ArticlePDF AvailableLiterature Review

Rheumatic Fever: From Sore Throat to Autoimmune Heart Lesions

Authors:

Abstract and Figures

Molecular mimicry between streptococci and heart components has been proposed as the triggering factor leading to autoimmunity in rheumatic heart disease (RHD). In this review, we present data from cellular autoimmune responses, focusing on the interactions between HLA class II molecules, streptococcal peptides and heart tissue proteins and T-cell receptor (TCR) usage. HLA-DR7DR53 associated with DQ molecules seem to be related with the development of valvular lesions in severe RHD patients. DR7DR53 molecules were also involved in the recognition of an immunodominant M5 peptide in these patients. T cells infiltrating RHD hearts displayed several oligoclonal expansions. Intralesional T-cell clones presenting identical TCR-BVBJ AVAJ and -CDR3 sequences were able to recognize several antigens with little or low homology, showing an intramolecular degenerate pattern of antigen recognition. Peripheral blood mononuclear cells of rheumatic fever (RF) patients produced proinflammatory cytokines, and intralesional mononuclear cells from severe RHD patients produced predominantly Th1-type cytokines. These results illustrate the complex mechanisms leading to heart tissue damage in RF/RHD patients.
Content may be subject to copyright.
Review
Int Arch Allergy Immunol 2004;134:56–64
DOI: 10.1159/000077915
Rheumatic Fever: From Sore Throat to
Autoimmune Heart Lesions
Luiza Guilherme
a, b
Jorge Kalil
a–c
a
Heart Institute-InCor, University of Sa˜ o Paulo School of Medicine;
b
Institute for Investigation in Immunology,
Millenium Institute, and
c
Division of Clinical Immunology and Allergy, Department of Clinical Medicine,
University of Sa˜ o Paulo School of Medicine, Sa˜ o Paulo, Brazil
Published online: April 16, 2004
Correspondence to: Dr. Luiza Guilherme
Laborato´rio de Imunologia, Instituto do Coraça˜o-HC-FMUSP
Av. Dr. Eneas de Carvalho Aguiar, 44-9 andar
Sa˜o Paulo, SP 05403-000 (Brazil)
Tel. +55 11 3069 5901/3082 7730, Fax +55 11 3082 9350, E-Mail luizagui@usp.br
ABC
Fax + 41 61 306 12 34
E-Mail karger@karger.ch
www.karger.com
© 2004 S. Karger AG, Basel
1018–2438/04/1341–0056$21.00/0
Accessible online at:
www.karger.com/iaa
Key Words
Autoimmunity
W Cytokines W Heart tissue proteins W
M protein W Rheumatic heart disease W T cell receptor
Abstract
Molecular mimicry between streptococci and heart com-
ponents has been proposed as the triggering factor lead-
ing to autoimmunity in rheumatic heart disease (RHD). In
this review, we present data from cellular autoimmune
responses, focusing on the interactions between HLA
class II molecules, streptococcal peptides and heart tis-
sue proteins and T-cell receptor (TCR) usage. HLA-
DR7DR53 associated with DQ molecules seem to be
related with the development of valvular lesions in se-
vere RHD patients. DR7DR53 molecules were also in-
volved in the recognition of an immunodominant M5
peptide in these patients. T cells infiltrating RHD hearts
displayed several oligoclonal expansions. Intralesional
T-cell clones presenting identical TCR-BVBJ AVAJ and
-CDR3 sequences were able to recognize several anti-
gens with little or low homology, showing an intramole-
cular degenerate pattern of antigen recognition. Periph-
eral blood mononuclear cells of rheumatic fever (RF)
patients produced proinflammatory cytokines, and in-
tralesional mononuclear cells from severe RHD patients
produced predominantly Th1-type cytokines. These re-
sults illustrate the complex mechanisms leading to heart
tissue damage in RF/RHD patients.
Copyright © 2004 S. Karger AG, Basel
Introduction
Rheumatic fever (RF) is an inflammatory disease me-
diated by humoral and cellular autoimmune responses
that occurs as a delayed sequelae of Streptococcus pyo-
genes infection in 3–4% of susceptible and untreated chil-
dren and adolescents (aged 5–18 years). Carditis affects
30–45% of RF patients and is the most serious manifesta-
tion of the disease, leading to valvular lesions mainly in
the mitral and aortic valves. It causes chronic rheumatic
heart disease (RHD) that still remains a major public
health problem in developing countries.
The pathogenic mechanisms involved in the develop-
ment of RF/RHD are not fully understood. It is be-
lieved that the molecular mimicry mechanism is respon-
sible for the cross-reactions between streptococcal anti-
gens and human tissue proteins, mainly heart tissue pro-
teins in susceptible individuals. It is now clear that the
Rheumatic Fever
Int Arch Allergy Imunol 2004;134:56–64
57
disease is mediated by both humoral and cellular im-
mune responses and that the cellular branch of the im-
mune response is more involved in the development of
RHD.
In this review, we will focus on the cellular branch of
autoimmune responses leading to heart lesions in RHD
patients. The three main components involved in the T-
cell responses: HLA class II molecules, antigenic peptides
and T-cell receptors (TCR; trimolecular complex) will be
discussed.
Trimolecular Complex
Early studies in animal models by Benacerraf [1]
showed that immunization with synthetic antigens were
able to induce the production of antibodies and that this
immune response was determined by certain MHC haplo-
types. Later experiments mapped the control of immune
responses to MHC class II genes [2]. Although several
HLA class II molecules have been associated with the
development of humoral or cellular-mediated diseases
and are considered as genetic markers of these diseases,
now we know that, in fact, the major role of HLA class II
molecules is to present antigens to Th (CD4+) cells. Anti-
gen-presenting cells (APC), e.g. macrophages, dendritic
cells and B lymphocytes, constitutively express HLA class
II molecules. When activated by specific cytokines such as
IFNÁ, these APCs express large amounts of HLA class II
molecules.
During throat infection by S. pyogenes, several strepto-
coccal peptides are generated by APC. These peptides,
mainly from the M protein, are associated with HLA class
II molecules and presented to Th cells triggering an
inflammatory or humoral response. In untreated individ-
uals with genetic predisposition to develop RF/RHD, T-
cell populations selected by some immunodominant
streptococcal peptides combined with HLA class II mole-
cules will be able to trigger an autoimmune reaction (dis-
cussed below – Molecular Mimicry).
