ArticlePDF Available

Gender Is a Major Factor in Determining the Severity of Mycoplasma Respiratory Disease in Mice

American Society for Microbiology
Infection and Immunity
Authors:

Abstract and Figures

Gender is a significant factor in determining the susceptibility to and severity of pulmonary diseases in both humans and animals. Murine respiratory mycoplasmosis (MRM), due to Mycoplasma pulmonisinfection, is an excellent animal model for evaluation of the role of various host factors on the development of acute or chronic inflammatory lung diseases. MRM has many similarities to mycoplasma respiratory disease in humans. The purpose of the present study was to determine whether gender has a significant impact on lung disease due to M. pulmonis infection in mice. It was demonstrated that male mice consistently developed more severe disease in the lung parenchyma than did female mice. There was no gender difference in disease severity along the airways or any difference in mycoplasma numbers in lungs of male and female mice. Furthermore, surgical removal of reproductive organs reduced the severity of mycoplasma disease and the numbers of mycoplasma organisms recovered from lungs. Thus, gender plays a significant role in determining the severity of M. pulmonis disease. In fact, the gender of the host was a major factor in determining whether an acute or chronic inflammatory lung disease developed after infection with M. pulmonis.
Content may be subject to copyright.
INFECTION AND IMMUNITY,
0019-9567/01/$04.000 DOI: 10.1128/IAI.69.5.2865–2871.2001
May 2001, p. 2865–2871 Vol. 69, No. 5
Copyright © 2001, American Society for Microbiology. All Rights Reserved.
Gender Is a Major Factor in Determining the Severity of
Mycoplasma Respiratory Disease in Mice
ANTHONY L. YANCEY,
1
HAROLD L. WATSON,
2
SAM C. CARTNER,
3
AND JERRY W. SIMECKA
4
*
Department of Microbiology
1
and Department of Comparative Medicine,
3
University of Alabama, Birmingham,
Alabama; Lilly Research Laboratories, Indianapolis, Indiana
2
; and Department of Molecular Biology
and Immunology, University of North Texas Health Science Center, Fort Worth, Texas
4
Received 6 November 2000/Returned for modification 19 January 2001/Accepted 24 January 2001
Gender is a significant factor in determining the susceptibility to and severity of pulmonary diseases in both
humans and animals. Murine respiratory mycoplasmosis (MRM), due to Mycoplasma pulmonis infection, is an
excellent animal model for evaluation of the role of various host factors on the development of acute or chronic
inflammatory lung diseases. MRM has many similarities to mycoplasma respiratory disease in humans. The
purpose of the present study was to determine whether gender has a significant impact on lung disease due to
M. pulmonis infection in mice. It was demonstrated that male mice consistently developed more severe disease
in the lung parenchyma than did female mice. There was no gender difference in disease severity along the
airways or any difference in mycoplasma numbers in lungs of male and female mice. Furthermore, surgical
removal of reproductive organs reduced the severity of mycoplasma disease and the numbers of mycoplasma
organisms recovered from lungs. Thus, gender plays a significant role in determining the severity of M. pul-
monis disease. In fact, the gender of the host was a major factor in determining whether an acute or chronic
inflammatory lung disease developed after infection with M. pulmonis.
Respiratory disease is a major health problem in the United
States, with males, in general, being more susceptible than
females to several major lung diseases (4, 11, 19, 26, 29).
Chronic obstructive pulmonary disease, which includes chronic
bronchitis, chronic asthma, and emphysema, is the fifth most
common cause of death. Chronic obstructive pulmonary dis-
ease is especially problematic in the elderly, with males being
affected more frequently than females (19). Males are also
more likely to develop community-acquired and nosocomial
bacterial pneumonias than are females (11, 26, 29). Further-
more, the severity of pneumonia appears greater in male pa-
tients, since as males have a higher risk of hospitalization and
mortality due to pneumonia (4, 19). In the adolescent patient
population, the same tendency exists in that males can also be
more susceptible to lung disease than females. This is demon-
strated in Mycoplasma pneumoniae disease, which is one of the
most prevalent respiratory infections in children and young
adults (12, 17, 18). Thus, gender has an effect on susceptibility
to several pulmonary diseases and may be an unappreciated
but significant factor when considering the diagnosis and treat-
ment of respiratory diseases in humans.
Gender also influences the development of infectious dis-
ease in animals. Male mice are either more susceptible to or
develop more severe disease after infection with Candida, cox-
sackievirus, and Leishmania (3, 23, 28). However, there are few
animal models of respiratory disease where gender has been
shown to influence host susceptibility. After infection with
Mycobacteria marinum or Mycobacteria intracellulare, male
mice developed more severe granulomatous lung lesions than
did females (39, 40). The difference in disease severity corre-
sponds to the numbers of M. intracellulare cells in the lungs. In
M. marinum infection, as well as the other models of infectious
diseases (28, 39), it was further demonstrated that testosterone
exacerbated disease severity. Although the results of the stud-
ies with mycobacteria are important, lung disease in humans is
not limited to the characteristic granulomatous lesions de-
scribed in these animal models. Therefore, here is a need to
establish additional animal models to investigate the influence
of gender on respiratory disease.
Murine respiratory mycoplasmosis (MRM) is an excellent
animal model for use in evaluation of the role of various
factors on the development of acute or chronic inflammatory
lung diseases. MRM is a naturally occurring respiratory disease
in rodents and results from infection with Mycoplasma pulmo-
nis (8, 25, 35). Although it is not an exact model of human
disease, there are similarities in the pathology and clinical signs
between the mycoplasma respiratory disease in humans and
M. pulmonis disease in mice. As in many human diseases, host
and environmental factors can affect the progression of M. pul-
monis respiratory disease (13–15, 25, 27, 30, 31). An additional
advantage of MRM is that both acute alveolar and chronic
peribronchial pneumonias are characteristic of M. pulmonis
disease in mice. Because of its similarity to human disease and
the presence of both acute and chronic inflammation, MRM
appears to be an ideal model to examine the effect of gender
on the pathogenesis of lung disease. The purpose of the pres-
ent study was to determine if gender does influence the sever-
ity of lung lesions due to M. pulmonis infection in mice.
MATERIALS AND METHODS
Animals. Six-week old, specific-pathogen-free C3H/HeN mice, reared and
maintained in Trexler-type plastic film isolators, were used in these experiments
(30). All retired breeders from the colony were examined for the presence of
serum immunoglobulin G (IgG) and IgM antibodies to M. pulmonis and Myco-
plasma arthritidis by enzyme-linked immunosorbent assays (ELISA). The ab-
* Corresponding author. Mailing address: Department of Molecular
Biology and Immunology, University of North Texas Health Science
Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107. E-mail:
jsimecka@hsc.unt.edu.
2865
sence of other murine pathogens was confirmed using bacterial fecal cultures,
necropsy, histological examination, and serologic tests for viruses. Sera from
mice were tested by hemagglutination inhibition, complement fixation, or ELISA
by Charles River Biotechnical Services (Wilmington, Mass.) for the following
pathogens: Sendai virus, pneumonia virus of mice, polyomavirus, minute virus of
mice, ectromelia virus, mouse hepatitis virus, reovirus type 3, Theiler’s GD-VII
virus, lymphocytic choriomeningitis virus, and mouse adenovirus. No murine
pathogens have been detected in this animal colony during the past 5 years.
Specific-pathogen-free C57BL/6N and DBA/2N mice were obtained from the
National Cancer Institute, Frederick Cancer Research Facility, Frederick, Md.
Health surveillance was similarly performed on these mice to exclude the pres-
ence of murine mycoplasma, viruses, bacteria, and parasites.
Experimental mice were maintained in microisolators with sterile bedding
(five to six mice per cage), and sterile food and water was provided ad libitum.
Prior to experimental manipulation, mice were anesthetized with a intramuscular
injection of 10 mg of ketamine hydrochloride (Bristol Laboratories, Syracuse,
N.Y.) per 100 g of body weight and 3.0 mg of xylazine (Haver-Lockhart, Shaw-
nee, Kans.) per 100 g of body weight.
