belongs to the nuclear hormone recep-
case highlights the need for rigorous assessment and careful tor superfamily of transcription factors. Ligand binding of PR
validation of autoimmune incidence following exogenous estrogen induces changes in gene expression through direct binding to pro-
exposure. This is of considerable importance, as estrogen-mediated moter elements or through protein–protein interactions with other
therapies may effectively alleviate complications with autoim- transcription factors. Of interest, recent observations highlight that
mune diseases. In this instance, unsubstantiated concerns regard- actions of progestins (including progesterone) may occur indepen-
ing the use of estrogen-containing oral contraceptives may in dent of the PR, therefore arguing for non-genomic interactions (Li
fact be to the detriment of women suffering from autoimmune et al., 2004). As summarized by Hughes (2012), the actions of pro-
conditions, and in particular SLE. gesterone on immunity are best described by the wide distribution
While the potential benefits of estrogen therapy in some auto- of PRs on immune cells, including granulocytes, NK cells, dendritic
immune diseases are recognized, it should be noted that conflicting cells, T-cells and B cells (Hughes, 2012), although non-genomic
observations make the interpretation of the extent of potential interactions may occur.
benefits difficult. Moreover, identification of potential beneficial In general, the effects of progesterone in immunity are anti-
effects of estrogen on autoimmune disease requires careful consid- inflammatory. Of these, known effects include the promotion of
eration of the type and severity of the condition. For example, apoptosis and inhibition of IFNc production from NK cells
while estrogen therapy in MS (Sicotte et al., 2002) and RA (Arruvito et al., 2008), the inhibition of macrophage activity
(Bijlsma et al., 1987) may be beneficial, estrogen appears to (Pisetsky and Spencer, 2011), the modulation of myeloid dendritic
enhance disease severity in SLE. Thus, blockade of ER in SLE is cell activity (reviewed in (Hughes and Clark, 2007)), the inhibition
beneficial (Abdou et al., 2008). Similarly, while the use of contra- of glucocorticoid-mediated thymocyte apoptosis (McMurray et al.,
ceptives may protect against RA (Vandenbroucke et al., 1982), 2000), the reduction of nitric oxide production (Miller et al., 1996)
some evidence suggest that the beneficial effects are limited to and the expression of toll-like receptors (TLR) by macrophages
females under 35 years of age (Spector et al., 1990), or to females (Jones et al., 2008), the differentiation of Th2 cells in vitro
that used contraceptives earlier in life (Hazes et al., 1990). In this (Piccinni et al., 1995), and the expression of the co-stimulatory
latter study, it was concluded that current use in older females molecules CD80, CD86 and CD40, as well as MHC II (Ivanova
has no benefits. Accordingly, estrogen-containing contraceptives et al., 2005; Liang et al., 2006). The role of progesterone in immu-
for the prevention of RA may only be of some benefit to females nity was recently reviewed (Hughes, 2012).
at childbearing age. Studies that directly assess this are however As with estrogens the potential actions of progesterone on
scarce, and current observations are not promising. For example, autoimmunity are more commonly defined in context of fluctua-
estrogen treatment (using lynestrenol plus ethinyl estradiol) of tions in endogenous progesterone levels, dictated by changes in
S.T. Ngo et al. / Frontiers in Neuroendocrinology 35 (2014) 347–369 357

reproductive function. The rise in progesterone in pregnancy is significantly improve disease severity (Gordon et al., 2008),
associated with remarkable changes in maternal immunity, and whereas the impact of dehydroepiandrosterone (DHEA) treatment
may contribute to changes in immune responses. In general, the in SLE was inconsistent. For example, while initial reports showed
effects of progesterone in immunity are anti-inflammatory. As with that oral DHEA treatment (200 mg/day for 6 months) improved SLE
estrogens, the potential effects of progesterone may also vary rel- Disease Activity Index (SLEDAI) scores and overall assessment
ative to disease severity and type. For example, progesterone pro- scores in patients (van Vollenhoven et al., 1994), subsequent dou-
tects against disease onset in gonadectomised lupus-prone NZB/W ble-blinded studies from the same group were unable to corrobo-
mice (a model wherein mice spontaneously develop an autoim- rate these results (van Vollenhoven et al., 1995, 1999). Of
mune disorder similar to SLE) (Roubinian et al., 1978), whereas interest, a larger multicenter randomized, double-blind placebo
in CIA rats (a common rat model for arthritis), progesterone alone controlled trial showed significant improvements in assessment
had no effect while reducing the beneficial effects of estrogen scores from DHEA treated female patients (200 mg/day for
(Ganesan et al., 2008). Similarly, continuous progesterone adminis- 6 months), corresponding with a significant reduction in disease
tration to pre-autoimmune NZB/W mice delayed the development flares (Chang et al., 2002). Importantly, DHEA is a precursor of both
of pathogenic serum anti-DNA antibodies and spontaneous auto- androgens and estrogens, and thus these modest effects may not
immune disease and death (Hughes and Clark, 2007). In humans, necessarily involve direct androgenic roles. In light of potential
combined estrogen and progesterone hormone replacement negative effects on HDL cholesterol (Casson et al., 1998), coupled
therapy may cause lupus flares in post-menopausal women with the increased risk of developing atherosclerotic heart disease
(Buyon et al., 2005), whereas a current clinical trial is assessing in SLE patients (van Leuven et al., 2006), it is critical that the long-
the potential benefits of progestin treatment to prevent post- term ramification on cardiovascular health be considered before
partum flares in women with MS (Vukusic et al., 2009) (Results recommending DHEA as a treatment option for SLE.
from this trial are pending (ClinicalTrials.gov Identifier:
NCT00127075)). Given this variable response, substantive research 3.4.4. Prolactin
is needed before we can define the contribution of or treatment of While predominantly secreted from the anterior pituitary gland,
autoimmune diseases using therapies targeting progesterone. immune cells including lymphocytes secrete prolactin (Chavez-
Rueda et al., 2005; Vera-Lastra et al., 2002). The prolactin receptor,
3.4.3. Androgens a member of the cytokine receptor superfamily, is found on mono-
Androgens are steroid hormones commonly associated with the cytes and T lymphocytes, and other cells of the immune system
development of masculine characteristics; implicated in testes for- (Jara et al., 1991). Thus, while prolactin predominantly acts on the
mation, spermatogenesis, and the deposition of lean muscle mass brain and within the mammary gland to ensure physiological adap-
at the expense of adipose. In males and females, androgens main- tation essential for sustained maternal behavior and lactation
tain bone mass while impacting behavior (including aggression). (Grattan et al., 2008), it may play an important role in immunity,
Importantly, androgens are the precursors of all estrogens. While and thus pregnancy associated changes in autoimmunity.
generally present in high levels in males, androgens decline with Functionally, the paracrine actions of prolactin produced within
increasing age (Mitchell et al., 1995), whereas low circulating lev- lymphocytes may mediate important immunological effects. For
els of androgens persist throughout life in females (Davison et al., example, prolactin is able to induce the proliferation of lympho-
2005). As with estrogen and progesterone, androgens may act via cytes, whereas inactivation of prolactin action through adminstra-
genomic (acting through the androgen receptor) and non-genomic tion of neutralizing antibodies significantly decreases the
mechanisms (Lutz et al., 2003). activation and proliferation of T lymphocytes (Chavez-Rueda
The actions of androgens on immune function may vary et al., 2005). Within the T lymphocytes, prolactin mediates T-bet,
depending on the type of androgen used, the dosage administered, an essential transcription factor involved in the production of
and the timing of administration. For example, some studies report Th1-type cytokines, including IFN (Tomio et al., 2008). Thus, prolac-
immunosuppressive effects (Fujii et al., 1975; Olsen and Kovacs, tin may stimulate T-cell mediated inflammatory reactions (Tomio
1996; Paavonen, 1994), whereas endogenous androgens are also et al., 2008). However, at high concentrations, prolactin may inhibit
thought to be immunostimulatory (Calabrese et al., 1989). Testos- the proinflammatory activation of T helper cells (Tomio et al., 2008).
terone reduces the proliferation and differentiation of lymphocytes Prolactin may enhance autoimmune disease, whereas serum
(Sakiani et al., 2013; Sthoeger et al., 1988; Wyle and Kent, 1977), levels of prolactin may increase as a consequences of autoimmune
and the use of androgens may suppress immunoglobulins, and in disease (Orbach and Shoenfeld, 2007). For example, hyperprolacti-
particular IgA (Calabrese et al., 1989; Hirota et al., 1976). Andro- nemia is seen in autoimmune rheumatic diseases (ARD) including
gens, including supraphysiological doses of testosterone may inhi- SLE, RA, systemic sclerosis, SS, T1D, GD, HT, celiac disease, and MS
bit the cytotoxic activity of NK cells (Callewaert et al., 1991; (Orbach et al., 2012). Elevated levels of prolactin was found in the
Marshall-Gradisnik et al., 2008; Sulke et al., 1985). Given that the CSF in SLE patients with neuropsychiatric lupus (Jara et al., 1998),
effects of androgens may vary considerably relative to the level while hyperprolactinemia is associated with activity of SLE in preg-
of exposure, and that multiple factors may contribute to the nancy (Jara et al., 2001). Of interest, hyperprolactinemia in patients
immune profile in females, the potential role of endogenous andro- with pituitary tumors or following therapy with antipsychotic
gens in gender specific immune function remains unknown. drugs is associated with raised levels of thyroid autoantibodies
Androgen levels are higher in males, and autoimmune incidence (Poyraz et al., 2008). Associations between prolactin and autoim-
is generally lower. Thus, it is thought that androgens may to some munity are somewhat corroborated by animal studies, wherein
extent protect against autoimmunity. Indeed, gonadectomy of prolactin treatment often worsens disease severity. For example,
male NZB/W mice result in overt autoimmunity and earlier death, disease severity in NZB/W mice increases following prolactin treat-
whereas treatment with androgens in ovariectomised female NZB/ ment, and improves following treatment with bromocriptine (an
W mice reduced mortality (Roubinian et al., 1977). In humans, ergoline derivative used to suppress endogenous prolactin produc-
transdermal treatment with testosterone for 1 year was of some tion) (McMurray et al., 1991).
benefit to men with MS, slowing cognitive decline and brain atro- Attempts to treat human autoimmune disease with bromocrip-
phy. Treatment, however, did not affect the number or volume of tine have produced some positive results. Notably, in some SLE
enhancing lesions (Sicotte et al., 2007). In females with SLE, patients bromocriptine treatment reduces disease flares (Alvarez-
testosterone administration via skin patches for 12 weeks did not Nemegyei et al., 1998; McMurray et al., 1995). More recently, it
358 S.T. Ngo et al. / Frontiers in Neuroendocrinology 35 (2014) 347–369

was found that bromocriptine treatment in postpartum lupus Vitamin D is an environmental factor that may underlie the
patients reduce disease flares (Yang et al., 2003), whereas this pilot development of autoimmunity. Vitamin D has immunomodulatory
clinical data suggest that bromocriptine treatment may prevent effects on T lymphocytes, B lymphocytes, and dendritic cells
complications in pregnancy including preterm birth and active dis- (Arnson et al., 2007; Deluca and Cantorna, 2001). By signaling
ease (Jara et al., 2007). Given the potential value of bromocriptine through the vitamin D receptor, vitamin D reduces the effector T
and other prolactin antagonists in the treatment of autoimmunity, cell response (Adorini, 2005; Gascon-Barre et al., 2003; Godfrey
further studies to delineate the mechanisms by which PRL affects et al., 2000; Lemire, 1992). Vitamin D also interacts with human
autoimmune disease activity are of critical importance. leukocyte antigen molecules, which predispose to MS
(Ramagopalan et al., 2009a). Evidence to support the a role for
sun exposure in autoimmunity is seen in SS (Erten et al., 2014),
3.5. The environment
RA (Gatenby et al., 2013), SLE (Kamen et al., 2006), and anti-
phospholipid syndrome (Andreoli et al., 2012; Piantoni et al.,
Environmental exposure can interact with the genotype of
2012) where vitamin D deficiency is associated with disease or
(Selmi et al., 2012), and can influence the prevalence and influence
increased risk of developing disease. Indeed, greater vitamin D3
the risk of developing an autoimmune disease or the severity of
synthetic rate resulting from higher intensity ultraviolet light at
disease. Environmental candidates that have been proposed to play
high altitudes, may account for lower MS rates at higher altitudes
a role in autoimmune diseases include infectious agents and chem-
(Hayes et al., 1997). It must be noted however, that within any geo-
icals (Chandran and Raychaudhuri, 2010; Costenbader et al., 2012;
graphical location, exposure to sunlight between men and women
Hemminki et al., 2010). Infectious agents are important in the
could vary, and this may be dependent on lifestyle factors, or occu-
mosaic of autoimmunity (Shoenfeld et al., 2008), and a number
pational choices. Given that men tend to have more unprotected
of autoimmune diseases have been linked to infection; rheumatic
sun exposure (Hall et al., 1997), gender-specific responses to sun-
fever presents after Streptococcus pyogenes infection (Fae et al.,
light may play a role in autoimmune disease. Another example of
2006), primary anti-phospholipid syndrome patients have high
differences in exposure to environmental triggers is the role of cos-
levels of IgM antibodies to rubella and Toxoplasma (Zinger et al.,
metics in PBC, where the exposure to cosmetics is much greater in
2009), diabetes is associated with Enterovirus (Richardson et al.,
women than men. Other environmental agents that are implicated
2009), and active infections with Helicobacter pylori have been
in autoimmunity and that may have differential exposures in
observed in autoimmune gastritis (Annibale et al., 2001). Chemi-
males and females are mercury, pesticides and solvents (Pollard,
cal/drug exposure, and immunotoxicity of such compounds are
2012).
linked to autoimmune disease. Chemicals that are commonly
found in cosmetics are associated with PBC (Rieger et al., 2006),
and acetaminophen may underlie the development of PBC by pro- 4. Genetic aspects of the sexual dimorphism in autoimmunity
moting antimitochondrial autoantibodies (Leung et al., 2013).
Exposure to pesticides has been suggested to underlie the develop- Genetic factors may contribute to the sexual dimorphism that is
ment of autoantibodies (McConnachie and Zahalsky, 1991, 1992; seen in autoimmune disease. Such factors that might influence the
Thrasher et al., 1993), and lifetime exposure to insecticides is asso- development of autoimmune disease may be related to susceptibil-
ciated with the presence of anti-nuclear antibodies (Rosenberg ity genes, chromosomal differences, or epigenetics. The presenta-
et al., 1999). More recently, it has been found that exposure to tion of multiple autoimmune diseases in a given individual or
organic solvents is associated with an increased risk of developing family suggests that common genetic factors may prejudice an
an autoimmune trait (Barragan-Martinez et al., 2012). Since these individual to developing autoimmunity. The clustering of suscepti-
exposures underlie the development of autoimmunity, gender dif- bility genes in conditions like MS and CD suggests that in some
ferences could occur if males and females have different types of cases, autoimmune diseases may be controlled by a set of suscep-
exposure or different biological response to these agents (Fig. 5). tibility genes (Becker et al., 1998).
We now discuss several examples.
4.1. Interaction of genes and gender

The HLA genes are located in a region that includes a large


number of genes that are involved in the immune response. Asso-
ciations exist between HLA genes and a number of autoimmune
diseases including GD (Li and Chen, 2013), MG (Avidan et al.,
2013), MS (Cree, 2014), RA (Jin et al., 2014), and SLE (Deng et al.,
2013) amongst many others (Horton et al., 2004). The majority of
these associations are with HLA-DR and HLA-DQ genes (Gough
and Simmonds, 2007), which encode for proteins that play a cen-
tral role in presenting antigens to CD4 + T cells (Schendel and
Johnson, 1985). The association between HLA genes and autoim-
mune disease generally shows a gender bias toward females. In
MG, single nucleotide polymorphisms in the HLA-locus support
the premise that there is a female gender bias for increased risk
for MG (Avidan et al., 2013). Similarly, being female may further
the risk associated with HLA DR4 alleles in type 1 autoimmune
hepatitis, since females have a higher frequency of HLA DR4 than
Fig. 5. The role of the environment in the sexual dimorphism in autoimmunity. men (Czaja and Donaldson, 2002). In MS, there is a strong associa-
Men and women are exposed to environmental factors to varying degrees (e.g. men tion between HLA-DR2, DQ6 in females (Celius et al., 2000), and in
tend to get more sunlight than women), and they have different physiological
responses to such factors. This differential exposure and response might influence
chronic inflammatory demyelinating polyneuropathy the associa-
the prevalence and the risk of developing an autoimmune disease, or the severity of tion with HLA-DR2 is greater for females (Czaja and Donaldson,
disease between men and women. 2002). In RA, there is an association between disease and females
S.T. Ngo et al. / Frontiers in Neuroendocrinology 35 (2014) 347–369 359