In the case of acute rheumatic carditis, Aschoff’s bod-
ies, the pathognomonic sign of the disease, develop in the
myocardium and/or endocardium [3]. The Aschoff body
is constituted by an agglomeration of several cells, e.g.
monocytes/macrophages and B lymphocytes that are
probably acting as APCs. Furthermore, mononuclear cells
that also include APC (macrophages and B lymphocytes)
and T lymphocytes infiltrate the heart in patients with
both acute and chronic RHD.
The third component of the trimolecular complex is
the T lymphocyte. The majority of T cells (90%) present
an antigen receptor (TCR) composed of ·- and ß-chains.
The TCR ·- and ß-chain are produced by the assembly of
variable (V), joining (J) and constant (C) gene segments
and also diverse (D) gene segments for ß-chain. Combina-
tions of these genes generate around 10
18
TCRs. Both
chains have three regions designated as complementarity-
determining region (CDR1, CDR2, CDR3). CDR1 and
CDR2 of the TCR interact with MHC molecules. The
peptide side chains of the MHC-peptide complex on the
surface of APC interact most closely with the hypervari-
able region of the TCR (CDR3) encoded by the V-D-J
region on the TCR-ß-chain. The combinations between
HLA class II molecules and antigens will drive the T-cell
repertoire.
Genetic Susceptibility
Several genetic markers of susceptibility to RF/RHD
have been studied [4], and associations with different
HLA class II antigens have been observed in several popu-
lations, e.g. DR4/DR9 in American Caucasians and DR2
in American blacks [5, 6], DR4 in Arabians [7], DR3 in
Indians [8], DR1 and DR6 in Africans [9] and DR5 in
Turks [10]. Interestingly, DR7 was found to be associated
with the disease in different countries [11–15]. Associa-
tions of HLA-DR7 with some HLA-DQ antigens seem to
be related to the presence of multivalvular lesions in RHD
patients [14–15]. HLA-DR53 is another class II molecule
always associated with DR4, DR7 and DR9 molecules. In
our studies, DR53 was also associated with the disease
[11, 12]. The fact that several HLA class II antigens are
associated with the development of RF/RHD in different
countries is consistent with the possibility that different
strains of group A streptococci could be implicated in the
development of RF/RHD in different countries. The vari-
able association may also be due to the important role that
HLA class II antigens play in antigen presentation to the
TCR.
In order to analyze the role of HLA-DR7 molecules in
T-cell immune responses against the N-terminal region of
the M5 protein, we studied the T-cell reactivity in the
peripheral blood of RHD patients. Amongst the M5 pep-
tides tested, it was possible to map the immunodominant
regions recognized by severe and mild RHD patients.
Mild RHD patients recognized preferentially the region
of amino acid residues 1–25 of M5 protein by several
HLA class II molecules. However, severe RHD patients
58
Int Arch Allergy Imunol 2004;134:56–64
Guilherme/Kalil
Table 1.
HLA-DR7 and RF/RHD
Country Population HLA Clinical picture
of the disease
Functional
study
References
Brazil Mulatto DR7, DR53
allogenotope
a
TaqI
DRß 13.81 kb
RF/RHD M5(81–96) peptide
preferentially recognized by
DR7
+
severe RHD patients;
capacity of binding with the
HLA-DR53 molecule
11, 12, 16
Brazil Caucasian DR7 RF/RHD 13
Egypt Egyptian DR7 DQ A1 02 01 RHD-MVL 14
Latvia Latvian DR7 DQ B1 03 02
DR7 DQ B1 04 01
RF/RHD-MVL
RF/MVR Sydeham’s
chorea
15
a
Defined by restriction fragment length polymorphism, 13.81-kb fragment corresponding to the HLA-DR53 anti-
gen [12].
MVL = Multivalvular lesions; MVR = mitral valve regurgitation.
recognized the region of amino acid residues 81–96,
defined as an immunodominant M5 epitope. Interest-
ingly, 70% of severe RHD patients that recognized this
M5 peptide were HLA-DR7+ DR53+, suggesting that
M5(81–96) peptide was preferentially presented to the T
cells in the context of DR7 and DR53 molecules. The
analysis of the capacity to bind streptococcal M5 peptides
showed that the M5(81–96) peptide was also able to bind
to the DR53 molecule [16]. These results are presented in
table 1.
Molecular Mimicry
Antigenic mimicry between streptococcal antigens,
mainly M-protein epitopes and heart components, has
been proposed as the triggering factor leading to autoim-
munity in RF. M protein is the major antigen of S. pyo-
genes and extends from the cell wall. It is composed of
approximately 450 amino acid residues showing antigenic
variations but high homology on the amino-terminal (N-
terminal) region, except for the 11 first amino acid resi-
dues that define the different serotypes, nowadays ap-
proximately 100 strains. The carboxyterminal (C-termi-
nal) region contains multiple repeat regions and is con-
served [17].
Molecular mimicry was first demonstrated in studies
on humoral immune responses. Anti-streptococcal anti-
bodies cross-reacted with several human tissues, includ-
ing the heart, skin, brain, glomerular basement mem-
brane, and striated and smooth muscles [18, 19].
One intersection point between humoral and cellular
immune responses in RHD patients could be the fact that
cross-reactive antibodies in the heart tissue may bind to
the valvular endothelium leading to inflammation, cellu-
lar infiltration and valve scarring [20]. Once activated, the
valvular endothelium expressed increased amounts of the
adhesion molecule VCAM-1, that facilitates the binding/
adhesion of T cells and consequently extravasation into
the valves, leading to the cycle of scarring, neovasculariza-
tion and infiltration of lymphocytes [21].
The presence of CD4+ T cells at lesion sites in the heart
of RHD patients has been demonstrated, suggesting a
direct role of these cells in the pathogenesis of RHD [22,
23].
We showed the significance of molecular mimicry
between ß-hemolytic streptococci and the heart tissue,
analyzing the T-cell repertoire leading to local tissue dam-
age in RHD. We demonstrated the capacity of infiltrating
T cell clones from heart lesions of severe RHD patients in
recognizing M protein peptides and heart tissue-derived
proteins. Our results pointed out three M5 immunodomi-
nant regions (residues 1–25, 81–103 and 163–177) that
cross-reacted to several heart protein fractions, mainly
those derived from valvular tissue with a molecular mass
of 95–150, 43–63 and 30–43 kD [24]. Peripheral T lym-
Rheumatic Fever
Int Arch Allergy Imunol 2004;134:56–64
59
Fig. 1.