Mycoplasma. M. pulmonis strain CT was derived from a naturally infected
mouse (14). A defined mixture of mycoplasma subclones, derived from the
parental M. pulmonis CT strain, was shown to consistently result in both peri-
bronchial and alveolar inflammatory disease (unpublished results). This organ-
ism stock is simply referred to as CTM, and M. pulmonis CTM was used in these
studies unless indicated otherwise. For experimental infection, anesthetized mice
were inoculated intranasally with 50 l of mycoplasma at a total dose of 10
6
CFU, unless otherwise noted. Medium and harvesting techniques were previ-
ously described (5, 6, 14).
Lung homogenates were cultured on Hayflick’s agar plates to determine the
numbers of mycoplasma CFU. Samples were diluted in broth from 10
-1
to 10
-6
dilutions. Portions (20 l) of the diluted broth were pipetted on the agar plates
and then incubated for 7 days at 37°C. Mycoplasma colonies were counted using
a dissecting microscope.
Surgical removal of reproductive organs. Anesthetized mice were prepared
for surgery by shaving the abdomen and swabbing it with Betadine Surgical Scrub
(Purdue Frederick, Norwalk, Conn.). A midline surgical incision was made in the
abdominal wall. For ovahysterectomy, the uterus, oviducts, and ovaries were
removed and ligatures were placed at the uterine-cervical junction. For orchiec-
tomies, the seminal vesicles and testicles were removed. For all surgeries, the
abdominal muscular layer was sutured using surgical chromic gut and the skin
was sutured with silk. For controls (sham neutered), a midline surgical incision
was made on the abdomen and a surgical probe was used by manipulate the
reproductive organs. The probe was removed, and the muscular layer and skin
were sutured as above. The mice were allowed to recover and observed for 2 to
4 h after surgery. Mice were infected with Mycoplasma 2 weeks after surgery.
Histological examination. Lungs and tracheas were removed and inflated with
cold 95% ethanol. The lung lobes were separated and placed in tissue cassettes
for embedding and subsequent hematoxylin and eosin staining. Each lung lobe
was scored subjectively for airway and alveolar disease (6). Airways were scored
as follows: 0, normal tissue; 1, few scattered neutrophils in the airways; 2, some
pooling of neutrophils in the bronchi and bronchioles; 3, major pooling of
neutrophils in the bronchi and bronchioles. Alveolar disease was scored using a
similar scale: 0, normal tissue; 1, inflammatory exudate affecting 1 to 25% of the
lung parenchyma; 2, inflammatory exudate affecting 26 to 50% of the lung
parenchyma; 3, inflammatory exudate affecting more than 50% of the lung
parenchyma. Slides were coded so that subjective scoring of lesions was done
blindly. Alveolar and airway scores for each lobe were summed after adjustment
for their relative contribution to the total lung weight.
Testosterone levels. Serum samples were collected at the time of sacrifice and
pooled. Serum testosterone levels were performed using radioimmunoassay (Tri-
Level Ligand Control; CIBA-Corning Diagnostics Corp., Irvine, Calif.).
Antibody levels. Anti-mycoplasma antibody titers in the sera were determined
using ELISA (6, 34). Serial dilutions of each serum sample were added in
triplicate to microtiter wells coated with M. pulmonis lysate at 10 g/ml. After
overnight incubation, the plates were washed with phosphate-buffered saline–
0.2% Tween 20. A secondary antibody, labeled with alkaline phosphatase, was
then added to each of the wells at a dilution shown to have minimal nonspecific
binding to antigen-coated wells. Next, the microtiter wells were incubated for 30
min at 37°C, and the optical density was read at 405 nm using a model 3550
microplate reader (Bio-Rad Laboratories, Richmond, Calif.). The relative anti-
body activities were determined relative to a standard composed of a pool of
high-titer sera from M. pulmonis-infected mice. The standard was given an
arbitrary activity of 3,000 and used to compare the activity of other sera. Com-
parisons between serum activities were made only with values obtained within a
single ELISA run to eliminate daily variation within the assay.
Statistical analysis. Statistics were performed using the SYSTAT program
(Systat, Inc., Evanston, Ill.). Arcsine transformation was performed on lesion
indices to normalize the data. Data were analyzed by analysis of variance fol-
lowed by post hoc tests for multigroup comparisons, as needed. Survival and
frequency of gross lesions were compared by Yates corrected chi-square analysis.
Testosterone levels were analyzed by Student’s independent ttests. A probability
(P) of less than 0.05 was accepted as significant.
RESULTS
Male mice develop more severe alveolar pneumonia than
female mice do. To determine if gender influences the severity
of M. pulmonis respiratory disease, male and female mice were
experimentally infected with M. pulmonis. Animals were ob-
served daily for clinical signs and mortality. At 14 days after
infection, mice were sacrificed and their lungs were collected
for histologic examination.
Male mice infected with M. pulmonis developed more severe
clinical disease than did female mice. The clinical signs seen
included ruffled fur, weight loss, and a hunched position. Fe-
male mice were consistently more active than male mice, and
those that survived even showed signs of improvement. Over-
all, a larger proportion of male mice than female mice died
after infection (P0.001). In three experiments, 60% of male
mice died (n22) compared to only 9% of the female mice
(n33).
There was also a histopathological difference in the lungs of
male and female mice inoculated with M. pulmonis (Fig. 1).
Lung lesions in male mice were associated predominantly with
an alveolar disease characterized by thickened alveolar walls,
edema, hemorrhage, and accumulation of acute inflammatory
cells. In addition, the airways and adjacent air spaces contained
inflammatory exudates. The character of the lesions in the
male mice was the same whether the mice survived or not. In
contrast to male mice, alveolar disease was minimal in female
mice but the severity of airway lesions was similar to that in
male mice.
FIG. 1. Lung disease in male and female mice after infection with
M. pulmonis. The graph shows the mean lesion index scores, along with
standard error bars, from male and female mice infected with M. pul-
monis 14 days previously. No significant difference was observed in the
severity of peribronchial lesions between male and female mice. How-
ever, there was a significant difference (P0.05) in the severity of
interstitial lung lesion between the sexes. The asterisk denotes a sig-
nificant difference in lesion indices between male and female mice.
2866 YANCEY ET AL. INFECT.IMMUN.
Neutering of mice reduces the severity of mycoplasma lung
disease. To examine the influence of the reproduction organs
on the development of lesions after infection, male and female
mice were neutered at 6 weeks of age. Sham-neutered mice
were included as controls. The mice were infected with M. pul-
monis after 14 days of recovery and then sacrificed at 14 days
after infection, and their lungs were removed for histological
examination.
Removal of the reproductive organs reduced the severity of
lung lesions in both male and female infected mice. As in
previous experiments, infected male mice developed an acute
interstitial pneumonia while infected female mice developed
a chronic bronchopneumonia (Fig. 2). Sham-neutered male
mice infected with M. pulmonis had more severe interstitial
lung disease than did neutered male mice. In neutered male
mice, the severity of inflammatory cell infiltrates was reduced.
Similarly, there was a reduction in the severity of the peri-
bronchial lesions in female mice after surgical removal of the
reproductive organs. There were no observable histological
changes in the lungs of uninfected male and female mice after
surgery.
Gender differences were also present earlier than 14 days.
For mice infected with M. pulmonis and sacrificed at 7 days
postinfection, male mice had an acute inflammatory disease of
the alveoli while the disease in female mice was localized to the
airways of the lungs. In both sexes, pulmonary disease was less
severe in mice that had undergone gonadectomy (Fig. 3).
Orchiectomies decreased the presurgical levels of testoster-
one in male mice. The mean level of testosterone in males
before surgery was 842 217 ng/ml (mean standard error).
Postsurgery, the level of testosterone was significantly reduced
to 67 43 ng/ml (P0.05).
