The role of Toxoplasma gondii as a possible inflammatory ...

19
Family Medicine and Community Health SYSTEMATIC REVIEW Family Medicine and Community Health 2016;4(4):44–62 44 www.fmch-journal.org DOI 10.15212/FMCH.2016.0128 © 2016 Family Medicine and Community Health REVIEW The role of Toxoplasma gondii as a possible inflammatory agent in the pathogenesis of type 2 diabetes mellitus in humans Aus Molan 1 , Kazunori Nosaka 1 , Michael Hunter 2,3 , Wei Wang 1 Abstract Type 2 diabetes mellitus (T2DM) continues to be a major challenge for public health authorities worldwide. While potential causes such as obesity, physical inactivity, and dietary patterns have been proposed to explain the growing epidemic, there may also be unidentified environmental determinants. An emerging field of research is starting to examine the association of infectious and environmental pathogens with diabetes. In particular, the potential of these pathogens to cause low- grade inflammation that facilitates the risk and development of T2DM. An understudied pathogen of potential interest is the protozoan parasite Toxoplasma gondii (T. gondii). There is limited clinical evidence supporting the association between chronic T. gondii infection and the development of many disorders, including T2DM, in both animals and humans. This review (1) addresses the exist- ing knowledge of the role of T. gondii in the inflammation process leading to T2DM, (2) examines the current studies describing the relationship between T. gondii and T2DM, and (3) makes recom- mendations for future studies to determine the role of T. gondii in the pathogenesis of T2DM. We believe that T. gondii may be an important target for T2DM intervention, and propose a new field of study, “toxoplasmic type 2 diabetes.” Keywords: Etiology; inflammation; Toxoplasma gondii infection; toxoplasmosis; diabetes mel- litus, type 2 1. School of Medical and Health Sciences, Edith Cowan Univer- sity, Joondalup, WA, Australia 2. Busselton Population and Med- ical Research Institute, Busselton, WA, Australia 3. School of Population Health, University of Western Australia, Nedlands, WA, Australia CORRESPONDING AUTHOR: Wei Wang, MD, PhD, FFPH School of Medical and Health Sciences, Edith Cowan Uni- versity, 270 Joondalup Drive, Joondalup WA 6027, Australia Tel.: +61-8-63043717 E-mail: [email protected] Received 24 August 2016; Accepted 18 September 2016 Introduction Diabetes mellitus is one of the most challeng- ing public health burdens of the 21st century [1]. It is a chronic disorder characterized by persistent hyperglycemia with disturbances in fat, carbohydrate, and protein metabo- lism resulting from abnormalities in insulin secretion, insulin action, or both [1, 2]. Type 2 diabetes mellitus (T2DM; International Classification of Diseases, Tenth Revision, Clinical Modification diagnosis code E11.9) is characterized by reduced insulin production and an inability of body tissues to fully respond to insulin (insulin resistance) [1–3], account- ing for approximately 90% of all diagnosed diabetes cases [2, 3]. The pathogenesis of T2DM has been recognized as an ongoing deterioration of the insulin secretory capacity of pancreatic β cells that does not allow com- pensation for an increased peripheral insulin demand [4, 5]. The prevalence of diabetes is reaching epidemic levels, with an estimated global prevalence of 9% among adults aged 18 years or older [6–12]. Additionally, dia- betes places excessively high human, social, and economic costs on all countries. In 2013, on November 8, 2021 by guest. Protected by copyright. http://fmch.bmj.com/ Fam Med Com Health: first published as 10.15212/FMCH.2016.0128 on 1 December 2016. Downloaded from

Transcript of The role of Toxoplasma gondii as a possible inflammatory ...

Family Medicine and Community Health

SYSTEMATIC REVIEW

Family Medicine and Community Health 2016;4(4):44–62 44www.fmch-journal.org DOI 10.15212/FMCH.2016.0128© 2016 Family Medicine and Community Health

RE

VIE

W

The role of Toxoplasma gondii as a possible inflammatory agent in the pathogenesis of type 2 diabetes mellitus in humans

Aus Molan1, Kazunori Nosaka1, Michael Hunter2,3, Wei Wang1

Abstract

Type 2 diabetes mellitus (T2DM) continues to be a major challenge for public health authorities

worldwide. While potential causes such as obesity, physical inactivity, and dietary patterns have

been proposed to explain the growing epidemic, there may also be unidentified environmental

determinants. An emerging field of research is starting to examine the association of infectious and

environmental pathogens with diabetes. In particular, the potential of these pathogens to cause low-

grade inflammation that facilitates the risk and development of T2DM. An understudied pathogen

of potential interest is the protozoan parasite Toxoplasma gondii (T. gondii). There is limited clinical

evidence supporting the association between chronic T. gondii infection and the development of

many disorders, including T2DM, in both animals and humans. This review (1) addresses the exist-

ing knowledge of the role of T. gondii in the inflammation process leading to T2DM, (2) examines

the current studies describing the relationship between T. gondii and T2DM, and (3) makes recom-

mendations for future studies to determine the role of T. gondii in the pathogenesis of T2DM. We

believe that T. gondii may be an important target for T2DM intervention, and propose a new field

of study, “toxoplasmic type 2 diabetes.”

Keywords: Etiology; inflammation; Toxoplasma gondii infection; toxoplasmosis; diabetes mel-

litus, type 2

1. School of Medical and Health

Sciences, Edith Cowan Univer-

sity, Joondalup, WA, Australia

2. Busselton Population and Med-

ical Research Institute, Busselton,

WA, Australia

3. School of Population Health,

University of Western Australia,

Nedlands, WA, Australia

CORRESPONDING AUTHOR:

Wei Wang, MD, PhD, FFPH

School of Medical and Health

Sciences, Edith Cowan Uni-

versity, 270 Joondalup Drive,

Joondalup WA 6027, Australia

Tel.: +61-8-63043717

E-mail: [email protected]

Received 24 August 2016;

Accepted 18 September 2016

Introduction

Diabetes mellitus is one of the most challeng-

ing public health burdens of the 21st century

[1]. It is a chronic disorder characterized by

persistent hyperglycemia with disturbances

in fat, carbohydrate, and protein metabo-

lism resulting from abnormalities in insulin

secretion, insulin action, or both [1, 2]. Type

2 diabetes mellitus (T2DM; International

Classification of Diseases, Tenth Revision,

Clinical Modification diagnosis code E11.9) is

characterized by reduced insulin production

and an inability of body tissues to fully respond

to insulin (insulin resistance) [1–3], account-

ing for approximately 90% of all diagnosed

diabetes cases [2, 3]. The pathogenesis of

T2DM has been recognized as an ongoing

deterioration of the insulin secretory capacity

of pancreatic β cells that does not allow com-

pensation for an increased peripheral insulin

demand [4, 5]. The prevalence of diabetes is

reaching epidemic levels, with an estimated

global prevalence of 9% among adults aged

18 years or older [6–12]. Additionally, dia-

betes places excessively high human, social,

and economic costs on all countries. In 2013,

on Novem

ber 8, 2021 by guest. Protected by copyright.

http://fmch.bm

j.com/

Fam

Med C

om H

ealth: first published as 10.15212/FM

CH

.2016.0128 on 1 Decem

ber 2016. Dow

nloaded from

Molan et al.

45 Family Medicine and Community Health 2016;4(4):44–62

RE

VIE

W

5.1 million deaths were caused by diabetes worldwide (one

person every 6 s) [6, 12]. Looking ahead, it is estimated that

642 million people will be living with diabetes by 2040, which

is a more than 50% increase from 415 million in 2015 [7]. This

is mainly due to the increase in the numbers of people with

T2DM as a result of the increase in life expectancy, genetic

predisposition, physical inactivity, dietary changes, the obe-

sity epidemic, and the decreased mortality rates in diabetic

individuals [1, 3, 6–11]. However, there may also be additional

unidentified novel risk factors, such as subclinical inflamma-

tion caused by infectious agents, that contribute to this rising

prevalence of T2DM [13].

There are epidemiological data that link inflammatory bio-

markers as important risk indicators for future development of

diabetes [14]. In this regard, an emerging field of research is

beginning to investigate the potential of infectious and envi-

ronmental pathogens to cause low-grade inflammation that

may facilitate the risk and development of various metabolic

conditions, including diabetes and obesity. An understud-

ied pathogen of potential interest is the protozoan parasite

Toxoplasma gondii (T. gondii) (International Classification

of Diseases, Tenth Revision, Clinical Modification diagnosis

code B58.9). First described in 1908 by Nicolle and Manceaux

[15], T. gondii infects approximately one third of the world’s

population and is considered one of the most successful human

parasites [16–19]. Humans acquire T. gondii by the ingestion

of food, water, or soil contaminated by oocysts from the defin-

itive hosts, cats (Fig. 1). T. gondii is also transmitted in people

vertically via the maternal placenta and horizontally via blood

transfusion [18, 21–23]. T. gondii is capable of infecting all

warm-blooded animals [24], and toxoplasmosis is a disease

of considerable public health impact – the global prevalence

rates of infection by this parasite are phenomenal, with figures

ranging from 15% to 85% depending on dietary habits, cli-

mate conditions, hygiene standards, and geographical regions

[21, 23, 25–27]. Although T. gondii has a worldwide distribu-

tion and possibly the widest host range of any parasite, there

is only one species (T. gondii) in the genus Toxoplasma [28],

and cats are the only definitive host in which T. gondii sexual

development is known to occur [25, 20].

Toxoplasmosis can present with various nonspecific signs

and symptoms, but most are similar to general flulike indicators

[22, 23, 27]. In immunocompetent individuals, it is thought

that the disease is asymptomatic and that the individual recov-

ers without treatment because of an efficient immune system

which limits the spread of the rapidly multiplying tachyzoites

[22]. Therefore people are not routinely screened for T. gondii

infection unless they are immunocompromised or pregnant

[4, 23–25]. In all cases the parasite remains detectable in the

serum throughout the life of the host, with dormant cysts being

formed in various anatomical sites, including the central nerv-

ous system, often establishing latent toxoplasmosis [18, 22].

T. gondii can infect and replicate in any nucleated host cells,

leading to the production of various inflammatory markers

via the innate acute inflammatory responses and antigen-spe-

cific adaptive immunity [4]. This facilitates a state of chronic

inflammation at various anatomical sites in the host [4].

Chronic T. gondii infection has been linked to several auto-

immune disorders, including thyroid disease, systemic sclero-

sis, rheumatoid arthritis, and inflammatory bowel syndrome

[29]. In addition, several reports have demonstrated a positive

correlation between T. gondii infection and numerous neu-

rological disorders and cancers [4, 22, 23, 28, 30]. However,

T. gondii infection in individuals with T2DM has received

little recognition, and human studies investigating T. gondii

infection and inflammatory biomarkers in T2DM subjects are

not available. This review (1) addresses the existing knowl-

edge of the role of T. gondii in the inflammation process lead-

ing to T2DM, (2) examines the current studies describing

the relationship between T. gondii and T2DM, and (3) makes

recommendations for further studies to determine the role of

T. gondii in the pathogenesis of T2DM using inflammation

as the main outcome. The association between T2DM and

T. gondii, if proven, will have important and significant public

health outcomes. Routine screening for T. gondii infection in

individuals with T2DM would add to the risk prediction for

diabetes to target individuals for early aggressive interven-

tion. Targeting T. gondii and reducing its risk factors could be

used as an Achilles heel to reduce inflammation, leading to

improved glucose tolerance and reduced insulin resistance in

diabetic individuals. We believe that T. gondii latent infection

may have excellent potential as a novel target for T2DM inter-

vention and may pave a path for a new field of study, “toxoplas-

mic type 2 diabetes.”

on Novem

ber 8, 2021 by guest. Protected by copyright.

http://fmch.bm

j.com/

Fam

Med C

om H

ealth: first published as 10.15212/FM

CH

.2016.0128 on 1 Decem

ber 2016. Dow

nloaded from

The role of Toxoplasma gondii as a possible inflammatory agent

Family Medicine and Community Health 2016;4(4):44–62 46

RE

VIE

W

Host inflammatory responses to T. gondii infection

The founding study by Frenkel [31] demonstrated that the

transfer of serum from infected to uninfected animals failed

to protect the latter against parasite infection. This knowl-

edge assisted in pointing toward T cells rather than antibod-

ies as the major effectors of resistance to T. gondii infection.