Shared sequences of human and murine M5 epitopes recognized by T cells. Several T-cell immunodominant
M5 protein peptides were described and here we presented only peptides with shared sequences. The sequences of M5
peptides [M5(1–25), M5(81–96), M5(83–103) and M5(163–177)] from references 16 and 24 were based on the
sequence of the M5 protein published by Manjula et al. [29]. Sequences used on murine studies from reference 27
(peptides NT4, NT5, NT6, B1B2, B2, B2B3A and B3A) were from Miller et al. [30] and reference 28 [M5(1–35)
peptide] refers to a mutant M5 protein published by the authors. Human and murine amino acid residues of overlap-
ping peptides are underlined; murine amino acid residues of overlapping peptides are in bold type.
phocytes also recognized these immunodominant M5
peptides and valve proteins. The M5(81–96) mentioned
above is included in the M5(81–103) region peptide and
was preferentially recognized by HLA-DR7+DR53+ in
severe RHD patients (table 1) [16]. By using a proteomic
approach, we were able to characterize some mitral-valve
proteins identified by molecular weight and isoelectric
point (pI). Several valve-derived proteins were recognized
by peripheral blood and intralesional T-cell clones from
severe RHD patients. Amongst them, we identified vi-
mentin (molecular weight 53 kD/pI 5.12, recognized
mainly by peripheral T cells) and other cytoskeleton pro-
teins (recognized by both peripheral and intralesional T
cells) [manuscript in preparation]. In line with these
results, previous work showed the recognition of a 50- to
54-kD myocardial-derived protein by peripheral T lym-
phocytes from RHD patients [25].
We have also analyzed the intralesional T-cell re-
sponses against synthetic peptides from human cardiac
myosin ß-chain, and we found that 29% of these intrale-
sional T-cell clones derived from both myocardium and
valves were reactive. Taken together, our results indicate
that several autoantigens were recognized, and vimentin
could be the initial target of RF lesions resulting from
polyarthritis reactions, while myosin could also be an
immunodominant target during carditis episodes. The
phenomena of epitope spreading described by Sercaz et
al. [26] that lead to a broad diversity of recognition and
triggers an amplification and diversification of immune
responses could explain these results.
Myosin/M5 protein cross-reactive T-cell epitopes were
also investigated in mice immunized with intact cardiac
myosin [27]. Lymph node T cells were tested against over-
lapping M5 peptides named NT5/6/7 and B1B2/B2 and
B2B3A/B3A aligned with the M5 regions identified by us,
the M5(81–96) and M5(163–177), respectively. Robinson
et al. [28] obtained lymph node T-cell clones from mice
immunized with recombinant M5 protein that were able
to recognize M5 epitopes. Amongst the M5 epitopes rec-
ognized by the mouse T-cell clones, only the M5(1–35)
60
Int Arch Allergy Imunol 2004;134:56–64
Guilherme/Kalil
Table 2.
TCR analysis of cross-reactive intralesional T cell clones
T cell clone
identification
TCR BV BJ CDR3
(N-D-N) sequences
Antigens recognized mitral-valve-derived
protein, LMM or M5 peptides
Lu 3.1.3 SGRQGRYEQY-10aa 35 kD/pI 8.84
Lu 3.1.8 BV13 BJ2S7
AV2 AV3
SGRQGRYEQY-10aa 35 kD/pI 8.84, LMM 28 (1647–1664)
LMM 28B (1660–1677), LMM 32 (1699–1716)
Lu 3.1.29 SGRQGRYEQY-10aa 56–53 kD/pI 6.76
Lu 3. 2. 12.9 BV13 BJ2S7
AV2 AV7
SGRQGRYEQY-10aa 56–53 kD/pI 6.76
Lu 3.1.85 BV3 BJ2S1
AV5
SFTGRLDNEQF-11aa 79 kD/pI 5.12
M5 (1–20), M5 (11–25), M5 (81–96)
M5(111–130), M5(121–140)
M5(163–177), M5( 183–201)
LMM = Light meromyosin peptides; LMM28 = SLQSLLKDTQIQLDDAVR; LMM28B = DDAVRANDDLKE-
NIAIVE; LMM32 = RSRKL
AEQELIETSERVQ; M5 peptides: 1–20 = VTRGTISDPQRAKEALDKY; 11–25 =
QRAKEALDKYELENH; 81–96 = DKLKQQRDTLSTQKET; 83–103 = QQRDTLSTQKETLEREVQN; 111–
130 = TRQELANKQQESKENEKALN; 121–140 = ESKENEKALNELLEKTVKDK; 163–177 = ETIGTLKKI-
LDETVK; 183–201 = LDETVKDKLAKEQKSJQNI; NT = not tested. Shared sequences are underlined (adapted
from Faé et al. [43]).
region aligns with the M5(1–25) region recognized by the
human infiltrating T-cell clones. Figure 1 summarizes
both human and murine reactivity against M protein, the
N-terminal portion [29, 30].
T-Cell Repertoire
In the 90s, some researchers described a superantigenic
effect of streptococcal M5 protein preparations (pepsin
cleaved fragment – pepM5) for human T cells expressing
TCR-BV2, BV4, and BV8 [31–35]. Superantigens are
proteins that polyclonally activate T cells by an MHC
class II-dependent, but haplotype-unrestricted mecha-
nism. Proliferative responses to superantigens are limited
to T cells expressing a particular TCR-BV gene but inde-
pendent of antigen specificity. M protein has an impor-
tant role in the host anti-streptococcal immune response,
and for this reason it has been ascribed superantigenic
properties. However, the superantigenic effect was later
dismissed by some studies showing that the superantige-
nicity of pepM1 and pepM5 were due to contamination
with pyrogenic exotoxins that had themselves a potent
superantigen effect on BV2-bearing human T cells [36–
39].