Influence of gender and neutering on mycoplasma numbers
in the lung. To determine if the effects of gender and surgery
were associated with a change in the numbers of mycoplasma
in the lungs, neutered and sham-neutered mice were infected
at 8 weeks old and sacrificed at 7 days postinfection. There was
no significant difference in the numbers of mycoplasma recov-
ered from the lungs of sham-neutered male and female mice
after infection (Table 1). However, surgical removal of repro-
ductive organs affected the numbers of mycoplasmas in lungs.
At 7 days after infection, neutered male mice had significantly
FIG. 2. Effect of neutering on the severity of M. pulmonis disease at 14 days after infection. Neutered male and female mice were infected with
M. pulmonis, and the severity of interstitial and peribronchial lesions was determined 14 days after infection. Sham-neutered mice were included
for comparison. (A) As in earlier studies, no significant difference was observed in the severity of peribronchial lesions between sham-neutered
male (n12) and female (n11) mice. However, there was a significant difference (P0.05) in the severity of the interstitial lung lesion between
the sexes. (B and C) Removal of reproductive organs from male (n8) and (B) female (n7) (C) mice significantly decreased (P0.05) the
severity of disease. Removal of the reproductive organs of infected male mice resulted in a significant decrease in alveolar inflammation, while
removal of the reproductive organs of female mice led to a decrease in the severity of both interstitial and peribronchial lesions. Gonadectomy
had no significant effect on the peribronchial inflammatory response. The graph shows the mean lesion index scores, along with standard error bars,
from lungs of infected mice. The asterisk denotes a significant difference in lesion indices between male and female mice (A) or reduction in
severity after surgery (B and C).
VOL. 69, 2001 GENDER DIFFERENCES IN MYCOPLASMA RESPIRATORY DISEASE 2867
FIG. 3. Effect of neutering on M. pulmonis disease in mice at 7 days after infection. Neutered male and female mice were infected with
M. pulmonis, and the severity of interstitial and peribronchial lesions was determined 7 days after infection. Sham-neutered mice were included
for comparison. These are representative sections from each of the experimental groups. Sham-neutered male mice (n11) had more severe
inflammatory response in the alveoli than did neutered male mice (n13). Similarly, sham-neutered female mice (n15) had more severe lung
lesions than did female mice with ovohysterectomies (n15).
2868 YANCEY ET AL. INFECT.IMMUN.
smaller numbers of mycoplasmas in lungs than did control
(sham-neutered) male mice. Similarly, smaller numbers of my-
coplasmas were recovered from the lungs of neutered female
mice than of sham-neutered female mice after infection. Last-
ly, there was a significant difference in the mycoplasma num-
bers obtained from infected male mice that were neutered and
from infected female mice that were neutered.
Serum anti-mycoplasma antibody levels in mice. We deter-
mined the serum antibody levels of surgically neutered mice
and sham-neutered mice infected with M. pulmonis at 7 days
postinfection. Only very low levels of M. pulmonis-specific IgA
and IgG were detected, although significant levels of M. pul-
monis-specific IgM were present. Sham-neutered male mice
had an IgM titer of 555 57, compared 570 75 for neutered
male mice. The mean IgM titers were 288 57) and 438 57
for sham-neutered and neutered female mice, respectively.
Overall, there was no significant difference in the IgM antibody
response between male and female mice in any of the experi-
mental groups.
Gender influences the severity of mycoplasma disease in
other mouse strains. To determine if gender influences the
severity of M. pulmonis respiratory disease in other strains of
mice, C57BL/6N and DBA/2N mice were infected with 2 10
4
CFU of M. pulmonis CT and mortality and the presence of
gross lung lesions were noted at 21 days after infection. Overall
significantly greater numbers of C57BL/6N male mice had
gross lung lesions than C57BL/6N female mice (Table 2). Also,
there was a higher mortality in DBA/2N male mice than DBA/
2N female mice.
DISCUSSION
Although gender is a significant factor that affects the sus-
ceptibility to and severity of respiratory disease in humans, there
are few animal models of infectious lung disease where gender
is shown to influence the outcome. MRM due to M. pulmonis
infection in mice is an excellent animal model of acute and
chronic inflammatory lung disease (7, 9). The purpose of the
present studies was to determine if M. pulmonis disease in mice
is affected by gender.
Male mice infected with M. pulmonis developed more severe
clinical disease and had higher mortality than did infected
female mice. Female C3H mice displayed a chronic wasting syn-
drome characterized by weight loss, ruffled fur, and hunched
appearance but with few deaths. However, male C3H mice
often exhibited a fatal shock-like syndrome within 4 to 6 days
after infection. Gender differences were also present in DBA/
2N and C57BL/6N mice infected with M. pulmonis. Thus, the
gender differences in disease susceptibility were not unique to
one strain of mice, indicating that the increased susceptibility
of male mice to severe mycoplasma respiratory disease is a
common phenomenon.
Pulmonary histopathological tests demonstrates that gender
affects the type and character of the inflammatory response in
C3H mice. In M. pulmonis-infected male mice, the pulmonary
lesion was an acute inflammatory response in the alveoli, char-
acterized by a predominantly neutrophilic infiltrate, edema,
and hemorrhage. In contrast, infected female mice developed
a chronic peribronchial inflammatory response with an infil-
tration of mononuclear cells and few neutrophils. This differ-
ence in the inflammatory response between the sexes was not
due to the number of organisms, because the mycoplasma
numbers in the lungs of male and female mice were similar.
Thus, the difference due to gender in mycoplasma disease was
independent of the clearance of the organism but was related
to the mechanisms which regulate the type and character of the
inflammatory response.
Sex hormones appear to directly or indirectly influence the
inflammatory process and host resistance to mycoplasma in the
lungs. M. pulmonis-infected male mice which were neutered
developed less severe lung lesions and had correspondingly
smaller numbers of mycoplasma in the lungs than did infected
control mice. A similar decrease in disease and infection was
observed in female mice after neutering. Surgical removal of
male gonads has been reported to increase protection against
other infections (28, 39). For example, gonadectomized BALB/c
male mice infected with M. marium had increased host resis-
tance, smaller numbers of organisms, and less severe lung le-
sions than did infected control males (39). In preliminary stud-
ies, we also found that testosterone treatment of neutered
female mice increased the severity of lung disease due to M.
pulmonis (unpublished results). Although sex hormones are
probably involved in mycoplasma disease, the mechanisms re-
sponsible for the gender differences in inflammatory lesions,
independent of the clearance of the organism, are unknown.
Since the severity of inflammation is regulated by lympho-
cyte and macrophage activities, gender differences may be linked
to one or both of these cell populations. In previous studies, we
demonstrated that lymphocyte activity has both beneficial and
deterimental effects on the pathogenesis of mycoplasma dis-
ease, including the development of pulmonary inflammation
(5). In support of lymphocyte responses contributing to the
gender differences in mycoplasma disease, female mice are
TABLE 2. Gender differences after infection with M. pulmonis
a
Outcome of
infection
% of mice showing outcome
b
C57BL/6 DBA/2N
Female Male Female Male
Gross lung lesions 0 (0/15) 53 (8/15)
c
100 (16/16) 100 (16/16)
Death 0 (0/15) 0 (0/15) 38 (6/16) 100 (16/16)
c
a
C57BL/6N and DBA/2N mice were infected with 2 10
4
CFU M. pulmonis
CT. Mortality and the presence of gross lung lesions were noted 21 days postin-
fection.
b
Percentage of affected mice (number of affected mice/total number of mice).
c
A significant difference between male and female mice was found (P0.05).
Experiments were performed twice.
TABLE 1. Influence of neutering on mycoplasma
numbers in the lungs
Gender Control mice Neutered mice
CFU
a
nCFU
a
n
Male 59 10
3
(1.4) 11 17 10
3
(1.7)
b
10
Female 33 10
3
(1.5) 11 4.4 10
3
(1.2)
b
12
a
Geometric mean (x/standard error) mycoplasma numbers recovered from
the lungs at 7 days postinfection.
b
Neutered male and female mice had a significantly smaller number of my-
coplasmas in their lungs than did sham-neutered mice (P0.05). There was no
significant difference between the mycoplasma numbers recovered from sham-
neutered male and female mice.