Direct

Indirect

Fig. 1. The complete life cycle of Toxoplasma gondii. Infection consists of an acute phase followed by a chronic phase. The life cycle is divided

into the sexual and asexual stages, where the former occurs in the intestine of the definitive host, while the latter occurs in the intermediate hosts.

After completion of the sexual phase, oocysts released in the feces undergo sporulation to form sporocysts that become a source of infection

for intermediate hosts. When the oocysts are ingested, the internal sporocysts pass through the stomach and excyst in the intestine to release

sporozoites. These sporozoites invade epithelial cells and differentiate into tachyzoites, an actively replicating form of T. gondii. Because of their

active multiplication, tachyzoites cause extensive tissue damage and may disseminate to different tissues of the host. T. gondii infects circulating

cells and can use them as a Trojan horse to gain access to protective tissues such as the brain, where entry of immune cells is restricted. The

tachyzoites eventually undergo stage conversion and become slowly multiplying bradyzoites within tissue cysts. If for some reason the tissue

cyst becomes activated, the bradyzoites can undergo stage conversion and become tachyzoites. The bradyzoite form is the only stage that can

infect feline enterocytes and lead to oocyst production, restarting the life cycle. (Adapted from Hunter and Sibley [20] with permission).

on Novem

ber 8, 2021 by guest. Protected by copyright.

http://fmch.bm

j.com/

Fam

Med C

om H

ealth: first published as 10.15212/FM

CH

.2016.0128 on 1 Decem

ber 2016. Dow

nloaded from

Molan et al.

47 Family Medicine and Community Health 2016;4(4):44–62

RE

VIE

W

More recently, it has become clear that T. gondii manipulates

and modulates host resistance mechanisms at multiple points

along the proinflammatory pathway which in turn dictate

parasite burden and disease (Fig. 2) [32]. During the acute

phase of this infection, which lasts for less than 10 days, T.

gondii elicits a very strong T-helper type 1 (Th1) cell medi-

ated inflammatory response (Table 1) [41]. The ability of the

host to control T. gondii depends on the subsequent produc-

tion of interleukin (IL)-12 by macrophages and dendritic

cells, resulting from the differentiation and clonal expansion

of host Th1 cells in response to the recognition of conserved

T. gondii microbe molecular structures (glycosylphosphati-

dylinositol anchors and profilin) by Toll-like receptors [32,

92]. In turn, IL-12 stimulates natural killer cells and CD4+

and CD8+ T cells to release interferon gamma (IFN-γ), which

is also central in host resistance to T. gondii infection [32].

The Th1 response, defined by the production of IL-12 and

IFN-γ, is a characteristic of infection with intracellular patho-

gens, including T. gondii [93]. In fact, a significant discovery

that allowed progress toward the identification of essential

recognition and effector mechanisms involved in host resist-

ance to T. gondii infection was the identification of IFN-γ

as the major regulator of cell-mediated immunity to T. gon-

dii in both mouse and human cells [94–97]. Studies show-

ing the importance of T cells in resisting T. gondii infection

are unequivocal. For example, mice which lack functional T

cells are extremely susceptible to T. gondii infection [94, 95].

Likewise, IL-12- or IFN-γ-deficient mice are extremely sus-

ceptible to infection by T. gondii and demonstrate an inabil-

ity to control parasite burden [96, 97]. Furthermore, several

murine models have demonstrated the protection of naïve

mice against T. gondii infection as a result of adoptive trans-

fer of immune T cells [31, 98–100]. With regard to T. gondii

infection in humans, the importance of the adaptive immune

response has been demonstrated by the increase of suscep-

tibility to T. gondii infection in individuals with primary or

acquired T-cell function defects [96].

In addition to the systemic effects, T. gondii also induces

the production of a number of proinflammatory cytokines

(tumor necrosis factor alpha, TNF-α; IL-1; IL-1β; IL-6;

IL-18; IL-12p40; IL-8; IL-17; IL-22; IL-15), anti-inflam-

matory cytokines (transforming growth factor beta, TGF-β;

IL-4; IL-10; IL-27), nitric oxide synthase, and reactive oxy-

gen species that are associated with inflammatory responses

in various sites and cells, including the brain, microglial

cells, astrocytes, and infiltrating CD4+ and CD8+ T cells

[30, 32, 41, 50, 61–67, 92, 101–108]. It is important to note

that excess production of proinflammatory cytokines ends

up causing damage to the host; therefore a delicate balance

between the above mentioned proinflammatory and anti-

inflammatory signals is necessary for the survival of the host

[32]. Maintaining this immune homeostasis during T. gon-

dii infection requires both the capacity to control the host

immune response and the capacity to limit the replication of

the pathogen [93]. To this extent, IL-10, TGF-β, and IL-27

are key downregulators that prevent the overproduction of

Th1-type cytokines, thereby regulating inflammation [32,

92, 103, 104]. More specifically, during T. gondii infection,

the in vivo synthesis of both IL-12 and IFN-γ is modulated

by IL-10 to avoid an excessive immune response that could

cause host tissue damage and widespread inflammation [45,

109]. Paradoxically, while the host immune response lim-

its the tissue spread of the parasite to ensure the survival of

the host, it is also assisting in the survival of the parasite

by converting it into a bradyzoite, an intracellular dormant

resistant form persisting in the muscles, central nervous sys-

tem, and brain tissues [106]. Given that a strong Th1 immune

response is elicited to maintain the dormant bradyzoite stage

[110] and that a Th1 immune response is also maintained to

promote long-lasting immunity [111], hosts infected with T.

gondii remain in an “asymptomatic” state of constant low-

grade inflammation.

T2DM as an inflammatory disease

Inflammation not only participates in host defenses against

environmental and infectious agents such as T. gondii but also

contributes to the pathophysiology of many chronic diseases

[112]. Increased circulatory acute-phase response markers

and proinflammatory cytokines in individuals with T2DM

were first reported in 1997 [113]. Since then, the ideas that

inflammation participates in the pathogenesis of T2DM and

that T2DM is a chronic inflammatory disease have gained

undeniable attention, with many reports describing changes

to immune cell function [4, 43, 53, 55, 114–117]. In addition,

on Novem

ber 8, 2021 by guest. Protected by copyright.

http://fmch.bm

j.com/

Fam

Med C

om H

ealth: first published as 10.15212/FM

CH

.2016.0128 on 1 Decem

ber 2016. Dow

nloaded from

The role of Toxoplasma gondii as a possible inflammatory agent

Family Medicine and Community Health 2016;4(4):44–62 48

RE

VIE

W

Fig. 2. Important inflammatory regulators and antiparasitic effector mechanisms during acute infection with Toxoplasma gondii. Early in

T. gondii infection, dendritic cells, monocytes, and macrophages are the first cells to respond. T. gondii profilin interacts with Toll-like

receptor (TLR) 11 and TLR12 on dendritic cells. This is important for the host production of interleukin (IL)-12. In addition, macrophages

also induce tumor necrosis factor alpha (TNF-α) in response to TLR2- and TLR4-mediated detection of glycosylphosphatidylinositol (GPI)-

anchored parasite proteins. The innate immune response results in the production of interferon gamma (IFN-γ) from natural killer (NK) cells.

Eventually, this induces inflammatory mediators that control T. gondii multiplication and direct the developing adaptive immune system,

resulting in the expansion of T-helper type 1 (Th1) cells and cytolytic CD8+ T cells that secrete IFN- γ. Th1 cell differentiation is induced by

the release of IL-12 and IL-18, while T-helper type 17 (Th17) cell differentiation and expansion is promoted by transforming growth factor

beta (TGF-β), IL-6, and IL-23. This is counterregulated by T-helper type 2 (Th2) cells and regulatory T (Treg) cells mainly through the

production of IL-4 and IL-10. IL-10 and IL-27 are key to modulating these pathways and prevent the overproduction of Th1-type cytokines.

The production of IFN-γ during the innate and adaptive phases is responsible for activating cells to control parasite infection. IFN-γ

signaling promotes the development of signal transducer and activator of transcription 1 (STAT1; a nuclear transcription factor that controls

the expression of many genes) dependent antiparasitic effector mechanisms, including reactive oxygen intermediates and p47 GTPase

upregulation. The cell-mediated immune response that results limits parasite replication. IFNγR, Interferon gamma receptor; MC, mast cells;

MP, macrophages; ROS, reactive oxygen species.

on Novem

ber 8, 2021 by guest. Protected by copyright.

http://fmch.bm

j.com/

Fam

Med C

om H

ealth: first published as 10.15212/FM

CH

.2016.0128 on 1 Decem

ber 2016. Dow

nloaded from

Molan et al.

49 Family Medicine and Community Health 2016;4(4):44–62

RE

VIE

W

Tabl

e 1.

Im

mun

e re

spon

se to

Tox

opla

sma

gond

ii a

nd ty

pe 2

dia

bete

s m

ellit

us in

hum

an a

nd a

nim

al m

odel

s

Ana

lyze

d

biom

arke

r

B

iolo

gica

l act

ivity

Sel

ecte

d re

fere

nces

Type

2 d

iabe

tes

mel

litus

res

pons

e

Toxo

plas

ma

gond

ii re

spon

se

IL-1

β

PI. M

edia

tor

of in

flam

mat

ory

resp

onse

. Inv

olve

d in

a v

arie

ty o

f

cellu

lar

activ

ities

(ce

ll pr

olif

erat

ion,

diff

eren

tiatio

n, a

nd a

popt

osis

)

Secr

eted

fro

m m

onoc

ytes

: Mos

ser

and

Edw

ards

[33

], G

iulie

tti e

t al.

[34]

, Hat

anak

a

et a

l. [3

5], P

icku

p [3

6], P

icku

p an

d C

rook

[37

]

Ars

enije

vic

et a

l. [3

8], F

abia

ni e

t al.

[39]

, Tom

asik

et a

l. [4

0]

IL-6

PI

cyt

okin

e an

d A

I m

yoki

ne

(inc

reas

ed d

urin

g ex

erci

se).

Stim

ulat

es im

mun

e re

spon

se

Secr

eted

fro

m m

onoc

ytes

: Mos

ser

and

Edw

ards

[33

], G

iulie

tti e

t al.

[34]

, Hat

anak

a

et a

l. [3

5], P

icku

p [3

6], P

icku

p an

d C

rook

[37

]

Ars

enije

vic

et a

l. [3

8], F

ilise

tti a

nd C

ando

lfi [

41],

Prad

a an

d N

go-T

u [4

2], P

arlo

g et

al.

[21]

, Fab

iani

et a

l. [3

9]

IL-8

PI

. CX

CL

8. I

nduc

es c

hem

otax

is a

nd

phag

ocyt

osis

Secr

eted

fro

m m

onoc

ytes

: Mos

ser

and

Edw

ards

[33

], G

iulie

tti e

t al.

[34]

, Hat

anak

a

et a

l. [3

5], P

icku

p [3

6], P

icku

p an

d C

rook

[37

].

Secr

eted

fro

m β

cel

ls: J

agan

nath

an e

t al.

[43]

Parl

og e

t al.

[21]

IL-1

0

AI.

Dow

nreg

ulat

es e

xpre

ssio

n of

Th1

cyto

kine

s. I

nvol

ved

in r

egul

atio

n of

the

JAK

–STA

T s

igna

ling

path

way

Secr

eted

fro

m β

cel

ls: J

agan

nath

an e

t al.

[43]

.

Not

wel

l stu

died

Ars

enije

vic

et a

l. [3

8, 4

4], F

ilise

tti a

nd C

ando

lfi

[41]

, Gaz

zine

lli e

t al.

[45]

, Fab

iani

et a

l. [3

9]

IL-1

2

PI. I

nvol

ved

in d

iffe

rent

iatio

n of

nai

ve

T c

ells

into

Th1

cel

ls. S

timul

ated

prod

uctio

n of

IFN

-γ a

nd T

NF-

α

Weg

ner

et a

l. [4

6], T

siav

ou e

t al.

[47]

, Win

er

et a

l. [4

8]

Blis

s et

al.