with a HLA DRB1 *0401/*0401 genotype (Meyer et al., 1996). In SLE development of autoimmune disease. Skewed X chromosome inac-
however, there is a reversal in gender bias, with a higher HLA asso- tivation is observed in HT, GD (Brix et al., 2005; Chabchoub et al.,
ciated genetic risk in men (Hughes et al., 2012). 2009; Ozcelik et al., 2006), sclerodoma (Ozbalkan et al., 2005; Uz
Non-HLA genes are also associated with susceptibility to auto- et al., 2008), RA (Chabchoub et al., 2009), and SS (Ozcelik, 2008),
immune disease. Polymorphisms in cytotoxic T-lymphocyte anti- but not in SLE (Huang et al., 1997) and MS (Knudsen, 2009;
gen-4 (CTLA-4), acid phosphatase locus 1 (ACP1), Discs large Knudsen et al., 2007). Data from a number of studies suggest that
homolog 5 (DLG5), interleukin-10 (IL-10), and apolipoprotein E X chromosome monosomy rates may also underlie the female pre-
(APOE) are commonly observed. Genetic A/G polymorphisms in dominance in autoimmune diseases. In disease such as PBC, HT,
CTLA-4 have been identified in AS (Huang et al., 2014), GD (Du and GD, enhanced X monosomy rates have been observed in
et al., 2013), and T/C polymorphisms may be associated with SLE peripheral blood cells (Invernizzi et al., 2004, 2005), but this is
risk (Zhu et al., 2013). By contrast, there is no evidence to suggest not the case for SLE (Invernizzi et al., 2007). Skewed X chromosome
that CTLA-4 polymorphisms confer susceptibility to MS (Gyu Song inactivation and monosomy appear to be more common in autoim-
and Ho Lee, 2013). While the majority of studies that investigate mune thyroid diseases (Brix et al., 2005; Chabchoub et al., 2009;
CTLA-4 polymorphisms in autoimmune disease do not necessarily Invernizzi et al., 2005; Ozcelik et al., 2006).
consider results relative to gender, CTLA-4 A/G polymorphisms Males with two X chromosomes (Chagnon et al., 2006), or males
can modify maternal inheritance of the HLA-DR3 (Fajardy et al., with two X chromosomes and a Y chromosome have also been
2002). known to present with SLE (Lahita, 1999). Interestingly, SLE males
Polymorphisms in ACP1 and DLG5 are associated with CD that are XXY, are similar to female SLE patients in that they have
(Browning et al., 2008; Stoll et al., 2004), and there is evidence of increased oxidation of testosterone (Lahita, 1999). These observa-
a reduced susceptibility to CD in females who possess ACP1*A tions support a role for the X chromosome in SLE.
alleles (Gloria-Bottini et al., 2007), and modest evidence of reduced
risk of CD in females with the DLG5 R30Q variant (Browning et al., 4.3. The Y chromosome in autoimmune disease
2008). Polymorphisms in IL-10 are linked to severity of HT (Inoue
et al., 2013), RA (Hee et al., 2007), and primary SS (Qin et al., Compared to the X chromosome, the Y chromosome has
2013). While these studies do not stratify risk according to gender, received little attention in human autoimmunity. The Y chromo-
an AA -1087 IL-10 genotype appears to be more common in women some accounts for a small portion of the human genome, and is
with RA (Padyukov et al., 2004). Polymorphisms in APOE are asso- associated with male fertility and testis formation (Quintana-
ciated with primary SS (Pertovaara et al., 2004) and MS (Cocco Murci et al., 2001). While the Y chromosome is linked to the inher-
et al., 2005; Rafiei et al., 2012). Again, while not considered relative itance of human coronary heart disease (Charchar et al., 2012), the
to gender, female carriers of the apolipoprotein E epsilon4 allele majority of studies that have assessed the contribution of the Y
had a tendency for earlier onset of primary SS when compared to chromosome to autoimmunity has been conducted in mouse mod-
female non-carriers (Pertovaara et al., 2004). In the case of MS els of SLE (Lin et al., 2010; Murphy and Roths, 1979; Santiago-
however, presence of the APOE alleles seems to favor females; Raber et al., 2008; Subramanian et al., 2006) and MS (Palaszynski
women carriers of the epsilon2 allele had reduced severity of dis- et al., 2005; Spach et al., 2009; Teuscher et al., 2006) (reviewed
ease (Kantarci et al., 2004), and cognitive decline is more promi- in (McCombe et al., 2009)). Recent studies have found an age-
nent in males who carry the epsilon4 allele (Savettieri et al., 2004). dependent loss of the Y chromosome in PBC (Lleo et al., 2013),
The observations from these studies provide strong evidence to and in HT and GD (Persani et al., 2012). These observations further
support the notion that genetic risks underlie susceptibility to support the role of the X chromosome in autoimmune disease.
some autoimmune diseases. While a large number of studies focus
on the involvement of HLA genes, it is becoming strikingly clear
that polymorphisms in non-HLA genes should also be taken into 5. Parental inheritance/transmission in autoimmunity
account when considering the relationship between genetic factors
that predispose to autoimmune disease. Given the strong associa- Maternal and paternal inheritance occurs in autoimmune dis-
tions between HLA genes and non-HLA genes and autoimmune dis- ease. While one study in Canada has found that there is equal
ease, future studies that investigate the genetic underpinning of inheritance of MS from affected fathers and mothers (Herrera
such diseases should be considered relative to gender. et al., 2007), and paternal inheritance in MS has been reported
(Marrosu et al., 2004), the majority of studies assessing parent-
4.2. The X chromosome in autoimmune disease of-origin effects in MS are more indicative of a strong maternal
effect for risk of inheritance (Ebers et al., 2004), and predominant
The X chromosome contains a large number of genes that are maternal inheritance (Chao et al., 2010; Ebers et al., 2004;
involved in immunity (reviewed in (Fish, 2008; Libert et al., Herrera et al., 2008; Hoppenbrouwers et al., 2008). Inheritance of
2010)). While females carry two X chromosomes, one X chromo- autoimmune disease in T1D however, takes on a paternal domi-
some is inactivated early in embryogenesis. This allows for equal nance (MacDonald et al., 1986). Islet autoimmune antibodies are
gene expression dosage between males and females, but also more frequently transmitted via diabetic fathers (Yu et al., 1995),
results in cellular mosaicism (Migeon, 2006). Consequently, paternal inheritance of the insulin gene B allele confers protection
females may avoid any effects that arise from harmful gene-muta- from T1D (Pugliese et al., 1994), while paternal inheritance of the
tions, and cellular mosaicism (arising from natural variations in 11p15.5 region of the insulin gene (IDDM2) confers susceptibility
one gene that might result in two different alleles) may result in to T1D (Bui et al., 1996).
added advantages in response to immune challenges (Migeon,
2006; Spolarics, 2007). 5.1. Causative factors for parental inheritance of autoimmunity
Autoimmune phenomena are observed in X-linked primary
immunodeficiencies. Thus it is likely that genes on the X chromo- The interaction of genetic and environmental factors may
some may also play a role in autoimmunity (Bussone and underlie parental inheritance of autoimmunity. In the case of
Mouthon, 2009; Carneiro-Sampaio and Coutinho, 2007; Mackay maternal inheritance, mitochondrial DNA could play a role. While
and Rose, 2001). It has been suggested that X chromosome inacti- studies addressing mitochondrial DNA variants with autoimmune
vation, loss of X chromosomes or X monosomy may underlie the disease are limited, MS has been associated with the mitochondrial
360 S.T. Ngo et al. / Frontiers in Neuroendocrinology 35 (2014) 347–369

haplotype K (Vyshkina et al., 2008) and haplotype U (Ban et al., (Sugiyama et al., 2010), and increased risk of MS (in women stud-
2008). Interestingly, variants in mitochondrial ND2 and ATP6 ied) (Hernan et al., 2001). Moreover, smoking has been found to be
genes have also been found to be associated with MS and SLE, sug- a risk factor for relapsing–remitting MS (Hernan et al., 2005). Expo-
gesting that these two diseases may share a common mitochon- sure to lower levels of sunlight (Hammond et al., 1988; Kurtzke
drial genetic background. et al., 1979; Taylor et al., 2010; van der Mei et al., 2003; Vukusic
In addition to genetic factors, environmental factors may also et al., 2007), season of birth (Dobson et al., 2013; Willer et al.,
contribute to parental inheritance of autoimmunity 2005), low maternal exposure to sunlight (Staples et al., 2010),
(Hoppenbrouwers et al., 2008). Some of the best examples of envi- and low levels of vitamin D (van der Mei et al., 2007) have been
ronmental influence in parental transmission in autoimmunity well established to be associated with increased risk for MS. Recent
have come from studies in MS cohorts. The development of MS studies assessing dietary intake of vitamin D in women with MS
in affected offspring may arise from in utero exposure to epigenetic report that higher levels of vitamin D are associated with reduced
changes that might result from environmental influences on the risk (Munger et al., 2004), and assessment of micronutrient intake
mother (e.g. timing of birth is associated with risk of MS) (Willer has provided insight into the geographical blueprint of autoim-
et al., 2005). This is discussed further below. mune disease in China (Qin et al., 2009). While it is easy to specu-
late that varying exposure to extrinsic triggers would occur
between males and females, earlier adoption of smoking in men
6. Epigenetics in autoimmunity
(Zang and Wynder, 1996), increased unprotected exposure to sun-
light in men (Hall et al., 1997), and different dietary habits
Epigenetics relates to the stable and heritable patterns of gene
between men and women (Dynesen et al., 2003; Ferraro and
expression that are not due to changes in the original DNA
Vieira, 2010; Friel et al., 2005) would support the notion that dif-
sequence. Altered gene expression may be persistent, but not
ferential exposure to extrinsic factors may play a role in gender
permanent, and the epigenome of the cell can be defined by meth-
bias in autoimmune disease.
ylation, histone modification and micro RNA (miRNA) profile
(Goldberg et al., 2007; Jiang et al., 2004; Lu, 2013). Epigenetic
changes that arise in autoimmune disease can be related to the 7. Conclusion
sex of the parent, or can result from external factors. With respect
to parental gender, epigenetic changes occur via genetic imprinting, The gender differences that exist in autoimmunity tend to fol-
whereby genes inherited either from the father or mother are low a female bias. A number of factors may underlie this striking
imprinted and inactivated, or when transcripts from either the gender difference. The clear differences between the female and
maternal or paternal chromosome are expressed in a monoallelic male immune systems would suggest that the increased immune
fashion (Camprubi and Monk, 2011). Extrinsic influences in autoim- reactivity in females might predispose them to developing autoim-
mune disease include tobacco smoke, exposure to sunlight, and diet mune disease. This may be explained in part by the effects of sex
(Ellis et al., 2014). Below, we discuss evidence that indicates a role hormones since estrogen suppresses Th1-dependent disease, but
for genomic imprinting and epigenetics in driving sexual dimor- potentiates Th2-dependent diseases. Accordingly, differences in
phism in autoimmune disease. reproductive function, and pregnancy in women, may also explain
gender differences in autoimmunity. While the impact of micro-
6.1. Genomic imprinting as a cause for autoimmunity chimerism during pregnancy in autoimmune disease remains to
be understood, parental inheritance, mitochondrial inheritance,
Genomic imprinting is the allele-specific expression of genes in genomic imprinting and chromosomal inactivation could also play
a parent-of-origin fashion. It has been suggested that the genetics a role. Finally, extrinsic epigenetic influences and more specifically,
underlying HLA susceptibility in T1D may exhibit parental inheri- the level of exposure to external factors and the subsequent
tance; Pani and colleagues suggest that the maternal HLA-DQ2 or response to such factors might influence susceptibility to autoim-
DQ8 alleles are risk factors for T1D (Pani et al., 2002). For psoriatic mune disease. In conclusion, the striking evidence of gender differ-
arthritis, genetic linkage analysis suggests that there is imprinted ences in autoimmune disease should dictate that future studies
transmission at chromosome 16q that is of paternal origin considering incidence, prevalence, severity, mortality, disease
(Karason et al., 2003). mechanisms and potential therapies in autoimmunity be stratified
Imprinting of miRNAs may also underlie autoimmunity. Micro according to gender.
RNAs play important roles in the post-transcriptional regulation
of gene expression. In this regard, a loss of miRNAs or the deregu- Acknowledgment
lation of miRNAs could contribute to altered autoimmune gene
expression that would result in the development of autoimmune Dr. S. Ngo acknowledges the support of the Motor Neurone
disease. Indeed, imprinted miRNAs that may regulate autoimmune Disease Research Institute of Australia.
disease associated genes have been identified for MS, RA, SLE, and
T1D (Camprubi and Monk, 2011). Given that there is differential
hormonal and chromosomal regulation of miRNAs between males References
and females (Sharma and Eghbali, 2014), it is highly likely that Abdou, N.I., Rider, V., Greenwell, C., Li, X., Kimler, B.F., 2008. Fulvestrant (Faslodex),
miRNA imprinting could contribute to the sexual dimorphism seen an estrogen selective receptor downregulator, in therapy of women with
in autoimmune disease. systemic lupus erythematosus. clinical, serologic, bone density, and T cell
activation marker studies: a double-blind placebo-controlled trial. J. Rheumatol.
35, 797.
6.2. Extrinsically driven epigenetic gene modifications in Adams Waldorf, K.M., Nelson, J.L., 2008. Autoimmune disease during pregnancy and
the microchimerism legacy of pregnancy. Immunol. Invest. 37, 631–644.
autoimmunity
Adorini, L., 2005. Intervention in autoimmunity: the potential of vitamin D receptor
agonists. Cell. Immunol. 233, 115–124.
External stimuli including tobacco smoke, sunlight, and diet Aggarwal, R., Malaviya, A.N., 2009. Clinical characteristics of patients with
that lead to DNA modification could explain gender differences in ankylosing spondylitis in India. Clin. Rheumatol. 28, 1199–1205.
Agret, F., Cosnes, J., Hassani, Z., Gornet, J.M., Gendre, J.P., Lemann, M., Beaugerie, L.,
autoimmune disease. Smoking is associated with an increased risk 2005. Impact of pregnancy on the clinical activity of Crohn’s disease. Aliment.
for RA in both men and women when compared to non-smokers Pharmacol. Ther. 21, 509–513.
S.T. Ngo et al. / Frontiers in Neuroendocrinology 35 (2014) 347–369 361

Al-Chalabi, T., Underhill, J.A., Portmann, B.C., McFarlane, I.G., Heneghan, M.A., 2008. Bentzen, J., Flachs, E.M., Stenager, E., Bronnum-Hansen, H., Koch-Henriksen, N.,
Impact of gender on the long-term outcome and survival of patients with 2010. Prevalence of multiple sclerosis in Denmark 1950–2005. Multip. Scl. 16,
autoimmune hepatitis. J. Hepatol. 48, 140–147. 520–525.
Alshekhlee, A., Hussain, Z., Sultan, B., Katirji, B., 2008. Guillain–Barre syndrome: Beretta, L., Caronni, M., Origgi, L., Ponti, A., Santaniello, A., Scorza, R., 2006. Hormone
incidence and mortality rates in US hospitals. Neurology 70, 1608–1613. replacement therapy may prevent the development of isolated pulmonary
Aluvihare, V.R., Kallikourdis, M., Betz, A.G., 2004. Regulatory T cells mediate hypertension in patients with systemic sclerosis and limited cutaneous
maternal tolerance to the fetus. Nat. Immunol. 5, 266–271. involvement. Scand. J. Rheumatol. 35, 468–471.
Alvarez-Nemegyei, J., Cobarrubias-Cobos, A., Escalante-Triay, F., Sosa-Munoz, J., Bernatsky, S., Boivin, J.F., Joseph, L., Manzi, S., Ginzler, E., Gladman, D.D., Urowitz, M.,
Miranda, J.M., Jara, L.J., 1998. Bromocriptine in systemic lupus erythematosus: a Fortin, P.R., Petri, M., Barr, S., Gordon, C., Bae, S.C., Isenberg, D., Zoma, A.,
double-blind, randomized, placebo-controlled study. Lupus 7, 414–419. Aranow, C., Dooley, M.A., Nived, O., Sturfelt, G., Steinsson, K., Alarcon, G.,
Amarapurkar, D.N., Patel, N.D., 2007. Spectrum of autoimmune liver diseases in Senecal, J.L., Zummer, M., Hanly, J., Ensworth, S., Pope, J., Edworthy, S., Rahman,
western India. J. Gastroenterol. Hepatol. 22, 2112–2117. A., Sibley, J., El-Gabalawy, H., McCarthy, T., St Pierre, Y., Clarke, A., Ramsey-
Anderson, S.G., Narayanan, R.P., Amlesh, J., Qureshi, M.Z., Heald, A.H., 2012. Type 1 Goldman, R., 2006. Mortality in systemic lupus erythematosus. Arthritis Rheum.
diabetes in Cheshire: cardiometabolic risk factor trends (2004–2009). Primary 54, 2550–2557.
Diabet. 6, 123–126. Bhadada, S.K., Kochhar, R., Bhansali, A., Dutta, U., Kumar, P.R., Poornachandra, K.S.,
Andreoli, L., Piantoni, S., Dall’Ara, F., Allegri, F., Meroni, P.L., Tincani, A., 2012. Vaiphei, K., Nain, C.K., Singh, K., 2011. Prevalence and clinical profile of celiac
Vitamin D and antiphospholipid syndrome. Lupus 21, 736–740. disease in type 1 diabetes mellitus in north India. J. Gastroenterol. Hepatol. 26,
Annibale, B., Negrini, R., Caruana, P., Lahner, E., Grossi, C., Bordi, C., Delle Fave, G., 378–381.
2001. Two-thirds of atrophic body gastritis patients have evidence of Bhattacharya, M., Kapoor, S., Dubey, A.P., 2013. Celiac disease presentation in a
Helicobacter pylori infection. Helicobacter 6, 225–233. tertiary referral centre in India: current scenario. Ind. J. Gastroenterol.: Off. J.
Ansar Ahmed, S., Penhale, W.J., Talal, N., 1985. Sex hormones, immune responses, Ind. Soc. Gastroenterol. 32, 98–102.
and autoimmune diseases. Mechanisms of sex hormone action. Am. J. Pathol. Bianchi, D.W., Zickwolf, G.K., Weil, G.J., Sylvester, S., DeMaria, M.A., 1996. Male fetal
121, 531–551. progenitor cells persist in maternal blood for as long as 27 years postpartum.
Arnson, Y., Amital, H., Shoenfeld, Y., 2007. Vitamin D and autoimmunity: new Proc. Nat. Acad. Sci. U.S.A. 93, 705–708.
aetiological and therapeutic considerations. Ann. Rheum. Dis. 66, 1137–1142. Bias, W.B., Reveille, J.D., Beaty, T.H., Meyers, D.A., Arnett, F.C., 1986. Evidence that
Arruvito, L., Giulianelli, S., Flores, A.C., Paladino, N., Barboza, M., Lanari, C., Fainboim, autoimmunity in man is a Mendelian dominant trait. Am. J. Hum. Genet. 39,
L., 2008. NK cells expressing a progesterone receptor are susceptible to 584–602.
progesterone-induced apoptosis. J. Immunol. 180, 5746–5753. Bijlsma, J.W., Huber-Bruning, O., Thijssen, J.H., 1987. Effect of oestrogen treatment
Artlett, C.M., Smith, J.B., Jimenez, S.A., 1998. Identification of fetal DNA and cells in on clinical and laboratory manifestations of rheumatoid arthritis. Ann. Rheum.
skin lesions from women with systemic sclerosis. New Engl. J. Med. 338, 1186– Dis. 46, 777–779.
1191. Biver, E., Beague, V., Verloop, D., Mollet, D., Lajugie, D., Baudens, G., Neirinck, P.,
Asakura, K., Nishiwaki, Y., Inoue, N., Hibi, T., Watanabe, M., Takebayashi, T., 2009. Flipo, R.M., 2009. Low and stable prevalence of rheumatoid arthritis in northern
Prevalence of ulcerative colitis and Crohn’s disease in Japan. J. Gastroenterol. 44, France. Joint, Bone, Spine : Revue du Rhumatisme 76, 497–500.
659–665. Blum, S., Reddel, S., Spies, J., McCombe, P., 2013. Clinical features of patients with
Asao, K., Sarti, C., Forsen, T., Hyttinen, V., Nishimura, R., Matsushima, M., Reunanen, Guillain–Barre syndrome at seven hospitals on the East Coast of Australia. J.
A., Tuomilehto, J., Tajima, N.Diabetes Epidemiology Research International Peripher. Nerv. Syst.: JPNS 18, 316–320.
Mortality Study, G., 2003. Long-term mortality in nationwide cohorts of Boddaert, J., Huong, D.L., Amoura, Z., Wechsler, B., Godeau, P., Piette, J.C., 2004. Late-
childhood-onset type 1 diabetes in Japan and Finland. Diabetes Care 26, onset systemic lupus erythematosus: a personal series of 47 patients and
2037–2042. pooled analysis of 714 cases in the literature. Medicine 83, 348–359.
Athreya, B.H., Pletcher, J., Zulian, F., Weiner, D.B., Williams, W.V., 1993. Subset- Boes, T., Levy, D., 2012. Influence of sex, estrous cycle, and estrogen on intracranial
specific effects of sex hormones and pituitary gonadotropins on human dural mast cells. Cephalalgia: Int. J. Headache 32, 924–931.
lymphocyte proliferation in vitro. Clin. Immunol. Immunopathol. 66, 201–211. Borchers, A.T., Naguwa, S.M., Keen, C.L., Gershwin, M.E., 2010. The implications of
Australia and New Zealand Multiple Sclerosis Genetics, C., 2009. Genome-wide autoimmunity and pregnancy. J. Autoimmun. 34, J287-99.
association study identifies new multiple sclerosis susceptibility loci on Bouman, A., Schipper, M., Heineman, M.J., Faas, M.M., 2004. Gender difference in the
chromosomes 12 and 20. Nat. Genet. 41, 824–828. non-specific and specific immune response in humans. Am. J. Reprod. Immunol.
Avidan, N., Le Panse, R., Berrih-Aknin, S., Miller, A., 2013. Genetic basis of 52, 19–26.
myasthenia gravis – a comprehensive review. J. Autoimmun., 11. Bouman, A., Heineman, M.J., Faas, M.M., 2005. Sex hormones and the immune
Bamigboye, A.A., Morris, J., 2003. Oestrogen Supplementation, Mainly response in humans. Hum. Reprod. Update 11, 411–423.
Diethylstilbestrol, for Preventing Miscarriages and Other Adverse Pregnancy Bove, R., 2013. Autoimmune diseases and reproductive aging. Clin. Immunol. 149,
Outcomes. The Cochrane Database of Systematic Reviews. CD004353. 251–264.
Ban, M., Elson, J., Walton, A., Turnbull, D., Compston, A., Chinnery, P., Sawcer, S., Brix, T.H., Knudsen, G.P., Kristiansen, M., Kyvik, K.O., Orstavik, K.H., Hegedus, L.,
2008. Investigation of the role of mitochondrial DNA in multiple sclerosis 2005. High frequency of skewed X-chromosome inactivation in females with
susceptibility. PLoS One 3, e2891. autoimmune thyroid disease: a possible explanation for the female
Banchereau, J., Steinman, R.M., 1998. Dendritic cells and the control of immunity. predisposition to thyroid autoimmunity. J. Clin. Endocrinol. Metab. 90, 5949–
Nature 392, 245–252. 5953.
Barragan-Martinez, C., Speck-Hernandez, C.A., Montoya-Ortiz, G., Mantilla, R.D., Browning, B.L., Annese, V., Barclay, M.L., Bingham, S.A., Brand, S., Buning, C., Castro,
Anaya, J.M., Rojas-Villarraga, A., 2012. Organic solvents as risk factor for M., Cucchiara, S., Dallapiccola, B., Drummond, H., Ferguson, L.R., Ferraris, A.,
autoimmune diseases: a systematic review and meta-analysis. PLoS One 7, Fisher, S.A., Gearry, R.B., Glas, J., Henckaerts, L., Huebner, C., Knafelz, D., Lakatos,
e51506. L., Lakatos, P.L., Latiano, A., Liu, X., Mathew, C., Muller-Myhsok, B., Newman,
Barrett, J.C., Clayton, D.G., Concannon, P., Akolkar, B., Cooper, J.D., Erlich, H.A., Julier, W.G., Nimmo, E.R., Noble, C.L., Palmieri, O., Parkes, M., Petermann, I., Rutgeerts,
C., Morahan, G., Nerup, J., Nierras, C., Plagnol, V., Pociot, F., Schuilenburg, H., P., Satsangi, J., Shelling, A.N., Siminovitch, K.A., Torok, H.P., Tremelling, M.,
Smyth, D.J., Stevens, H., Todd, J.A., Walker, N.M., Rich, S.S.Type 1 Diabetes Vermeire, S., Valvano, M.R., Witt, H., 2008. Gender-stratified analysis of DLG5
Genetics, C., 2009. Genome-wide association study and meta-analysis find that R30Q in 4707 patients with Crohn disease and 4973 controls from 12 Caucasian
over 40 loci affect risk of type 1 diabetes. Nat. Genet. 41, 703–707. cohorts. J. Med. Genet. 45, 36–42.
Basso, O., Campi, R., Frydenberg, M., Koch-Henriksen, N., Bronnum-Hansen, Brucato, A., Frassi, M., Franceschini, F., Cimaz, R., Faden, D., Pisoni, M.P., Muscara, M.,
H., Olsen, J., 2004. Multiple sclerosis in women having children by Vignati, G., Stramba-Badiale, M., Catelli, L., Lojacono, A., Cavazzana, I.,
multiple partners. A population-based study in Denmark. Multip. Scl. 10, Ghirardello, A., Vescovi, F., Gambari, P.F., Doria, A., Meroni, P.L., Tincani, A.,
621–625. 2001. Risk of congenital complete heart block in newborns of mothers with
Batocchi, A.P., Majolini, L., Evoli, A., Lino, M.M., Minisci, C., Tonali, P., 1999. Course anti-Ro/SSA antibodies detected by counterimmunoelectrophoresis: a
and treatment of myasthenia gravis during pregnancy. Neurology 52, 447–452. prospective study of 100 women. Arthritis Rheum. 44, 1832–1835.
Batra, C.M., 2013. Fetal and neonatal thyrotoxicosis. Ind. J. Endocrinol. Metabol. 17, Brunton, P.J., Russell, J.A., 2010. Endocrine induced changes in brain function during
S50–S54. pregnancy. Brain Res. 1364, 198–215.
Bayir, H., Marion, D.W., Puccio, A.M., Wisniewski, S.R., Janesko, K.L., Clark, R.S., Buckley, O.E., 1954. Introduction. In: Miner, R.W. (Ed.), The Status of Multiple
Kochanek, P.M., 2004. Marked gender effect on lipid peroxidation after severe Sclerosis. Ann NY Acad Sci, pp. 541–720.
traumatic brain injury in adult patients. J. Neurotrauma 21, 1–8. Bui, M.M., Luo, D.F., She, J.Y., Maclaren, N.K., Muir, A., Thomson, G., She, J.X., 1996.
Bebo Jr., B.F., Fyfe-Johnson, A., Adlard, K., Beam, A.G., Vandenbark, A.A., Offner, H., Paternally transmitted IDDM2 influences diabetes susceptibility despite
2001. Low-dose estrogen therapy ameliorates experimental autoimmune biallelic expression of the insulin gene in human pancreas. J. Autoimmun. 9,
encephalomyelitis in two different inbred mouse strains. J. Immunol. 166, 97–103.
2080–2089. Bulloch, K., Miller, M.M., Gal-Toth, J., Milner, T.A., Gottfried-Blackmore, A., Waters,
Becker, K.G., Simon, R.M., Bailey-Wilson, J.E., Freidlin, B., Biddison, W.E., McFarland, E.M., Kaunzner, U.W., Liu, K., Lindquist, R., Nussenzweig, M.C., Steinman, R.M.,
H.F., Trent, J.M., 1998. Clustering of non-major histocompatibility complex McEwen, B.S., 2008. CD11c/EYFP transgene illuminates a discrete network of
susceptibility candidate loci in human autoimmune diseases. Proc. Nat. Acad. dendritic cells within the embryonic, neonatal, adult, and injured mouse brain.
Sci. U.S.A. 95, 9979–9984. J. Comp. Neurol. 508, 687–710.
Ben-Chetrit, A., Ben-Chetrit, E., 1994. Systemic lupus erythematosus induced by Bulow Pedersen, I., Laurberg, P., Knudsen, N., Jorgensen, T., Perrild, H., Ovesen, L.,
ovulation induction treatment. Arthritis Rheum. 37, 1614–1617. Rasmussen, L.B., 2006. Lack of association between thyroid autoantibodies and
362 S.T. Ngo et al. / Frontiers in Neuroendocrinology 35 (2014) 347–369