In a recent work, we compared the TCR-BV usage in
peripheral blood and heart-infiltrating T cell lines (HIL)
from severe RHD patients, looking for oligoclonal ß-
chain expansions in line with antigen-driven immune
responses. T-cell receptor ß-chain family (TCR-BV) usage
and the degree of clonality were assessed by the analysis of
the length of the ß-chain CDR3. Our results showed
expansion of several BV families with oligoclonal profiles,
mainly in heart-infiltrating T-cell lines, and favor no
superantigenicity of M proteins in RHD patients. Few
oligoclonal BV expansions were shared by mitral valve-
and left atrium-derived T-cell lines in the same individu-
al. However, in this study, we described a case of 1 patient
that presented a BV5 expansion with the same BJ2S3 seg-
ment in both valve and myocardium tissues. However,
these T cells presented different amino acid CDR3 se-
quences, suggesting that different antigenic peptides
could be predominantly recognized by T cells that infil-
trate mitral valve and myocardium tissues [40]. The high
frequency and the persistence of T-cell oligoclonal expan-
sions in the damaged heart valves seem to be associated
with the progression of the disease [41], probably due to
the T-cell recognition of several heart tissue proteins
exposed by local lesions. In agreement with these data, it
has been described that it is possible to detect T-cell
Rheumatic Fever
Int Arch Allergy Imunol 2004;134:56–64
61
expansions in damaged heart valves even 20 years after
the acute RF episode [42].
Recently, we described intralesional T-cell clones with
a degenerate pattern of reactivity [43]. Five heart tissue-
derived T-cell clones (three from the mitral valve and one
from the myocardium) obtained from a patient with
severe RHD presented the same TCR-BV13 BJ2S7 with
identical CDR3 sequences. They expressed two ·-chains
at the RNA level and recognized M5 epitopes or human
cardiac ß-chain synthetic myosin peptides or mitral valve-
derived proteins. Interestingly, a mitral valve-derived
protein (53–56 kD/pI7.76) was recognized by two intrale-
sional T-cell clones, one from mitral valve tissue and the
other from myocardial tissue. These T-cell clones ex-
pressed only one different ·-chain. We also found other
T-cell clones that recognized several different antigens
bearing the same TCR-BV3 BJ2S1. These results are sum-
marized in table 2. Our data are in agreement with those
done by Mason [44, 45], in which the flexibility of T-cell
antigen recognition was evaluated by the analysis of the
immune response pattern against pathogens in experi-
mental models. Using a mathematical approach, he esti-
mated that T cells can react with a very large number of
peptides. The high frequency of cross-reactivity was pos-
tulated as essential for keeping the T-cell repertoire active
against the large number of foreign antigens that an indi-
vidual can encounter and respond to in his/her life [44,
45]. Thus, it appears that the major role of degeneracy of
T cells is to maintain the physiological immunity. How-
ever, our report is the first to identify intramolecularly
degenerate pattern of recognition.
It is known that among autoreactive T cells it is possi-
ble to differentiate T-cell subsets triggering pathological
compared to what could be called ‘physiological autoim-
munity’ [46–48]. However, the mechanism used by de-
generate T cells to recognize selected peptides without
pathological potential and, on the other hand, the mecha-
nism of degenerate T-cell reactivity leading to pathologi-
cal autoimmunity in individuals with genetic susceptibili-
ty remain unknown.
Cytokines
The cytokine pattern produced by Th cells in response
to defined antigens is crucial to drive the humoral or cellu-
lar immune responses. In addition, the concept of patho-
logical or physiological autoimmune reactions depends on
the cytokine produced in response to the autoantigens
that are being recognized by T cells. RF manifests differ-
ent clinical pictures such as arthritis, chorea, carditis, ery-
thema marginatum and/or subcutaneous nodules [49]. All
these manifestations involve particular autoantigens as
targets of pathological autoimmunity. Arthritis, chorea
and mild RHD are in part due to a pathological autoim-
mune reaction probably mediated by Th2-type cytokines,
leading to an exacerbated humoral response, as reported
in several studies [19]. On the other hand, severe RHD is
mediated mainly by T lymphocytes [16, 22–24]. The pro-
duction of TNF·, IL-1 and IL-2 in the peripheral blood of
acute RF and active RHD patients have been described
[50, 51]. Other authors have confirmed these findings and
have also noted increased plasma levels of TNF· in RF/
RHD patients [52–54].
In heart lesions during the acute phase of RHD, the
production of IL-1, TNF· and IL-2 was correlated with
progression of the Aschoff nodule [55] localized mainly in
the endocardium, subendocardium or perivascular re-
gions of the myocardial interstitium.
Recently, we have shown that intralesional mononu-
clear cells from heart lesions predominantly secret IFNÁ
and TNF· in both acute RF and chronic RHD patients,
with a scarce production of IL-4 [submitted]. When stim-
ulated with streptococcal M5 antigens, mitral valve-
derived intralesional T-cell lines produced IFNÁ but not
IL-4, while myocardial intralesional T-cell lines produced
IFNÁ, IL-10 and IL-4. The predominant Th1-type cyto-
kine produced mainly by CD4+ T cells infiltrating valve
tissue could mediate the severe RHD valve lesions, and
the fact that myocardial-infiltrating cells were able to pro-
duce regulatory cytokines could have a role in the mild-
ness of myocardial damage in RHD.
Conclusions
The development of RF/RHD involves a complex net-
work of autoimmune reactions comprising the following
major points. (1) Molecular mimicry between streptococ-
cal antigens and human tissues, mainly heart tissue, leads
to rheumatic heart lesions in RHD patients. (2) CD4+
T lymphocytes are the major effectors of heart lesions
and display a degenerate pattern of antigen recognition.
(3) Several streptococcal immunodominant peptides gen-
erate cross-recognition of vimentin, myosin and several
mitral valve-derived proteins, possibly resulting from an
epitope-spreading mechanism. (4) Several HLA class II
molecules are associated with the disease, and HLA-DR7/
DR53 combined with some HLA-DQ molecules seem to
be associated with the development of multiple valvular
62
Int Arch Allergy Imunol 2004;134:56–64
Guilherme/Kalil
Fig. 2.
Model of the development of RF/RHD. After group A strep-
tococcal throat infection, untreated susceptible individuals (5–18
years old) developed RF/RHD. Humoral and cellular immune
responses against S. pyogenes trigger an autoimmune attack to
human tissues by molecular mimicry. The autoimmune reaction is
initiated in the periphery where T cells recognize immunodominant
M5 peptides presented by APC (macrophages/monocytes) in the con-
text of HLA class II molecules. Proinflammatory cytokines were pro-
duced in the periphery. Activated T CD4+ cell clones expanded and
migrate to the heart (myocardium and valvular tissues as shown in
the picture), and several heart tissue proteins are recognized by
molecular mimicry. Epitope spreading amplifies the autoimmune
response. Several autoreactive T cell clones are generated and display
degenerate TCR capable of recognizing several different antigens.