VOL. 69, 2001 GENDER DIFFERENCES IN MYCOPLASMA RESPIRATORY DISEASE 2869
known to develop higher antibody responses to various anti-
gens after immunization (1, 33). Also, sex hormones regulate
T-cell-mediated immune function (24, 37, 38). However, we
found no significant difference in the levels of anti-M. pulmonis
antibody in serum in male and female mice at 7 days after
infection. At this time point, the predominant antibody re-
sponse was of the IgM class, with low levels of IgG and IgA.
Differences in disease due to gender were observed within the
first week after infection, further suggesting that adaptive im-
mune responses are not responsible for these phenomena. In
support, preliminary studies demonstrate that these gender
differences are observed in mice with severe combined immu-
nodeficiency (SCID mice), which lack functional T and B cells
(unpublished results). In addition, sex hormones are able to
modulate the activation of macrophages, including the produc-
tion of proinflammatory cytokines (2, 32, 36). Thus, the differ-
ences in the early stage of M. pulmonis disease in male and
female mice may be due to the proinflammatory activity of
macrophages in the lungs (2, 32, 36) rather than to gender
differences in their anti-mycoplasmal activity in the lungs (10,
16, 20–22, 36, 39, 40). However, additional work is needed
before we can further understand the mechanisms through
which gender affects the susceptibility to respiratory diseases.
In summary, this is the first study to demonstrate that gender
does play a significant role in determining the severity of my-
coplasma respiratory disease in mice. In fact, the gender of the
host was a major factor in determining whether an acute or
chronic inflammatory lung disease developed after infection
with M. pulmonis. This is in contrast to the gender differences
described for mycobacterial infection of mice, where only gran-
ulomatous lung lesions are present (39, 40). The results of
these studies also suggest that modulation of innate host de-
fenses and proinflammatory responses by sex hormones is a
major factor in these gender-related differences. Although
gender is a significant factor in lung disease in humans and
animals, the mechanisms for these differences are not clearly
understood; however, mycoplasma disease in mice will serve as
an excellent animal model to further delineate the role of sex
hormones on both acute and chronic inflammatory lung dis-
eases.
ACKNOWLEDGMENTS
The technical support of Heather Baker, Eugene Arms, Chandra E.
Frank, and Yvette Hale is greatly appreciated. We thank J. Russell
Lindsey and Gail H. Cassell for their useful discussion and support
during the performance of these studies.
This work and preparation of the manuscript was supported by
Public Health Service grants HL19741 (to G.H.C.), AI 42075 (to J.W.S.),
and RR11105 (to J. R. Lindsey) from the National Institutes of Health.
REFERENCES
1. Ahmed, S. A., and N. Talal. 1990. Sex hormones and the immune system.
Part 2. Animal data. Baillieres Clin. Rheumatol. 4:13–31.
2. Angele, M. K., M. W. Knoferl, M. G. Schwacha, A. Ayala, W. G. Cioffi, K. I.
Bland, and I. H. Chaudry. 1999. Sex steroids regulate pro- and anti-inflam-
matory cytokine release by macrophages after trauma-hemorrhage. Am. J.
Physiol. 277:C35–C42.
3. Ashman, R. B., P. H. Kay, D. M. Lynch, and J. M. Papadimitriou. 1991.
Murine candidiasis: sex differences in the severity of tissue lesions are not
associated with levels of serum C3 and C5. Immunol. Cell Biol. 69:7–10.
4. Callahan, C. M., and F. D. Wolinsky. 1996. Hospitalization for pneumonia
among older adults. J. Gerontol. Ser. A 51:M276–M282.
5. Cartner, S. C., J. R. Lindsey, J. Gibbs-Erwin, G. H. Cassell, and J. W.
Simecka. 1998. Roles of innate and adaptive immunity in respiratory myco-
plasmosis. Infect. Immun. 66:3485–3491.
6. Cartner, S. C., J. W. Simecka, J. R. Lindsey, G. H. Cassell, and J. K. Davis.
1995. Chronic respiratory mycoplasmosis in C3H/HeN and C57BL/6N mice:
lesion severity and antibody response. Infect. Immun. 63:4138–4142.
7. Cassell, G. H. 1982. Derrick Edward Award Lecture. The pathogenic po-
tential of mycoplasmas: Mycoplasma pulmonis as a model. Rev. Infect. Dis.
4(Suppl.):S18–S34.
8. Cassell, G. H., J. K. Davis, and J. R. Lindsey. 1981. Control of Mycoplasma
pulmonis infection in rats and mice: detection and elimination vs. vaccina-
tion. Isr. J. Med. Sci. 17:674–677.
9. Cassell, G. H., J. R. Lindsey, R. G. Overcash, and H. J. Baker. 1973. Murine
mycoplasma respiratory disease. Ann. N. Y. Acad. Sci. 225:395–412.
10. Chao, T. C., A. Phuangsab, P. J. Van Alten, and R. J. Walter. 1996. Steroid
sex hormones and macrophage function: regulation of chemiluminescence
and phagocytosis. Am. J. Reprod. Immunol. 35:106–113.
11. Chen, C. W., G. M. Jong, J. J. Shiau, T. R. Hsiue, H. Y. Chang, Y. C. Chuang,
and C. R. Chen. 1992. Adult bacteremic pneumonia: bacteriology and prog-
nostic factors. J. Formosan Med. Assoc. 91:754–759.
12. Cherry, J. D., E. S. Hurwitz, and R. C. Welliver. 1975. Mycoplasma pneu-
moniae infections and exanthems. J. Pediatr. 87:369–373.
13. Davis, J. K., and G. H. Cassell. 1982. Murine respiratory mycoplasmosis in
LEW and F344 rats: strain differences in lesion severity. Vet. Pathol. 19:280–
293.
14. Davis, J. K., R. F. Parker, H. White, D. Dziedzic, G. Taylor, M. K. Davidson,
N. R. Cox, and G. H. Cassell. 1985. Strain differences in susceptibility to
murine respiratory mycoplasmosis in C57BL/6 and C3H/HeN mice. Infect.
Immun. 50:647–654.
15. Davis, J. K., R. B. Thorp, P. A. Maddox, M. B. Brown, and G. H. Cassell.
1982. Murine respiratory mycoplasmosis in F344 and LEW rats: evolution of
lesions and lung lymphoid cell populations. Infect. Immun. 36:720–729.
16. Ferrandez, M. D., and M. de la Fuente. 1999. Effects of age, sex and physical
exercise on the phagocytic process of murine peritoneal macrophages. Acta
Physiol. Scand. 166:47–53.
17. Foy, H. M. 1993. Infections caused by Mycoplasma pneumoniae and possible
carrier state in different populations of patients. Clin. Infect. Dis. 17(Suppl.
1):S37–S46.
18. Foy, H. M., G. E. Kenny, M. K. Cooney, and I. D. Allan. 1979. Long-term
epidemiology of infections with Mycoplasma pneumoniae. J. Infect. Dis. 139:
681–687.
19. Gordon, H. S., and G. E. Rosenthal. 1999. The relationship of gender and
in-hospital death: increased risk of death in men. Med. Care 37:318–24.
20. Hickman-Davis, J., J. Gibbs-Erwin, J. R. Lindsey, and S. Matalon. 1999.
Surfactant protein A mediates mycoplasmacidal activity of alveolar macro-
phages by production of peroxynitrite. Proc. Natl. Acad. Sci. USA. 96:4953–
4958.
21. Hickman-Davis, J. M., J. R. Lindsey, S. Zhu, and S. Matalon. 1998. Surfac-
tant protein A mediates mycoplasmacidal activity of alveolar macrophages.
Am. J. Physiol. 274:L270–L277.
22. Hickman-Davis, J. M., S. M. Michalek, J. Gibbs-Erwin, and J. R. Lindsey.
1997. Depletion of alveolar macrophages exacerbates respiratory mycoplas-
mosis in mycoplasma-resistant C57BL mice but not mycoplasma-susceptible
C3H mice. Infect. Immun. 65:2278–2282.