parity in a population study argues against microchimerism as a trigger of Chitnis, T., 2013. Role of puberty in multiple sclerosis risk and course. Clin.
thyroid autoimmunity. Eur. J. Endocrinol./Eur. Fed. Endocr. Soc. 154, 39–45. Immunol. 149, 192–200.
Bussone, G., Mouthon, L., 2009. Autoimmune manifestations in primary immune Chitnis, T., Glanz, B., Jaffin, S., Healy, B., 2009. Demographics of pediatric-onset
deficiencies. Autoimmun. Rev. 8, 332–336. multiple sclerosis in an MS center population from the Northeastern United
Butterworth, M., McClellan, B., Allansmith, M., 1967. Influence of sex in States. Multip. Scl. 15, 627–631.
immunoglobulin levels. Nature 214, 1224–1225. Clayton, D.G., 2009. Prediction and interaction in complex disease genetics:
Buyon, J.P., Petri, M.A., Kim, M.Y., Kalunian, K.C., Grossman, J., Hahn, B.H., Merrill, experience in type 1 diabetes. PLoS Genet. 5, e1000540.
J.T., Sammaritano, L., Lockshin, M., Alarcon, G.S., Manzi, S., Belmont, H.M., Cocco, E., Sotgiu, A., Costa, G., Murru, M.R., Mancosu, C., Murru, R., Lai, M., Contu, P.,
Askanase, A.D., Sigler, L., Dooley, M.A., Von Feldt, J., McCune, W.J., Friedman, A., Marrosu, M.G., 2005. HLA-DR, DQ and APOE genotypes and gender influence in
Wachs, J., Cronin, M., Hearth-Holmes, M., Tan, M., Licciardi, F., 2005. The effect Sardinian primary progressive MS. Neurology 64, 564–566.
of combined estrogen and progesterone hormone replacement therapy on Confavreux, C., Aimard, G., Devic, M., 1980. Course and prognosis of multiple
disease activity in systemic lupus erythematosus: a randomized trial. Ann. sclerosis assessed by the computerized data processing of 349 patients. Brain: J.
Intern. Med. 142, 953–962. Neurol. 103, 281–300.
Calabrese, L.H., Kleiner, S.M., Barna, B.P., Skibinski, C.I., Kirkendall, D.T., Lahita, R.G., Confavreux, C., Hutchinson, M., Hours, M.M., Cortinovis-Tourniaire, P., Moreau, T.,
Lombardo, J.A., 1989. The effects of anabolic steroids and strength training on 1998. Rate of pregnancy-related relapse in multiple sclerosis. Pregnancy in
the human immune response. Med. Sci. Sports Exerc. 21, 386–392. Multiple Sclerosis Group. New Engl. J. Med. 339, 285–291.
Callewaert, D.M., Moudgil, V.K., Radcliff, G., Waite, R., 1991. Hormone specific Cook, I.F., 2008. Sexual dimorphism of humoral immunity with human vaccines.
regulation of natural killer cells by cortisol. Direct inactivation of the cytotoxic Vaccine 26, 3551–3555.
function of cloned human NK cells without an effect on cellular proliferation. Cooper, G.S., Dooley, M.A., Treadwell, E.L., St Clair, E.W., Gilkeson, G.S., 2002.
FEBS Lett. 285, 108–110. Hormonal and reproductive risk factors for development of systemic lupus
Camprubi, C., Monk, D., 2011. Does genomic imprinting play a role in erythematosus: results of a population-based, case-control study. Arth. Rheum.
autoimmunity? In: Ballestar, E. (Ed.), Epigenetic Contributions in 46, 1830–1839.
Autoimmune Disease. Springer, New York, USA. Correale, J., Farez, M.F., Ysrraelit, M.C., 2012. Increase in multiple sclerosis activity
Cao, W., Xu, W., Chen, T., Wang, X., Wang, X., Qiu, J., Chen, N., Mao, Y., 2014. after assisted reproduction technology. Ann. Neurol. 72, 682–694.
CD4CD25FoxP3 regulatory T cells and cytokines interact with estradiol in cases Costa, M., Colia, D., 2008. Treating infertility in autoimmune patients.
of missed abortion. Exp. Therap. Med. 7, 417–422. Rheumatology 47 (Suppl. 3), iii38-41.
Carle, A., Pedersen, I.B., Knudsen, N., Perrild, H., Ovesen, L., Rasmussen, L.B., Costenbader, K.H., Feskanich, D., Stampfer, M.J., Karlson, E.W., 2007. Reproductive
Laurberg, P., 2011. Epidemiology of subtypes of hyperthyroidism in and menopausal factors and risk of systemic lupus erythematosus in women.
Denmark: a population-based study. Eur. J. Endocrinol./Eur. Fed. Endocr. Arth. Rheum. 56, 1251–1262.
Soc. 164, 801–809. Costenbader, K.H., Gay, S., Alarcon-Riquelme, M.E., Iaccarino, L., Doria, A., 2012.
Carneiro-Sampaio, M., Coutinho, A., 2007. Tolerance and autoimmunity: lessons at Genes, epigenetic regulation and environmental factors: which is the most
the bedside of primary immunodeficiencies. Adv. Immunol. 95, 51–82. relevant in developing autoimmune diseases? Autoimmun. Rev. 11, 604–609.
Casoli, P., Tumiati, B., La Sala, G., 1997. Fatal exacerbation of systemic lupus Courvoisier, N., Dougados, M., Cantagrel, A., Goupille, P., Meyer, O., Sibilia, J., Daures,
erythematosus after induction of ovulation. J. Rheumatol. 24, 1639–1640. J.P., Combe, B., 2008. Prognostic factors of 10-year radiographic outcome in
Casson, P.R., Santoro, N., Elkind-Hirsch, K., Carson, S.A., Hornsby, P.J., Abraham, G., early rheumatoid arthritis: a prospective study. Arth. Res. Ther. 10, R106.
Buster, J.E., 1998. Postmenopausal dehydroepiandrosterone administration Cree, B.A., 2014. Multiple sclerosis genetics. Handbook Clin. Neurol. 122, 193–209.
increases free insulin-like growth factor-I and decreases high-density Crosslin, K.L., Wiginton, K.L., 2011. Sex differences in disease severity among
lipoprotein: a six-month trial. Fertil. Steril. 70, 107–110. patients with systemic lupus erythematosus. Gend. Med. 8, 365–371.
Celius, E.G., Harbo, H.F., Egeland, T., Vartdal, F., Vandvik, B., Spurkiand, A., 2000. Sex Cutolo, M., Villaggio, B., Seriolo, B., Montagna, P., Capellino, S., Straub, R.H., Sulli, A.,
and age at diagnosis are correlated with the HLA-DR2, DQ6 haplotype in 2004. Synovial fluid estrogens in rheumatoid arthritis. Autoimmun. Rev. 3, 193–
multiple sclerosis. J. Neurol. Sci. 178, 132–135. 198.
Chabchoub, G., Uz, E., Maalej, A., Mustafa, C.A., Rebai, A., Mnif, M., Bahloul, Z., Farid, Czaja, A.J., Donaldson, P.T., 2002. Gender effects and synergisms with
N.R., Ozcelik, T., Ayadi, H., 2009. Analysis of skewed X-chromosome inactivation histocompatibility leukocyte antigens in type 1 autoimmune hepatitis. Am. J.
in females with rheumatoid arthritis and autoimmune thyroid diseases. Arth. Gastroenterol. 97, 2051–2057.
Res. Ther. 11, R106. Darmochwal-Kolarz, D., Kludka-Sternik, M., Tabarkiewicz, J., Kolarz, B., Rolinski, J.,
Chagnon, P., Schneider, R., Hebert, J., Fortin, P.R., Provost, S., Belisle, C., Gingras, M., Leszczynska-Gorzelak, B., Oleszczuk, J., 2012. The predominance of Th17
Bolduc, V., Perreault, C., Silverman, E., Busque, L., 2006. Identification and lymphocytes and decreased number and function of Treg cells in
characterization of an Xp22.33;Yp11.2 translocation causing a triplication of preeclampsia. J. Reprod. Immunol. 93, 75–81.
several genes of the pseudoautosomal region 1 in an XX male patient with Dasgupta, S., Jana, M., Liu, X., Pahan, K., 2005. Myelin basic protein-primed T cells of
severe systemic lupus erythematosus. Arth. Rheum. 54, 1270–1278. female but not male mice induce nitric-oxide synthase and proinflammatory
Chandran, V., Raychaudhuri, S.P., 2010. Geoepidemiology and environmental factors cytokines in microglia: implications for gender bias in multiple sclerosis. J. Biol.
of psoriasis and psoriatic arthritis. J. Autoimmun. 34, J314-21. Chem. 280, 32609–32617.
Chang, D.M., Lan, J.L., Lin, H.Y., Luo, S.F., 2002. Dehydroepiandrosterone treatment of Davison, S.L., Bell, R., Donath, S., Montalto, J.G., Davis, S.R., 2005. Androgen levels in
women with mild-to-moderate systemic lupus erythematosus: a multicenter adult females: changes with age, menopause, and oophorectomy. J. Clin.
randomized, double-blind, placebo-controlled trial. Arth. Rheum. 46, 2924– Endocrinol. Metab. 90, 3847–3853.
2927. de Andres, C., Aristimuno, C., Bartolome, M., de Las Heras, V., Martinez-Gines, M.L.,
Chao, M.J., Herrera, B.M., Ramagopalan, S.V., Deluca, G., Handunetthi, L., Orton, S.M., Arroyo, R., Fernandez-Cruz, E., Sanchez-Ramon, S., 2009. Clinical response to
Lincoln, M.R., Sadovnick, A.D., Ebers, G.C., 2010. Parent-of-origin effects at the interferon-beta-1a may be linked to low baseline circulating BDCA1 myeloid
major histocompatibility complex in multiple sclerosis. Hum. Mol. Genet. 19, dendritic cells differential role of circulating dendritic cells and CD4+ regulatory
3679–3689. T-cells in relapsing-remitting multiple sclerosis: a 1-year longitudinal study. J.
Charchar, F.J., Bloomer, L.D., Barnes, T.A., Cowley, M.J., Nelson, C.P., Wang, Y., Neuroimmunol. 212, 112–120.
Denniff, M., Debiec, R., Christofidou, P., Nankervis, S., Dominiczak, A.F., Bani- Dean, G., Elian, M., 1997. Age at immigration to England of Asian and Caribbean
Mustafa, A., Balmforth, A.J., Hall, A.S., Erdmann, J., Cambien, F., Deloukas, P., immigrants and the risk of developing multiple sclerosis. J. Neurol. Neurosurg.
Hengstenberg, C., Packard, C., Schunkert, H., Ouwehand, W.H., Ford, I., Goodall, Psychiat. 63, 565–568.
A.H., Jobling, M.A., Samani, N.J., Tomaszewski, M., 2012. Inheritance of coronary Degu, G., Mengistu, G., Jones, J., 2002. Some factors affecting prevalence of and
artery disease in men: an analysis of the role of the Y chromosome. Lancet 379, immune responses to Schistosoma mansoni in schoolchildren in Gorgora,
915–922. northwest Ethiopia. Ethiop. Med. J. 40, 345–352.
Chavez-Rueda, K., Hernandez, J., Zenteno, E., Leanos-Miranda, A., Legorreta-Haquet, Deluca, H.F., Cantorna, M.T., 2001. Vitamin D: its role and uses in immunology.
M.V., Blanco-Favela, F., 2005. Identification of prolactin as a novel FASEB J: Off. Publ. Fed. Am. Soc. Exp. Biol. 15, 2579–2585.
immunomodulator on the expression of co-stimulatory molecules and Demkow, U., Filewska, M., Michalowska-Mitczuk, D., Kus, J., Jagodzinski, J., Zielonka,
cytokine secretions on T and B human lymphocytes. Clin. Immunol. 116, 182– T., Zwolska, Z., Wasik, M., Rowinska-Zakrzewska, E., 2007. Heterogeneity of
191. antibody response to myobacterial antigens in different clinical manifestations
Chen, X.L., Kunsch, C., 2004. Induction of cytoprotective genes through Nrf2/ of pulmonary tuberculosis. J. Physiol. Pharmacol.: Off. J. Polish Physiol. Soc. 58
antioxidant response element pathway: a new therapeutic approach for the (Suppl 5), 117–127.
treatment of inflammatory diseases. Curr. Pharm. Des. 10, 879–891. Deng, F.Y., Lei, S.F., Zhang, Y.H., Zhang, Z.L., Guo, Y.F., 2013. Functional relevance for
Chen, Y., Ma, F., Zhang, J., Chu, X., Xu, Y., 2014. Population incidence of Guillain– associations between genetic variants and systemic lupus erythematosus. PLoS
Barre syndrome in parts of China: three large populations in Jiangsu province, One 8, e53037.
2008–2010. Eur. J. Neurol.: Off. J. Eur. Fed. Neurol. Soc. 21, 124–129. Deroo, B.J., Korach, K.S., 2006. Estrogen receptors and human disease. J. Clin. Invest.
Cheng, Q., Cheng, X.J., Jiang, G.X., 2009. Multiple sclerosis in China–history and 116, 561–570.
future. Multip. Scl. 15, 655–660. Dhadke, S.V., Dhadke, V.N., Bangar, S.S., Korade, M.B., 2013. Clinical profile of
Chiche, L., Jourde, N., Ulmann, C., Mancini, J., Darque, A., Bardin, N., Dicostanzo, M.P., Guillain Barre syndrome. J. Assoc. Phys. India 61, 168–172.
Thomas, G., Harle, J.R., Vienne, J., Loukos, H., Bornet, C., 2012. Seasonal D’Hooghe, M.B., Nagels, G., Uitdehaag, B.M., 2010. Long-term effects of childbirth in
variations of systemic lupus erythematosus flares in southern France. Eur. J. MS. J. Neurol. Neurosurg. Psychiatry 81, 38–41.
Internal Med. 23, 250–254. Dobson, R., Giovannoni, G., Ramagopalan, S., 2013. The month of birth effect in
Chighizola, C., Meroni, P.L., 2012. The role of environmental estrogens and multiple sclerosis: systematic review, meta-analysis and effect of latitude. J.
autoimmunity. Autoimmun. Rev. 11. A493-501. Neurol. Neurosurg. Psychiat. 84, 427–432.
S.T. Ngo et al. / Frontiers in Neuroendocrinology 35 (2014) 347–369 363