Intralesional mononuclear cells also produced predominant Th1-
type cytokines (IFNÁ and TNF·). Mitral valve picture from 1 RHD
patient shows verruca lesions as indicated by the arrow. On the lower
left, a fragment of the mitral valve lesions (HE, !20) shows infiltrat-
ing mononuclear cells in the endocardium. In vitro growing of T cells
shows lymphoblasts as ‘flowers’. !100.
lesions in RHD patients. (5) T-cell recognition displays an
intramolecular degenerate reactivity against streptococcal
and human protein epitopes with low homology. (6) Th1-
type cytokines seem to be predominant in heart lesions,
especially valvular lesions.
All these points extend the knowledge on the develop-
ment of RHD (summarized in fig. 2) and may open new
possibilities of immunotherapy.
Rheumatic Fever
Int Arch Allergy Imunol 2004;134:56–64
63
References
1 Benacerraf B, Green I, Paul WE: Immune re-
sponse of guinea pigs to hapten-poly-L-lysine
conjugates as an example of the genetic control
of the recognition of antigenicity. Cold Spring
Harbor Symp Quantit Biol 1967:32:569–575.
2 Benacerraf B, McDevitt HO: Histocompatibil-
ity-linked immune response genes. Science
1972;175:273–279.
3 Virmani R, Farb A, Burke AP, Narula J: Pa-
thology of acute rheumatic carditis; in Narula
J, Virmani R, Reddy KS, Tandon R (eds):
Rheumatic Fever. Washington, American Reg-
istry Pathology, 1999, pp 217–234.
4 Gibofsky A, Khanna A, Suh E, Zabriskie JB:
The genetics of rheumatic fever: Relationship
to streptococcal infection and autoimmune dis-
ease. J Rheumatol Suppl 1991;30:1–5.
5 Ayoub EM, Barrett DJ, Maclaren NK, Krisch-
er JP: Association of class II human histocom-
patibility leukocyte antigens with rheumatic fe-
ver. J Clin Invest 1986;77:2019–2026.
6 Anastasiou-Nana MI, Anderson JL, Carlquist
JF, Nanas JN: HLA-DR typing and lympho-
cyte subset evaluation in rheumatic heart dis-
ease: A search for immune response factors.
Am Heart J 1986;112:992–997.
7 Rajapakse CN, Halim K, Al Orainey I, Al Noz-
ha M, Al Aska AK: A genetic marker for rheu-
matic heart disease. Br Heart J 1987;58:659–
662.
8 Jhinghan B, Mehra NK, Reddy KS, Taneja V,
Vaidya MC, Bhatia ML: HLA, blood groups
and secretor status in patients with established
rheumatic fever and rheumatic heart disease.
Tissue Antigens 1986;27:172–178.
9 Maharaj B, Hammond MG, Appadoo B, Leary
WP, Pudifin DJ: HLA-A, B, DR, and DQ anti-
gens in black patients with severe chronic rheu-
matic heart disease. Circulation 1987;76:259–
261.
10 Olmez U, Turgay M, Ozenirler S, Tutkak H,
Duzgun N, Duman M, et al: Association of
HLA class I and class II antigens with rheumat-
ic fever in a Turkish population. Scand J Rheu-
matol 1993;22:49–52.
11 Guilherme L, Weidebach W, Kiss MH, Snit-
cowsky R, Kalil J: Association of human leuko-
cyte class II antigens with rheumatic fever or
rheumatic heart disease in a Brazilian popula-
tion. Circulation 1991;83:1995–1998.
12 Weidebach W, Goldberg AC, Chiarella JM,
Guilherme L, Snitcowsky R, Pileggi F, et al:
HLA class II antigens in rheumatic fever. Anal-
ysis of the DR locus by restriction fragment-
length polymorphism and oligotyping. Hum
Immunol 1994;40:253–258.
13 Guedez Y, Kotby A, El Demellawy M, Galal A,
Thomson G, Zaher S, et al: HLA class II asso-
ciations with rheumatic heart disease are more
evident and consistent among clinically homo-
geneous patients. Circulation 1999;99:2784–
2790.
14 Visentainer JE, Pereira FC, Dalalio MM,
Tsuneto, LT, Donadio PR, Moliterno RA: As-
sociation of HLA-DR7 with rheumatic fever in
the Brazilian population. J Rheumatol 2000;
27:1518–1520.
15 Stanevecchia V, Eglite J, Sochevs A, Gardovs-
ka D, Zavadska D, Shantere R: HLA class II
associations with rheumatic heart disease
among clinically homogeneous patients in chil-
dren in Latvia. Arthritis Res Ther 2003;5:340–
346.
16 Guilherme L, Oshiro SE, Fae KC, Cunha-Neto
E, Renesto G, Goldberg AC, et al: T-cell reac-
tivity against streptococcal antigens in the pe-
riphery mirrors reactivity of heart-infiltrating
T lymphocytes in rheumatic heart disease pa-
tients. Infect Immun 2001;69:5345–5351.
17 Fishetti V: Streptococcal M protein. Sci Am
1991;264:32–39.
18 Stollerman GH: Rheumatogenic streptococci
and autoimmunity. Clin Immunol Immunopa-
thol 1991;61(2 pt 1):131–142.
19 Cunningham MW: Pathogenesis of group A
streptococcal infections. Clin Microbiol Rev
2000;13:470–511.
20 Galvin JE, Hemric ME, Ward K, Cunningham
MW: Cytotoxic mAb from rheumatic carditis
recognizes heart valves and laminin. J Clin
Invest 2000;106:217–224.
21 Roberts S, Kosanke S, Dunn TS, Jankelow D,
Duran CMG, Cunningham MW: Pathogenic
mechanisms in rheumatic carditis: Focus on
valvular endothelium. J Infect Dis 2001;183:
501–511.
22 Raizada V, Williams RC Jr, Chopra P, Gopi-
nath N, Prakash K, Sharma KB, et al: Tissue
distribution of lymphocytes in rheumatic heart
valves as defined by monoclonal anti-T cell
antibodies. Am J Med 1983;74:90–96.
23 Kemeny E, Grieve T, Marcus R, Sareli P,
Zabriskie JB: Identification of mononuclear
cells and T cell subsets in rheumatic valvulitis.
Clin Immunol Immunopathol 1989;52:225–
237.