[49]

, Fili

setti

and

Can

dolfi

[41

], G

azzi

nelli

et a

l. [5

0, 4

5], N

guye

n et

al.

[51]

, Par

log

et a

l. [2

1],

Fabi

ani e

t al.

[39]

, Lie

berm

an e

t al.

[60]

IL-1

7

PI. I

nduc

ed a

nd m

edia

tes

PI r

espo

nses

(pro

duct

ion

of I

L-6

, G-C

SF, G

M-C

SF,

IL-1

β, T

GF-

β, T

NF-

α)

Jaga

nnat

han

et a

l. [5

3], W

iner

et a

l. [5

4],

Her

der

et a

l. [5

5], A

ndri

anka

ja e

t al.

[56]

,

Osb

orn

et a

l. [5

7], Y

ang

et a

l. [5

8], A

cost

a-

Rod

rigu

ez e

t al.

[59]

Fabi

ani e

t al.

[39]

, Kel

ly e

t al.

[55]

TG

F-β

A

I. C

ontr

ols

prol

ifer

atio

n, c

ellu

lar

dif f

eren

tiatio

n

Lev

el in

crea

ses

in th

e pr

edia

betic

sta

ge

Fi

liset

ti an

d C

ando

lfi [

41],

Fis

cher

et a

l. [6

1],

Gaz

zine

lli e

t al.

[50]

, Nag

inen

i et a

l. [6

2]

IFN

PI. A

ctiv

ator

of

mac

roph

ages

and

indu

cer

of M

HC

cla

ss I

I m

olec

ule

expr

essi

on. A

berr

ant I

FN-γ

exp

ress

ion

is a

ssoc

iate

d w

ith a

num

ber

of

auto

infla

mm

ator

y an

d au

toim

mun

e

dise

ases

Jaga

nnat

han

et a

l. [5

3], F

euer

er e

t al.

[63]

,

Win

er e

t al.

[48]

Ara

ujo

and

Slif

er [

64],

Ars

enije

vic

et a

l. [6

5],

Filis

etti

and

Can

dolfi

[41

], G

avri

lesc

u an

d D

enke

rs

[66]

, Gaz

zine

lli e

t al.

[45]

, Her

mes

et a

l. [6

7], K

han

et a

l. [6

8], M

ordu

e et

al.

[69]

, Ngu

yen

et a

l. [5

1],

Peng

et a

l. [1

6], P

arlo

g et

al.

[21]

, Fab

iani

et a

l.

[39]

, Tom

asik

et a

l. [4

0]

TN

F-α

PI

. Inv

olve

d in

sys

tem

ic in

flam

mat

ion

Secr

eted

fro

m m

onoc

ytes

: Mos

ser

and

Edw

ards

[33

], G

iulie

tti e

t al.

[34]

, Hat

anak

a

et a

l. [3

5], P

icku

p [3

6], P

icku

p an

d C

rook

[37

]

Ars

enije

vic

et a

l. [4

4], B

liss

et a

l. [6

1], F

ilise

tti a

nd

Can

dolfi

[41

], G

azzi

nelli

et a

l. [5

0, 4

5], K

han

et a

l.

[68]

, Pra

da a

nd N

go-T

u [4

2], P

arlo

g et

al.

[21]

,

Fabi

ani e

t al.

[39]

on Novem

ber 8, 2021 by guest. Protected by copyright.

http://fmch.bm

j.com/

Fam

Med C

om H

ealth: first published as 10.15212/FM

CH

.2016.0128 on 1 Decem

ber 2016. Dow

nloaded from

The role of Toxoplasma gondii as a possible inflammatory agent

Family Medicine and Community Health 2016;4(4):44–62 50

RE

VIE

W

Ana

lyze

d

biom

arke

r

B

iolo

gica

l act

ivity

Sel

ecte

d re

fere

nces

Type

2 d

iabe

tes

mel

litus

res

pons

e

Toxo

plas

ma

gond

ii re

spon

se

IL-1

RA

A

I. M

embe

r of

the

IL-1

fam

ily.

Secr

eted

by

vari

ous

type

s of

cel

ls,

incl

udin

g im

mun

e ce

lls, e

pith

elia

l

cells

, and

adi

pocy

tes,

and

is a

nat

ural

inhi

bito

r of

the

PI e

ffec

t of

IL-1

β

Lev

el in

crea

ses

in th

e pr

edia

betic

sta

ge

IL-2

7

Bel

ongs

to th

e IL

-12

fam

ily.

Reg

ulat

es th

e ac

tivity

of

B a

nd T

lym

phoc

ytes

Lev

el in

crea

ses

in th

e pr

edia

betic

sta

ge

H

all e

t al.

[70]

IL-4

AI.

Ind

uces

dif

fere

ntia

tion

of n

aive

help

er T

cel

ls to

Th2

cel

ls

Ars

enije

vic

et a

l. [3

8], F

ilise

tti a

nd C

ando

lfi [

41],

Parl

og e

t al.

[21]

, Fab

iani

et a

l. [3

9]

Lep

tin

“S

atie

ty h

orm

one.

” M

ade

by a

dipo

se

cells

and

hel

ps to

reg

ulat

e en

ergy

bala

nce

by in

hibi

ting

hung

er

Ver

y hi

gh le

vel i

n pe

ople

with

type

2 d

iabe

tes

mel

litus

and

obe

sity

(hi

gh, >

15 n

g/m

L):

Mitt

endo

rfer

et a

l. [7

2], M

oon

et a

l. [7

3]

Bal

taci

and

Mog

ulko

c [7

1]

Adi

pone

ctin

M

odul

atio

n of

glu

cose

and

lipi

d

met

abol

ism

in in

sulin

-sen

sitiv

e tis

sues

Lev

el d

ecre

ases

in th

e pr

edia

betic

sta

ge

M

ilova

ncov

ic e

t al.

[74]

(un

affe

cted

)

PAI-

1

Key

reg

ulat

or o

f fib

rino

lysi

s,

thus

atte

nuat

es th

e ac

tivity

of

the

fibri

noly

tic s

yste

m

Auw

erx

et a

l. [7

5], V

ague

et a

l. [7

6], J

anss

on

et a

l. [7

7], S

amps

on e

t al.

[78]

, Juh

an-V

ague

et a

l. [7

9], P

ando

lfi e

t al.

[80]

, Gra

nt e

t al.

[81]

,

Nor

dt e

t al.

[82]

, Sch

neid

er a

nd S

obel

[83

],

Sobe

l et a

l. [8

4], A

less

i et a

l. [8

5], C

arm

assi

et a

l. [8

6], C

alle

s-E

scan

don

et a

l. [8

7], M

eigs

et a

l. [8

8]

Tom

asik

et a

l. [4

0]

IL-2

3

Secr

eted

by

activ

ated

den

driti

c ce

lls

and

activ

ated

mac

roph

ages

. IL

-23

func

tions

in in

nate

and

ada

ptiv

e

imm

unity

to r

egul

ate

Th1

7 fu

nctio

n

and

prol

ifer

atio

n

Men

sah-

Bro

wn

et a

l. [8

9], L

ukic

et a

l. [9

0]

(IL

-23

lead

s to

dia

bete

s in

duct

ion)

Muñ

oz e

t al.

[91]

, Lie

berm

an e

t al.

[52]

AI,

Ant

i-in

flam

mat

ory;

G-C

SF, g

ranu

locy

te c

olon

y st

imul

atin

g fa

ctor

; GM

-CSF

, gra

nulo

cyte

–mac

roph

age

colo

ny s

timul

atin

g fa

ctor

; IL

, int

erle

ukin

; IL

-1R

A, i

nter

leuk

in-1

rece

ptor

ant

agon

ist;

IFN

-γ, i

nter

fero

n ga

mm

a; J

AK

, Jan

us k

inas

e; P

AI-

1, p

lasm

inog

en a

ctiv

ator

inhi

bito

r 1;

PI,

pro

infla

mm

ator

y; S

TAT,

sig

nal t

rans

duce

r an

d ac

tivat

or o

f

tran

scri

ptio

n; T

GF-

β tr

ansf

orm

ing

grow

th f

acto

r be

ta; T

h1, T

hel

per

type

1; T

h2, T

hel

per

type

2; T

h17,

T h

elpe

r ty

pe 1

; TN

F-α,

tum

or n

ecro

sis

fact

or a

lpha

.

Tabl

e 1

(con

tinue

d)

on Novem

ber 8, 2021 by guest. Protected by copyright.

http://fmch.bm

j.com/

Fam

Med C

om H

ealth: first published as 10.15212/FM

CH

.2016.0128 on 1 Decem

ber 2016. Dow

nloaded from

Molan et al.

51 Family Medicine and Community Health 2016;4(4):44–62

RE

VIE

W

there are also experimental data that provide evidence for a

direct link between inflammation and T2DM, which supports

the notion that this disease is, at least in part, an inflammatory

condition [13, 112].

Serum-based inflammatory biomarkers as a measure-

ment of subclinical inflammation have been implicated in

the development of T2DM, with several cross-sectional and

prospective studies describing elevated circulating levels of

acute-phase proteins as well as chemokines and cytokines

(Table 1). For example, elevated levels of IL-6 (a cytokine

that induces differentiation of Th17 cells), IL-8, IL-17, TNF-

α, TGF-β, and IL-1β secreted from monocytes in T2DM

patients have been reported [33–35, 43, 53–59]. Elevated lev-

els of IL-1β, IL-6, and C-reactive protein are predictive of

T2DM [115, 117, 118], while elevated levels of IL-1 recep-

tor antagonist are observed during obesity and prediabetes

[117]. Moreover, patients with T2DM also have elevated

serum levels of IL-12, a cytokine that promotes Th1 cell dif-

ferentiation and increased INF-γ production [47, 48, 59, 63].

In addition, proinflammatory cytokines are increasingly

thought to contribute to the dysfunction and death of β cells

during the progression of T2DM [119]. More specifically, it

has been established that pancreatic β cells, as well as neural

cells, can be destroyed by several toxic agents and noxious

stimuli, such as reactive oxygen species, NO, and cytokines

(TNF-α, IL-1β, and IFN-γ) [4]. The subsequent proinflam-

matory cytokine balance has been directly linked to T2DM

by several in vivo studies showing that the inhibition of key

inflammatory cytokines protects rats from insulin resistance

[120–124]. These reports suggest that increased cytokine pro-

duction not only precedes but also maintains insulin resist-

ance in animals, hence adding detail to the current model of

T2DM being an inflammatory disease.

Spranger et al. [115] prospectively examined the effects

of IL-1β, IL-6, and TNF-α on the development of T2DM in

more than 27,000 individuals. In addition to observing ele-

vated levels of IL-6 and TNF-α in individuals with T2DM,

they also found that IL-6 was an independent predictor of

T2DM. Their data also showed that patterns of these cytokines

could modify the risk of T2DM by elevating the levels of both

IL-1β and IL-6, rather than elevating the level of IL-6 alone,

which increases the risk of T2DM. This strongly supports the

hypothesis that a subclinical inflammatory reaction has a role

in the pathogenesis of T2DM.

Possible association between obesity, T2DM, and

T. gondii infection

In addition to T2DM, obesity is also thought to be associ-

ated with a chronic systemic inflammatory response that

is characterized by altered cytokine production and activa-

tion of inflammatory signaling pathways [125]. Obesity has

previously been referred to as “a state of chronic inflamma-

tion” [26]. The increase in the production of inflammatory

cytokines, such TNF-α and IL-6, and certain adipokines

during the inflammatory process in obesity has been linked

to the development of insulin resistance by several reports

[126–128]. It is well established that insulin resistance pre-

cedes the onset of T2DM, at which point impaired glucose

tolerance occurs because of insulin deficiency as a result of

β-cell decomposition [129]. While there are several factors

such as genetics, environmental influences, and obesity that

link the development of insulin resistance with T2DM [129],

the possibility that obesity may enhance the release of inflam-

matory markers that trigger the development of insulin resist-

ance has generated interest in the field of diabetes for a number

of reasons. Predominantly, most individuals with T2DM are

overweight or obese, at least in the early phase, and obesity

is an established risk factor for the development of T2DM [2,

130, 131]. The significant increase in the incidence and preva-

lence of diabetes in the last 2 decades can be largely explained

by the global epidemic of obesity [131]. Furthermore, chronic

inflammation is associated with obesity, insulin resistance,

and T2DM, all of which belong to the metabolic diseases

known as “metabolic syndrome” [132].