Donaldson, J.O., Penn, A.S., Lisak, R.P., 1981. Antiaceytlcholine receptor antibody in Forsum, E., Lof, M., 2007. Energy metabolism during human pregnancy. Annu. Rev.
neonatal myasthenia gravis. Am. J. Dis. Child. 135, 222–226. Nutr. 27, 277–292.
Doria, A., Cutolo, M., Ghirardello, A., Zampieri, S., Vescovi, F., Sulli, A., Giusti, M., Fraser, P.A., Ding, W.Z., Mohseni, M., Treadwell, E.L., Dooley, M.A., St Clair, E.W.,
Piccoli, A., Grella, P., Gambari, P.F., 2002. Steroid hormones and disease activity Gilkeson, G.S., Cooper, G.S., 2003. Glutathione S-transferase M null
during pregnancy in systemic lupus erythematosus. Arth. Rheum. 47, 202–209. homozygosity and risk of systemic lupus erythematosus associated with sun
Doria, A., Ghirardello, A., Iaccarino, L., Zampieri, S., Punzi, L., Tarricone, E., Ruffatti, exposure: a possible gene-environment interaction for autoimmunity. J.
A., Sulli, A., Sarzi-Puttini, P.C., Gambari, P.F., Cutolo, M., 2004. Pregnancy, Rheumatol. 30, 276–282.
cytokines, and disease activity in systemic lupus erythematosus. Arth. Rheum. Friedrich, N., Schwarz, S., Thonack, J., John, U., Wallaschofski, H., Volzke, H., 2008.
51, 989–995. Association between parity and autoimmune thyroiditis in a general female
Doria, A., Iaccarino, L., Sarzi-Puttini, P., Ghirardello, A., Zampieri, S., Arienti, S., population. Autoimmunity 41, 174–180.
Cutolo, M., Todesco, S., 2006. Estrogens in pregnancy and systemic lupus Friel, S., Newell, J., Kelleher, C., 2005. Who eats four or more servings of fruit and
erythematosus. Ann. N. Y. Acad. Sci. 1069, 247–256. vegetables per day? Multivariate classification tree analysis of data from the
Du, L., Yang, J., Huang, J., Ma, Y., Wang, H., Xiong, T., Xiang, Z., Zhang, Y., Huang, J., 1998 Survey of Lifestyle, Attitudes and Nutrition in the Republic of Ireland.
2013. The associations between the polymorphisms in the CTLA-4 gene and the Public Health Nutrit. 8, 159–169.
risk of Graves’ disease in the Chinese population. BMC Med. Genet. 14, 46. Fujii, H., Nawa, Y., Tsuchiya, H., Matsuno, K., Fukumoto, T., Fukuda, S., Kotani, M.,
Duquette, P., Murray, T.J., Pleines, J., Ebers, G.C., Sadovnick, D., Weldon, P., Warren, 1975. Effect of a single administration of testosterone on the immune response
S., Paty, D.W., Upton, A., Hader, W., et al., 1987. Multiple sclerosis in childhood: and lymphoid tissues in mice. Cell. Immunol. 20, 315–326.
clinical profile in 125 patients. J. Pediat. 111, 359–363. Gameiro, C., Romao, F., 2010. Changes in the immune system during menopause
Dynesen, A.W., Haraldsdottir, J., Holm, L., Astrup, A., 2003. Sociodemographic and aging. Front. Biosci. 2, 1299–1303.
differences in dietary habits described by food frequency questions–results Gameiro, C.M., Romao, F., Castelo-Branco, C., 2010. Menopause and aging: changes
from Denmark. Eur. J. Clin. Nutr. 57, 1586–1597. in the immune system – a review. Maturitas 67, 316–320.
Eaton, W.W., Rose, N.R., Kalaydjian, A., Pedersen, M.G., Mortensen, P.B., 2007. Ganesan, K., Balachandran, C., Manohar, B.M., Puvanakrishnan, R., 2008.
Epidemiology of autoimmune diseases in Denmark. J. Autoimmun. 29, 1–9. Comparative studies on the interplay of testosterone, estrogen and
Ebers, G.C., Sadovnick, A.D., Dyment, D.A., Yee, I.M., Willer, C.J., Risch, N., 2004. progesterone in collagen induced arthritis in rats. Bone 43, 758–765.
Parent-of-origin effect in multiple sclerosis: observations in half-siblings. Gascon-Barre, M., Demers, C., Mirshahi, A., Neron, S., Zalzal, S., Nanci, A., 2003. The
Lancet 363, 1773–1774. normal liver harbors the vitamin D nuclear receptor in nonparenchymal and
El Adssi, H., Debouverie, M., Guillemin, F.Group, L., 2012. Estimating the prevalence biliary epithelial cells. Hepatology 37, 1034–1042.
and incidence of multiple sclerosis in the Lorraine region, France, by the Gatenby, P., Lucas, R., Swaminathan, A., 2013. Vitamin D deficiency and risk for
capture-recapture method. Multip. Scl. 18, 1244–1250. rheumatic diseases: an update. Curr. Opin. Rheumatol. 25, 184–191.
Ellis, J.A., Kemp, A.S., Ponsonby, A.L., 2014. Gene–environment interaction in Gathungu, G., Zhang, C.K., Zhang, W., Cho, J.H., 2012. A two-marker haplotype in the
autoimmune disease. Expert Rev. Mol. Med. 16, e4. IRF5 gene is associated with inflammatory bowel disease in a North American
Erten, S., Sahin, A., Altunoglu, A., Gemcioglu, E., Koca, C., 2014. Comparison of cohort. Genes Immun. 13, 351–355.
plasma vitamin D levels in patients with Sjogren’s syndrome and healthy Gaujoux, S., Leenhardt, L., Tresallet, C., Rouxel, A., Hoang, C., Jublanc, C., Chigot, J.P.,
subjects. Int. J.Rheum. Dis. http://dx.doi.org/10.1111/1756-185X.12298. Menegaux, F., 2006. Extensive thyroidectomy in Graves’ disease. J. Am. Coll.
European Paediatric Hepatitis, C.V.N., 2005. A significant sex – but not elective Surg. 202, 868–873.
cesarean section – effect on mother-to-child transmission of hepatitis C virus Ghezzi, A., Deplano, V., Faroni, J., Grasso, M.G., Liguori, M., Marrosu, G., Pozzilli, C.,
infection. J. Infect. Dis. 192, 1872–1879. Simone, I.L., Zaffaroni, M., 1997. Multiple sclerosis in childhood: clinical
Evans, P.C., Lambert, N., Maloney, S., Furst, D.E., Moore, J.M., Nelson, J.L., 1999. Long- features of 149 cases. Multip. Scl. 3, 43–46.
term fetal microchimerism in peripheral blood mononuclear cell subsets in Giglio, T., Imro, M.A., Filaci, G., Scudeletti, M., Puppo, F., De Cecco, L., Indiveri, F.,
healthy women and women with scleroderma. Blood 93, 2033–2037. Costantini, S., 1994. Immune cell circulating subsets are affected by gonadal
Eymard, B., Vernet-der Garabedian, B., Berrih-Aknin, S., Pannier, C., Bach, J.F., Morel, function. Life Sci. 54, 1305–1312.
E., 1991. Anti-acetylcholine receptor antibodies in neonatal myasthenia gravis: Gimsa, U., Mitchison, N.A., Brunner-Weinzierl, M.C., 2013. Immune privilege as an
heterogeneity and pathogenic significance. J. Autoimmun. 4, 185–195. intrinsic CNS property: astrocytes protect the CNS against T-cell-mediated
Fae, K.C., da Silva, D.D., Oshiro, S.E., Tanaka, A.C., Pomerantzeff, P.M., Douay, C., neuroinflammation. Med. Inflamm. 2013, 320519.
Charron, D., Toubert, A., Cunningham, M.W., Kalil, J., Guilherme, L., 2006. Giordano, G., Tait, L., Furlong, C.E., Cole, T.B., Kavanagh, T.J., Costa, L.G., 2013. Gender
Mimicry in recognition of cardiac myosin peptides by heart-intralesional T cell differences in brain susceptibility to oxidative stress are mediated by levels of
clones from rheumatic heart disease. J. Immunol. 176, 5662–5670. paraoxonase-2 expression. Free Rad. Biol. Med. 58, 98–108.
Fairweather, D., Frisancho-Kiss, S., Rose, N.R., 2008. Sex differences in autoimmune Gloria-Bottini, F., Bottini, N., Renzetti, G., Bottini, E., 2007. ACP1 and Th class of
disease from a pathological perspective. Am. J. Pathol. 173, 600–609. immunological disease: evidence of interaction with gender. Int. Arch. Allergy
Fajardy, I., Vambergue, A., Stuckens, C., Weill, J., Danze, P.M., Fontaine, P., 2002. Immunol. 143, 170–176.
CTLA-4 49 A/G dimorphism and type 1 diabetes susceptibility: a French case- Glynn, J.R., Crampin, A.C., Ngwira, B.M., Ndhlovu, R., Mwanyongo, O., Fine, P.E., 2008.
control study and segregation analysis. Evidence of a maternal effect. Eur. J. Herpes simplex virus type 2 trends in relation to the HIV epidemic in northern
Immunogen.: Off. J. Brit. Soc. Histocompat. Immunogen. 29, 251–257. Malawi. Sex. Transmitt. Infect. 84, 356–360.
Farace, E., Alves, W.M., 2000. Do women fare worse: a metaanalysis of gender Goble, F.C., Konopka, E.A., 1973. Sex as a factor in infectious disease. Trans. N.Y.
differences in traumatic brain injury outcome. J. Neurosurg. 93, 539–545. Acad. Sci. 35, 325–346.
Farina, C., Aloisi, F., Meinl, E., 2007. Astrocytes are active players in cerebral innate Godfrey, D.I., Hammond, K.J., Poulton, L.D., Smyth, M.J., Baxter, A.G., 2000. NKT cells:
immunity. Trends Immunol. 28, 138–145. facts, functions and fallacies. Immunol. Today 21, 573–583.
Feldman, C.H., Hiraki, L.T., Liu, J., Fischer, M.A., Solomon, D.H., Alarcon, G.S., Goldberg, A.D., Allis, C.D., Bernstein, E., 2007. Epigenetics: a landscape takes shape.
Winkelmayer, W.C., Costenbader, K.H., 2013. Epidemiology and Cell 128, 635–638.
sociodemographics of systemic lupus erythematosus and lupus nephritis Gordon, C., Wallace, D.J., Shinada, S., Kalunian, K.C., Forbess, L., Braunstein, G.D.,
among US adults with Medicaid coverage, 2000–2004. Arth. Rheum. 65, 753– Weisman, M.H., 2008. Testosterone patches in the management of patients with
763. mild/moderate systemic lupus erythematosus. Rheumatology 47, 334–338.
Fernandez, M., Calvo-Alen, J., Alarcon, G.S., Roseman, J.M., Bastian, H.M., Fessler, B.J., Gough, S.C., Simmonds, M.J., 2007. The HLA region and autoimmune disease:
McGwin Jr., G., Vila, L.M., Sanchez, M.L., Reveille, J.D., 2005. Systemic lupus associations and mechanisms of action. Curr. Genom. 8, 453–465.
erythematosus in a multiethnic US cohort (LUMINA): XXI. Disease activity, Granger, J.P., 2002. Maternal and fetal adaptations during pregnancy: lessons in
damage accrual, and vascular events in pre- and postmenopausal women. Arth. regulatory and integrative physiology. Am. J. Physiol. Regul. Integ. Comp.
Rheum. 52, 1655–1664. Physiol. 283, R1289-92.
Fernandez, M., Calvo-Alen, J., Bertoli, A.M., Bastian, H.M., Fessler, B.J., McGwin Jr., G., Grattan, D.R., Steyn, F.J., Kokay, I.C., Anderson, G.M., Bunn, S.J., 2008. Pregnancy-
Reveille, J.D., Vila, L.M., Alarcon, G.S., 2007. Systemic lupus erythematosus in a induced adaptation in the neuroendocrine control of prolactin secretion. J.
multiethnic US cohort (LUMINA L II): relationship between vascular events and Neuroendocrinol. 20, 497–507.
the use of hormone replacement therapy in postmenopausal women. J. Clin. Gratwohl, A.A., Moutsopoulos, H.M., Chused, T.M., Akizuki, M., Wolf, R.O., Sweet, J.B.,
Rheumatol.: Pract. Rep. Rheum. Musculoskel. Dis. 13, 261–265. Deisseroth, A.B., 1977. Sjogren-type syndrome after allogeneic bone-marrow
Ferraro, M., Vieira, A.R., 2010. Explaining gender differences in caries: a transplantation. Ann. Intern. Med. 87, 703–706.
multifactorial approach to a multifactorial disease. Int. J. Dentistry 2010, Greer, J.M., Csurhes, P.A., Pender, M.P., McCombe, P.A., 2004. Effect of gender on T-
649643. cell proliferative responses to myelin proteolipid protein antigens in patients
Ferreira, R., Barreto, M., Santos, E., Pereira, C., Martins, B., Andreia, R., Crespo, F., with multiple sclerosis and controls. J. Autoimmun. 22, 345–352.
Viana, J.F., Vasconcelos, C., Ferreira, C., Vicente, A.M., Fesel, C., 2005. Heritable Grimaldi, C.M., Cleary, J., Dagtas, A.S., Moussai, D., Diamond, B., 2002. Estrogen alters
factors shape natural human IgM reactivity to Ro60/SS-A and may predispose thresholds for B cell apoptosis and activation. J. Clin. Invest. 109, 1625–1633.
for SLE-associated IgG anti-Ro and anti-La autoantibody production. J. Groswasser, Z., Cohen, M., Keren, O., 1998. Female TBI patients recover better than
Autoimmun. 25, 155–163. males. Brain Injury: [BI] 12, 805–808.
Fish, E.N., 2008. The X-files in immunity: sex-based differences predispose immune Grytten Torkildsen, N., Lie, S.A., Aarseth, J.H., Nyland, H., Myhr, K.M., 2008. Survival
responses. Nat. Rev. Immunol. 8, 737–744. and cause of death in multiple sclerosis: results from a 50-year follow-up in
Forger, F., Marcoli, N., Gadola, S., Moller, B., Villiger, P.M., Ostensen, M., 2008. Western Norway. Multip. Scl. 14, 1191–1198.
Pregnancy induces numerical and functional changes of CD4+CD25 high Guillot, P.V., Gotherstrom, C., Chan, J., Kurata, H., Fisk, N.M., 2007. Human first-
regulatory T cells in patients with rheumatoid arthritis. Ann. Rheum. Dis. 67, trimester fetal MSC express pluripotency markers and grow faster and have
984–990. longer telomeres than adult MSC. Stem Cells 25, 646–654.
364 S.T. Ngo et al. / Frontiers in Neuroendocrinology 35 (2014) 347–369