24 Guilherme L, Cunha-Neto E, Coelho V, Snit-
cowsky R, Pomerantzeff PMA, Assis RV, Pe-
dra F, Neumann J, Goldberg A, Patarroyo ME,
Pillegi F, Kalil J: Human heart-infiltrating T-
cell clones from rheumatic heart disease pa-
tients recognized both streptococcal and car-
diac proteins. Circulation 1995;92:415–420.
25 El-Demellawy M, El-Ridi R, Guirguis NI, Alim
MA, Kotby A, Kotb M: Preferential recogni-
tion of human myocardial antigens by T lym-
phocytes from rheumatic heart disease pa-
tients. Infect Immun 1997;65:2197–2205.
26 Sercaz EE, Lehmann PV, Ametami A, Beni-
chou G, Miller A, Mougdil K: Dominance and
crypticity of T-cell antigenic determinants.
Annu Rev Immunol 1993;11:729–766.
27 Cunningham MW, Antone SM, Smart M, Liu
R, Kosanke S: Molecular analysis of human
cardiac myosin-cross-reactive B and T-cell epi-
topes of the group A streptococcal M5 protein.
Infect Immun 1997;65:3913–3923.
28 Robinson JH, Atherton MC, Goodacre JA
´
,
Pinkney M, Weightman H, Kehoe MA: Map-
ping T-cell epitopes in group A streptococcal
type 5 M protein. Infect Immun 1991;59:
4324–4331.
29 Manjula BN, Acharya AS, Mische MS, Fairwell
T, Fischetti VA: The complete amino acid
sequence of a biologically active 197-residue
fragment of M protein isolated from type 5
group A streptococci. J Biol Chem 1984;259:
3686–3693.
30 Miller LC, Gray ED, Beachey EH, Kehoe MA:
Antigenic variation among group A streptococ-
cal M proteins: Nucleotide sequence of the
serotype 5 M protein gene and its relationship
with genes encoding types 6 and 24 M proteins.
J Biol Chem 1988;263:5668–5673.
31 Kotb M, Majumdar G, Tomai M, Beachey EH:
Accessory cell-independent stimulation of hu-
man T cells by streptococcal M protein super-
antigen. J Immunol 1990;145:1332–1336.
32 Tomai M, Kotb M, Majumdar G, Beachey EH:
Superantigenicity of streptococcal M protein. J
Exp Med 1990;172:359–362.
33 Tomai MA, Schlievert PM, Kotb M: Distinct
T-cell receptor Vß gene usage by human T lym-
phocytes stimulated with the streptococcal py-
rogenic exotoxins and pep M5 protein. Infect
Immun 1992;60:701–705.
34 Watanabe-Ohnishi R, Aelion J, Legros L, To-
mai MA, Sokurenko EV, Newton D, Takahara
J, Irino S, Rashed S, Kotb M: Characterization
of unique human TCR Vß specificities for a
family of streptococcal superantigens repre-
sented by rheumatogenic serotypes of M pro-
tein. J Immunol 1994;152:2066–2073.
35 Tomai MA, Aelion J, Dockter ME, Majumdar
G, Spinella DG, Kotb M: T cell receptor V gene
usage by human T cells stimulated with the
superantigen streptococcal M protein. J Exp
Med 1991;174:285–288.
36 Wang B, Schlievert PM, Gaber AO, Kotb M:
Localization of an immunologically functional
region of the streptococcal superantigen pep-
sin-extracted fragment of type 5 M protein. J
Immunol 1993;151:1419.
37 Degnan B, Taylor J, Hawkes C, O’Shea U,
Smith J, Robinson JH, Kehoe MA, Boylston A,
Goodacre JA: Streptococcus pyogenes type 5M
protein is an antigen, not a superantigen, for
human T cells. Hum Immunol 1997;53:206–
215.
38 Fleischer B, Schmidt KH, Erlach D, Kohler W:
Separation of T cell stimulating activity from
streptococcal M protein. Infect Immun1992;1:
767–772.
39 Li PLL, Tiedemann RE, Moffat LS, Fraser JD:
The superantigen streptococcal pyrogenic exo-
toxin C (SPE-C) exhibits a novel mode of
action. J Exp Med 1997;186:375–391.
40 Guilherme L, Dulphy N, Douay C, Coelho V,
Cunha-Neto E, Oshiro SE, Assis RV, Tanaka
AC, Pomerantzeff PMA, Charron D, Toubert
A, Kalil J: Molecular evidence for antigen-driv-
en immune responses in cardiac lesions of
rheumatic heart disease patients. Int Immunol
2000;12:1063–1074.
41 Guilherme L, Cunha-Neto E, Tanaka AC, Dul-
phy N, Toubert A, Kalil J: Heart-directed auto-
immunity: The case of rheumatic fever. J Au-
toimmun 2001;16:363–367.
64
Int Arch Allergy Imunol 2004;134:56–64
Guilherme/Kalil
42 Figueroa F, Gonzalez M, Carrion F, Lobos C,
Turner F, Lasagna N, Valdes F: Restriction in
the usage of variable ß regions in T-cells infil-
trating valvular tissue from rheumatic heart
disease patients. J Autoimmun 2002;19:233–
240.
43 Faé K, Kalil J, Toubert A, Guilherme L: Heart
infiltrating T-cell clones from a rheumatic
heart disease patient display a common TCR
usage and a degenerate antigen recognition pat-
tern. Mol Immunol 2004;40:1129–1135.
44 Mason D: A very high level of crossreactivity is
an essential feature of the T-cell receptor. Im-
munol Today 1998;19:395–404.
45 Mason D: Antigen cross-reactivity: Essential in
the function of TCRs. Immunologist 1998;6:
220–222.
46 Cohen IR, Young DB: Autoimmunity, micro-
bial immunity and the immunological homun-
culus. Immunol Today 1991;12:105–110.
47 Cohen IR: The cognitive paradigm and the
immunological homunculus. Immunol Today
1992:13:490–494.
48 Cohen IR: Antigenic mimicry, clonal selection
and autoimmunity. J Autoimmun 2001;16:
337–340.
49 Stollerman GH: Rheumatic and heritable con-
nective tissue diseases of cardiovascular sys-
tem; in Braunwald E (ed): Heart Disease. Phila-
delphia, Saunders,1988, vol 11, pp 1706–
1734.