The first molecular study to establish the link between

obesity, inflammation, and insulin resistance was published

in 1993 by Hotamisligil et al. [133]. They demonstrated that

the inflammatory cytokine TNF-α is constitutively expressed

in adipose tissue and overexpressed in obese rodents [133].

Furthermore, the inhibition of TNF-α in these obese rodents

significantly increased insulin sensitivity by increasing the

peripheral uptake of glucose in response to insulin. Two years

later, Hotamisligil et al. [134] examined the expression patterns

of TNF-α messenger RNA (mRNA) in the adipose tissues of

on Novem

ber 8, 2021 by guest. Protected by copyright.

http://fmch.bm

j.com/

Fam

Med C

om H

ealth: first published as 10.15212/FM

CH

.2016.0128 on 1 Decem

ber 2016. Dow

nloaded from

The role of Toxoplasma gondii as a possible inflammatory agent

Family Medicine and Community Health 2016;4(4):44–62 52

RE

VIE

W

19 obese and 18 control premenopausal women using Northern

blot analysis and found that the obese group expressed sig-

nificantly greater (2.5-fold, P<0.001) TNF-α mRNA in their

adipose tissue than the lean, control group. In addition, the

expression levels of TNF-α mRNA in adipose tissue showed

a strong correlation with the level of hyperinsulinemia, which

is an indirect measure of insulin resistance [134]. Lastly, they

also observed an increase in insulin sensitivity and a 45%

decrease in TNF-α mRNA expression (P<0.001) in adipose

tissue when the obese individuals reduced their body weight.

In support of a direct role for inflammation in the develop-

ment of obesity, the above-mentioned results were validated

by a human study that found a significant increase in adipose

TNF-α mRNA levels with increasing adiposity and that a sig-

nificant correlation exists between TNF-α mRNA levels and

percent body fat (P<0.05) [135]. It was also found that TNF-α

levels markedly decreased with weight loss and in some indi-

viduals the decreases were up to 46% of initial levels (P<0.02).

Further studies established TNF-α as a potent insulin receptor

inhibitor, hence further implicating this cytokine in the insulin

resistance of T2DM and obesity [136].

With use of animal models it has also been shown that

diet-induced obese mice have more T cells in adipose tissue

than lean, control mice [137], and diet-controlled weight loss

is associated with reduced adipose tissue inflammation [137].

In addition, the development of insulin resistance and other

metabolic abnormalities, including reduced metabolic rates,

has also been associated with chronic inflammation [138,

139]. Accordingly, it is possible that the chronic proinflamma-

tory state induced by latent T. gondii infection precipitates (in

nonobese individuals), perpetuates, or exacerbates (in obese

individuals) the inflammation-related weight gain. Reeves

et al. [130] reported a positive association between T. gondii

seropositivity and obesity from a sample of 999 human adults.

Their results showed that individuals with positive T. gon-

dii serological findings had twice the odds of being obese

(BMI >30 kg/m2) than seronegative individuals (P=0.01).

Furthermore, it has been proposed that infection with

T. gondii may be associated with obesity because of the abil-

ity of the organism to alter inflammatory fat distribution as

it adjusts and resides in fatty tissues [29]. Certainly, exces-

sive gestational weight gain has been previously reported

from a study comparing pregnant woman infected with T.

gondii with uninfected pregnant woman [120]. In that study,

979 mothers (mean age 30 years, range 19–44 years) were

examined, and 194 (19.2%) tested positive for T. gondii infec-

tion and 758 (80.2%) tested negative for T. gondii infection

during the 16th week of pregnancy. The study authors col-

lected data on maternal weight and other parameters before

pregnancy and at the 16th, 20th, 30th, and 36th weeks of

pregnancy. One of the main findings of the study was that

the mean maternal weight of the T. gondii-infected mothers

before pregnancy was much higher (63.6 kg) than that of the

uninfected mothers (61.5 kg). In addition, the effect of T. gon-

dii infection on weight gain was significant in the 16th week

(P=0.006) and the 20th week (P=0.049) of pregnancy and

nearly significant in the 30th week (P=0.073). Moreover, the

extra weight remained until the end of the pregnancies. In

another study, a murine pregnancy model was used to mimic

toxoplasmosis complications in humans and it was found that

dams infected with high doses of T. gondii tachyzoites had

significant excess body weight gain that led to fetal abortion

or mummified embryos [121]. Consistent with the conclusions

of the study authors, excessive weight gain during pregnancy,

especially in the first trimester, usually indicates a negative

effect of some internal (genetic or epigenetic) or external

(environmental) factor(s) [120].

Since obesity in humans is characterized by hyperleptine-

mia and a body-weight-suppressing response to exogenous

leptin [122], it is very interesting to note that the hormone

leptin has also been implicated in the T. gondii host inflamma-

tory response [29]. Using a rat model, Baltaci and Mogulkoc

[71] found that infection with T. gondii caused a significant

increase of plasma leptin concentrations (P<0.01), with no

change in body weight of the animals. They concluded that

this biomarker, similar in structure to IL-2, exerts a proinflam-

matory action in obese and diabetic individuals.

T. gondii may also influence motivation- and reward-driven

behaviors (e.g., overeating) via altered dopamine pathways.

Animal models have shown that T. gondii influences both

dopamine release and availability of the rate-limiting enzyme

in dopamine synthesis [121]. Dopamine antagonists can block

behavioral changes in T. gondii infected rats, and thus para-

site-induced host behavioral changes that may be driven by

on Novem

ber 8, 2021 by guest. Protected by copyright.

http://fmch.bm

j.com/

Fam

Med C

om H

ealth: first published as 10.15212/FM

CH

.2016.0128 on 1 Decem

ber 2016. Dow

nloaded from

Molan et al.

53 Family Medicine and Community Health 2016;4(4):44–62

RE

VIE

W

survival needs may have inadvertent effects on eating patterns

that promote obesity [130].

Accordingly, it can be concluded that the association

between T. gondii infection and obesity is very significant and

could alter the current views on obesity management and other

important public health issues related to obesity, especially

diabetes.

Studies describing the association between

diabetes and T. gondii infection

Only a handful of case–control epidemiological studies that

demonstrate the possible association between T. gondii infec-

tion and T2DM have been published [123, 124, 140, 141].

Most of these studies were conducted in less developed coun-

tries such as Iran, and showed an overall positive association

between T. gondii infection and T2DM (P≤0.001) (Table 2).

Gokce et al. [123] investigated the seropositivity rates

of anti-T. gondii antibodies in 807 individuals with T2DM

(351 men, 456 woman, age range 15–88 years, mean age and

standard deviation 52.8±14.01 years) in comparison with 250

healthy sex- and aged-matched controls (110 men, 140 woman,

age range 18–75 years, mean age and standard deviation

51.9±13.4 years). Using commercial IgG and IgM ELISA kits,

they found IgG antibodies more frequently in the T2DM group

(n=457, 56.6%) than in healthy, control group (n=56, 22.4%,

P<0.001). IgM antibodies were also found in 19 of the patients

with T2DM (2.4%) and only in four controls (1.6%, P=0.3).

Furthermore, a striking correlation was observed between the

duration of diabetes and T. gondii IgG seropositivity (Table 3).

In another serological study, anti-T. gondii IgG antibod-

ies were detected in 60.4% of diabetic patients (n=91) com-

pared with 37.7% of individuals in the healthy, control group

(n=93) [124]. In the same study, the risk of T. gondii infection

in diabetic patients was two-fold higher in comparison with

the healthy controls (odds ratio 2.21, 95% confidence interval

1.6–3.7, P=0.001). However, a similar study also conducted in

2013, by Siyadatpanah et al. [140], did not find a statistically

significant difference in the prevalence of T. gondii in diabetic

(52.6%) and nondiabetic (50.6%) individuals when they meas-

ured the IgG and IgM levels in a cohort of 300 individuals.

Most recently, Saki et al. [141] investigated the prevalence

of anti-T. gondii IgG and IgM antibodies in a cohort of 110 Tabl

e 2.

Des

crip

tion

of d

ata

extr

acte

d fr

om th

e ca

se–c

ontr

ol s

tudi

es d

escr

ibin

g as

soci

atio

n be

twee

n To

xopl

asm

a go

ndii

infe

ctio

n an

d ty

pe 2

dia

bete

s m

ellit

us

Ref

eren

ce

N

Age

M

atch

inga

D

efini

tion

M

easu

re o

f Tox

opla

sma

gond

ii ex

posu

re

Ser

opre

vale

nce

P

T2D

M

posi

tive

(n)

T2D

M

nega

tive

(n)

T2D

M p

ositi

ve

and

Toxo

plas

ma

gond

ii po

sitiv

e (n

) T

2DM

neg

ativ

e

and

Toxo

plas

ma

gond

ii po

sitiv

e (n

)

Saki

et a

l. [1

41]

(201

6, I

ran)

22

0

Adu

lts

No

U

nspe

cifie

d Ig

G/I

gM E

LIS

A (T

rini

ty B

iote

ch,

USA

) and

IFA

kit

(Eur

oim

mun

,

Uni

ted

Kin

gdom

)

11

0 11

0 50

(45

.4%

) 24

(21

.8%

) <0

.001

Siya

datp

anah

et a

l.

[140

] (20

13, I

ran)

30

0

Adu

lts

Uns

peci

fied

Uns

peci

fied

IgG

/IgM

EL

ISA

(V

IRO

,

Ger

man

y)

15

0 15

0 79

(52

.7%

) 76

(50

.6%

) 0.

729

Shir

bazo

u et

al.

[124

] (2

013,

Ira

n)

184

A

dults

Y

es

Uns

peci

fied

IgG

EL

ISA

(Pi

shta

z Te

b Z

aman

Dia

gnos

tics,

Ira

n)

91

93

55

(60

.4%

) 36

(38

.7%

) 0.

003

Gok

ce e

t al.

[123

]

(200

8, T

urke

y)

10

57

15–8

8 ye

ars

Yes

A

DA

Ig

G/I

gM E

LIS

A a

nd I

FAT,

imm

unofl

uore

scen

ce a

ntib

ody

test

(E

uroi

mm

un)

80

7 25

0 45

7 (5

6.6%

) 56

(22

.4%

) <0

.001

Tota

l

1761

11

58

603

641

(55.

4%)

192

(31.

8%)

<0.0

01

AD

A, A

mer

ican

Dia

bete

s A

ssoc

iatio

n; I

FA, i

mm

unofl

uore

scen

ce; I

FAT,

; T

2DM

, typ

e 2

diab

etes

mel

litus

.a M

atch

ing

is b

y ag

e an

d se

x al

one.

on Novem

ber 8, 2021 by guest. Protected by copyright.

http://fmch.bm

j.com/

Fam

Med C

om H

ealth: first published as 10.15212/FM

CH

.2016.0128 on 1 Decem

ber 2016. Dow

nloaded from

The role of Toxoplasma gondii as a possible inflammatory agent

Family Medicine and Community Health 2016;4(4):44–62 54

RE

VIE

W

diabetic pregnant woman and 110 nondiabetic controls and

found that the prevalence rate of both IgG antibody and IgM

antibody in the former group (42.7% and 2.7% respectively)

was significantly higher than in the latter group (21.8% and

0% respectively). They also examined the prevalence rate of

IgG antibody with the duration of diabetes and reported find-

ings similar to those of Gokce et al. [123] in that the preva-

lence rate of IgG antibody was directly associated with the

duration of diabetes.

An animal study has also shown a positive correlation

between both the T. gondii parasite load and the IgM antibody

titer and blood glucose levels in diabetic rats infected with

T. gondii and monitored at 15-day intervals for 60 days after

induction of diabetes and 105 days after T. gondii infection

[142]. During this period, Hassanain et al. [142] observed a sig-

nificant increase in the number of T. gondii cysts (in the brain)

and blood glucose levels in the infected diabetic group when

compared with the uninfected diabetic group. In addition, the

elevated brain parasite load and the IgM titer corresponded

to the elevated glucose levels in the infected diabetic group,

which led to the view that T. gondii infection can increase the

risk of developing diabetes [142].