Guo, T., Huo, Y., Zhu, W., Xu, F., Liu, C., Liu, N., Cao, M., Cui, B., Ning, G., 2013. Genetic Holmqvist, P., Wallberg, M., Hammar, M., Landtblom, A.M., Brynhildsen, J., 2006.
association between IL-17F gene polymorphisms and the pathogenesis of Symptoms of multiple sclerosis in women in relation to sex steroid exposure.
Graves’ Disease in the Han Chinese population. Gene 512, 300–304. Maturitas 54, 149–153.
Gupta, N., Bostrom, A.G., Kirschner, B.S., Ferry, G.D., Winter, H.S., Baldassano, R.N., Hoppenbrouwers, I.A., Liu, F., Aulchenko, Y.S., Ebers, G.C., Oostra, B.A., van Duijn,
Gold, B.D., Abramson, O., Smith, T., Cohen, S.A., Heyman, M.B., 2007. Gender C.M., Hintzen, R.Q., 2008. Maternal transmission of multiple sclerosis in a dutch
differences in presentation and course of disease in pediatric patients with population. Arch. Neurol. 65, 345–348.
Crohn disease. Pediatrics 120, e1418-25. Horton, R., Wilming, L., Rand, V., Lovering, R.C., Bruford, E.A., Khodiyar, V.K., Lush,
Guptill, J.T., Marano, A., Krueger, A., Sanders, D.B., 2011. Cost analysis of myasthenia M.J., Povey, S., Talbot Jr., C.C., Wright, M.W., Wain, H.M., Trowsdale, J., Ziegler, A.,
gravis from a large U.S. insurance database. Muscle Nerve 44, 907–911. Beck, S., 2004. Gene map of the extended human MHC. Nat. Rev. Genet. 5, 889–
Guy-Coichard, C., Nguyen, D.T., Delorme, T., Boureau, F., 2008. Pain in hereditary 899.
neuromuscular disorders and myasthenia gravis: a national survey of Howard, M., Sellors, J.W., Jang, D., Robinson, N.J., Fearon, M., Kaczorowski, J.,
frequency, characteristics, and impact. J. Pain Sympt. Manage. 35, 40–50. Chernesky, M., 2003. Regional distribution of antibodies to herpes simplex virus
Gveric-Ahmetasevic, S., Colic, A., Elvedji-Gasparovic, V., Gveric, T., Vukelic, V., 2008. type 1 (HSV-1) and HSV-2 in men and women in Ontario, Canada. J. Clin.
Can neonatal myasthenia gravis be predicted? J. Perinat. Med. 36, 503–506. Microbiol. 41, 84–89.
Gyu Song, G., Ho Lee, Y., 2013. CTLA-4 +49 A/G and -318 C/T polymorphisms and Huang, Q., Parfitt, A., Grennan, D.M., Manolios, N., 1997. X-chromosome inactivation
susceptibility to multiple sclerosis: a meta-analysis. Immunol. Invest. 42, 409– in monozygotic twins with systemic lupus erythematosus. Autoimmunity 26,
422. 85–93.
Hadjimichael, O., Vollmer, T., Oleen-Burkey, M.North American Research Huang, C.H., Wei, J.C., Chen, C.C., Chuang, C.S., Chou, C.H., Lin, Y.J., Wang, M.F., Wong,
Committee on Multiple, S., 2008. Fatigue characteristics in multiple sclerosis: R.H., 2014. Associations of the PTPN22 and CTLA-4 genetic polymorphisms with
the North American Research Committee on Multiple Sclerosis (NARCOMS) Taiwanese ankylosing spondylitis. Rheumatol. Int. 34, 683–691.
survey. Health Qual. Life Outcomes 6, 100. Huang, X., Liu, W.B., Men, L.N., Feng, H.Y., Li, Y., Luo, C.M., Qiu, L., 2013. Clinical
Hall, H.I., May, D.S., Lew, R.A., Koh, H.K., Nadel, M., 1997. Sun protection behaviors of features of myasthenia gravis in southern China: a retrospective review of 2,154
the U.S. white population. Prev. Med. 26, 401–407. cases over 22 years. Neurol. Sci.: Off. J. Italian Neurol. Soc. Ital. Soc. Clin.
Hamlyn, A.N., Sherlock, S., 1974. The epidemiology of primary biliary cirrhosis: a Neurophysiol. 34, 911–917.
survey of mortality in England and Wales. Gut 15, 473–479. Hughes, G.C., 2012. Progesterone and autoimmune disease. Autoimmun. Rev. 11.
Hammond, S.R., McLeod, J.G., Millingen, K.S., Stewart-Wynne, E.G., English, D., A502-14.
Holland, J.T., McCall, M.G., 1988. The epidemiology of multiple sclerosis in three Hughes, G.C., Clark, E.A., 2007. Regulation of dendritic cells by female sex steroids:
Australian cities: Perth, Newcastle and Hobart. Brain: J. Neurol. 111 (Pt 1), 1–25. relevance to immunity and autoimmunity. Autoimmunity 40, 470–481.
Hantai, D., Richard, P., Koenig, J., Eymard, B., 2004. Congenital myasthenic Hughes, T., Adler, A., Merrill, J.T., Kelly, J.A., Kaufman, K.M., Williams, A., Langefeld,
syndromes. Curr. Opin. Neurol. 17, 539–551. C.D., Gilkeson, G.S., Sanchez, E., Martin, J., Boackle, S.A., Stevens, A.M., Alarcon,
Harada, K., Hirohara, J., Ueno, Y., Nakano, T., Kakuda, Y., Tsubouchi, H., Ichida, T., G.S., Niewold, T.B., Brown, E.E., Kimberly, R.P., Edberg, J.C., Ramsey-Goldman, R.,
Nakanuma, Y., 2013. Incidence of and risk factors for hepatocellular carcinoma Petri, M., Reveille, J.D., Criswell, L.A., Vila, L.M., Jacob, C.O., Gaffney, P.M., Moser,
in primary biliary cirrhosis: national data from Japan. Hepatology 57, 1942– K.L., Vyse, T.J., Alarcon-Riquelme, M.E., Network, B., James, J.A., Tsao, B.P.,
1949. Scofield, R.H., Harley, J.B., Richardson, B.C., Sawalha, A.H., 2012. Analysis of
Hayes, C.E., Cantorna, M.T., DeLuca, H.F., 1997. Vitamin D and multiple sclerosis. autosomal genes reveals gene–sex interactions and higher total genetic risk in
Proc. Soc. Exp. Biol. Med. Soc. Exp. Biol. Med. 216, 21–27. men with systemic lupus erythematosus. Ann. Rheum. Dis. 71, 694–699.
Hazes, J.M., Dijkmans, B.A., Vandenbroucke, J.P., Cats, A., 1989. Oral contraceptive Ichikawa, N., Kotake, S., Hakoda, M., Kamatani, N., 2001. Microchimerism in
treatment for rheumatoid arthritis: an open study in 10 female patients. Br. J. Japanese patients with systemic sclerosis. Arth. Rheum. 44, 1226–1228.
Rheumatol. 28 (Suppl. 1), 28–30. Inagaki, T., Nishii, Y., Suzuki, N., Suzuki, S., Koizumi, Y., Aizawa, T., Hashizume, K.,
Hazes, J.M., Dijkmans, B.C., Vandenbroucke, J.P., de Vries, R.R., Cats, A., 1990. 2002. Fulminant diabetes mellitus associated with pregnancy: case reports and
Reduction of the risk of rheumatoid arthritis among women who take oral literature review. Endocr. J. 49, 319–322.
contraceptives. Arth. Rheum. 33, 173–179. Inoue, N., Watanabe, M., Wada, M., Morita, M., Hidaka, Y., Iwatani, Y., 2013. IL-10 -
Hee, C.S., Gun, S.C., Naidu, R., Gupta, E., Somnath, S.D., Radhakrishnan, A.K., 2007. 592A/C polymorphism is associated with severity of Hashimoto’s disease.
Comparison of single nucleotide polymorphisms in the human interleukin-10 Cytokine 64, 370–374.
gene promoter between rheumatoid arthritis patients and normal subjects in Invernizzi, P., Miozzo, M., Battezzati, P.M., Bianchi, I., Grati, F.R., Simoni, G., Selmi, C.,
Malaysia. Mod. Rheumatol./Jpn. Rheumatism Assoc. 17, 429–435. Watnik, M., Gershwin, M.E., Podda, M., 2004. Frequency of monosomy X in
Hellwig, K., Beste, C., Brune, N., Haghikia, A., Muller, T., Schimrigk, S., Gold, R., 2008. women with primary biliary cirrhosis. Lancet 363, 533–535.
Increased MS relapse rate during assisted reproduction technique. J. Neurol. Invernizzi, P., Miozzo, M., Selmi, C., Persani, L., Battezzati, P.M., Zuin, M., Lucchi, S.,
255, 592–593. Meroni, P.L., Marasini, B., Zeni, S., Watnik, M., Grati, F.R., Simoni, G., Gershwin,
Hemminki, K., Li, X., Sundquist, J., Sundquist, K., 2010. The epidemiology of Graves’ M.E., Podda, M., 2005. X chromosome monosomy: a common mechanism for
disease: evidence of a genetic and an environmental contribution. J. autoimmune diseases. J. Immunol. 175, 575–578.
Autoimmun. 34, J307-13. Invernizzi, P., Miozzo, M., Oertelt-Prigione, S., Meroni, P.L., Persani, L., Selmi, C.,
Hench, P.S., 1983. The ameliorating effect of pregnancy on chronic atrophic Battezzati, P.M., Zuin, M., Lucchi, S., Marasini, B., Zeni, S., Watnik, M., Tabano, S.,
(infectious rheumatoid) arthritis, fibrositis, and intermittent hydrathrosis. Maitz, S., Pasini, S., Gershwin, M.E., Podda, M., 2007. X monosomy in female
Proc. Staff Meet. Mayo Clin. 13, 161–167. systemic lupus erythematosus. Ann. N. Y. Acad. Sci. 1110, 84–91.
Henriques, A., Ines, L., Carvalheiro, T., Couto, M., Andrade, A., Pedreiro, S., Laranjeira, Ito, A., Bebo Jr., B.F., Matejuk, A., Zamora, A., Silverman, M., Fyfe-Johnson, A., Offner,
P., Morgado, J.M., Pais, M.L., da Silva, J.A., Paiva, A., 2012. Functional H., 2001. Estrogen treatment down-regulates TNF-alpha production and
characterization of peripheral blood dendritic cells and monocytes in reduces the severity of experimental autoimmune encephalomyelitis in
systemic lupus erythematosus. Rheumatol. Int. 32, 863–869. cytokine knockout mice. J. Immunol. 167, 542–552.
Herbst, A.L., Ulfelder, H., Poskanzer, D.C., 1971. Adenocarcinoma of the vagina. Ivanova, E., Kyurkchiev, D., Altankova, I., Dimitrov, J., Binakova, E., Kyurkchiev, S.,
Association of maternal stilbestrol therapy with tumor appearance in young 2005. CD83 monocyte-derived dendritic cells are present in human decidua and
women. New Engl. J. Med. 284, 878–881. progesterone induces their differentiation in vitro. Am. J. Reprod. Immunol. 53,
Hernan, M.A., Olek, M.J., Ascherio, A., 2001. Cigarette smoking and incidence of 199–205.
multiple sclerosis. Am. J. Epidemiol. 154, 69–74. Jain, A.K., Sircar, S., Jain, M., Adkar, S., Waghmare, C., 2012. Inflammatory bowel
Hernan, M.A., Jick, S.S., Logroscino, G., Olek, M.J., Ascherio, A., Jick, H., 2005. Cigarette disease in central India: a single centre experience over five years. Trop. Doct.
smoking and the progression of multiple sclerosis. Brain: J. Neurol. 128, 1461– 42, 198–199.
1465. Jansson, L., Olsson, T., Holmdahl, R., 1994. Estrogen induces a potent suppression of
Herrera, B.M., Ramagopalan, S.V., Orton, S., Chao, M.J., Yee, I.M., Sadovnick, A.D., experimental autoimmune encephalomyelitis and collagen-induced arthritis in
Ebers, G.C., 2007. Parental transmission of MS in a population-based Canadian mice. J. Neuroimmunol. 53, 203–207.
cohort. Neurology 69, 1208–1212. Jara, L.J., Lavalle, C., Fraga, A., Gomez-Sanchez, C., Silveira, L.H., Martinez-Osuna, P.,
Herrera, B.M., Ramagopalan, S.V., Lincoln, M.R., Orton, S.M., Chao, M.J., Sadovnick, Germain, B.F., Espinoza, L.R., 1991. Prolactin, immunoregulation, and
A.D., Ebers, G.C., 2008. Parent-of-origin effects in MS: observations from autoimmune diseases. Semin. Arth. Rheum. 20, 273–284.
avuncular pairs. Neurology 71, 799–803. Jara, L.J., Irigoyen, L., Ortiz, M.J., Zazueta, B., Bravo, G., Espinoza, L.R., 1998. Prolactin
Hewagama, A., Patel, D., Yarlagadda, S., Strickland, F.M., Richardson, B.C., 2009. and interleukin-6 in neuropsychiatric lupus erythematosus. Clin. Rheumatol.
Stronger inflammatory/cytotoxic T-cell response in women identified by 17, 110–114.
microarray analysis. Genes Immun. 10, 509–516. Jara, L.J., Vera-Lastra, O., Miranda, J.M., Alcala, M., Alvarez-Nemegyei, J., 2001.
Hill, N.J., Hultcrantz, M., Sarvetnick, N., Flodstrom-Tullberg, M., 2007. The target Prolactin in human systemic lupus erythematosus. Lupus 10, 748–756.
tissue in autoimmunity – an influential niche. Eur. J. Immunol. 37, 589–597. Jara, L.J., Cruz-Cruz, P., Saavedra, M.A., Medina, G., Garcia-Flores, A., Angeles, U.,
Hirota, Y., Suzuki, T., Chazono, Y., Bito, Y., 1976. Humoral immune responses Miranda-Limon, J.M., 2007. Bromocriptine during pregnancy in systemic lupus
characteristic of testosterone-propionate-treated chickens. Immunology 30, erythematosus: a pilot clinical trial. Ann. N. Y. Acad. Sci. 1110, 297–304.
341–348. Jensen, C.J., Massie, A., De Keyser, J., 2013. Immune players in the CNS: the astrocyte.
Ho, H.N., Wu, M.Y., Chen, H.F., Chao, K.H., Yang, Y.S., Huang, S.C., Lee, T.Y., Gill 3rd, J. Neuroimmun. Pharmacol.: Off. J. Soc. NeuroImmune Pharmacol. 8, 824–839.
T.J., 1995. In vivo CD3+CD25+ lymphocyte subpopulation is down-regulated Jess, T., Riis, L., Vind, I., Winther, K.V., Borg, S., Binder, V., Langholz, E., Thomsen, O.O.,
without increased serum-soluble interleukin-2 receptor (sIL-2R) by Munkholm, P., 2007. Changes in clinical characteristics, course, and prognosis of
gonadotropin releasing hormone agonist (GnRH-a). Am. J. Reprod. Immunol. inflammatory bowel disease during the last 5 decades: a population-based
33, 134–139. study from Copenhagen, Denmark. Inflamm. Bowel Dis. 13, 481–489.
S.T. Ngo et al. / Frontiers in Neuroendocrinology 35 (2014) 347–369 365

Jiang, Y.H., Bressler, J., Beaudet, A.L., 2004. Epigenetics and human disease. Annu. autoimmune inflammation in female mice by limiting CD4+ T-cell homing into
Rev. Genom. Hum. Genet. 5, 479–510. the CNS. Eur. J. Immunol. 40, 3489–3498.
Jin, H., Arase, N., Hirayasu, K., Kohyama, M., Suenaga, T., Saito, F., Tanimura, K., Lemire, J.M., 1992. Immunomodulatory role of 1,25-dihydroxyvitamin D3. J. Cell.
Matsuoka, S., Ebina, K., Shi, K., Toyama-Sorimachi, N., Yasuda, S., Horita, T., Biochem. 49, 26–31.
Hiwa, R., Takasugi, K., Ohmura, K., Yoshikawa, H., Saito, T., Atsumi, T., Sasazuki, Lepez, T., Vandewoestyne, M., Hussain, S., Van Nieuwerburgh, F., Poppe, K.,
T., Katayama, I., Lanier, L.L., Arase, H., 2014. Autoantibodies to IgG/HLA class II Velkeniers, B., Kaufman, J.M., Deforce, D., 2011. Fetal microchimeric cells in
complexes are associated with rheumatoid arthritis susceptibility. Proc. Natl. blood of women with an autoimmune thyroid disease. PLoS One 6, e29646.
Acad. Sci. U.S.A. 111, 3787–3792. Lepez, T., Vandewoestyne, M., Deforce, D., 2012. Fetal microchimeric cells in blood
Johnson, B., 2013. Overview of neonatal lupus. J. Pediat. Health Care: Off., Publ. Natl. and thyroid glands of women with an autoimmune thyroid disease. Chimerism
Assoc. Pediat. Nurse Associat. Practition. http://dx.doi.org/10.1016/ 3, 21–23.
j.pedhc.2013.07.015. Leung, P.S., Wang, J., Naiyanetr, P., Kenny, T.P., Lam, K.S., Kurth, M.J., Gershwin, M.E.,
Johnson, K.L., Bianchi, D.W., 2004. Fetal cells in maternal tissue following 2013. Environment and primary biliary cirrhosis: electrophilic drugs and the
pregnancy: what are the consequences? Hum. Reprod. Update 10, 497–502. induction of AMA. J. Autoimmun. 41, 79–86.
Jones, L.A., Anthony, J.P., Henriquez, F.L., Lyons, R.E., Nickdel, M.B., Carter, K.C., Lewis, J.E., Fu, S.M., Gaskin, F., 2013. Autoimmunity, end organ damage, and the
Alexander, J., Roberts, C.W., 2008. Toll-like receptor-4-mediated macrophage origin of autoantibodies and autoreactive T cells in systemic lupus
activation is differentially regulated by progesterone via the glucocorticoid and erythematosus. Discov. Med. 15, 85–92.
progesterone receptors. Immunology 125, 59–69. Lezama-Davila, C.M., Oghumu, S., Satoskar, A.R., Isaac-Marquez, A.P., 2007. Sex-
Kamen, D.L., Cooper, G.S., Bouali, H., Shaftman, S.R., Hollis, B.W., Gilkeson, G.S., 2006. associated susceptibility in humans with chiclero’s ulcer: resistance in females
Vitamin D deficiency in systemic lupus erythematosus. Autoimmun. Rev. 5, is associated with increased serum-levels of GM-CSF. Scand. J. Immunol. 65,
114–117. 210–211.
Kanda, N., Tamaki, K., 1999. Estrogen enhances immunoglobulin production by Li, H., Chen, Q., 2013. Genetic susceptibility to Grave’s disease. Front. Biosci.
human PBMCs. J. Allergy Clin. Immunol. 103, 282–288. (Landmark Ed) 18, 1080–1087.
Kane, S.V., Reddy, D., 2008. Hormonal replacement therapy after menopause is Li, X., Lonard, D.M., O’Malley, B.W., 2004. A contemporary understanding of
protective of disease activity in women with inflammatory bowel disease. Am. J. progesterone receptor function. Mech. Ageing Dev. 125, 669–678.
Gastroenterol. 103, 1193–1196. Li, R., Sun, J., Ren, L.M., Wang, H.Y., Liu, W.H., Zhang, X.W., Chen, S., Mu, R., He, J.,
Kantarci, O.H., Hebrink, D.D., Achenbach, S.J., Pittock, S.J., Altintas, A., Schaefer-Klein, Zhao, Y., Long, L., Liu, Y.Y., Liu, X., Lu, X.L., Li, Y.H., Wang, S.Y., Pan, S.S., Li, C.,
J.L., Atkinson, E.J., De Andrade, M., McMurray, C.T., Rodriguez, M., Weinshenker, Wang, H.Y., Li, Z.G., 2012. Epidemiology of eight common rheumatic diseases in
B.G., 2004. Association of APOE polymorphisms with disease severity in MS is China: a large-scale cross-sectional survey in Beijing. Rheumatology 51, 721–
limited to women. Neurology 62, 811–814. 729.
Karason, A., Gudjonsson, J.E., Upmanyu, R., Antonsdottir, A.A., Hauksson, V.B., Liang, J., Sun, L., Wang, Q., Hou, Y., 2006. Progesterone regulates mouse dendritic
Runasdottir, E.H., Jonsson, H.H., Gudbjartsson, D.F., Frigge, M.L., Kong, A., cells differentiation and maturation. Int. Immunopharmacol. 6, 830–838.
Stefansson, K., Valdimarsson, H., Gulcher, J.R., 2003. A susceptibility gene for Libert, C., Dejager, L., Pinheiro, I., 2010. The X chromosome in immune functions:
psoriatic arthritis maps to chromosome 16q: evidence for imprinting. Am. J. when a chromosome makes the difference. Nat. Rev. Immunol. 10, 594–604.
Hum. Genet. 72, 125–131. Lichtman, M.A., Vaughan, J.H., Hames, C.G., 1967. The distribution of serum
Kaur, G., Han, S.J., Yang, I., Crane, C., 2010. Microglia and central nervous system immunoglobulins, anti-gamma-G globulins (‘‘rheumatoid factors’’) and
immunity. Neurosurg. Clin. N. Am. 21, 43–51. antinuclear antibodies in White and Negro subjects in Evans County, Georgia.
Kawasaki, E., Eguchi, K., 2004. Is Type 1 diabetes in the Japanese population the Arth. Rheumat. 10, 204–215.
same as among Caucasians? Ann. N. Y. Acad. Sci. 1037, 96–103. Lin, Q., Hou, R., Sato, A., Ohtsuji, M., Ohtsuji, N., Nishikawa, K., Tsurui, H., Amano, H.,
Khor, B., Gardet, A., Xavier, R.J., 2011. Genetics and pathogenesis of inflammatory Amano, E., Sudo, K., Nishimura, H., Shirai, T., Hirose, S., 2010. Inhibitory IgG Fc
bowel disease. Nature 474, 307–317. receptor promoter region polymorphism is a key genetic element for murine
Khosrotehrani, K., Bianchi, D.W., 2005. Multi-lineage potential of fetal cells in systemic lupus erythematosus. J. Autoimmun. 34, 356–363.
maternal tissue: a legacy in reverse. J. Cell Sci. 118, 1559–1563. Lindholm, E., Hallengren, B., Agardh, C.D., 2004. Gender differences in GAD
Khosrotehrani, K., Johnson, K.L., Cha, D.H., Salomon, R.N., Bianchi, D.W., 2004. antibody-positive diabetes mellitus in relation to age at onset, C-peptide and
Transfer of fetal cells with multilineage potential to maternal tissue. JAMA, J. other endocrine autoimmune diseases. Diabet./Metab. Res. Rev. 20, 158–164.
Am. Med. Assoc. 292, 75–80. Lista, P., Straface, E., Brunelleschi, S., Franconi, F., Malorni, W., 2011. On the role of
Knudsen, G.P., 2009. Gender bias in autoimmune diseases: X chromosome autophagy in human diseases: a gender perspective. J. Cell Mol. Med. 15, 1443–
inactivation in women with multiple sclerosis. J. Neurol. Sci. 286, 43–46. 1457.
Knudsen, G.P., Harbo, H.F., Smestad, C., Celius, E.G., Akesson, E., Oturai, A., Ryder, Lleo, A., Oertelt-Prigione, S., Bianchi, I., Caliari, L., Finelli, P., Miozzo, M., Lazzari, R.,
L.P., Spurkland, A., Orstavik, K.H., 2007. X chromosome inactivation in females Floreani, A., Donato, F., Colombo, M., Gershwin, M.E., Podda, M., Invernizzi, P.,
with multiple sclerosis. Eur. J. Neurol.: Off. J. Eur. Fed. Neurol. Soc. 14, 1392– 2013. Y chromosome loss in male patients with primary biliary cirrhosis. J.
1396. Autoimmun. 41, 87–91.
Koch, M., Uyttenboogaart, M., Heersema, D., Steen, C., De Keyser, J., 2009. Parity and Lo, Y.M., Lo, E.S., Watson, N., Noakes, L., Sargent, I.L., Thilaganathan, B., Wainscoat,
secondary progression in multiple sclerosis. J. Neurol. Neurosurg. Psychiat. 80, J.S., 1996. Two-way cell traffic between mother and fetus: biologic and clinical
676–678. implications. Blood 88, 4390–4395.
Kuiper, S., van Gestel, A.M., Swinkels, H.L., de Boo, T.M., da Silva, J.A., van Riel, P.L., Lu, Q., 2013. The critical importance of epigenetics in autoimmunity. J. Autoimmun.
2001. Influence of sex, age, and menopausal state on the course of early 41, 1–5.
rheumatoid arthritis. J. Rheumatol. 28, 1809–1816. Luchetti, S., van Eden, C.G., Schuurman, K., van Strien, M.E., Swaab, D.F., Huitinga, I.,
Kurtzke, J.F., Beebe, G.W., Norman Jr., J.E., 1979. Epidemiology of multiple sclerosis 2014. Gender differences in multiple sclerosis: induction of estrogen signaling
in U.S. veterans: 1. Race, sex, and geographic distribution. Neurology 29, 1228– in male and progesterone signaling in female lesions. J. Neuropathol. Exp.
1235. Neurol. 73, 123–135.
Kuusisto, H., Kaprio, J., Kinnunen, E., Luukkaala, T., Koskenvuo, M., Elovaara, I., 2008. Lui, N.L., Haroon, N., Carty, A., Shen, H., Cook, R.J., Shanmugarajah, S., Gladman, D.D.,
Concordance and heritability of multiple sclerosis in Finland: study on a Inman, R.D., 2011. Effect of pregnancy on ankylosing spondylitis: a case-control
nationwide series of twins. Eur. J. Neurol.: Off. J. Eur. Fed. Neurol. Soc. 15, 1106– study. J. Rheumatol. 38, 2442–2444.
1110. Luster, M.I., Faith, R.E., McLachlan, J.A., 1978. Alterations of the antibody response
Lahita, R.G., 1999. Emerging concepts for sexual predilection in the disease systemic following in utero exposure to diethylstilbestrol. Bull. Environ. Contam. Toxicol.
lupus erythematosus. Ann. N. Y. Acad. Sci. 876, 64–69, discussion 69-70. 20, 433–437.
Lain, K.Y., Catalano, P.M., 2007. Metabolic changes in pregnancy. Clin. Obstet. Lutz, L.B., Jamnongjit, M., Yang, W.H., Jahani, D., Gill, A., Hammes, S.R., 2003.
Gynecol. 50, 938–948. Selective modulation of genomic and nongenomic androgen responses by
Lamb, M.M., Myers, M.A., Barriga, K., Zimmet, P.Z., Rewers, M., Norris, J.M., 2008. androgen receptor ligands. Mol. Endocrinol. 17, 1106–1116.
Maternal diet during pregnancy and islet autoimmunity in offspring. Pediat. MacDonald, M.J., Gottschall, J., Hunter, J.B., Winter, K.L., 1986. HLA-DR4 in insulin-
Diabet. 9, 135–141. dependent diabetic parents and their diabetic offspring: a clue to dominant
Laplaud, D.A., Leray, E., Barriere, P., Wiertlewski, S., Moreau, T., 2006. Increase in inheritance. Proc. Natl. Acad. Sci. U.S.A. 83, 7049–7053.
multiple sclerosis relapse rate following in vitro fertilization. Neurology 66, Mackay, I.R., 2001. Tolerance and autoimmunity. West J. Med. 174, 118–123.
1280–1281. Mackay, I.R., Burnet, F.M., 1963. Autoimmune Diseases, Pathogenesis, Chemistry
Lateef, A., Petri, M., 2012. Management of pregnancy in systemic lupus and Therapy. Thomas, Springfield.
erythematosus. Nat. Rev. Rheumatol. 8, 710–718. Mackay, I.R., Rose, N.R., 2001. Autoimmunity and lymphoma: tribulations of B cells.
Lauszus, F.F., Klebe, J.G., Bek, T., Flyvbjerg, A., 2003. Increased serum IGF-I during Nat. Immunol. 2, 793–795.
pregnancy is associated with progression of diabetic retinopathy. Diabetes 52, Maloney, S., Smith, A., Furst, D.E., Myerson, D., Rupert, K., Evans, P.C., Nelson, J.L.,
852–856. 1999. Microchimerism of maternal origin persists into adult life. J. Clin. Invest.
Lee, L.A., 2009. The clinical spectrum of neonatal lupus. Arch. Dermatol. Res. 301, 104, 41–47.
107–110. Manwani, B., Liu, F., Scranton, V., Hammond, M.D., Sansing, L.H., McCullough, L.D.,
Lee, J.M., Li, J., Johnson, D.A., Stein, T.D., Kraft, A.D., Calkins, M.J., Jakel, R.J., Johnson, 2013. Differential effects of aging and sex on stroke induced inflammation
J.A., 2005. Nrf2, a multi-organ protector? FASEB J.: Off. Publ. Fed. Am. Soc. Exp. across the lifespan. Exp. Neurol. 249, 120–131.
Biol. 19, 1061–1066. Marrosu, M.G., Sardu, C., Cocco, E., Costa, G., Murru, M.R., Mancosu, C., Murru, R., Lai,
Lelu, K., Delpy, L., Robert, V., Foulon, E., Laffont, S., Pelletier, L., Engelhardt, B., Guery, M., Contu, P., 2004. Bias in parental transmission of the HLA-DR3 allele in
J.C., 2010. Endogenous estrogens, through estrogen receptor alpha, constrain Sardinian multiple sclerosis. Neurology 63, 1084–1086.
366 S.T. Ngo et al. / Frontiers in Neuroendocrinology 35 (2014) 347–369