23. Huber, S. A., L. P. Job, and K. R. Auld. 1982. Influence of sex hormones on
Coxsackie B-3 virus infection in Balb/c mice. Cell. Immunol. 67:173–179.
24. Krzych, U., H. R. Strausser, J. P. Bressler, and A. L. Goldstein. 1981. Effects
of sex hormones on some T and B cell functions, evidenced by differential
immune expression between male and female mice and cyclic pattern of
immune responsiveness during the estrous cycle in female mice. Am. J.
Reprod. Immunol. 1:73–77.
25. Lindsey, J. R., H. J. Baker, R. G. Overcash, G. H. Cassell, and C. E. Hunt.
1971. Murine chronic respiratory disease. Significance as a research compli-
cation and experimental production with Mycoplasma pulmonis. Am. J.
Pathol. 64:675–708.
26. Loeb, M., A. McGeer, M. McArthur, S. Walter, and A. E. Simor. 1999. Risk
factors for pneumonia and other lower respiratory tract infections in elderly
residents of long-term care facilities. Arch. Intern. Med. 159:2058–2064.
27. McIntosh, J. C., J. W. Simecka, S. E. Ross, J. K. Davis, E. J. Miller, and
G. H. Cassell. 1992. Infection-induced airway fibrosis in two rat strains with
differential susceptibility. Infect. Immun. 60:2936–2942.
28. Mock, B. A., and C. A. Nacy. 1988. Hormonal modulation of sex differences
in resistance to Leishmania major systemic infections. Infect. Immun. 56:
3316–3319.
29. Offner, P. J., E. E. Moore, and W. L. Biffl. 1999. Male gender is a risk factor
for major infections after surgery. Arch. Surg. 134:935–938, discussion 938–
940.
30. Parker, R. F., J. K. Davis, D. K. Blalock, R. B. Thorp, J. W. Simecka, and
G. H. Cassell. 1987. Pulmonary clearance of Mycoplasma pulmonis in
C57BL/6N and C3H/HeN mice. Infect Immun. 55:2631–2635.
31. Parker, R. F., J. K. Davis, G. H. Cassell, H. White, D. Dziedzic, D. K.
Blalock, R. B. Thorp, and J. W. Simecka. 1989. Short-term exposure to
nitrogen dioxide enhances susceptibility to murine respiratory mycoplasmo-
2870 YANCEY ET AL. INFECT.IMMUN.
sis and decreases intrapulmonary killing of Mycoplasma pulmonis. Am. Rev.
Respir. Dis. 140:502–512.
32. Savita, and U. Rai. 1998. Sex steroid hormones modulate the activation of
murine peritoneal macrophages: receptor mediated modulation. Comp. Bio-
chem. Physiol. Ser. C 119:199–204.
33. Schuurs, A. H., and H. A. Verheul. 1990. Effects of gender and sex steroids
on the immune response. J. Steroid Biochem. 35:157–172.
34. Simecka, J. W., and G. H. Cassell. 1987. Serum antibody and cellular re-
sponses in LEW and F344 rats after immunization with Mycoplasma pulmo-
nis antigens. Infect. Immun. 55:731–735.
35. Simecka, J. W., J. K. Davis, M. K. Davidson, S. E. Ross, C. Stadtlander, and
G. H. Cassell. 1992. Mycoplasma diseases of animals, p. 391–415. In J.
Maniloff, R. N. McElhaney, L. R. Finch, and J. B. Baseman (ed.), Myco-
plasmas. Molecular biology and pathogenesis. American Society for Micro-
biology, Washington, D.C.
36. Spitzer, J. A. 1999. Gender differences in some host defense mechanisms.
Lupus 8:380–383.
37. Wang, Y., H. D. Campbell, and I. G. Young. 1993. Sex hormones and dexa-
methasone modulate interleukin-5 gene expression in T lymphocytes. J.
Steroid Biochem. Mol. Biol. 44:203–210.
38. Wyle, F. A., and J. R. Kent. 1977. Immunosuppression by sex steroid hor-
mones. The effect upon PHA- and PPD-stimulated lymphocytes. Clin. Exp.
Immunol. 27:407–415.
39. Yamamoto, Y., H. Saito, T. Setogawa, and H. Tomioka. 1991. Sex differences
in host resistance to Mycobacterium marinum infection in mice. Infect. Im-
mun. 59:4089–4096.
40. Yamamoto, Y., H. Tomioka, K. Sato, H. Saito, Y. Yamada, and T. Setogawa.
1990. Sex differences in the susceptibility of mice to infection induced by
Mycobacterium intracellulare. Am. Rev. Respir. Dis. 142:430–433.
Editor: E. I. Tuomanen
VOL. 69, 2001 GENDER DIFFERENCES IN MYCOPLASMA RESPIRATORY DISEASE 2871
... A similar trend has been reported in mouse models treated with Mycoplasma pulmonis, where the mortality of males was increased compared to females and was correlated with dense inflammatory cell infiltrates within the pulmonary alveoli of males [33]. Interestingly, there were no big differences in IgM serum levels between male and female mice. ...
... In search of the underlying mechanisms that define the sex bias in infection severity and pathogenicity of the virus, several preliminary studies have proposed different biological mechanisms regulating immune responses in a sexbiased manner, contributing to viral defense mechanisms [29][30][31][32][33][34][35]. ...
Article
Full-text available
Sex presents a vital determinant of a person’s physiology, anatomy, and development. Recent clinical studies indicate that sex is also involved in the differential manifestation of various diseases, affecting both clinical outcome as well as response to therapy. Genetic and epigenetic changes are implicated in sex bias and regulate disease onset, including the inactivation of the X chromosome as well as sex chromosome aneuploidy. The differential expression of X-linked genes, along with the presence of sex-specific hormones, exhibits a significant impact on immune system function. Several studies have revealed differences between the two sexes in response to infections, including respiratory diseases and COVID-19 infection, autoimmune disorders, liver fibrosis, neuropsychiatric diseases, and cancer susceptibility, which can be explained by sex-biased immune responses. In the present review, we explore the input of genetic and epigenetic interplay in the sex bias underlying disease manifestation and discuss their effects along with sex hormones on disease development and progression, aiming to reveal potential new therapeutic targets. Key messages Sex is involved in the differential manifestation of various diseases. Epigenetic modifications influence X-linked gene expression, affecting immune response to infections, including COVID-19. Epigenetic mechanisms are responsible for the sex bias observed in several respiratory and autoimmune disorders, liver fibrosis, neuropsychiatric diseases, and cancer.
... Males typically exhibit weaker humoral and cell-mediated immune responses (15), and delayed lung maturation (16,17) compared to females. It has also been observed that the number and the activity of cells involved in innate immunity differ between sexes (18,19) as well as in lung diseases (20)(21)(22)(23)(24). Animal models of respiratory infection have shown that sex influences susceptibility, and severity of disease (25)(26)(27)(28)(29)(30)(31)(32) and that sex hormones play a role (33). Therefore, it is important to identify and study the factors that can influence the incidence, susceptibility, and severity of lung diseases. ...
... Previous studies have shown sex differences in lung function and disease susceptibility (29,31,32,72,73), and in risk, incidence, and pathogenesis of various lung diseases (74,75). Human and animal studies have shown an increased incidence of respiratory infections and severity of pneumonia in males (23,27). Furthermore, sexdependent survival was observed in wild type and SP-A-KO mice in response to K. pneumoniae infection, with females exhibiting higher survival compared to males, and that pattern reversed after oxidative stress (29), with females exhibiting lower survival compared to males. ...