Modrek et al. [143] reported a significantly high T. gondii

seroprevalence rate of 70.7% among diabetic patients in Iran

(n=205; age range 13–60 years). In addition, the difference

between the presence of toxoplasma antibodies and fasting

blood glucose levels was statistically significant (P<0.05).

While this study lacked a control group, a meta-analysis of

35 reports to determine the seroprevalence rate of T. gondii

infection in the Iranian general population reported an over-

all seroprevalence rate of 39.3% (95% confidence interval

33.0%–45.7%) [144]. This supports the suggestion of an asso-

ciation between T. gondii infection and diabetes in humans.

In addition, there is also evidence of possible indirect

associations between T. gondii infection and certain forms of

diabetes. For example, in 2015, researchers from the Czech

Republic reported significant findings from a cross-sectional

study looking at latent toxoplasma infection and the levels of

blood glucose in pregnant woman [27]. Serum samples for the

determination of T. gondii seropositivity were collected during

the 9th–12th gestational weeks from two subsets of woman

who were later screened for gestational diabetes mellitus in

the 24th–28th gestational weeks at the General University

Hospital in Prague in 2008. Pregnant women with latent toxo-

plasmosis had significantly higher (P=0.010) blood glucose

levels and a higher prevalence (19.5%) of gestational diabetes

mellitus (n=532, P=0.033) in the 24th–28th gestational weeks

than pregnant women without toxoplasmosis (healthy controls,

12%) (Table 4).

The role of T. gondii in the pathogenesis of T2DM remains

an enigma. One possible explanation could be the inflamma-

tory-mediated destruction of pancreatic β cells which leads to

the reduction in β-cell mass that ultimately contributes to the

failure of the β cell to produce enough insulin. This in turn

would increase the risk of developing acute and chronic pancre-

atitis as well as diabetes [5, 145–147]. Additionally, it is known

that T2DM is associated with an increased risk of developing

acute pancreatitis [148] and that T. gondii infection can cause

pancreatic tissue necrosis [149]. IL-18 and IL-1β are partly

responsible for the development of pancreatic inflammation,

and the neutralization of either of these cytokines decreases

inflammation [93]. Accordingly, individuals infected with T.

gondii may be at greater risk of developing diabetes than unin-

fected individuals. Certainly, insulin has been shown to have a

stimulatory effect on the in vitro replication of T. gondii [131].

Insulin and d-glucose have been shown to have a synergistic

Table 3. Relationship between the seropositivity rates of anti-Toxoplasma gondii antibodies analyzed in 807 individuals with type 2 diabetes

mellitus and duration of the disease (according to Gokce et al. [123])

Duration of T2DM (years) IgG-positive individuals (n) IgG-negative individuals (n)

0–5 39 (16.0%) 193 (83.2%)

6–10 149 (51.6%) 140 (48.4%)

>11 269 (94.1%) 17 (5.9%)

T2DM, type 2 diabetes mellitus.

on Novem

ber 8, 2021 by guest. Protected by copyright.

http://fmch.bm

j.com/

Fam

Med C

om H

ealth: first published as 10.15212/FM

CH

.2016.0128 on 1 Decem

ber 2016. Dow

nloaded from

Molan et al.

55 Family Medicine and Community Health 2016;4(4):44–62

RE

VIE

W

dose-responsive stimulating effect on the replication of T. gon-

dii tachyzoites in vitro [150].

Lastly, it has been argued that the simple explanation for

the relationship between T. gondii and T2DM may be that

diabetic patients are more susceptible to parasitic infections

because of the possibility of a suppressed immune system,

decreased arterial perfusion, and neuropathy [151–153].

However, there is evidence to suggest otherwise. A study

looking at parasitic infections in 200 diabetic individuals (16

with type 1 diabetes mellitus and 184 with T2DM) and 1024

nondiabetic individuals found no significant differences with

regard to the incidences of the presence of intestinal parasites

between the T2DM group and the control group (47% and

56% respectively) [154]. In light of above-mentioned reports,

there is a strong clinical and laboratory evidence to support

the notion of a genuine association between chronic latent T.

gondii infection and development of T2DM.

Desirable criteria for good-quality studies

on this topic

If the serological and inflammatory associations between T.

gondii and T2DM are further replicated, this will warrant the

need for further research to clarify whether T. gondii increases

the risk of developing T2DM, and the mechanisms that drive

this relationship, if any. Current reports are limited to associa-

tion studies, meaning that the authors are not able to determine

if there is a causal relationship between T. gondii and T2DM

or obesity, including possible reverse causality (i.e., T2DM

increasing the risk of T. gondii infection) or shared causal-

ity (i.e., common factor causing both T2DM and T. gondii

infection). In addition, studies measuring only T. gondii IgG

and IgM seropositivity cannot shed any light on the possible

inflammatory association between T2DM and T. gondii infec-

tion. Therefore future studies specifically designed to investi-

gate the role of T. gondii in T2DM using inflammation as the

main outcome are urgently needed so as to provide a more

robust link between T. gondii infection and T2DM. To this

extent, studies examining selected inflammatory biomark-

ers that are altered in the serum during and in the absence of

chronic T. gondii infection in individuals with T2DM versus

healthy controls without T2DM will be highly valuable.

Good-quality epidemiological research is warranted to

further help draw conclusions on this important subject. The

design should be a population-based case–control study with

samples representative of all age-groups matched for all pos-

sible confounding parameters. These studies should also use

standard criteria to define various parameters, especially

the diagnosis of T2DM, in addition to setting inclusion and

Table 4. Description of data highlighting the possible indirect association between latent Toxoplasma gondii infection with blood glucose levels

and gestational diabetes mellitus (according to Kankova et al. [27])

Subset 1 (n=191 pregnant women)

OGTT in the 24th–28th GW With Toxoplasma gondii Without Toxoplasma gondii P

Mean baseline BGL (mmol/L) after 8-h fast 5.04 (95% CI 4.92–5.17) 4.88 (95% CI 4.82–4.95) 0.026

Mean BGL (mmol/L) 60 min after administration of

glucose solution

7.73 (95% CI 7.09–8.37) 6.89 (95% CI 6.61–7.16) 0.007

Mean BGL (mmol/L) 120 min after administration of

glucose solution

6.43 (95% CI 6.03–6.83) 5.74 (95% CI 5.55–5.93) 0.001

0.010

Subset 2 (n=532 pregnant women)

GDM in the 24th–28th GW With Toxoplasma gondii (n) Without Toxoplasma gondii (n) P

24/123 (19.5%) 49/409 (12%) 0.033

Blood samples for Toxoplasma gondii analysis were collected in the 9th–12th gestational weeks.

BGL, Blood glucose level; CI, confidence interval; GDM, gestational diabetes mellitus; GW, gestational weeks; OGTT, oral glucose

tolerance test.

on Novem

ber 8, 2021 by guest. Protected by copyright.

http://fmch.bm

j.com/

Fam

Med C

om H

ealth: first published as 10.15212/FM

CH

.2016.0128 on 1 Decem

ber 2016. Dow

nloaded from

The role of Toxoplasma gondii as a possible inflammatory agent

Family Medicine and Community Health 2016;4(4):44–62 56

RE

VIE

W

exclusion criteria according to latest definitions of diabetes by

the World Health Organization. Lastly, individuals with psy-

chiatric conditions, including personality disorders, should

be excluded from these studies because of the association

between positive T. gondii serological findings and various

forms of serious mental illness, as previously reported [18,

21, 115].

Concluding remarks

T2DM is characterized by impaired pancreatic β-cell

function resulting in insulin resistance with an underlying

subclinical inflammation. Many reports suggest that obe-

sity and insulin resistance are associated with elevated

circulating levels of inflammatory cytokines such as IL-2,

IL-6, IL-12, IFN-γ, and TNF-α, raising the possibility that

metabolic abnormalities in diabetes may originate from or

be exacerbated by cytokine overproduction. A possible con-

tributor to this prolonged low-grade inflammation that sub-

sequently leads to the clinical expression of T2DM is the

protozoan parasite T. gondii. Cellular immunity has been

established as the key component of the host’s immune

response to infection by T. gondii. A wealth of data points

to the major role of T lymphocytes and type 1 cytokines

during murine T. gondii infection. In the clinical setting,

this has been confirmed by the emergence of T. gondii as

a major opportunistic infectious pathogen during the pro-

gression of many disorders and diseases. Thus T. gondii is

rapidly emerging as a pathogen of potential interest in obe-

sity and diabetes research. Much progress has been made

in recent years in determining how early innate responses,

namely, production of cytokines such as IL-12 and TNF-α,

drive the development of the strong Th1 response to T. gon-

dii. While this response is crucial to the control of infection,

it is becoming increasingly appreciated that an excessively

vigorous response induced by the parasite can lead to patho-

logical changes in the host. Therefore it is important for the

host and parasite to maintain tight control of the inflamma-

tory response, thereby prolonging it to the detriment of the

host. Human studies investigating inflammatory biomarkers

specifically in T2DM individuals with T. gondii infection

are urgently needed to provide a more robust link between

T. gondii infection and T2DM.

Conflict of interest

The authors declare no conflict of interest.

Funding

This work was partly supported by an Australia–China

international joint research grant (NHMRC-APP1112767-

NSFC-81561128020).

References1. Shaw J, Tanamas S. Diabetes: the silent pandemic and its impact

on Australia. Diabetes Australia [Internet]. [accessed 2016

Jul 12]. Available from: www.healthinfonet.ecu.edu.au/key-

resources/bibliography/?lid=22970.

2. American Diabetes Association. Diagnosis and classification of

diabetes mellitus. Diabetes Care 2008;31:S55–60.

3. Diabetes Australia. [Internet] Diabetes in Australia. [accessed

2016 Jul 1]. Available from: www.diabetesaustralia.com.au/dia-

betes-in-australia.

4. Prandota J. T. gondii infection acquired during pregnancy

and/or after birth may be responsible for development of both

type 1 and 2 diabetes mellitus. J Diabetes Metab 2013;4(2):

1000241.

5. Donath MY, Ehses JA, Maedler K, Schumann DM, Ellingsgaard

H, Eppler R, et al. Mechanisms of β-cell death in type 2 diabetes.

Diabetes 2005;54(Suppl 2):S108.

6. International Diabetes Federation. [Internet] IDF diabetes atlas

(sixth edition). [accessed 2016 Jul 1]. Available from: www.idf.

org/sites/default/files/EN_6E_Atlas_Full_0.pdf.

7. International Diabetes Federation. [Internet] IDF diabetes atlas

(seventh edition) [Internet]. [accessed 2016 Jul 1]. Available

from: www.diabetesatlas.org/.

8. Wild S, Roglic G, Green A, Sicree R, King H. Global prevalence

of diabetes: estimates for the year 2000 and projections for 2030.

Diabetes Care 2004;27(5):1047–53.

9. Lipscombe LL, Hux JE. Trends in diabetes prevalence, incidence,

and mortality in Ontario, Canada 1995–2005: a population-based

study. Lancet 2007;369(9563):750–6.

10. World Health Organization. [Internet] Global status report on

noncommunicable diseases 2014. Geneva: World Health Organi-

zation. [accessed 2016 Jul 12]. Available from: www.who.int/

nmh/publications/ncd-status-report-2014/en/.

11. World Health Organization. [Internet] Diabetes program.

Geneva: World Health Organization. [accessed 2016 Jul 12].

Available from: www.who.int/diabetes/en/.

on Novem

ber 8, 2021 by guest. Protected by copyright.

http://fmch.bm

j.com/

Fam

Med C

om H

ealth: first published as 10.15212/FM

CH

.2016.0128 on 1 Decem

ber 2016. Dow

nloaded from

Molan et al.

57 Family Medicine and Community Health 2016;4(4):44–62

RE

VIE

W

12. World Health Organization. [Internet] Global Health Estimates:

Deaths by Cause, Age, Sex and Country, 2000–2012. Geneva,

WHO. [accessed 2016 July 12]. Available from: www.who.int/

healthinfo/global_burden_disease/en/.