Marshall-Gradisnik, S., Weatherby, R.P., Deakin, G.B., Coutts, R.A., Meir, R., Murphy, E.D., Roths, J.B., 1979. A Y chromosome associated factor in strain BXSB
Connellan, P., Stevenson, L.M., Rogerson, S., Zhou, S., 2008. Natural killer cell producing accelerated autoimmunity and lymphoproliferation. Arth. Rheum.
activity following 6 weeks of strength training in healthy young males with/ 22, 1188–1194.
without testosterone enanthate adminstration. J. Exerc. Sci. Fit. 6, 106–114. Myasoedova, E., Crowson, C.S., Turesson, C., Gabriel, S.E., Matteson, E.L., 2011.
Martel, C., Huynh le, H., Garnier, A., Ventura-Clapier, R., Brenner, C., 2012. Inhibition Incidence of extraarticular rheumatoid arthritis in Olmsted County, Minnesota,
of the mitochondrial permeability transition for cytoprotection: direct versus in 1995–2007 versus 1985–1994: a population-based study. J. Rheumatol. 38,
indirect mechanisms. Biochem. Res. Int. 2012, 213403. 983–989.
McCombe, P.A., 2003. Gender issues in multiple sclerosis. Expert Rev. Neurother. 3, Nannini, C., Jebakumar, A.J., Crowson, C.S., Ryu, J.H., Matteson, E.L., 2013. Primary
649–660. Sjogren’s syndrome 1976–2005 and associated interstitial lung disease: a
McCombe, P.A., Greer, J.M., 2013a. Sexual dimorphism in the immune system. In: population-based study of incidence and mortality. BMJ Open. 3, e003569.
Rose, N.R., Mackay, I.R. (Eds.), The Autoimmune Diseases. Academic Press, pp. Nelson, J.L., 1999. Microchimerism: implications for autoimmune disease. Lupus 8,
319–328. 370–374.
McCombe, P.A., Greer, J.M., 2013b. Female reproductive issues in multiple sclerosis. Nelson, J.L., 2002. Microchimerism and human autoimmune diseases. Lupus 11,
Multip. Scler. 19, 392–402. 651–654.
McCombe, P.A., Chalk, J.B., Pender, M.P., 1990. Familial occurrence of multiple Nelson, J.L., Ostensen, M., 1997. Pregnancy and rheumatoid arthritis. Rheum. Dis.
sclerosis with thyroid disease and systemic lupus erythematosus. J. Neurol. Sci. Clin. North Am. 23, 195–212.
97, 163–171. Nerich, V., Monnet, E., Etienne, A., Louafi, S., Ramee, C., Rican, S., Weill, A., Vallier, N.,
McCombe, P.A., Greer, J.M., Mackay, I.R., 2009. Sexual dimorphism in autoimmune Vanbockstael, V., Auleley, G.R., Allemand, H., Carbonnel, F., 2006. Geographical
disease. Curr. Mol. Med. 9, 1058–1079. variations of inflammatory bowel disease in France: a study based on national
McConnachie, P.R., Zahalsky, A.C., 1991. Immunological consequences of exposure health insurance data. Inflamm. Bowel Dis. 12, 218–226.
to pentachlorophenol. Arch. Environ. Health 46, 249–253. Newsom-Davis, J., 2007. The emerging diversity of neuromuscular junction
McConnachie, P.R., Zahalsky, A.C., 1992. Immune alterations in humans exposed to disorders. Acta Myol.: Myopathies Cardiomyopathies: Off. J. Mediterran. Soc.
the termiticide technical chlordane. Arch. Environ. Health 47, 295–301. Myol./edited by the Gaetano Conte Academy for the Study of Striated Muscle
McKean, K.A., Nunney, L., 2005. Bateman’s principle and immunity: phenotypically Diseases 26, 5–10.
plastic reproductive strategies predict changes in immunological sex Ng, S.C., Tang, W., Ching, J.Y., Wong, M., Chow, C.M., Hui, A.J., Wong, T.C., Leung, V.K.,
differences. Evol. Int. J. Org. Evol. 59, 1510–1517. Tsang, S.W., Yu, H.H., Li, M.F., Ng, K.K., Kamm, M.A., Studd, C., Bell, S., Leong, R.,
McMurray, R., Keisler, D., Kanuckel, K., Izui, S., Walker, S.E., 1991. Prolactin de Silva, H.J., Kasturiratne, A., Mufeena, M.N., Ling, K.L., Ooi, C.J., Tan, P.S., Ong,
influences autoimmune disease activity in the female B/W mouse. J. Immunol. D., Goh, K.L., Hilmi, I., Pisespongsa, P., Manatsathit, S., Rerknimitr, R., Aniwan, S.,
147, 3780–3787. Wang, Y.F., Ouyang, Q., Zeng, Z., Zhu, Z., Chen, M.H., Hu, P.J., Wu, K., Wang, X.,
McMurray, R.W., Weidensaul, D., Allen, S.H., Walker, S.E., 1995. Efficacy of Simadibrata, M., Abdullah, M., Wu, J.C., Sung, J.J., Chan, F.K., Asia-Pacific,
bromocriptine in an open label therapeutic trial for systemic lupus C.S.Colitis Epidemiologic Study Study, G., 2013. Incidence and phenotype of
erythematosus. J. Rheumatol. 22, 2084–2091. inflammatory bowel disease based on results from the Asia–Pacific Crohn’s and
McMurray, R.W., Wilson, J.G., Bigler, L., Xiang, L., Lagoo, A., 2000. Progesterone colitis epidemiology study. Gastroenterology 145, 158-165 e2.
inhibits glucocorticoid-induced murine thymocyte apoptosis. Int. J. Nijhawan, S., Katiyar, P., Nagaich, N., Saradava, V., Nijhawan, M., Gupta, G., Mathur,
Immunopharmacol. 22, 955–965. A., Sharma, R., Nepalia, S., 2013. Prevalence of associated disorders in Indian
McNallan, K.T., Aponte, C., el-Azhary, R., Mason, T., Nelson, A.M., Paat, J.J., Crowson, patients with celiac disease. Ind. J. Gastroenterol.: Off. J. Ind. Soc. Gastroenterol.
C.S., Reed, A.M., 2007. Immunophenotyping of chimeric cells in localized 32, 330–334.
scleroderma. Rheumatology 46, 398–402. Noller, K.L., Blair, P.B., O’Brien, P.C., Melton 3rd, L.J., Offord, J.R., Kaufman, R.H.,
McNally, R.J., James, P.W., Ducker, S., James, O.F., 2011. Seasonal variation in the Colton, T., 1988. Increased occurrence of autoimmune disease among women
patient diagnosis of primary biliary cirrhosis: further evidence for an exposed in utero to diethylstilbestrol. Fertil. Steril. 49, 1080–1082.
environmental component to etiology. Hepatology 54, 2099–2103. Nomura, K., Sumida, Y., Yoh, T., Morita, A., Matsumoto, Y., Taji, S., Yoshida, N.,
Medina, K.L., Strasser, A., Kincade, P.W., 2000. Estrogen influences the Minami, M., Itoh, Y., Horiike, S., Kataoka, K., Taniwaki, M., Okanoue, T., 2004.
differentiation, proliferation, and survival of early B-lineage precursors. Blood Lack of evidence for leukocyte maternal microchimerism in primary biliary
95, 2059–2067. cirrhosis. World J. Gastroenterol.: WJG 10, 2415–2416.
Menke, A., Orchard, T.J., Imperatore, G., Bullard, K.M., Mayer-Davis, E., Cowie, C.C., Oldroyd, J., Schachna, L., Buchbinder, R., Staples, M., Murphy, B., Bond, M., Briggs, A.,
2013. The prevalence of type 1 diabetes in the United States. Epidemiology 24, Lassere, M., March, L., 2009. Ankylosing spondylitis patients commencing
773–774. biologic therapy have high baseline levels of comorbidity: a report from the
Meyer, J.M., Han, J., Singh, R., Moxley, G., 1996. Sex influences on the penetrance of Australian rheumatology association database. Int. J. Rheumatol. 2009, 268569.
HLA shared-epitope genotypes for rheumatoid arthritis. Am. J. Hum. Genet. 58, Olsen, N.J., Kovacs, W.J., 1996. Gonadal steroids and immunity. Endocr. Rev. 17,
371–383. 369–384.
Migeon, B.R., 2006. The role of X inactivation and cellular mosaicism in women’s Orbach, H., Shoenfeld, Y., 2007. Hyperprolactinemia and autoimmune diseases.
health and sex-specific diseases. JAMA, J. Am. Med. Assoc. 295, 1428–1433. Autoimmun. Rev. 6, 537–542.
Miller, L., Alley, E.W., Murphy, W.J., Russell, S.W., Hunt, J.S., 1996. Progesterone Orbach, H., Zandman-Goddard, G., Boaz, M., Agmon-Levin, N., Amital, H., Szekanecz,
inhibits inducible nitric oxide synthase gene expression and nitric oxide Z., Szucs, G., Rovensky, J., Kiss, E., Doria, A., Ghirardello, A., Gomez-Arbesu, J.,
production in murine macrophages. J. Leukoc. Biol. 59, 442–450. Stojanovich, L., Ingegnoli, F., Meroni, P.L., Rozman, B., Blank, M., Shoenfeld, Y.,
Misra, R., Hissaria, P., Tandon, V., Aggarwal, A., Krishnani, N., Dabadghao, S., 2003. 2012. Prolactin and autoimmunity: hyperprolactinemia correlates with
Primary Sjogren’s syndrome: rarity in India. J. Assoc. Phys. Ind. 51, 859–862. serositis and anemia in SLE patients. Clin. Rev. Allergy Immunol. 42, 189–198.
Mitchell, R., Hollis, S., Rothwell, C., Robertson, W.R., 1995. Age related changes in the Ormarsdottir, S., Mallmin, H., Naessen, T., Petren-Mallmin, M., Broome, U.,
pituitary-testicular axis in normal men; lower serum testosterone results from Hultcrantz, R., Loof, L., 2004. An open, randomized, controlled study of
decreased bioactive LH drive. Clin. Endocrinol. 42, 501–507. transdermal hormone replacement therapy on the rate of bone loss in
Mitsui, Y., Kusunoki, S., Arimura, K., Kaji, R., Kanda, T., Kuwabara, S., Sonoo, M., primary biliary cirrhosis. J. Intern. Med. 256, 63–69.
Takada, K., the Japanese, G.B.S.S.G., 2013. A multicentre prospective study of Osoegawa, M., Kira, J., Fukazawa, T., Fujihara, K., Kikuchi, S., Matsui, M., Kohriyama,
Guillain–Barre Syndrome in Japan: a focus on the incidence of subtypes. J. T., Sobue, G., Yamamura, T., Itoyama, Y., Saida, T., Sakata, K., Ochi, H., Matsuoka,
Neurol. Neurosurg. Psychiat. T.Research Committee of Neuroimmunological, D., 2009. Temporal changes and
Mok, C.C., 2008. Hormone replacement therapy in systemic lupus erythematosus. geographical differences in multiple sclerosis phenotypes in Japanese:
Nat. Clin. Pract. Rheumatol. 4, 60–61. nationwide survey results over 30 years. Multip. Scl. 15, 159–173.
Molvarec, A., Szarka, A., Walentin, S., Beko, G., Karadi, I., Prohaszka, Z., Rigo Jr., J., Ostensen, M., Ostensen, H., 1998. Ankylosing spondylitis – the female aspect. J.
2011. Serum leptin levels in relation to circulating cytokines, chemokines, Rheumatol. 25, 120–124.
adhesion molecules and angiogenic factors in normal pregnancy and Ostensen, M., Villiger, P.M., 2007. The remission of rheumatoid arthritis during
preeclampsia. Reprod. Biol. Endocrinol.: RB&E 9, 124. pregnancy. Semin. Immunopathol. 29, 185–191.
Morton, H., Rolfe, B., Clunie, G.J., 1977. An early pregnancy factor detected in human Ostensen, M., Forger, F., Nelson, J.L., Schuhmacher, A., Hebisch, G., Villiger, P.M.,
serum by the rosette inhibition test. Lancet 1, 394–397. 2005. Pregnancy in patients with rheumatic disease: anti-inflammatory
Muller, A.F., Drexhage, H.A., Berghout, A., 2001. Postpartum thyroiditis and cytokines increase in pregnancy and decrease post partum. Ann. Rheum. Dis.
autoimmune thyroiditis in women of childbearing age: recent insights and 64, 839–844.
consequences for antenatal and postnatal care. Endocr. Rev. 22, 605–630. Ozbalkan, Z., Bagislar, S., Kiraz, S., Akyerli, C.B., Ozer, H.T., Yavuz, S., Birlik, A.M.,
Munger, K.L., Zhang, S.M., O’Reilly, E., Hernan, M.A., Olek, M.J., Willett, W.C., Calguneri, M., Ozcelik, T., 2005. Skewed X chromosome inactivation in blood
Ascherio, A., 2004. Vitamin D intake and incidence of multiple sclerosis. cells of women with scleroderma. Arth. Rheum. 52, 1564–1570.
Neurolog. 62, 60–65. Ozcelik, T., 2008. X chromosome inactivation and female predisposition to
Munoz-Suano, A., Hamilton, A.B., Betz, A.G., 2011. Gimme shelter: the immune autoimmunity. Clin. Rev. Allergy Immunol. 34, 348–351.
system during pregnancy. Immunol. Rev. 241, 20–38. Ozcelik, T., Uz, E., Akyerli, C.B., Bagislar, S., Mustafa, C.A., Gursoy, A., Akarsu, N.,
Murase, J.E., Chan, K.K., Garite, T.J., Cooper, D.M., Weinstein, G.D., 2005. Hormonal Toruner, G., Kamel, N., Gullu, S., 2006. Evidence from autoimmune thyroiditis of
effect on psoriasis in pregnancy and post partum. Arch. Dermatol. 141, 601– skewed X-chromosome inactivation in female predisposition to autoimmunity.
606. Eur. J. Hum. Gen.: EJHG 14, 791–797.
Murata, H., Nakauchi, H., Sumida, T., 1999. Microchimerism in Japanese women Paavonen, T., 1994. Hormonal regulation of immune responses. Ann. Med. 26, 255–
patients with systemic sclerosis. Lancet 354, 220. 258.
S.T. Ngo et al. / Frontiers in Neuroendocrinology 35 (2014) 347–369 367