Article
Full-text available
Surfactant protein A (SP-A) in addition to its surfactant-related functions interacts with alveolar macrophages (AM), the guardian cells of innate immunity in the lungs, and regulates many of its functions under basal condition and in response to various pressures, such as infection and oxidative stress. The human SP-A locus consists of two functional genes, SFTPA1 and SFTPA2, and one pseudogene. The functional genes encode human SP-A1 and SP-A2 proteins, respectively, and each has been identified with several genetic variants. SP-A variants differ in their ability to regulate lung function mechanics and survival in response to bacterial infection. Here, we investigated the effect of hSP-A variants on the AM gene expression profile in response to Klebsiella pneumoniae infection. We used four humanized transgenic (hTG) mice that each carried SP-A1 (6A2, 6A4) or SP-A2 (1A0, 1A3), and KO. AM gene expression profiling was performed after 6 h post-infection. We found: (a) significant sex differences in the expression of AM genes; (b) in response to infection, 858 (KO), 196 (6A2), 494 (6A4), 276 (1A0), and 397 (1A3) genes were identified (P < 0.05) and some of these were differentially expressed with ≥2 fold, specific to either males or females; (c) significant SP-A1 and SP-A2 variant-specific differences in AM gene expression; (d) via Ingenuity Pathway Analysis (IPA), key pathways and molecules were identified that had direct interaction with TP53, TNF, and cell cycle signaling nodes; (e) of the three pathways (TNF, TP-53, and cell cycle signaling nodes) studied here, all variants except SP-A2 (1A3) female, showed significance for at least 2 of these pathways, and KO male showed significance for all three pathways; (f) validation of key molecules exhibited variant-specific significant differences in the expression between sexes and a similarity in gene expression profile was observed between KO and SP-A1. These results reveal for the first time a large number of biologically relevant functional pathways influenced in a sex-specific manner by SP-A variants in response to infection. These data may assist in studying molecular mechanisms of SP-A-mediated AM gene regulation and potentially identify novel therapeutic targets for K. pneumoniae infection.
... In terms of gender differences in disease susceptibility, the mice challenge experiment conducted with M. pulmonis demonstrated that male mice had more severe lung parenchymal inflammatory damage than female mice [22]. The finding suggests that there is a possibility of gender differences in terms of the severity of Mycoplasma infection, therefore explaining our study finding of more male cases observed in the children group, similar to another study performed during the COVID-19 pandemic [23]. ...
Article
Full-text available
The rebound characteristics of respiratory infections after lifting pandemic control measures were uncertain. From January to November 2023, patients presenting at a teaching hospital were tested for common respiratory viruses and Mycoplasma pneumoniae using a combination of antigen, nucleic acid amplification, and targeted next-generation sequencing (tNGS) tests. The number and rate of positive tests per month and clinical and microbiological characteristics were analyzed. A rapid rebound of SARS-CoV-2 was followed by a slower rebound of M. pneumoniae, with an interval of 5 months between their peaks. The hospitalization rate was higher, with infections caused by respiratory viruses compared to M. pneumoniae. Though the pediatric hospitalization rate of respiratory viruses (66.1%) was higher than that of M. pneumoniae (34.0%), the 4094 cases of M. pneumoniae within 6 months posed a huge burden on healthcare services. Multivariate analysis revealed that M. pneumoniae-infected adults had more fatigue, comorbidities, and higher serum C-reactive protein, whereas children had a higher incidence of other respiratory pathogens detected by tNGS or pathogen-specific PCR, fever, and were more likely to be female. A total of 85% of M. pneumoniae-positive specimens had mutations detected at the 23rRNA gene, with 99.7% showing A2063G mutation. Days to defervescence were longer in those not treated by effective antibiotics and those requiring a change in antibiotic treatment. A delayed but significant rebound of M. pneumoniae was observed after the complete relaxation of pandemic control measures. No unusual, unexplained, or unresponsive cases of respiratory infections which warrant further investigation were identified.
... As for our case, the patient was a male also diagnosed with multiple comorbidities, including diabetes mellitus, hypothyroidism, and adrenal insufficiency, and having a possible underlying UTI infection, suggesting an increased risk of E. coli pneumonia from bacteremia spread in critically ill patients. Males may also be more susceptible to more severe pneumonia symptoms due to hormonal influences [13]. Furthermore, the age range reported in the studies was 52-73 years old while our patient at age 44 years old was below the age range [5]. ...
Article
Full-text available
Escherichia (E.) coli pneumonia is a rare infection commonly presenting with a cavitary lesion. We report a case of a 44-year-old Hispanic male with comorbidities who was admitted to our facility with multiple falls for two days, shortness of breath, continuous diarrhea, and urinary urgency. Lab results showed leukocytosis with neutrophil predominance, anemia, and respiratory alkalosis. The patient was also noted to have uncontrolled diabetes mellitus with an A1c of 17.6%. Prior to admission to the medical intensive care unit (MICU), the patient was administered vancomycin and cefepime. The patient was then started on fluconazole while admitted to the MICU. In addition, a chest X-ray was conducted, showing patchy airspace opacities in the right upper lobe. A chest and abdominal CT also showed multiple cavitary lesions, pulmonary nodules, and nodular liver contour. Bronchoscopy with bronchoalveolar lavage conferred trimethoprim/sulfamethoxazole-resistant E. coli without fungal or acid-fast bacilli growth and was subsequently started on ampicillin/sulbactam. Infectious disease was consulted and advised to begin ertapenem. The patient developed increased respiratory demands and was subsequently started on mechanical ventilation with vasopressors. The patient was successfully weaned off and downgraded to the telemetry floor. The patient was successfully discharged in stable condition. This case highlights a severe and uncommon complication of E. coli infection causing pneumonia with cavitary lesions.
... The role of sex and sex hormones on lung immunity in both humans and animals has been previously documented (71)(72)(73)(74)(75)(76)(77)(78)(79)(80)(81). A number of animal models have shown differences in survival after infection as a function of sex (13,21,32,44,59,82) as well as in disease susceptibility and severity (32,44,59,(83)(84)(85)(86)(87). In humans, both prematurely born males vs. females exhibit higher susceptibility to neonatal respiratory distress syndrome (RDS) (73,74), and adult males exhibit a higher susceptibility in Idiopathic pulmonary fibrosis (IPF), and Chronic obstructive pulmonary disease (COPD) (77,88) and others, as well as in different types of pneumonia (79,80,88,89). ...
Article
Full-text available
Human surfactant protein (SP)-A1 and SP-A2 exhibit differential qualitative and quantitative effects on the alveolar macrophage (AM), including a differential impact on the AM miRNome. Moreover, SP-A rescue (treatment) of SP-A-knockout (KO) infected mice impoves survival. Here, we studied for the first time the role of exogenous SP-A protein treatment on the regulation of lung alveolar cell (LAC) miRNome, the miRNA-RNA targets, and gene expression of SP-A-KO infected mice of both sexes. Toward this, SP-A-KO mice of both sexes were infected with Klebsiella pneumoniae, and half of them were also treated with SP-A2 (1A0). After 6 h of infection/SP-A treatment, the expression levels and pathways of LAC miRNAs, genes, and target miRNA-mRNAs were studied in both groups. We found 1) significant differences in the LAC miRNome, genes, and miRNA-mRNA targets in terms of sex, infection, and infection plus SP-A2 (1A0) protein rescue; 2) an increase in the majority of miRNA-mRNA targets in both study groups in KO male vs. female mice and involvement of the miRNA-mRNA targets in pathways of inflammation, antiapoptosis, and cell cycle; 3) genes with significant changes to be involved in TP-53, tumor necrosis factor (TNF), and cell cycle signaling nodes; 4) when significant changes in the expression of molecules from all analyses (miRNAs, miRNA-mRNA targets, and genes) were considered, two signaling pathways, the TNF and cell cycle, referred to as “integrated pathways” were shown to be significant; 5) the cell cycle pathway to be present in all comparisons made. Because SP-A could be used therapeutically in pulmonary diseases, it is important to understand the molecules and pathways involved in response to an SP-A acute treatment. The information obtained contributes to this end and may help to gain insight especially in the case of infection.
... Male house finches have been known to have a greater susceptibility to MG infection during an MG pandemic (Nolan et al., 1998). In mice infected with Mycoplasma pulmonis, males had more severe respiratory disease symptoms than females, and neutered male mice showed a reduction in disease symptoms (Yancey et al., 2001). The effects of testosterone on the adaptive immune system of chickens has also been shown. ...