13. Pradhan A. Obesity, metabolic syndrome, and type 2 diabetes:

inflammatory basis of glucose metabolic disorders. Nutr Rev

2007;65(12 Pt 2):S152–6.

14. Shoelson SE, Lee J, Goldfine AB. Inflammation and insulin

resistance. J Clin Invest 2006;116(7):1793–801.

15. Nicolle C, Manceaux L. Sur une infection à corps de Leishman

(ou organismes voisins) du gondi. C R Acad Sci 1908;147:763–6.

16. Peng HJ, Chen XC, Lindsay DS. A review: competence, compro-

mise, and concomitance-reaction of the host cell to Toxoplasma

gondii infection and development. J Parasitol 2011;94(4):620–8.

17. Centers for Disease Control and Prevention. [Internet] Parasites

– toxoplasmosis (toxoplasma infection). Centers for Disease

Control and Prevention. [accessed 2016 Jul 12]. Available from:

www.cdc.gov/parasites/toxoplasmosis/epi.html.

18. Halonen SK, Weiss LM. Toxoplasmosis. In: Garcia HH, Tanow-

itz HB, Del Brutto OH, editors. Neuroparasitology and tropical

neurology. Handbook of clinical neurology, vol. 114 (3rd series).

Amsterdam: Elsevier; 2013. pp. 125–45.

19. Pappas G, Roussos N, Falagas ME. Toxoplasmosis snapshots:

global status of Toxoplasma gondii seroprevalence and implica-

tions for pregnancy and congenital toxoplasmosis. Int J Parasitol

2009;39(12):1385–94.

20. Hunter CA, Sibley LD. Modulation of innate immunity by Toxo-

plasma gondii virulence effectors. Nat Rev 2012;10(11):766–88.

21. Parlog A, Schluter D, Dunay IR. Toxoplasma gondii-induced

neuronal alterations. Parasite Immunol 2015;37(3):159–70.

22. Cong W, Liu G, Meng Q, Dong W, Qin S, Zhang F, et al. Toxo-

plasma gondii infection in cancer patients: Prevalence, risk fac-

tors, genotypes and association with clinical diagnosis. Cancer

Lett 2015;359(2):307–13.

23. Tenter A, Heckeroth A, Weiss M. Toxoplasma gondii: from ani-

mals to humans. Int J Parasitol 2000;30(12–13):1217–58.

24. Robert-Gangneux F, Dardé ML. Epidemiology of and diag-

nostic strategies for toxoplasmosis. Clin Microbiol Rev

2012;25(2):264–96.

25. Albuquerque MC, Aleixo AL, Benchimol EI, Leandro AC, das

Neves LB, Vicente RT, et al. The IFN-γ +874T/A gene polymor-

phism is associated with retinochoroiditis toxoplasmosis suscep-

tibility. Mem Inst Oswaldo Cruz 2009;104(3):451–5.

26. Dandona P, Aljada A, Bandyopadhyay A. Inflammation: the link

between insulin resistance, obesity and diabetes. Trends Immu-

nol 2004;25(1):4–7.

27. Kankova S, Flegr J, Calda P. An elevated blood glucose level and

increased incidence of gestational diabetes mellitus in pregnant

woman with latent toxoplasmosis. Folia Parasitol 2015;62. pii:

2015.056.

28. Dupey JP. The history of Toxoplasma gondii – the first 100 years.

J Eukaryot Microbiol 2008;55(6):467–75.

29. Carter CJ. Toxoplasmosis and polygenic disease susceptibility

genes: extensive Toxoplasma gondii host/pathogen interactome

enrichment in nine psychiatric or neurological disorders. J Pat-

hog 2013;2013:965046.

30. Henriques SA, Brett R, Alexander J, Pratt J, Roberts CW. Neu-

ropsychiatric disease and Toxoplasma gondii infection. Neuro-

immunomodulation 2009;16(2):122–33.

31. Frenkel JK. Adoptive immunity to intracellular infection. J

Immunol 1967;98(6):1309–19.

32. Melo MB, Jensen KD, Saeij J. Toxoplasma gondii effectors are

master regulators of the inflammatory response. Trends Parasitol

2011;27(11):487–95.

33. Mosser DM, Edwards JP. Exploring the full spectrum of mac-

rophage activation. Nat Rev Immunol 2008;8(12):958–69.

34. Giulietti A, van Etten E, Overbergh L, Stoffels K, Bouillon R,

Mathieu C. Monocytes from type 2 diabetic patients have a pro-

inflammatory profile. 1,25-Dihydroxyvitamin D3 works as anti-

inflammatory. Diabetes Res Clin Pract 2007;77(1):47–57.

35. Hatanaka E, Monteagudo PT, Marrocos MS, Campa A. Neutrophils

and monocytes as potentially important sources of proinflamma-

tory cytokines in diabetes. Clin Exp Immunol 2006;146(3):443–7.

36. Pickup JC. Inflammation and activated innate immunity in the

pathogenesis of type 2 diabetes. Diabetes Care 2004;27(3):813–23.

37. Pickup JC, Crook MA. Is type II diabetes mellitus a disease of the

innate immune system? Diabetologia 1998;41(10):1241–8.

38. Arsenijevic D, Girardier L, Seydoux J, Chang HR, Dulloo AG.

Altered energy balance and cytokine gene expression in a murine

model of chronic infection with Toxoplasma gondii. Am J Phys-

iol 1997;272(5 Pt 1):E908–17.

39. Fabiani S, Pinto B, Bonuccelli, Bruschi F. Neurobiological stud-

ies on the relationship between toxoplasmosis and neuropsychi-

atric diseases. J Neurol Sci 2015;351(1-2):3–8.

40. Tomasik J, Schultz TL, Kluge W, Yolken RH, Bahn S, Carruthers

VB. Shared Immune and Repair Markers During Experimental

Toxoplasma Chronic Brain Infection and Schizophrenia. Schizophr

Bull 2016; 42(4):386–95.

41. Filisetti D, Candolfi E. Immune response to Toxoplasma gondii.

Ann Inst Super Sanita 2004;40(1):71–80.

42. Prada J, Ngo-Tu T. Overexpression and protective role of

interleukin6, tumor necrosis factor-alpha and nitric oxides in

on Novem

ber 8, 2021 by guest. Protected by copyright.

http://fmch.bm

j.com/

Fam

Med C

om H

ealth: first published as 10.15212/FM

CH

.2016.0128 on 1 Decem

ber 2016. Dow

nloaded from

The role of Toxoplasma gondii as a possible inflammatory agent

Family Medicine and Community Health 2016;4(4):44–62 58

RE

VIE

W

experimental and clinical ocular toxoplasmosis. Int J Infect Dis

2008;12(1):e388.

43. Jagannathan M, McDonnell M, Liang Y, Hasturk H, Hetzel J,

Rubin D, et al. Toll-like receptors regulate B cell cytokine produc-

tion in patients with diabetes. Diabetologia 2010;53(7):1461–71.

44. Arsenijevic D, Girardier L, Seydoux J, Pechere JC, Garcia I,

Lucas R, et al. Metabolic-cytokine responses to a second immu-

nological challenge with LPS in mice with T. gondii infection.

Am J Physiol 1998;274(3 Pt 1):E439–45.

45. Gazzinelli RT, Wysocka M, Hieny S, Scharton-Kersten T,

Cheever A, Kuhn R, et al. In the absence of endogenous IL-10,

mice acutely infected with Toxoplasma gondii succumb to a

lethal immune response dependent upon CD4+ T cells and

accompanied by overproduction of IL-12, IFN-γ, and TNF-α. J

Immunol 1996;157(2):798–805.

46. Wegner M, Winiarska H, Bobkiewicz-Kozłowska T, Dworacka

M. IL-12 serum levels in patients with type 2 diabetes treated

with sulphonylureas. Cytokine 2008;42(3):312–16.

47. Tsiavou A, Degiannis D, Hatziagelaki E, Koniavitou K, Raptis

SA. Intracellular IFN-γ production and IL-12 serum levels in

latent autoimmune diabetes of adults (LADA) and in type 2 dia-

betes. J Interferon Cytokine Res 2004;24(7):381–7.

48. Winer S, Chan Y, Paltser G, Truong D, Tsui H, Bahrami J, et al.

Normalization of obesity associated insulin resistance through

immunotherapy. Nat Med 2009;15(8):921–9.

49. Bliss SK, Marshall AJ, Zhang Y, Denkers EY. Human poly-

morphonuclear leukocytes produce IL-12, TNF-alpha and

the chemokines macrophageinflammatory protein-1a and

-1b in response to Toxoplasma gondii antigens. J Immunol

1999;162(12):7369–75.

50. Gazzinelli RT, Amichay D, Scharton-Kersten T, Grunwald E,

Farber JM, Sher A. Role of macrophage-derived cytokines in

the induction and regulation of cell-mediated immunity to Toxo-

plasma gondii. Curr Top Microbiol Immunol 1996;219:127–39.

51. Nguyen TD, Bigaignon G, Markine-Goriaynoff D, Heremans

H, Nguyen TN, Warnier G, et al. Virulent Toxoplasma gondii

strain RH promotes T-cellin dependent overproduction of pro-

inflammatory cytokines IL12 and g-interferon. J Med Microbiol

2003;52(Pt 10):869–76.

52. Lieberman LA, Cardillo F, Owyang AM, Rennick DM, Cua

DJ, Kastelein RA, et al. IL-23 provides a limited mechanism

of resistance to acute toxoplasmosis in the absence of IL-121.

J Immunol 2004;173(3):1887–93.

53. Jagannathan M, McDonnell ME, Shin H, Rehman Q, Hasturk

H, Apovian CM, et al. Elevated proinflammatory cytokine pro-

duction by a skewed T cell compartment requires monocytes

and promotes inflammation in type 2 diabetes. J Immunol

2011;186(2):1162–72.

54. Winer S, Paltser G, Chan Y, Tsui H, Engleman E, Winer D, et al.

Obesity predisposes to Th17 bias. Eur J Immunol 2009;39(9):

2629–35.

55. Herder C, Zierer A, Koenig W, Roden M, Meisinger C, Thorand

B. Transforming growth factor-β1 and incident type 2 diabetes:

results from the MONICA/KORA case-cohort study, 1984–2002.

Diabetes Care 2009;32(10):1921–3.

56. Andriankaja OM, Barros SP, Moss K, Panagakos FS, DeVizio

W, Beck J, et al. Levels of serum interleukin (IL)-6 and gingival

crevicular fluid of IL-1β and prostaglandin E2 among non-smok-

ing subjects with gingivitis and type 2 diabetes. J Periodontol

2009;80(2):307–16.

57. Osborn O, Brownell SE, Sanchez-Alavez M, Salomon D, Gram

H, Bartfai T. Treatment with an interleukin 1 beta antibody

improves glycemic control in diet-induced obesity. Cytokine

2008;44(1):141–8.

58. Yang L, Anderson DE, Baecher-Allan C, Hastings WD,

Bettelli E, Oukka M, et al. IL-21 and TGF-β are

required for differentiation of human TH

17 cells. Nature

2008;454(7202):350–2.

59. Acosta-Rodriguez EV, Napolitani G, Lanzavecchia A, Sallusto

F. Interleukins 1β and 6 but not transforming growth factor-β

are essential for the differentiation of interleukin 17–producing

human T helper cells. Nat Immunol 2007;8(9):942–9.

60. Kelly MN, Kolls JK, Happel K, Schwartzman JD, Schwarzen-

berger P, Combe C, et al. Interleukin-17/interleukin-17 receptor-

mediated signaling is important for generation of an optimal

polymorphonuclear response against Toxoplasma gondii infec-

tion. Infect Immun 2005;73(1):617–21.

61. Fischer HG, Nitzgen B, Reichmann G, Hadding U. Cytokine

responses induced by Toxoplasma gondii in astrocytes and

microglial cells. Eur J Immunol 1997;27(6):1539–48.

62. Nagineni CN, Detrick B, Hooks JJ. Transforming growth

factor-beta expression in human retinal pigment epithelial

cells is enhanced by Toxoplasma gondii: a possible role in the

immunopathogenesis of retinochoroiditis. Clin Exp Immunol

2002;128(2):372–8.