Padyukov, L., Hytonen, A.M., Smolnikova, M., Hahn-Zoric, M., Nilsson, N., Hanson, Pugliese, A., Awdeh, Z.L., Alper, C.A., Jackson, R.A., Eisenbarth, G.S., 1994. The
L.A., Tarkowski, A., Klareskog, L., 2004. Polymorphism in promoter region of paternally inherited insulin gene B allele (1,428 FokI site) confers protection
IL10 gene is associated with rheumatoid arthritis in women. J. Rheumatol. 31, from insulin-dependent diabetes in families. J. Autoimmun. 7, 687–694.
422–425. Qin, Y., Melse-Boonstra, A., Shi, Z., Pan, X., Yuan, B., Dai, Y., Zhao, J., Zimmermann,
Palaszynski, K.M., Smith, D.L., Kamrava, S., Burgoyne, P.S., Arnold, A.P., Voskuhl, R.R., M.B., Kok, F.J., Zhou, M., 2009. Dietary intake of zinc in the population of Jiangsu
2005. A yin-yang effect between sex chromosome complement and sex Province, China. Asia Pacif. J. Clin. Nutrit. 18, 193–199.
hormones on the immune response. Endocrinology 146, 3280–3285. Qin, B., Wang, J., Liang, Y., Yang, Z., Zhong, R., 2013. The association between TNF-
Pandit, L., Kundapur, R., 2014. Prevalence and patterns of demyelinating central alpha, IL-10 gene polymorphisms and primary Sjogren’s syndrome: a meta-
nervous system disorders in urban Mangalore, South India. Multip. Scler. http:// analysis and systemic review. PLoS One 8, e63401.
dx.doi.org/10.1177/1352458514521503. Quintana-Murci, L., Krausz, C., McElreavey, K., 2001. The human Y chromosome:
Pani, M.A., Van Autreve, J., Van der Auwera, B.J., Gorus, F.K., Badenhoop, K., 2002. function, evolution and disease. Forensic Sci. Int. 118, 169–181.
Non-transmitted maternal HLA DQ2 or DQ8 alleles and risk of Type I diabetes in Rabenau, H.F., Buxbaum, S., Preiser, W., Weber, B., Doerr, H.W., 2002.
offspring: the importance of foetal or post partum exposure to diabetogenic Seroprevalence of herpes simplex virus types 1 and type 2 in the Frankfurt
molecules. Diabetologia 45, 1340–1343. am Main area, Germany. Med. Microbiol. Immunol. 190, 153–160.
Papenfuss, T.L., Powell, N.D., McClain, M.A., Bedarf, A., Singh, A., Gienapp, I.E., Radetti, G., Zavallone, A., Gentili, L., Beck-Peccoz, P., Bona, G., 2002. Foetal and
Shawler, T., Whitacre, C.C., 2011. Estriol generates tolerogenic dendritic cells neonatal thyroid disorders. Minerva Pediatr. 54, 383–400.
in vivo that protect against autoimmunity. J. Immunol. 186, 3346–3355. Rafiei, M., Zarif Yeganeh, M., Sheikholeslami, S., Gozalpour, E., Ghaffarpour, M.,
Pedersen, J.K., Svendsen, A.J., Horslev-Petersen, K., 2011. Prevalence of rheumatoid Hedayati, M., 2012. Apolipoprotein E polymorphisms status in Iranian patients
arthritis in the southern part of denmark. Open Rheumatol. J. 5, 91–97. with multiple sclerosis. J. Neurol. Sci. 320, 22–25.
Pedersen, E.G., Hallas, J., Hansen, K., Jensen, P.E., Gaist, D., 2013. Late-onset Raghupathy, R., 2001. Pregnancy: success and failure within the Th1/Th2/Th3
myasthenia not on the increase: a nationwide register study in Denmark, 1996– paradigm. Semin. Immunol. 13, 219–227.
2009. Eur. J. Neurol.: Off. J. Eur. Fed. Neurol. Soc. 20, 309–314. Ramagopalan, S.V., Maugeri, N.J., Handunnetthi, L., Lincoln, M.R., Orton, S.M.,
Pelfrey, C.M., Cotleur, A.C., Lee, J.C., Rudick, R.A., 2002. Sex differences in cytokine Dyment, D.A., Deluca, G.C., Herrera, B.M., Chao, M.J., Sadovnick, A.D., Ebers,
responses to myelin peptides in multiple sclerosis. J. Neuroimmunol. 130, 211– G.C., Knight, J.C., 2009a. Expression of the multiple sclerosis-associated MHC
223. class II Allele HLA-DRB1*1501 is regulated by vitamin D. PLoS Genet. 5,
Pembrey, L., Newell, M.L., Tovo, P.A.European Paediatric Hepatitis, C.V.N., 2008. Age- e1000369.
related lymphocyte and neutrophil levels in children of hepatitis C-infected Ramagopalan, S.V., Valdar, W., Criscuoli, M., DeLuca, G.C., Dyment, D.A., Orton, S.M.,
women. Pediatr. Infect. Dis. J. 27, 800–807. Yee, I.M., Ebers, G.C., Sadovnick, A.D.Canadian Collaborative Study, G., 2009b.
Pender, M.P., Greer, J.M., 2007. Immunology of multiple sclerosis. Curr. Aller. Age of puberty and the risk of multiple sclerosis: a population based study. Eur.
Asthma Rep. 7, 285–292. J. Neurol.: Off. J. Eur. Fed. Neurol. Soc. 16, 342–347.
Pernis, A.B., 2007. Estrogen and CD4+ T cells. Curr. Opin. Rheumatol. 19, 414–420. Ramagopalan, S.V., Byrnes, J.K., Orton, S.M., Dyment, D.A., Guimond, C., Yee, I.M.,
Perry, V.H., Andersson, P.B., Gordon, S., 1993. Macrophages and inflammation in the Ebers, G.C., Sadovnick, A.D., 2010. Sex ratio of multiple sclerosis and clinical
central nervous system. Trends Neurosci. 16, 268–273. phenotype. Eur. J. Neurol.: Off. J. Eur. Fed. Neurol. Soc. 17, 634–637.
Persani, L., Bonomi, M., Lleo, A., Pasini, S., Civardi, F., Bianchi, I., Campi, I., Finelli, P., Ramagopalan, S.V., Goldacre, R., Skingsley, A., Conlon, C., Goldacre, M.J., 2013.
Miozzo, M., Castronovo, C., Sirchia, S., Gershwin, M.E., Invernizzi, P., 2012. Associations between selected immune-mediated diseases and tuberculosis:
Increased loss of the Y chromosome in peripheral blood cells in male patients record-linkage studies. BMC Med. 11, 97.
with autoimmune thyroiditis. J. Autoimmun. 38, J193-6. Renne, C., Ramos Lopez, E., Steimle-Grauer, S.A., Ziolkowski, P., Pani, M.A., Luther, C.,
Pertovaara, M., Lehtimaki, T., Rontu, R., Antonen, J., Pasternack, A., Hurme, M., 2004. Holzer, K., Encke, A., Wahl, R.A., Bechstein, W.O., Usadel, K.H., Hansmann, M.L.,
Presence of apolipoprotein E epsilon4 allele predisposes to early onset of Badenhoop, K., 2004. Thyroid fetal male microchimerisms in mothers with
primary Sjogren’s syndrome. Rheumatology 43, 1484–1487. thyroid disorders: presence of Y-chromosomal immunofluorescence in thyroid-
Phitayakorn, R., Morales-Garcia, D., Wanderer, J., Lubitz, C.C., Gaz, R.D., Stephen, infiltrating lymphocytes is more prevalent in Hashimoto’s thyroiditis and
A.E., Ehrenfeld, J.M., Daniels, G.H., Hodin, R.A., Parangi, S., 2013. Surgery for Graves’ disease than in follicular adenomas. J. Clin. Endocrinol. Metab. 89,
Graves’ disease: a 25-year perspective. Am. J. Surg. 206, 669–673. 5810–5814.
Piantoni, S., Andreoli, L., Allegri, F., Meroni, P.L., Tincani, A., 2012. Low levels of Reveille, J.D., 2011. Epidemiology of spondyloarthritis in North America. Am. J. Med.
vitamin D are common in primary antiphospholipid syndrome with thrombotic Sci. 341, 284–286.
disease. Reumatismo 64, 307–313. Richardson, S.J., Willcox, A., Bone, A.J., Foulis, A.K., Morgan, N.G., 2009. The
Piccinni, M.P., Giudizi, M.G., Biagiotti, R., Beloni, L., Giannarini, L., Sampognaro, S., prevalence of enteroviral capsid protein vp1 immunostaining in pancreatic
Parronchi, P., Manetti, R., Annunziato, F., Livi, C., et al., 1995. Progesterone favors islets in human type 1 diabetes. Diabetologia 52, 1143–1151.
the development of human T helper cells producing Th2-type cytokines and Rider, V., Li, X., Peterson, G., Dawson, J., Kimler, B.F., Abdou, N.I., 2006. Differential
promotes both IL-4 production and membrane CD30 expression in established expression of estrogen receptors in women with systemic lupus erythematosus.
Th1 cell clones. J. Immunol. 155, 128–133. J. Rheumatol. 33, 1093–1101.
Pierdominici, M., Ortona, E., 2013. Estrogen impact on autoimmunity onset and Rieger, R., Gershwin, M.E., 2007. The X and why of xenobiotics in primary biliary
progression: the paradigm of systemic lupus erythematosus. Int. Trends cirrhosis. J. Autoimmun. 28, 76–84.
Immun. 1, 24–34. Rieger, R., Leung, P.S., Jeddeloh, M.R., Kurth, M.J., Nantz, M.H., Lam, K.S., Barsky, D.,
Pierdominici, M., Maselli, A., Colasanti, T., Giammarioli, A.M., Delunardo, F., Vacirca, Ansari, A.A., Coppel, R.L., Mackay, I.R., Gershwin, M.E., 2006. Identification of 2-
D., Sanchez, M., Giovannetti, A., Malorni, W., Ortona, E., 2010. Estrogen receptor nonynoic acid, a cosmetic component, as a potential trigger of primary biliary
profiles in human peripheral blood lymphocytes. Immunol. Lett. 132, 79–85. cirrhosis. J. Autoimmun. 27, 7–16.
Pikwer, M., Bergstrom, U., Nilsson, J.A., Jacobsson, L., Turesson, C., 2012a. Early Roof, R.L., Hall, E.D., 2000. Gender differences in acute CNS trauma and stroke:
menopause is an independent predictor of rheumatoid arthritis. Ann. Rheum. neuroprotective effects of estrogen and progesterone. J. Neurotrauma 17, 367–
Dis. 71, 378–381. 388.
Pikwer, M., Nilsson, J.A., Bergstrom, U., Jacobsson, L.T., Turesson, C., 2012b. Early Rose, N.R., Bona, C., 1993. Defining criteria for autoimmune diseases (Witebsky’s
menopause and severity of rheumatoid arthritis in women older than 45 years. postulates revisited). Immunol. Today 14, 426–430.
Arth. Res. Ther. 14, R190. Rosenberg, A.M., Semchuk, K.M., McDuffie, H.H., Ledingham, D.L., Cordeiro, D.M.,
Pisetsky, D.S., Spencer, D.M., 2011. Effects of progesterone and estradiol sex Cessna, A.J., Irvine, D.G., Senthilselvan, A., Dosman, J.A., 1999. Prevalence of
hormones on the release of microparticles by RAW 264.7 macrophages antinuclear antibodies in a rural population. J. Toxicol. Environ. Health Part A
stimulated by Poly(I:C). Clin. Vaccine Immunol: CVI 18, 1420–1426. 57, 225–236.
Pivneva, T.A., 2008. Microglia in normal condition and pathology. Fiziolohichnyi Roubinian, J.R., Papoian, R., Talal, N., 1977. Androgenic hormones modulate
zhurnal 54, 81–89. autoantibody responses and improve survival in murine lupus. J. Clin. Invest.
Pollard, K.M., 2012. Gender differences in autoimmunity associated with exposure 59, 1066–1070.
to environmental factors. J. Autoimmun. 38, J177-86. Roubinian, J.R., Talal, N., Greenspan, J.S., Goodman, J.R., Siiteri, P.K., 1978. Effect
Ponsonby, A.L., Lucas, R.M., van der Mei, I.A., 2005. UVR, vitamin D and three of castration and sex hormone treatment on survival, anti-nucleic acid
autoimmune diseases–multiple sclerosis, type 1 diabetes, rheumatoid arthritis. antibodies, and glomerulonephritis in NZB/NZW F1 mice. J. Exp. Med. 147,
Photochem. Photobiol. 81, 1267–1275. 1568–1583.
Poppe, K., Glinoer, D., Tournaye, H., Devroey, P., van Steirteghem, A., Kaufman, L., Rowley, M.J., Mackay, I.R., 1969. Measurement of antibody-producing capacity in
Velkeniers, B., 2003. Assisted reproduction and thyroid autoimmunity: an man. I. The normal response to flagellin from Salmonella adelaide. Clin. Exp.
unfortunate combination? J. Clin. Endocrinol. Metab. 88, 4149–4152. Immunol. 5, 407–418.
Poynard, T., Bedossa, P., Opolon, P., 1997. Natural history of liver fibrosis Ruiz, E., Ramalle-Gomara, E., Elena, A., Quinones, C., Alonso, V., Posada, M., Spain,
progression in patients with chronic hepatitis C. The OBSVIRC, METAVIR, R.D.R.W.g., 2014. Trends in systemic lupus erythematosus mortality in Spain
CLINIVIR, and DOSVIRC groups. Lancet 349, 825–832. from 1981 to 2010. Lupus 23, 431–435.
Poyraz, B.C., Aksoy, C., Balcioglu, I., 2008. Increased incidence of autoimmune Ruiz-Irastorza, G., Lima, F., Alves, J., Khamashta, M.A., Simpson, J., Hughes, G.R.,
thyroiditis in patients with antipsychotic-induced hyperprolactinemia. Eur. Buchanan, N.M., 1996. Increased rate of lupus flare during pregnancy and the
Neuropsychopharmacol.: J. Eur. College Neuropsychopharmacol. 18, 667–672. puerperium: a prospective study of 78 pregnancies. Br. J. Rheumatol. 35, 133–
Premawardhana, L.D., Parkes, A.B., John, R., Harris, B., Lazarus, J.H., 2004. Thyroid 138.
peroxidase antibodies in early pregnancy: utility for prediction of postpartum Runmarker, B., Andersen, O., 1993. Prognostic factors in a multiple sclerosis
thyroid dysfunction and implications for screening. Thyroid : Off. J. Am. Thyroid incidence cohort with twenty-five years of follow-up. Brain: J. Neurol. 116 (Pt
Assoc. 14, 610–615. 1), 117–134.
368 S.T. Ngo et al. / Frontiers in Neuroendocrinology 35 (2014) 347–369

Runmarker, B., Andersen, O., 1995. Pregnancy is associated with a lower risk of Spector, T.D., Roman, E., Silman, A.J., 1990. The pill, parity, and rheumatoid arthritis.
onset and a better prognosis in multiple sclerosis. Brain: J. Neurol. 118 (Pt 1), Arth. Rheum. 33, 782–789.
253–261. Speight, J., Browne, J.L., Holmes-Truscott, E., Hendrieckx, C., Pouwer, F., 2012.
Saito, S., Sasaki, Y., Sakai, M., 2005. CD4(+)CD25high regulatory T cells in human Diabetes MILES–Australia (management and impact for long-term
pregnancy. J. Reprod. Immunol. 65, 111–120. empowerment and success): methods and sample characteristics of a
Saito, S., Nakashima, A., Ito, M., Shima, T., 2011. Clinical implication of recent national survey of the psychological aspects of living with type 1 or type 2
advances in our understanding of IL-17 and reproductive immunology. Expert diabetes in Australian adults. BMC Publ. Health 12, 120.
Rev. Clin. Immunol. 7, 649–657. Spence, R.D., Hamby, M.E., Umeda, E., Itoh, N., Du, S., Wisdom, A.J., Cao, Y., Bondar,
Sakai, R., Matsui, S., Fukushima, M., Yasuda, H., Miyauchi, H., Miyachi, Y., 2005. G., Lam, J., Ao, Y., Sandoval, F., Suriany, S., Sofroniew, M.V., Voskuhl, R.R., 2011.
Prognostic factor analysis for plaque psoriasis. Dermatology 211, 103–106. Neuroprotection mediated through estrogen receptor-alpha in astrocytes. Proc.
Sakiani, S., Olsen, N.J., Kovacs, W.J., 2013. Gonadal steroids and humoral immunity. Nat. Acad. Sci. U.S.A. 108, 8867–8872.
Nat. Rev. Endocrinol. 9, 56–62. Spolarics, Z., 2007. The X-files of inflammation: cellular mosaicism of X-linked
Sanchez-Guerrero, J., Villegas, A., Mendoza-Fuentes, A., Romero-Diaz, J., Moreno- polymorphic genes and the female advantage in the host response to injury and
Coutino, G., Cravioto, M.C., 2001. Disease activity during the premenopausal and infection. Shock 27, 597–604.
postmenopausal periods in women with systemic lupus erythematosus. Am. J. St John, A.L., Abraham, S.N., 2013. Innate immunity and its regulation by mast cells.
Med. 111, 464–468. J. Immunol. 190, 4458–4463.
Sanchez-Guerrero, J., Gonzalez-Perez, M., Durand-Carbajal, M., Lara-Reyes, P., Staples, J., Ponsonby, A.L., Lim, L., 2010. Low maternal exposure to ultraviolet
Jimenez-Santana, L., Romero-Diaz, J., Cravioto, M.D., 2007. Menopause radiation in pregnancy, month of birth, and risk of multiple sclerosis in
hormonal therapy in women with systemic lupus erythematosus. Arth. offspring: longitudinal analysis. BMJ 340, c1640.
Rheum. 56, 3070–3079. Stark, A.K., Petersen, A.O., Gardi, J., Gundersen, H.J., Pakkenberg, B., 2007. Spatial
Santiago-Raber, M.L., Kikuchi, S., Borel, P., Uematsu, S., Akira, S., Kotzin, B.L., Izui, S., distribution of human neocortical neurons and glial cells according to sex and
2008. Evidence for genes in addition to Tlr7 in the Yaa translocation linked with age measured by the saucer method. J. Neurosci. Methods 164, 19–26.
acceleration of systemic lupus erythematosus. J. Immunol. 181, 1556–1562. Stevens, A.M., Hermes, H.M., Rutledge, J.C., Buyon, J.P., Nelson, J.L., 2003.
Savettieri, G., Messina, D., Andreoli, V., Bonavita, S., Caltagirone, C., Cittadella, R., Myocardial-tissue-specific phenotype of maternal microchimerism in
Farina, D., Fazio, M.C., Girlanda, P., Le Pira, F., Liguori, M., Lugaresi, A., Nocentini, neonatal lupus congenital heart block. Lancet 362, 1617–1623.
U., Reggio, A., Salemi, G., Tedeschi, G., Trojano, M., Valentino, P., Quattrone, A., Steyn, F.J., Anderson, G.M., Grattan, D.R., 2008. Hormonal regulation of suppressors
2004. Gender-related effect of clinical and genetic variables on the cognitive of cytokine signaling (SOCS) messenger ribonucleic acid in the arcuate nucleus
impairment in multiple sclerosis. J. Neurol. 251, 1208–1214. during late pregnancy. Endocrinology 149, 3206–3214.
Schendel, D.J., Johnson, J.P., 1985. T cells specific for different antigens express Sthoeger, Z.M., Chiorazzi, N., Lahita, R.G., 1988. Regulation of the immune response
different HLA-D region products. Eur. J. Immunol. 15, 1239–1243. by sex hormones. I. In vitro effects of estradiol and testosterone on pokeweed
Selmi, C., Lu, Q., Humble, M.C., 2012. Heritability versus the role of the environment mitogen-induced human B cell differentiation. J. Immunol. 141, 91–98.
in autoimmunity. J. Autoimmun. 39, 249–252. Stojan, G., Baer, A.N., 2012. Flares of systemic lupus erythematosus during
Seror, R., Gottenberg, J.E., Devauchelle-Pensec, V., Dubost, J.J., Le Guern, V., Hayem, pregnancy and the puerperium: prevention, diagnosis and management.
G., Fauchais, A.L., Goeb, V., Hachulla, E., Hatron, P.Y., Larroche, C., Morel, J., Expert Rev. Clin. Immunol. 8, 439–453.
Pedriger, A., Puechal, X., Rist, S., Saraux, A., Sene, D., Sibilia, J., Vittecoq, O., Stoll, M., Corneliussen, B., Costello, C.M., Waetzig, G.H., Mellgard, B., Koch, W.A.,
Zarnitsky, C., Labetoulle, M., Ravaud, P., Mariette, X., 2013. European league Rosenstiel, P., Albrecht, M., Croucher, P.J., Seegert, D., Nikolaus, S., Hampe, J.,
against rheumatism Sjogren’s syndrome disease activity index and european Lengauer, T., Pierrou, S., Foelsch, U.R., Mathew, C.G., Lagerstrom-Fermer, M.,
league against rheumatism Sjogren’s syndrome patient-reported index: a Schreiber, S., 2004. Genetic variation in DLG5 is associated with inflammatory
complete picture of primary Sjogren’s syndrome patients. Arth. Care Res. 65, bowel disease. Nat. Genet. 36, 476–480.
1358–1364. Straub, R.H., 2007. The complex role of estrogens in inflammation. Endocr. Rev. 28,
Sharma, S., Eghbali, M., 2014. Influence of sex differences on microRNA gene 521–574.
regulation in disease. Biol. Sex diff. 5, 3. Stringer, E.M., Kaseba, C., Levy, J., Sinkala, M., Goldenberg, R.L., Chi, B.H., Matongo, I.,
Shen, M., Zhang, F., Zhang, X., 2009. Pulmonary hypertension in primary biliary Vermund, S.H., Mwanahamuntu, M., Stringer, J.S., 2007. A randomized trial of
cirrhosis: a prospective study in 178 patients. Scand. J. Gastroenterol. 44, 219– the intrauterine contraceptive device vs hormonal contraception in women
223. who are infected with the human immunodeficiency virus. Am. J. Obstet.
Shoenfeld, Y., Blank, M., Abu-Shakra, M., Amital, H., Barzilai, O., Berkun, Y., Bizzaro, Gynecol. 197 (144), e1-8.
N., Gilburd, B., Zandman-Goddard, G., Katz, U., Krause, I., Langevitz, P., Mackay, Strohsnitter, W.C., Noller, K.L., Troisi, R., Robboy, S.J., Hatch, E.E., Titus-Ernstoff, L.,
I.R., Orbach, H., Ram, M., Sherer, Y., Toubi, E., Gershwin, M.E., 2008. The mosaic Kaufman, R.H., Palmer, J.R., Anderson, D., Hoover, R.N., 2010. Autoimmune
of autoimmunity: prediction, autoantibodies, and therapy in autoimmune disease incidence among women prenatally exposed to diethylstilbestrol. J.
diseases–2008. Israel Med. Assoc. J.: IMAJ 10, 13–19. Rheumatol. 37, 2167–2173.
Shohat, T., Green, M.S., Nakar, O., Ballin, A., Duvdevani, P., Cohen, A., Shohat, M., Subramanian, S., Tus, K., Li, Q.Z., Wang, A., Tian, X.H., Zhou, J., Liang, C., Bartov, G.,
2000. Gender differences in the reactogenicity of measles-mumps-rubella McDaniel, L.D., Zhou, X.J., Schultz, R.A., Wakeland, E.K., 2006. A Tlr7
vaccine. Israel Med. Assoc. J.: IMAJ 2, 192–195. translocation accelerates systemic autoimmunity in murine lupus. Proc. Nat.
Si, M.L., Al-Sharafi, B., Lai, C.C., Khardori, R., Chang, C., Su, C.Y., 2001. Gender Acad. Sci. U.S.A. 103, 9970–9975.
difference in cytoprotection induced by estrogen on female and male bovine Sugiyama, D., Nishimura, K., Tamaki, K., Tsuji, G., Nakazawa, T., Morinobu, A.,
aortic endothelial cells. Endocrine 15, 255–262. Kumagai, S., 2010. Impact of smoking as a risk factor for developing
Sicotte, N.L., Liva, S.M., Klutch, R., Pfeiffer, P., Bouvier, S., Odesa, S., Wu, T.C., Voskuhl, rheumatoid arthritis: a meta-analysis of observational studies. Ann. Rheum.
R.R., 2002. Treatment of multiple sclerosis with the pregnancy hormone estriol. Dis. 69, 70–81.
Ann. Neurol. 52, 421–428. Sukenik-Halevy, R., Ellis, M.H., Fejgin, M.D., 2008. Management of immune
Sicotte, N.L., Giesser, B.S., Tandon, V., Klutch, R., Steiner, B., Drain, A.E., Shattuck, thrombocytopenic purpura in pregnancy. Obstet. Gynecol. Surv. 63, 182–188.
D.W., Hull, L., Wang, H.J., Elashoff, R.M., Swerdloff, R.S., Voskuhl, R.R., 2007. Sulke, A.N., Jones, D.B., Wood, P.J., 1985. Hormonal modulation of human natural
Testosterone treatment in multiple sclerosis: a pilot study. Arch. Neurol. 64, killer cell activity in vitro. J. Reprod. Immunol. 7, 105–110.
683–688. Suzuki, Y., Utsugisawa, K., Suzuki, S., Nagane, Y., Masuda, M., Kabasawa, C., Shimizu,
Silverman, A.J., Sutherland, A.K., Wilhelm, M., Silver, R., 2000. Mast cells migrate Y., Utsumi, H., Uchiyama, S., Fujihara, K., Suzuki, N., 2011. Factors associated
from blood to brain. J. Neurosci.: Off. J. Soc. Neurosci. 20, 401–408. with depressive state in patients with myasthenia gravis: a multicentre cross-
Singhal, B.S., Bhatia, N.S., Umesh, T., Menon, S., 2008. Myasthenia gravis: a study sectional study. BMJ Open 1, e000313.
from India. Neurol. Ind. 56, 352–355. Takao, T., Kumagai, C., Hisakawa, N., Matsumoto, R., Hashimoto, K., 2005. Effect of
Sjöberg, L., He, L., Tuomilehto, J., Kaaja, R., Pitkäniemi, J., 2013. Mortality and Having 17beta-estradiol on tumor necrosis factor-alpha-induced cytotoxicity in the
Offspring in Type 1 Diabetes, European Association for the Study of Diabetes, human peripheral T lymphocytes. J. Endocrinol. 184, 191–197.
Barcelona. Tan, E.K., Tan, E.L., 2013. Alterations in physiology and anatomy during pregnancy.
Smith, R., Studd, J.W., 1992. A pilot study of the effect upon multiple sclerosis of the Best Pract. Res. Clin. Obstet. Gynaecol. 27, 791–802.
menopause, hormone replacement therapy and the menstrual cycle. J. R. Soc. Taylor, B.V., Pearson, J.F., Clarke, G., Mason, D.F., Abernethy, D.A., Willoughby, E.,
Med. 85, 612–613. Sabel, C., 2010. MS prevalence in New Zealand, an ethnically and latitudinally
Smithson, G., Couse, J.F., Lubahn, D.B., Korach, K.S., Kincade, P.W., 1998. The role of diverse country. Multip. Scler. 16, 1422–1431.
estrogen receptors and androgen receptors in sex steroid regulation of B Tedeschi, S.K., Bermas, B., Costenbader, K.H., 2013. Sexual disparities in the
lymphopoiesis. J. Immunol. 161, 27–34. incidence and course of SLE and RA. Clin. Immunol. 149, 211–218.
Somerset, D.A., Zheng, Y., Kilby, M.D., Sansom, D.M., Drayson, M.T., 2004. Normal Teuscher, C., Noubade, R., Spach, K., McElvany, B., Bunn, J.Y., Fillmore, P.D., Zachary,
human pregnancy is associated with an elevation in the immune suppressive J.F., Blankenhorn, E.P., 2006. Evidence that the Y chromosome influences
CD25+ CD4+ regulatory T-cell subset. Immunology 112, 38–43. autoimmune disease in male and female mice. Proc. Nat. Acad. Sci. U.S.A. 103,
Sood, S., Gow, P.J., Christie, J.M., Angus, P.W., 2004. Epidemiology of primary biliary 8024–8029.
cirrhosis in Victoria, Australia: high prevalence in migrant populations. Thompson, C., Farid, N.R., 1985. Post-partum thyroiditis and goistrous
Gastroenterology 127, 470–475. (Hashimoto’s) thyroiditis are associated with HLA-DR4. Immunol. Lett. 11,
Spach, K.M., Blake, M., Bunn, J.Y., McElvany, B., Noubade, R., Blankenhorn, E.P., 301–303.
Teuscher, C., 2009. Cutting edge: the Y chromosome controls the age-dependent Thrasher, J.D., Madison, R., Broughton, A., 1993. Immunologic abnormalities in
experimental allergic encephalomyelitis sexual dimorphism in SJL/J mice. J. humans exposed to chlorpyrifos: preliminary observations. Arch. Environ.
Immunol. 182, 1789–1793. Health 48, 89–93.
S.T. Ngo et al. / Frontiers in Neuroendocrinology 35 (2014) 347–369 369