Article
Full-text available
Previous trials in which layers were in ovo-vaccinated against strain F Mycoplasma gallisepticum (FMG) showed that nearly 50% of the birds produced IgM antibody against FMG at 6 wk of age (WOA). Standard FMG vaccination application at 9 or 10 woa, result in this percentage at approximately 15 woa. This study investigated when FMG in ovo-vaccinated birds initiate a humoral immune response prior to 6 wk, and if sex influences this response. Hy-Line W-36 embryonated eggs were either not vaccinated (controls) or in-ovo vaccinated with a 50 µL volume of a 10⁻⁶ dilution of Poulvac MycoF vaccine (Zoetis). For each treatment group, 384 straight-run chicks were reared. At hatch and at 2, 3, 5, 7, 14, 21, and 28 d post-hatch, 54 birds per treatment were individually weighed and a blood sample was collected for Mycoplasma gallisepticum (MG) IgM antibody detection. ELISA was run on blood samples at 14, 21, and 28 d to distinguish IgG antibody production. At each age, BW was not different between vaccinated and control chicks (all P > 0.19). Males, however, outweighed females starting at d 5 (P = 0.02). Mortality was 1.0% for the control birds and 12.2% for the FMG birds during the first 2 wk. The majority (72.3%) of the mortalities in the FMG group were male. The percentage of control and FMG in ovo-vaccinated birds with IgM antibody production was 0% and 1.9% on d 7, 0% and 31.5% on d 14, 1.9% and 55.9% on d 21, and 0% and 60.6% on d 28, respectively. IgG antibody production in the FMG in ovo-vaccinated birds was 0.0% at 14 d, 2.9% at 21 d, and 21.2% at 28 d of age. All control birds tested negative for FMG-IgG production. In conclusion, the earliest detection of MG antibodies after in ovo vaccination with live FMG occurred at 7 d. Male layer chickens were more susceptible to the effects of an in ovo FMG vaccine than females.
... On the other hand, Yancey, Watson, Cartner and Simecka [30] noticed that there was no gender difference in disease severity along the airways or any difference in mycoplasma numbers in lungs of male and female mice despite, surgical removal of reproductive organs reduced the severity of mycoplasma disease and the numbers of mycoplasma organisms recovered from lungs. ...
... 3 Only four case reports of MIRM in adults were found in the literature, including three men (26-, 27-, and 42years-old) with classic MIRM and one 46-year-old man with MIRM skin rash. 4,5,6,7 There is a known male predominance in MIRM, with 66% of the identified cases occurring in males, and this predominance may even be more pronounced in the adult population. There is some evidence that men develop more severe lung disease in response to M pneumoniae infections as compared to women. ...
... Males and females differ in their immunological responses to pathogens, and males generally show higher susceptibility, prevalence, and severity of infection than females, including respiratory tract infection (20)(21)(22). Observed in mice infected with Mycoplasma pulmonis, the pulmonary parenchyma disease of male mice is always more serious than that of female mice (23). On the other hand, smoking is generally more prevalent among men than women. ...
Article
Full-text available
This study was performed to describe the epidemiologic characteristics of coronavirus disease 2019 (COVID-19) and explore risk factors for severe infection. Data of all 131 confirmed cases in Tianjin before February 20 were collected. By February 20, a total of 14/16 districts reported COVID-19 cases, with Baodi district reporting the most cases (n = 56). A total of 22 (16.8%) cases had a Wuhan-related exposure. Fever was the most common symptom (82.4%). The median duration of symptom onset to treatment was [1.0 (0.0–4.0) days], the duration of symptom onset to isolation [2.0 (0.0–6.0) days], and the duration of symptom onset to diagnosis [5.0 (2.0–8.0) days]. The analysis of the transmission chain showed two cluster infections with 62 cases infected. Transmission from a family member constituted 42%, usually at the end of transmission chain. Compared with patients with non-severe infections, patients with severe infections were more likely to be male (46.2 vs. 77.3%, P = 0.009) and had a Wuhan-related exposure (14.0 vs. 40.9%, P = 0.004). Multivariate logistic regression showed that male (OR 3.913, 95% CI 1.206, 12.696; P = 0.023) was an independent risk factor for severe infection. This study provides evidence on the epidemic of COVID-19 by analyzing the epidemiological characteristics of confirmed cases in Tianjin. Self-quarantine at an outbreak's early stage, especially for those with high-risk exposures, is conducive to prevent the transmission of infection. Further investigation is needed to confirm the risk factors for severe COVID-19 infection and investigate the mechanisms involved.
Article
Background The purpose of this study is to evaluate risk factors for pneumonia following THA and TKA. Methods Patients were identified from the American College of Surgeons National Quality Improvement Database (NSQIP) who experienced postoperative pneumonia after undergoing primary THA and TKA. Results Many characteristics including old age, anemia, diabetes, cardiac comorbidities, dialysis, and smoking were independent risk factors for postoperative pneumonia after THA or TKA. Conclusion This analysis offers new evidence on risk factors associated with the development of pneumonia after THA and TKA. These risk factors can help guide clinicians in preventing postoperative pneumonia after THA and TKA.
Article
Full-text available
Mice deficient in the fifth component of complement are known to be extremely susceptible to lethal challenge with Candida albicans. However, male mice, that have significantly higher concentrations of serum C5 than females, were markedly more susceptible to infection. This difference was observed in both susceptible (CBA/H) and resistant (BALB/c) mice. Levels of serum C3 likewise showed no correlation with susceptibility.
Article
Full-text available
Sex hormones have physiological and pathological (autoimmune conditions) effects on the immune system. Studies in experimental animal models of human autoimmune diseases have clearly shown that sex hormones regulate the expression, severity and course of autoimmune diseases.Sex hormones affect the function of T, B and NK cells, and macrophages. Precisely how sex hormones affect lymphocytes is a highly complex question. Sex hormones can modulate the immune system, perhaps directly (e.g. thymic reticular tissue), or indirectly via host and many oestrogen target tissues, including the central nervous system hypothalamic-pituitary axis (the neuroendocrine tissues) (Figure 1). The effects of sex hormones on the immune system (immunosuppression or immunopotentiation) may vary, even with the same hormone. For example, oestrogen can increase IgA levels in the uterus, but decrease IgA levels in the vagina or have no effect in lacrimal tissues (Sullivan, 1989). Therefore the effects of sex hormones on the immune system cannot be generalized but must be evaluated independently.Some of the reasons for variability in results have been reviewed in detail elsewhere (Steinberg et al, 1979; Ansar Ahmed et al, 1985b). These include, dose of hormones, age and sex-hormonal status of animals, route and time of administration, the immunocompetence of the host, stress, the metabolism of hormones (e.g. metabolism of testosterone to oestrogen) resulting in alteration of biological activity, and differential response to various antigens. The initial encounter of sex hormones with the type of target cells, the variability of secondary messengers and gene activation events are other important considerations.The effects of sex hormones on the immune system to modulate immune responses are unequivocal. The burgeoning advances in cellular immunology, endocrinology and molecular biology, should provide a better understanding of: (1) the interactions of hormones with the immune system; (2) how hormones activate specific genes; and (3) how hormones influence intracellular communication. In a clinical situation, it is hoped that androgenic compounds which lack virilizing effects, but possessing the desired immunomodulatory effects, will eventually be synthesized. These hormone analogues, in combination with specific (non-toxic) oestrogen antagonists, may offer new therapeutic avenues.