63. Feuerer M, Herrero L, Cipolletta D, Naaz A, Wong J, Nayer A,

et al. Lean, but not obese, fat is enriched for a unique population

of regulatory T cells that affect metabolic parameters. Nat Med

2009;15(8):930–9.

64. Araujo FG, Slifer T. Different strains of Toxoplasma gondii

induce different cytokine responses in CBA/Ca mice. Infect

Immun 2003;71(7):4171–4.

on Novem

ber 8, 2021 by guest. Protected by copyright.

http://fmch.bm

j.com/

Fam

Med C

om H

ealth: first published as 10.15212/FM

CH

.2016.0128 on 1 Decem

ber 2016. Dow

nloaded from

Molan et al.

59 Family Medicine and Community Health 2016;4(4):44–62

RE

VIE

W

65. Arsenijevic D, Bilbao FD, Giannakopoulos P, Girardier L,

Samec S, Richard D. A role for interferon-gamma in the hyper-

metabolic response to murine toxoplasmosis. Eur Cytokine Netw

2001;12(3):518–27.

66. Gavrilescu LC, Denkers EY. IFN-γ overproduction and high level

apoptosis are associated with high but not low virulence Toxo-

plasma gondii infection. J Immunol 2001;167(2):902–9.

67. Hermes G, Ajioka JW, Kelly KA, Mui E, Roberts F, Kasza K,

et al. Neurological and behavioral abnormalities, ventricular

dilatation, altered cellular functions, inflammation, and neuronal

injury in brains of mice due to common, persistent, parasitic

infection. J Neuroinflammation 2008;5:48.

68. Khan IA, Schwartzman JD, Matsuura T, Kasper L. A dichotomous

role for nitric oxide during acute Toxoplasma gondii infection in

mice. Proc Natl Acad Sci USA 1997;94(25):13960–95.

69. Mordue DG, Monroy F, La Regina M, Dinarello CA, Sibley

LD. Acute toxoplasmosis leads to lethal overproduction of Th1

cytokines. J Immunol 2001;167(8):4574–84.

70. Hall AO, Beiting DP, Tato C, John B, Oldenhove G, Lombana

CG, et al. The cytokines interleukin 27 and interferon-γ promote

distinct Treg cell populations required to limit infection-induced

pathology. Immunology 2012; 37(3):511–23.

71. Baltaci AK, Mogulkoc R. Plasma leptin levels in rats with induced

Toxoplasma gondii infection. Bratisl Med J 2012;113(2):67–9.

72. Mittendorfer B, Horowitz JF, DePaoli AM, McCamish MA, Pat-

terson BW, Klein S. Recombinant human leptin treatment does

not improve insulin action in obese subjects with type 2 diabetes.

Diabetes 2011;60(5):1474–7.

73. Moon HS, Matarese G, Brennan AM, Chamberland JP, Liu X,

Fiorenza CG, et al. Efficacy of metreleptin in obese patients with

type 2 diabetes: cellular and molecular pathways underlying lep-

tin tolerance. Diabetes 2011;60(6):1647–56.

74. Milovanovi I, Vujani M, Klun I, Bobi B, Nikoli A, Ivovi

V, et al. Toxoplasma gondii infection induces lipid metabolism

alterations in the murine host. Memórias do Instituto Oswaldo

Cruz. 2009;104(2):175–8.

75. Auwerx J, Bouillon R, Collen D, Geboers J. Tissue-type

plasminogen activator antigen and plasminogen activator inhibi-

tor in diabetes mellitus. Arteriosclerosis 1988;8(1):68–72.

76. Vague P, Juhan-Vague I, Aillaud MF, Badier C, Viard R, Alessi

MC, et al. Correlation between blood fibrinolytic activity, plas-

minogen activator inhibitor level, plasma insulin level, and

relative body weight in normal and obese subjects. Metabolism

1986;35(3):250–3.

77. Jansson JH, Johansson B, Boman K, Nilsson TK. Hypo-

fibrinolysis in patients with hypertension and elevated choles-

terol. J Intern Med 1991;229(4):309–16.

78. Sampson M, Kong C, Patel A, Unwin R, Jacobs HS. Ambulatory

blood pressure profiles and plasminogen activator inhibitor (PAI-

1) activity in lean women with and without the polycystic ovary

syndrome. Clin Endocrinol (Oxf) 1996;45(5):623–9.

79. Juhan-Vague I, Vague P, Alessi MC, Badier C, Valadie J, Aillaud

MF, Alan C. Relationships between plasma insulin triglyceride,

body mass index, and plasminogen activator inhibitor 1. Diabetes

Metab 1987;13(3 Pt 2):331–6.

80. Pandolfi A, Cetrullo D, Polishuck R, Alberta MM, Calafiore

A, Pellegrini G, et al Plasminogen activator inhibitor type 1 is

increased in the arterial wall of type II diabetic subjects. Arterio-

scler Thromb Vasc Biol 2001;21(8):1378–82.

81. Grant MB, Ellis EA, Caballero S, Mames RN. Plasminogen

activator inhibitor-1 overexpression in nonproliferative diabetic

retinopathy. Exp Eye Res 1996;63(3):233–44.

82. Nordt TK, Sawa H, Fujii S, Sobel BE. Induction of plasminogen

activator inhibitor type-1 (PAI-1) by proinsulin and insulin in

vivo. Circulation 1995;91(3):764–70.

83. Schneider DJ, Sobel BE. PAI-1 and diabetes: a journey from the

bench to the bedside. Diabetes Care 2012;35(10):1961–7.

84. Sobel BE, Woodcock-Mitchell J, Schneider DJ, Holt RE,

Marutsuka K, Gold H. Increased plasminogen activator inhibi-

tor type 1 in coronary artery atherectomy specimens from type

2 diabetic compared with nondiabetic patients: a potential fac-

tor predisposing to thrombosis and its persistence. Circulation

1998;97(22):2213–21.

85. Alessi MC, Peiretti F, Morange P, Henry M, Nalbone G, Juhan-

Vague I. Production of plasminogen activator inhibitor 1 by

human adipose tissue: possible link between visceral fat accumu-

lation and vascular disease. Diabetes 1997;46(5):860–7.

86. Carmassi F, Morale M, Ferrini L, Dell’Omo G, Ferdeghini M,

Pedrinelli R, et al. Local insulin infusion stimulates expression

of plasminogen activator inhibitor-1 and tissue-type plasmi-

nogen activator in normal subjects. Am J Med 1999;107(4):

344–50.

87. Calles-Escandon J, Mirza SA, Sobel BE, Schneider DJ. Induc-

tion of hyperinsulinemia combined with hyperglycemia and

hypertriglyceridemia increases plasminogen activator inhibitor 1

in blood in normal human subjects. Diabetes 1998;47(2):290–3.

88. Meigs JB, Wilson PW, Fox CS, Vasan RS, Nathan DM,

Sullivan LM, et al. Body mass index, metabolic syndrome, and

on Novem

ber 8, 2021 by guest. Protected by copyright.

http://fmch.bm

j.com/

Fam

Med C

om H

ealth: first published as 10.15212/FM

CH

.2016.0128 on 1 Decem

ber 2016. Dow

nloaded from

The role of Toxoplasma gondii as a possible inflammatory agent

Family Medicine and Community Health 2016;4(4):44–62 60

RE

VIE

W

risk of type 2 diabetes or cardiovascular disease. J Clin Endo-

crinol Metab 2006;91(8):2906–12.

89. Mensah-Brown EP, Shahin A, Al-Shamisi M, Wei X, Lukic ML.

IL-23 leads to diabetes induction after subdiabetogenic treat-

ment with multiple low doses of streptozotocin. Eur J Immunol

2006;36(1):216–23.

90. Lukic ML, Shahin A, Mensah-Brown E, Al-Hakim A. Il-23

enhances diabetes induction in the wild type but not in INF-

gamma –/– mice. J Immunol 2007;178:129–36.

91. Muñoz M, Heimesaat MM, Danker K, Struck D, Lohmann

U, Plickert R, et al. Interleukin (IL)-23 mediates Toxoplasma

gondii–induced immunopathology in the gut via matrixmetal-

loproteinase-2 and IL-22 but independent of IL-17. J Exp Med

2009;206(13):3047.

92. Lang C, Grob U, Luder CGK. Subversion of innate and adap-

tive immune responses by Toxoplasma gondii. Parasitol Res

2007;100(2):191–203.

93. Dupont CD, Christian DA, Hunter CA. Immune response and

immunopathology during toxoplasmosis. Semin Immunopathol

2012;34(6):793–813.

94. Gazzinelli RT, Hieny A, Wynn T, Wolf S, Sher A. IL-12 is

required for the T-lymphocyte-independent induction of inter-

feron γ by an intracellular parasite and induces resistance in

T-cell-deficient hosts. Proc Natl Acad Sci USA 1993;90(13):

6115–9.

95. Lindberg RE, Frenkel JK. Toxoplasmosis in nude mice. J Parasi-

tol 1977;63(2):219–21.

96. Gazzinelli RT, Wysocka M, Hayashi S, Denkers EY, Hieny S,

Caspar P, et al. Parasite-induced IL-12 stimulates early IFN-

gamma synthesis and resistance during acute infection with Tox-

oplasma gondii. J Immunol 1994;153(6):2533–43.

97. Suzuki Y, Orellana MA, Schreiber RD, Remington JS. Inter-

feron-gamma: the major mediator of resistance against Toxo-

plasma gondii. Science 1988;240(4851):516–8.

98. Parker SJ, Roberts CW, Alexander J. CD81 T cells are the major

lymphocyte subpopulation involved in the protective immune

response to Toxoplasma gondii in mice. Clin Exp Immunol

1991;84(2):207–12.

99. Suzuki Y, Remington JS. Dual regulation of resistance against

Toxoplasma gondii infection by Lyt-21 and Lyt-11, L3T41 T

cells in mice. J Immunol 1988;140(11):3943–6.

100. Suzuki Y, Remington JS. The effect of anti-IFN-gamma antibody

on the protective effect of lyt-21 immune T cells against toxo-

plasmosis in mice. J Immunol 1990;144(5):1954–6.

101. Montoyoa JG. Laboratory diagnosis of Toxoplasma gondii infec-

tion and toxoplasmosis. J Infect Dis 2002;85(1):S73–82.

102. Walpole IR, Hodgen N, Bower C. Congenital toxoplasmosis: a

large survey in Western Australia. Med J Aust 1991;154(11):720–4.

103. Opsteegh M, Haveman R, Swart AN, Mensink-Beerepoot ME,

Hofhuis A, Langelaar MFM, et al. Seroprevalence and risk fac-

tors for Toxoplasma gondii infection in domestic cats in the

Netherlands. Prev Vet Med 2012;104(3–4):317–26.

104. Kolbekova P, Kourbatova E, Novotna M, Kodym P, Flegr J. New

and old risk-factors for Toxoplasma gondii infection: prospec-

tive cross-sectional study among military personnel in the Czech

Republic. Clin Microbiol Infect 2007;13(10):1012–7.

105. Villena I, Aubert D, Gomis P, Ferté H, Inglard JC, Denis-Bisiaux

H, et al. Evaluation of a strategy for Toxoplasma gondii oocyst

detection in water. Appl Environ Microbiol 2004;70(7):4035–9.

106. VanWormer E, Miller MA, Conrad PA, Grigg ME, Rejmanek

D, Carpenter TE, et al. Using molecular epidemiology to track

Toxoplasma gondii from terrestrial carnivores to marine hosts:

implications for public health and conservation. PLoS Negl Trop

Dis 2014;8(5):e2852.

107. Yap G, Pesin M, Sher A. Cutting edge: IL-12 is required for

the maintenance of IFN-gamma production in T cells mediat-

ing chronic resistance to the intracellular pathogen, Toxoplasma

gondii. J Immunol 2000;165(2):628–31.

108. Gaddi PJ, Yap S. Cytokine regulation of immunopathology in

toxoplasmosis. Immunol and Cell Biol 2007;85(2):155–9.