Tomio, A., Schust, D.J., Kawana, K., Yasugi, T., Kawana, Y., Mahalingaiah, S., Fujii, T., Weetman, A.P., McGregor, A.M., Smith, B.R., Hall, R., 1981. Sex hormones enhance
Taketani, Y., 2008. Prolactin can modulate CD4+ T-cell response through immunoglobulin synthesis by human peripheral blood lymphocytes. Immunol.
receptor-mediated alterations in the expression of T-bet. Immunol. Cell Biol. 86, Lett. 3, 343–346.
616–621. Weinshenker, B.G., Rice, G.P., Noseworthy, J.H., Carriere, W., Baskerville, J., Ebers,
Tripanichkul, W., Sripanichkulchai, K., Finkelstein, D.I., 2006. Estrogen down- G.C., 1991. The natural history of multiple sclerosis: a geographically based
regulates glial activation in male mice following 1-methyl-4-phenyl-1,2,3,6- study. 3. Multivariate analysis of predictive factors and models of outcome.
tetrahydropyridine intoxication. Brain Res. 1084, 28–37. Brain: J. Neurol. 114 (Pt 2), 1045–1056.
Tsuboi, H., Asashima, H., Takai, C., Hagiwara, S., Hagiya, C., Yokosawa, M., Hirota, T., Werner, M., Prytz, H., Ohlsson, B., Almer, S., Bjornsson, E., Bergquist, A., Wallerstedt,
Umehara, H., Kawakami, A., Nakamura, H., Sano, H., Tsubota, K., Ogawa, Y., S., Sandberg-Gertzen, H., Hultcrantz, R., Sangfelt, P., Weiland, O., Danielsson, A.,
Takamura, E., Saito, I., Inoue, H., Nakamura, S., Moriyama, M., Takeuchi, T., Tanaka, 2008. Epidemiology and the initial presentation of autoimmune hepatitis in
Y., Hirata, S., Mimori, T., Yoshifuji, H., Ohta, A., Matsumoto, I., Sumida, T., 2014. Sweden: a nationwide study. Scand. J. Gastroenterol. 43, 1232–1240.
Primary and secondary surveys on epidemiology of Sjogren’s syndrome in Japan. Wheeler, C., Nabors, L.B., Barnum, S., Yang, X., Hu, X., Schoeb, T.R., Chen, D., Ardelt,
Mod. Rheumatol./Jpn. Rheumat. Assoc 24, 464–470. A.A., King, P.H., 2012. Sex hormone-dependent attenuation of EAE in a
Urowitz, M.B., Ibanez, D., Jerome, D., Gladman, D.D., 2006. The effect of menopause on transgenic mouse with astrocytic expression of the RNA regulator HuR. J.
disease activity in systemic lupus erythematosus. J. Rheumatol. 33, 2192–2198. Neuroimmunol. 246, 34–37.
Uvnas-Moberg, K., Widstrom, A.M., Werner, S., Matthiesen, A.S., Winberg, J., 1990. Whitacre, C.C., Reingold, S.C., O’Looney, P.A., 1999. A gender gap in autoimmunity.
Oxytocin and prolactin levels in breast-feeding women. Correlation with milk Science 283, 1277–1278.
yield and duration of breast-feeding. Acta Obstet. Gynecol. Scand. 69, 301–306. Willer, C.J., Dyment, D.A., Risch, N.J., Sadovnick, A.D., Ebers, G.C.Canadian
Uz, E., Loubiere, L.S., Gadi, V.K., Ozbalkan, Z., Stewart, J., Nelson, J.L., Ozcelik, T., 2008. Collaborative Study, G., 2003. Twin concordance and sibling recurrence rates
Skewed X-chromosome inactivation in scleroderma. Clin. Rev. Allergy Immunol. in multiple sclerosis. Proc. Nat. Acad. Sci. U.S.A. 100, 12877–12882.
34, 352–355. Willer, C.J., Dyment, D.A., Sadovnick, A.D., Rothwell, P.M., Murray, T.J., Ebers,
van der Lugt, N.M., van Kampen, A., Walther, F.J., Brand, A., Lopriore, E., 2013. G.C.Canadian Collaborative Study, G., 2005. Timing of birth and risk of multiple
Outcome and management in neonatal thrombocytopenia due to maternal sclerosis: population based study. BMJ 330, 120.
idiopathic thrombocytopenic purpura. Vox Sang. 105, 236–243. Willer, C.J., Herrera, B.M., Morrison, K.M., Sadovnick, A.D., Ebers, G.C.Canadian
van der Mei, I.A., Ponsonby, A.L., Dwyer, T., Blizzard, L., Simmons, R., Taylor, B.V., Collaborative Study on Genetic Susceptibility to Multiple, S., 2006. Association
Butzkueven, H., Kilpatrick, T., 2003. Past exposure to sun, skin phenotype, and between microchimerism and multiple sclerosis in Canadian twins. J.
risk of multiple sclerosis: case-control study. BMJ 327, 316. Neuroimmunol. 179., 145–151.
van der Mei, I.A., Ponsonby, A.L., Dwyer, T., Blizzard, L., Taylor, B.V., Kilpatrick, T., Wirenfeldt, M., Babcock, A.A., Vinters, H.V., 2011. Microglia – insights into immune
Butzkueven, H., McMichael, A.J., 2007. Vitamin D levels in people with multiple system structure, function, and reactivity in the central nervous system. Histol.
sclerosis and community controls in Tasmania, Australia. J. Neurol. 254, 581–590. Histopathol. 26, 519–530.
van Leuven, S.I., Kastelein, J.J., D’Cruz, D.P., Hughes, G.R., Stroes, E.S., 2006. Witebsky, E., Rose, N.R., Terplan, K., Paine, J.R., Egan, R.W., 1957. Chronic thyroiditis
Atherogenesis in rheumatology. Lupus 15, 117–121. and autoimmunization. J. Am. Med. Assoc. 164, 1439–1447.
van Vollenhoven, R.F., Engleman, E.G., McGuire, J.L., 1994. An open study of Wundes, A., Amtmann, D., Brown, T., Christian, S., 2011. Menopause in women with
dehydroepiandrosterone in systemic lupus erythematosus. Arth. Rheum. 37, multiple sclerosis. Int. J. MS Care 12, 47.
1305–1310. Wyle, F.A., Kent, J.R., 1977. Immunosuppression by sex steroid hormones. The effect
van Vollenhoven, R.F., Engleman, E.G., McGuire, J.L., 1995. Dehydroepiandrosterone upon PHA- and PPD-stimulated lymphocytes. Clin. Exp. Immunol. 27, 407–415.
in systemic lupus erythematosus. Results of a double-blind, placebo-controlled, Xiang, Y.J., Dai, S.M., 2009. Prevalence of rheumatic diseases and disability in China.
randomized clinical trial. Arth. Rheum. 38, 1826–1831. Rheumatol. Int. 29, 481–490.
van Vollenhoven, R.F., Park, J.L., Genovese, M.C., West, J.P., McGuire, J.L., 1999. A Yamanaka, H., Sugiyama, N., Inoue, E., Taniguchi, A., Momohara, S., 2013. Estimates
double-blind, placebo-controlled, clinical trial of dehydroepiandrosterone in of the prevalence of and current treatment practices for rheumatoid arthritis in
severe systemic lupus erythematosus. Lupus 8, 181–187. Japan using reimbursement data from health insurance societies and the IORRA
Vanags, D., Williams, B., Johnson, B., Hall, S., Nash, P., Taylor, A., Weiss, J., Feeney, D., cohort (I). Mod. Rheumatol./Jpn. Rheumat. Assoc.
2006. Therapeutic efficacy and safety of chaperonin 10 in patients with Yan, J., Greer, J.M., Hull, R., O’Sullivan, J.D., Henderson, R.D., Read, S.J., McCombe,
rheumatoid arthritis: a double-blind randomised trial. Lancet 368, 855–863. P.A., 2010. The effect of ageing on human lymphocyte subsets: comparison of
Vandenberg, L.N., Colborn, T., Hayes, T.B., Heindel, J.J., Jacobs Jr., D.R., Lee, D.H., males and females. Immunity Ageing: I & A 7, 4.
Shioda, T., Soto, A.M., vom Saal, F.S., Welshons, W.V., Zoeller, R.T., Myers, J.P., Yang, J.H., Chen, C.D., Wu, M.Y., Chao, K.H., Yang, Y.S., Ho, H.N., 2000. Hormone
2012. Hormones and endocrine-disrupting chemicals: low-dose effects and replacement therapy reverses the decrease in natural killer cytotoxicity but
nonmonotonic dose responses. Endocr. Rev. 33, 378–455. does not reverse the decreases in the T-cell subpopulation or interferon-gamma
Vandenbroucke, J.P., Valkenburg, H.A., Boersma, J.W., Cats, A., Festen, J.J., Huber- production in postmenopausal women. Fertil. Steril. 74, 261–267.
Bruning, O., Rasker, J.J., 1982. Oral contraceptives and rheumatoid arthritis: Yang, X.Y., Liang, L.Q., Xu, H.S., He, M., Yao, S.Z., Zhan, Z.P., Ye, Y.J., 2003. Efficacy of
further evidence for a preventive effect. Lancet 2, 839–842. oral bromocriptine in protecting the postpartum systemic lupus erythematosus
Veenstra van Nieuwenhoven, A.L., Heineman, M.J., Faas, M.M., 2003. The patients from disease relapse. Zhonghua nei ke za zhi. 42, 621–624.
immunology of successful pregnancy. Hum. Reprod. Update 9, 347–357. Yildirim, A., Tunaoolu, F.S., Karaaoac, A.T., 2013. Neonatal congenital heart block.
Vera-Lastra, O., Jara, L.J., Espinoza, L.R., 2002. Prolactin and autoimmunity. Ind. Pediatr. 50, 483–488.
Autoimmun. Rev. 1, 360–364. Yip, L., McCluskey, J., Sinclair, R., 2006. Immunological aspects of pregnancy. Clin.
Verdru, P., Theys, P., D’Hooghe, M.B., Carton, H., 1994. Pregnancy and multiple Dermatol. 24, 84–87.
sclerosis: the influence on long term disability. Clin. Neurol. Neurosurg. 96, 38–41. Yu, L., Chase, H.P., Falorni, A., Rewers, M., Lernmark, A., Eisenbarth, G.S., 1995.
Vogelsang, P., Brun, J.G., Oijordsbakken, G., Skarstein, K., Jonsson, R., Appel, S., 2010. Sexual dimorphism in transmission of expression of islet autoantibodies to
Levels of plasmacytoid dendritic cells and type-2 myeloid dendritic cells are offspring. Diabetologia 38, 1353–1357.
reduced in peripheral blood of patients with primary Sjogren’s syndrome. Ann. Zandman-Goddard, G., Peeva, E., Shoenfeld, Y., 2007. Gender and autoimmunity.
Rheum. Dis. 69, 1235–1238. Autoimmun. Rev. 6, 366–372.
Vukusic, S., Van Bockstael, V., Gosselin, S., Confavreux, C., 2007. Regional variations Zanetti, A.R., Tanzi, E., Romano, L., Zuin, G., Minola, E., Vecchi, L., Principi, N., 1998. A
in the prevalence of multiple sclerosis in French farmers. J. Neurol. Neurosurg. prospective study on mother-to-infant transmission of hepatitis C virus.
Psychiat. 78, 707–709. Intervirology 41, 208–212.
Vukusic, S., Ionescu, I., El-Etr, M., Schumacher, M., Baulieu, E.E., Cornu, C., Zang, E.A., Wynder, E.L., 1996. Differences in lung cancer risk between men and
Confavreux, C.Prevention of Post-Partum Relapses with, P., and Estradiol in women: examination of the evidence. J. Natl. Cancer Inst. 88, 183–192.
Multiple Sclerosis Study, G., 2009. The Prevention of Post-Partum Relapses with Zhang, L., Li, H., Ji, Q.H., Zhu, Y.X., Wang, Z.Y., Wang, Y., Huang, C.P., Shen, Q., Li, D.S.,
Progestin and Estradiol in Multiple Sclerosis (POPART’MUS) trial: rationale, Wu, Y., 2012. The clinical features of papillary thyroid cancer in Hashimoto’s
objectives and state of advancement. J. Neurol. Sci. 286, 114–118. thyroiditis patients from an area with a high prevalence of Hashimoto’s disease.
Vyshkina, T., Sylvester, A., Sadiq, S., Bonilla, E., Canter, J.A., Perl, A., Kalman, B., 2008. BMC Cancer 12, 610.
Association of common mitochondrial DNA variants with multiple sclerosis and Zhao, J., Ng, S.C., Lei, Y., Yi, F., Li, J., Yu, L., Zou, K., Dan, Z., Dai, M., Ding, Y., Song, M.,
systemic lupus erythematosus. Clin. Immunol. 129, 31–35. Mei, Q., Fang, X., Liu, H., Shi, Z., Zhou, R., Xia, M., Wu, Q., Xiong, Z., Zhu, W., Deng,
Wagner, A.K., Sasser, H.C., Hammond, F.M., Wiercisiewski, D., Alexander, J., 2000. L., Kamm, M.A., Xia, B., 2013. First prospective, population-based inflammatory
Intentional traumatic brain injury: epidemiology, risk factors, and associations bowel disease incidence study in mainland of China: the emergence of
with injury severity and mortality. J. Trauma 49, 404–410. ‘‘western’’ disease. Inflamm. Bowel Dis. 19, 1839–1845.
Wahren-Herlenius, M., Sonesson, S.E., 2006. Specificity and effector mechanisms of Zhu, J.M., Li, B.K., Chen, G.M., Feng, C.C., Cen, H., Fan, Y.G., Wang, B., Pan, H.F., Ye,
autoantibodies in congenital heart block. Curr. Opin. Immunol. 18, 690–696. D.Q., 2013. CTLA-4 -1722T/C polymorphism and systemic lupus erythematosus
Walitt, B., Pettinger, M., Weinstein, A., Katz, J., Torner, J., Wasko, M.C., Howard, susceptibility: a meta-analysis involving ten separate studies. Immunol. Invest.
B.V.Women’s Health Initiative, I., 2008. Effects of postmenopausal hormone 42, 91–105.
therapy on rheumatoid arthritis: the women’s health initiative randomized Zinger, H., Sherer, Y., Goddard, G., Berkun, Y., Barzilai, O., Agmon-Levin, N., Ram, M.,
controlled trials. Arthritis Rheum. 59, 302–310. Blank, M., Tincani, A., Rozman, B., Cervera, R., Shoenfeld, Y., 2009. Common
Walker, S.E., 2007. The importance of sex hormones in lupus. In: Wallace, D.J., Hahn, infectious agents prevalence in antiphospholipid syndrome. Lupus 18, 1149–
G.H. (Eds.), Dubois Lupus Erythematosus Williams and Wilkins, Baltimore. 1153.
Wardlaw, A.J., Moqbel, R., Cromwell, O., Kay, A.B., 1986. Platelet-activating factor. A Zoller, A.L., Kersh, G.J., 2006. Estrogen induces thymic atrophy by eliminating early
potent chemotactic and chemokinetic factor for human eosinophils. J. Clin. thymic progenitors and inhibiting proliferation of beta-selected thymocytes. J.
Invest. 78, 1701–1706. Immunol. 176, 7371–7378.

You might also like