Article
Full-text available
Mycoplasma pulmonis causes a chronic respiratory disease in rats which is more severe in LEW than in F344 rats. This study compared the ability of each of these rat strains to produce specific immune responses to M. pulmonis antigens. By an enzyme-linked immunosorbent assay, LEW rats were found to produce approximately 10 times lower levels of specific immunoglobulin G (IgG) after immunization with M. pulmonis antigens than F344 rats, while no significant difference was found in the levels of IgM. The difference in IgG levels was due to much greater levels of specific IgG2b (about 50 times) in F344 rats; no differences were found in other subclasses. Nonimmune LEW rats were found to have as much total IgG2b in their sera as unimmunized F344 rats by a single radial immunodiffusion test; thus, the difference was not due to the inability of LEW rats to produce IgG2b. In contrast to the antibody response to M. pulmonis antigens, anti-keyhole limpet hemocyanin IgG responses in LEW and F344 rats were similar, but F344 rats produced significantly more (about 21 times) IgG2b than was found in M. pulmonis responses. Antisera from F344 rats recognized several additional M. pulmonis antigens than antisera from LEW rats; however, this could not explain the differences in the level of IgG2b in LEW and F344 rats. In vitro stimulation of splenic lymphocytes with M. pulmonis antigens from immunized F344 rats produced much greater proliferative responses than in LEW and nonimmune F344 cells. Thus, the susceptible rat strain LEW produced lower cellular and humoral immune responses to M. pulmonis antigens than the resistant rat strain F344 after immunization.
Article
Objectives. The prognostic importance of gender in hospitalized patients has been poorly studied. The current study compared in-hospital death rates between men and women after adjusting for severity of illness. Design. Retrospective cohort study. Patients. 89,793 eligible patients with 6 common nonsurgical diagnoses who were discharged from 30 hospitals in Northeast Ohio in 1991 to 1993. Methods. Admission severity of illness (ie, predicted risk of death) was calculated using multivariable models that were based on data abstracted from patients' clinical records (ROC curve areas, 0.83-0.90). In hospital death rates were then adjusted for predicted risks of death and other covariates using logistic regression analysis. Results. Adjusted odds of death were higher (P < 0.05) in men, compared with women, for 4 diagnoses (stroke [OR, 1.60]; obstructive airway disease [OR, 1.38]; gastrointestinal hemorrhage [OR 1.32]; pneumonia [OR, 1.18]) and similar for two diagnoses (congestive heart failure [OR, 1.12]; and acute myocardial infarction [OR, 0.97]). These differences were somewhat attenuated by excluding patients discharged to skilled nursing facilities or other hospitals from analysis; nonetheless, the odds of death in men remained higher for 3 diagnoses. Conclusions. The findings indicate that in-hospital death rates are generally higher in men than in women, after adjusting for severity of illness. In addition, the risk of in-hospital death in men and women was influenced by diagnosis. These differences may reflect gender-related variation in the utilization of hospital services, the effectiveness of care, over- or underestimation of severity of illness, or biological differences in men and women.
Article
For years the ubiquity of Mycoplasma pulmonis in rodents overshadowed its pathogenic potential. Its etiologic significance in murine chronic respiratory disease was established only by recognition of the delicate equilibrium between organism and host. Environmental factors and genetic predisposition of the host rather than microbial virulence are the critical determinants of disease. The prevalence of M. pulmonis is undoubtedly related to the recently demonstrated in utero transmission and the ability of the organism to colonize and produce disease in the genital tract. The etiologic significance of Ureaplasma urealyticum in human genitourinary disease, like its murine counterpart, has been surrounded by controversy. Recent studies indicate that only a subgroup of colonized individuals develop clinical manifestations of disease, ranging from infertility to fetal wastage. While the natural occurrence of both respiratory and genital mycoplasmoses seriously restricts the usefulness of rats and mice for other research purposes, they represent useful models for the study of human disease. The recognized morphologic similarities and similar natural histories of chronic bronchitis, bronchiectasis, and emphysema in humans and of M. pulmonis respiratory disease in rats and mice make the latter a particularly useful model for study of the pathogenesis of chronic pulmonary inflammation. At the same time, murine genital mycoplasmosis represents one of the few naturally occurring genital tract diseases in laboratory animals and therefore makes an attractive model for elucidating those subtle host-parasite interactions that predispose to genital disease and subsequent reproductive failure.
Article
Few diseases of laboratory animals have been as troublesome to research projects or as enigmatic to microbiologists as murine mycoplasma respiratory disease. Its entire history has been characterized by continuing disagreement about possible etiologies and endless proliferation of new terms for its pathological identification. Its ubiquity as an indigenous infection in most animal facilities has made the study of the problem by many investigators impractical, or even impossible. Only very recently has there been significant success in reproducing all lesions of the natural disease by inoculation of a single agent, M. pulmonis, into rats and mice known to be free of other pathogens. The purpose of the present paper is: (1) to summarize recent progress in understanding pathogenesis and host response of experimental M. pulmonis infection in mice and rats, (2) to present data that may help explain past difficulties of reproducing lower respiratory tract disease with this agent in the rat, and (3) to place the experimental and natural diseases of both species in proper perspective.
Article
Pneumonia due to Mycoplasma pneumoniae was monitored in a large prepaid medical-care group in Seattle, Washington, between 1963 and 1975. The disease was diagnosed by isolation of M. pneumoniae and/or significant rises in titer of complementfixing (antilipid) antibody in paired sera. Infection was endemic without significant seasonal fluctuations. Two epidemics occurred: the first peaked in January 1967, the second late in the summer of 1974. Total rates of pneumonia infection in children increased during M. pneumoniae epidemics, but epidemics of infection with respiratory syncytial virus had a greater effect. Age-specific attack rates for M. pneumoniae pneumonia among children aged five to nine years (about six per 1,000) were about twice the rates for younger children and four times those for adults. Serologic study of healthy schoolchildren showed annual rates of infection that paralleled but greatly exceeded rates of recognized M. pneumoniae pneumonia. Infection rates varied from 2% in endemic years to 35% in epidemic periods. A higher proportion of infections among children aged five to nine years than among adolescents aged 15–19 years resulted in pneumonia.
Article
Progesterone, estradiol, testosterone, cortisol, and 11-desoxycortisol (compound S) were added to cultures of human lymphocytes stimulated with phytohaemagglutinin (PHA) and purified protein derivative (PPD). The immunosuppressive effect of cortisol was verified and the three sex-steroid hormones also were found to inhibit lymphocyte transformation although at concentrations higher than for cortisol. Compound S, a steroid of low biological potency, also had immunosuppressive activity. At concentrations (0-01-1-0 microng/ml), progesterone, oestrogen, testosterone, and Compound S augmented the transformation response to PPD but not to PHA. Marked variation from individual to individual in the suppressive effects of all the steroids were noted. The clinical implications of immunosuppression by the sex steroid hormones are discussed.
Article
A review of the medical literature and two case reports of M. pneumoniae infections with exanthems are presented. Erythematous maculopapular and vesicular exanthems were most common. The duration of rash was more than seven days in the majority of instances, and most patients had associated pneumonia. A striking difference in prevalence and clinical symptomatology by sex was noted; 16 of 20 patients analyzed were males, and they frequently dad severe mucocutaneous syndromes. In contrast, severe conjunctivitis, generalized ulcerative stomatitis, and vesicular or bullous exanthems were not seen in females. Clinicians should suspect infection with M. pneumoniae in patients with exanthem and pneumonia, although other etiologic possibilities should also be considered.
Article
Bacteremic pneumonia is a highly specified subgroup of pneumonia that is potentially life-threatening. In order to find out the prognostic factors in this subgroup of pneumonia, we conducted a 40-month retrospective analysis of 70 cases in our hospital. The male to female ratio was 54:16. Forty-one cases were community-acquired bacteremic pneumonia (CABP), and 29 cases were nosocomial bacteremic pneumonia (NBP). Both CABP and NBP were predominated by gram-negative bacteria. Klebsiella pneumoniae was the most common microorganism isolated in both CABP and NBP. The overall mortality was 62.9% (44/70). There was no significant difference in the mortality between CABP (61.0%) and NBP (65.5%). After univariate analysis of all possible prognostic factors, 10 variables were found to have significantly poor prognostic values. They were: 1) the presence of septic shock; 2) the use of ventilatory support; 3) the presence of radiologic spread; 4) treatment in an intensive care unit; 5) male gender; 6) the development of adult respiratory distress syndrome; 7) Klebsiella bacteremic pneumonia in patients with an alcohol habit; 8) patients with ultimately fatal underlying diseases; 9) an initial AaDO2 > 200 mmHg; and 10) an initial arterial pH < 7.25.