109. Neyer LE, Grunig G, Fort M, Remington JS, Rennick D, Hunter

CA. Role of interleukin-10 in regulation of T-cell-dependent and

T-cell-independent mechanisms of resistance to Toxoplasma

gondii. Infect Immun 1997;65(5):1675–82.

110. Denkers EY, Gazzinelli RT. Regulation and function of T-cell-

mediated immunity during Toxoplasma gondii infection. Clin

Microbiol Rev 1998;11(4):569–88.

111. Gigley JP, Fox BA, Bzik DJ. Cell-mediated immunity to Toxo-

plasma gondii develops primarily by local Th1 host immune

responses in the absence of parasite replication. J Immunol

2009;182(2):1069–78.

112. Chagas CEA, Borges MC, Martini LA, Rogero MM. Focus

on vitamin D, inflammation and type 2 diabetes. Nutrients

2012;4(1):52–67.

113. Pickup JC, Mattock MB, Chuaney GD, Burt D. NIDDM as a

disease of the innate immune system: association of acute-phase

reactants and interleukin-6 with metabolic syndrome X. Diabeto-

logia 1997;40(11):1286–92.

on Novem

ber 8, 2021 by guest. Protected by copyright.

http://fmch.bm

j.com/

Fam

Med C

om H

ealth: first published as 10.15212/FM

CH

.2016.0128 on 1 Decem

ber 2016. Dow

nloaded from

Molan et al.

61 Family Medicine and Community Health 2016;4(4):44–62

RE

VIE

W

114. Tilg H, Moschen AR. Inflammatory mechanisms in the regula-

tion of insulin resistance. Mol Med 2008;14(3–4):222–31.

115. Spranger J, Kroke A, Möhlig M, Hoffmann K, Bergmann MM,

Ristow M, et al. Inflammatory cytokines and the risk to develop

type 2 diabetes: results of the prospective population-based Euro-

pean Prospective Investigation into Cancer and Nutrition (EPIC)-

Potsdam Study. Diabetes 2003;52(3):812–7.

116. Grimble RF. Inflammatory status and insulin resistance. Curr

Opin Clin Nutr Metab Care 2002;5(5):551–9.

117. Donath MY, Shoelson SE. Type 2 diabetes as an inflammatory

disease. Nat Rev Immunol 2011;11(2):98–107.

118. Pradhan AD, Manson JE, Rifai N, Buring JE, Ridker PM. C-reac-

tive protein, interleukin 6, and risk of developing type 2 diabetes

mellitus. J Am Med Assoc 2001;286(3):327–34.

119. Ramadan JW, Steiner SR, O’Neill CM, Nunemaker CS. The cen-

tral role of calcium in the effects of cytokines on beta-cell func-

tion: implications for type 1 and type 2 diabetes. Cell Calcium

2011;50(6):481–90.

120. Flegr J. Influence of latent Toxoplasma infection on human per-

sonality, physiology and morphology: pros and cons of the Toxo-

plasma–human model in studying the manipulation hypothesis.

J Exp Biol 2013;216(Pt 1):127–33.

121. Oz HS, Tobin T. Atovaquone ameliorate gastrointestinal toxo-

plasmosis complications in a pregnancy model. Med Sci Monit

2012;18(9):BR337–45.

122. Coppari R, Bjørbæk C. The potential of leptin for treating

diabetes and its mechanism of action. Nat Rev Drug Discov

2012;11(9):692–70.

123. Gokce C, Yazar S, Bayram F, Gundogan K, Yaman O, Sahin I.

Anti-Toxoplasma gondii antibodies in type 2 diabetes. Natl Med

J India 2008;21(1):51.

124. Shirbazou S, Delpisheh A, Mokhetari R, Tavakoli G. Serologic

detection of anti Toxoplasma gondii infection in diabetic patients.

Iran Red Crescent Med J 2013;15(8):701–3.

125. Shih YL, Ho KT, Tsao CH, Chang YH, Shiau MY, Huang CN,

et al. Role of cyotkines in metabolism and type 2 diabetes mel-

litus. Int J Biomed Lab Sci 2013;2(1):1–6.

126. Andersson CX, Gustafson B, Hammarstedt A, Hedjazifar S,

Smith U. Inflamed adipose tissue, insulin resistance and vascular

injury. Diabetes Metab Res Rev 2008;24(8):595–603.

127. Gustafson B, Hammarstedt A, Andersson CX, Smith U.

Inflamed adipose tissue: a culprit underlying the metabolic

syndrome and atherosclerosis. Arterioscler Thromb Vasc Biol

2007;27(11):2276–83.

128. Maury E, Brichard SM. Adipokine dysregulation, adipose tis-

sue inflammation and metabolic syndrome. Mol Cell Endocrinol

2010;314(1):1–16.

129. King GL. The role of inflammatory cytokines in diabetes and its

complications. J Periodontol 2008;79(8 Suppl):1527–34.

130. Reeves GM, Mazaheri S, Snitker S, Langenberg P, Giegling I,

Hartmann A, et al. A positive association between T. gondii sero-

positivity and obesity. Front Public Health 2013;1:73.

131. Oz H. Toxoplasmosis, pancreatitis, obesity, and drug discovery.

Pancreat Disord Ther 2014;4(2):138.

132. Hotamisligil GS. Inflammation and metabolic disorders. Nature

2006;444(7121):860–7.

133. Hotamisligil GS, Shargill NS, Spiegelman BM. Adipose expres-

sion of tumor necrosis factor-alpha: direct role in obesity-linked

insulin resistance. Science 1993;259(5091):87–91.

134. Hotamisligil GS, Arner P, Caro JF, Atkinson RL, Spiegelman

BM. Increased adipose tissue expression of tumor necrosis fac-

tor-alpha in human obesity and insulin resistance. J Clin Invest

1992;95(5):2409–15.

135. Kern PA, Saghizadeh M, Ong JM, Bosch RJ, Deem R, Simsolo

RB. The expression of tumor necrosis factor in human adipose

tissue. Regulation by obesity, weight loss, and relationship to

lipoprotein lipase. J Clin Invest 1995;95(5):2111–9.

136. Hotamisligil GS, Spiegelman BM. Tumor necrosis fac-

tor a: a key component of the obesity-diabetes link. Diabetes

1994;43(11):1271–8.

137. Rocha VZ, Folco EJ, Sukhova G, Shimizu K, Gotsman I, Ver-

non AH. Interferon-gamma, a Th1 cytokine, regulates fat

inflammation: a role for adaptive immunity in obesity. Circ Res

2008;103(5):467–76.

138. Priceman SJ, Kujawski M, Shen S, Cherryholmes GA, Lee H,

Zhang C. Regulation of adipose tissue T cell subsets by Stat3 is

crucial for diet-induced obesity and insulin resistance. Proc Natl

Acad Sci USA 2013;110(32):13079–84.

139. Gregor MF, Hotamisligil GS. Inflammatory mechanisms in

obesity. Annu Rev Immunol 2011;29:415–45.

140. Siyadatpanah A, Tabatabaie F, Oormazdi H, Meamar AH, Razm-

jou E, Hadighi R, et al. Comparison of anti-toxoplasma IgG and

IgM antibodies determined by ELISA method in diabetic and

non-diabetic individuals in west Mazandaran province, Iran,

2011–2012. Ann Biol Res 2013;4(6):281–5.

141. Saki J, Shafieenia S, Foroutan-Rad M. Seroprevalence of toxo-

plasmosis in diabetic pregnant women in southwestern of Iran.

J Parasit Dis 2016;40(4):1586.

on Novem

ber 8, 2021 by guest. Protected by copyright.

http://fmch.bm

j.com/

Fam

Med C

om H

ealth: first published as 10.15212/FM

CH

.2016.0128 on 1 Decem

ber 2016. Dow

nloaded from

The role of Toxoplasma gondii as a possible inflammatory agent

Family Medicine and Community Health 2016;4(4):44–62 62

RE

VIE

W

142. Hassanain MA, El-Fadaly HA, Hassanain NA. Toxoplasma gon-

dii parasite load elevation in diabetic rats as latent opportunistic

character. Ann Trop Med Public Health 2014;7(2):110–5.

143. Modrek M J, Saravani R, Mousavi M, Khorashad AS, Piri M.

Investigation of IgG and IgM antibodies against Toxoplasma

gondii among diabetic patients. Int J Infect 2015;2(3):e27595.

144. Daryani A, Sarvi S, Aarabi M, Mizani A, Ahmadpour E, Shokri

A, et al. Seroprevalence of Toxoplasma gondii in the Iranian

general population: a systematic review and meta-analysis. Acta

Trop 2014;137:185–94.

145. Butler AE, Janson J, Bonner-Weir S, Ritzel R, Rizza RA, Butler

PC. Beta-cell deficit and increased beta-cell apoptosis in humans

with type 2 diabetes. Diabetes 2013;52(1):102–10.

146. Sakuraba H, Mizukami H, Yagihashi N, Wada R, Hanyu C,

Yagihashi S. Reduced beta-cell mass and expression of oxida-

tive stress-related DNA damage in the islet of Japanese type II

diabetic patients. Diabetologia 2002;45(1):85–96.

147. Yoon KH, Ko SH, Cho JH, Lee JM, Ahn YB, Song KH, et al.

Selective beta-cell loss and alpha-cell expansion in patients

with type 2 diabetes mellitus in Korea. J Clin Endocrinol Metab

2003;88(5):2300–8.

148. Gonzalez-Perez A, Schlienger RG, Rodríguez LA. Acute pancre-

atitis in association with type 2 diabetes and antidiabetic drugs.

Diabetes Care 2010;33(12):2580–5.

149. Waree P. Toxoplasmosis: pathogenesis and immune response.

Thammasat Med J 2008;8(4):487–96.

150. Zhu S, Lai D, Li S, Lun Z. Stimulative effects of insulin on

Toxoplasma gondii replication in 3T3-L1 cells. Cell Biol Int

2006;30(2):149–53.

151. Akinbo FO, Olujobi SO, Omoregie R, Egbe E. Intestinal parasitic

infections among diabetes mellitus patients. Biomark Genomic

Med 2013;5(1–2):44–7.

152. Mohtashamipour M, Hoseini SG, Pestehchian N, Yousefi H, Fal-

lah E, Hazratatian T. Intestinal parasitic infections in patients

with diabetes mellitus: a case-control study. J Anal Res Clin Med

2015;3(3):157–63.

153. Bessman AN, Sapico FL. Infections in the diabetic patient: the

role of immune dysfunction and pathogen virulence factors.

J Diabetes Complications 1992;6(4):258–62.

154. Nazligul Y, Sabuncu T, Ozbilge H. Is there a predisposition

to intestinal parasitosis in diabetic patients? Diabetes Care

2001;24(8):1503–4.

Related Information

Family Medicine and Community Health can provide you more papers focusing on diabetes mellitus. Please find them below.

• Correlation between KCNQ1 gene polymorphism and type 2 diabetes mellitus in Huaihai region of China

http://www.ingentaconnect.com/content/cscript/fmch/2014/00000002/00000001/art00003

• Four-year dynamic observation and study on standardized management of elderly patients with type 2 diabetes in Beijing

Yongding Road Community

http://www.ingentaconnect.com/content/cscript/fmch/2014/00000002/00000002/art00005

• Hypomagnesemia is a risk factor for metabolic syndrome and type 2 diabetes mellitus in native Balinese

http://www.ingentaconnect.com/content/cscript/fmch/2013/00000001/00000001/art00003

• Availability and social determinants of community health management service for patients with chronic diseases: An

empirical analysis on elderly hypertensive and diabetic patients in an eastern metropolis of China

http://www.ingentaconnect.com/content/cscript/fmch/2015/00000003/00000001/art00003

• Exploring point-of-care transformation in diabetic care: A quality improvement approach

http://www.ingentaconnect.com/content/cscript/fmch/2015/00000003/00000002/art00004

• Longitudinal study of a community hospital integrated model for diabetes management in the Beijing Jingsong community

http://www.ingentaconnect.com/content/cscript/fmch/2014/00000002/00000001/art00004

on Novem

ber 8, 2021 by guest. Protected by copyright.

http://fmch.bm

j.com/

Fam

Med C

om H

ealth: first published as 10.15212/FM

CH

.2016.0128 on 1 Decem

ber 2016. Dow

nloaded from