50 Miller LC, Gray ED, Mansour M, Abdin ZH,
Kamel R, Zaher S, Regelmann WE: Cytokines
and immunoglobulin in rheumatic heart dis-
ease: Production by blood and tonsillar mono-
nuclear cells. J Rheumatol 1989;16:1436–
1442.
51 Morris K, Mohan C, Wahi PL, Anand IS, Gan-
guly NK: Enhancement of IL-1, IL-2 produc-
tion and IL-2 receptor generation in patients
with acute rheumatic fever and active rheumat-
ic heart disease: A prospective study. Clin Exp
Immunol 1993;91:429–436.
52 Narin N, Kütükçüler N, Özyürek R, Bakiler
AR, Parlar A, Arcasoy M: Lymphocyte subsets
and plasma IL-1 ·, IL-2, and TNF-· concentra-
tions in acute rheumatic fever and chronic
rheumatic heart disease. Clin Immunol Immu-
nopathol 1995;77:172–176.
53 Samsonov MY, Tilz GP, Pisklakov VP, Reib-
negger G, Nassonov EL, Nassonova VA,
Wachter H, Fuchs D: Serum-soluble receptors
for tumor necrosis factor-· and interleukin-2
and neopterin in acute rheumatic fever. Clin
Immunol Immunopathol 1995;74:31–34.
54 Yegin O, Coskun M, Ertug H: Cytokines in
acute rheumatic fever. Eur J Pediatr 1997;156:
25–29.
55 Fraser WJ, Haffejee Z, Jankelow D, Wadee A,
Cooper K: Rheumatic Aschoff nodules revi-
sited. II. Cytokine expression corroborates re-
cently proposed sequential stages. Histopathol-
ogy 1997;31:460–464.
... Rheumatic fever (RF) is an inflammatory disease mediated by humoral and cellular autoimmune responses that represent delayed sequelae to Streptococcus pyogenes infection in 3% to 4% of susceptible and untreated children or adolescents (aged 5-18 years). Group A streptococcal (GAS) infections of the pharynx are the main cause of RF. 1 The latter is characterized by the involvement of the heart, joints, central nervous system, subcutaneous tissue, and skin. 2 Development of RF is likely related to a molecular mimicry mechanism, resulting from similarities between streptococcal antigens and human tissue proteins, particularly in the cardiac tissue of susceptible individuals. 1 The most severe complication of RF is carditis, leading to rheumatic heart disease (RHD) in 30% to 45% of affected patients that involves mainly the mitral and aortic valves. 1 Appropriate antibiotic treatment of streptococcal pharyngitis prevents the development of acute RF in most instances. ...
... Group A streptococcal (GAS) infections of the pharynx are the main cause of RF. 1 The latter is characterized by the involvement of the heart, joints, central nervous system, subcutaneous tissue, and skin. 2 Development of RF is likely related to a molecular mimicry mechanism, resulting from similarities between streptococcal antigens and human tissue proteins, particularly in the cardiac tissue of susceptible individuals. 1 The most severe complication of RF is carditis, leading to rheumatic heart disease (RHD) in 30% to 45% of affected patients that involves mainly the mitral and aortic valves. 1 Appropriate antibiotic treatment of streptococcal pharyngitis prevents the development of acute RF in most instances. 3 Therefore, the primary prevention of RF consists of adequate antibiotic therapy for GAS-associated upper respiratory infections in order to reduce the incidence of an initial attack of acute RF. 2 Patients with previous episodes of RF are at higher risk of recurrence, and therefore, require continuous secondary prophylaxis. ...
... 1 The most severe complication of RF is carditis, leading to rheumatic heart disease (RHD) in 30% to 45% of affected patients that involves mainly the mitral and aortic valves. 1 Appropriate antibiotic treatment of streptococcal pharyngitis prevents the development of acute RF in most instances. 3 Therefore, the primary prevention of RF consists of adequate antibiotic therapy for GAS-associated upper respiratory infections in order to reduce the incidence of an initial attack of acute RF. 2 Patients with previous episodes of RF are at higher risk of recurrence, and therefore, require continuous secondary prophylaxis. ...
Article
Full-text available
Background Secondary prevention of recurrent rheumatic fever in individuals with rheumatic heart disease (RHD) requires continuous antibiotic prophylaxis. However, the impact of antibiotic prophylaxis on the outcome of patients with severe RHD who underwent heart valve replacement is unknown. The objective of the study was to assess the relationship between the use of antibiotics as secondary prophylaxis in RHD patients who underwent valve replacement and clinical outcomes including mortality, reoperation, and valve-related hospitalization. Methods We retrospectively compared outcomes of adult patients who underwent heart valve replacement for RHD at our institution from 1990 through 2014 and who received secondary antibiotic prophylaxis (prophylaxis group) with those who did not receive prophylaxis (nonprophylaxis group) using propensity score matching analysis. Results A total of 1094 patients (56% females, median age 40 years, range 31-53 years) were included with a median follow-up of 9.6 years (range 2.9-12.6 years). Antibiotic prophylaxis was prescribed in 201 patients (18%). Propensity score matching analysis demonstrated no significant difference in overall survival (95% [92%-98%] vs 97% [95%-99%], respectively; P = .7), valve-related hospitalization-independent survival (72% [range 65%-78%] vs 81% [range 76%-88%]; P = .25), and redo valve surgery-independent survival [76% [range 70%-83%] vs 75% [range 72%-79%]; P = .41) at 10-year follow-up in the nonantibiotic prophylaxis versus the antibiotic prophylaxis group. Conclusion Secondary antibiotic prophylaxis among adult RHD patients following valve replacement is not associated with improved clinical outcomes.
... Cytomegalovirus is spread through contact with infected body fluids, such as urine or saliva. Infection among pregnant women most frequently occur through close contact immuncompetent individuals, but can serious disease among HIV-infected persons, organ transplant recipients on immunosuprresive therapy, and fetuses (4). Cytomegalovirus is the most common congenital infection and the leading cause of sensorineural hearing loss, mental retardation, retinal disease, and cerebral palsy (5).Cytomegalovirus is the largest of the herpesviruses, and has a diameter of 200 nm with douple-stranded DNA viral genome of 240 kb in a 64 nm core enclosed by an icoshedral capsid composed of 162 capsomers. ...
... The incidence of infection in pregnancy is estimated to be as high as 1 to 200 pregnancies, of which around 40% will result in fetal infections (3). Congenital Cytomegalovirus occurs in 0.05%-1.5% of births (4) . Higher rate of infection among populations with lower economic standard of living. ...
Article
Full-text available
In this study indirect ELISA test was conducted on 118 serum samples for Cytomegalovirus collected from women in different ages and cases in Bent AL-Huda hospital. Indirect ELISA was applied on all these samples depending on the optical density (OD) value of sera previously prepared from blood of well know healthy and non-vaccinated women. According to ages groups the higher ratio are (56.5%) was observed in first age group. A highly significant differences were observed in the seropositivity concerning to age groups (P< 0.01). According to Cases groups the higher ratio of seropositivity were found in women (75.61%) was observed in Recurrent abortion and then in Bleeding (75 %). The Quantitative ELISA test was used to detect the Luteinizing hormone as antigens in same serum samples. the higher rate of Luteinizing hormone positivity was observed in the last age groups (83.33%). there was a highly significant difference between the ages groups. Depending on Cases the higher rate of Luteinizing hormone positivity was (87.50%) in bleeding case , there was highly significant difference in the Luteinizing hormone in Cases groups of Quantitative ELISA.
... Romatizmal ateş (RA), A grubu β-hemolitik streptokokların neden olduğu boğaz enfeksiyonundan sonra antijenler ile konak doku proteinleri arasındaki moleküler taklitten kaynaklanır (1)(2)(3). RA'nın en ciddi belirtilerinden olan kardit, hastaların %30-45'inde ilk atak sırasında gelişebilse de, RKKH çoğunlukla tekrarlayan semptomatik akut RA epizodlarına bağlı kümülatif kapak hasarından kaynaklanmaktadır (4,5). Özellikle gelişmekte olan ülkelerde oldukça ciddi bir sağlık problemi haline gelen RKKH, çocuklarda ve genç yetişkinlerde kalp yetmezliğinin önde gelen nedeni olup, sakatlık ve/ veya erken ölümle sonuçlanabilir. ...
... [2] RA'nın en ciddi belirtilerinden olan kardit, hastaların %30-45'inde ilk atak sırasında gelişebilse de, RKH çoğunlukla tekrarlayan semptomatik akut RA epizodlarına bağlı kümülatif kapak hasarından kaynaklanmaktadır. [5,6] RA'nın meydana getirdiği bu inflamatuvar hasar kalp kapaklarının fonksiyonlarının bozulmasına (kapak yetersizliği ya da darlığı) yol açar. Fonksiyonları bozulmuş kapaklar ise, hemodinamik değişikliklere neden olur. ...
... In such a condition it is known that after a viral infection a chronic condition can develop [2], characterized by sometimes persistent debilitating fatigue, with muscular weakness, mild fever, tender lymph nodes, headache and depression. Another example is acute viral hepatitis, which may continue as a hepatic derangement and finally cirrhosis [3], as well as the acute rheumatic fever which ends in chronic heart condition [4][5][6][7][8][9][10][11][12]. It is also known that elderly persons have a lower ability to raise a high fever after an exposure to an infectious agent [1,13]. ...
Article
Full-text available
This essay's theme was inspired by a question asked by a child: 'Why do I get ill?' The question is very interesting, but has no easy answer. This paper discusses a few possible answers to this difficult question. Through the life of a person, from birth to death, there is a "continuum" in the pathological conditions a person may experience. The body, as a whole, suffers deeply any time there is an acute or a chronic condition that is either maltreated or neglected. Chronic and acute diseases in the medical history of a person constitute a rigidly related chain of immune responses in the form of a real "continuum" that at every point in time indicates the end result of this continuum. The idea promoted here is that suppression of diseases, through excess of chemical drugs or other means, many times overwhelms the body's natural defenses and forces the immune system to compromise and start a deeper line of defense, which then constitute the beginning of a new chronic condition. Thus, the original inflammation of an acute condition may continue as a sub-acute inflammatory process on a deeper level. Acute inflammatory conditions must therefore be treated very carefully from their beginnings in childhood in order not to force the immune system to compromise. It is also suggested here that all chronic degenerative conditions have a sub-acute inflammatory character, and that "inflammation" constitutes the main common parameter of all diseases.
... Exaggerated antibody response to group A carbohydrate has been observed in ARF patients, which remained elevated in individuals with residual mitral valve disease supporting the suggestion that these antibodies cause valvular damage [52,53]. It is not clear whether the initial valvular injury results from antibody or cell mediated autoimmune damage but T-cell and macrophage infiltration are involved in subsequent damages [54,55]. ...
Article
Rheumatic heart failure (RHF) is a potentially preventable cardiac complication of rheumatic fever (RF) characterized by scarring or stretching in one or more cardiac valves leading to low cardiac output. Over the past five decades, the epidemiology of RHF has shifted markedly, reducing in high-income countries while increasing in low/middle-income countries. However, the shift has lacked a matching proportional expansion in research and public health practices. Prophylactic and curative therapies combined with targeted public health control programs have been associated with the reducing prevalence in high-income countries but such strategies either have not been adequately implemented or have not translated into reduced burden in low-income countries. Further, the lack of recent research has undermined access to evidence-based data to support the development of treatment guidelines appropriate for low/middle-income countries. In the present paper, we review published data to provide a comprehensive update on epidemiology, pathophysiology, diagnosis, and management strategies for RHF.
Article
Full-text available
Recent research suggests that T-cell receptor (TCR) sequences expanded during human immunodeficiency virus and SARS-CoV-2 infections unexpectedly mimic these viruses. The hypothesis tested here is that TCR sequences expanded in patients with type 1 diabetes mellitus (T1DM) and autoimmune myocarditis (AM) mimic the infectious triggers of these diseases. Indeed, TCR sequences mimicking coxsackieviruses, which are implicated as triggers of both diseases, are statistically significantly increased in both T1DM and AM patients. However, TCRs mimicking Clostridia antigens are significantly expanded in T1DM, whereas TCRs mimicking Streptococcal antigens are expanded in AM. Notably, Clostridia antigens mimic T1DM autoantigens, such as insulin and glutamic acid decarboxylase, whereas Streptococcal antigens mimic cardiac autoantigens, such as myosin and laminins. Thus, T1DM may be triggered by combined infections of coxsackieviruses with Clostridia bacteria, while AM may be triggered by coxsackieviruses with Streptococci. These TCR results are consistent with both epidemiological and clinical data and recent experimental studies of cross-reactivities of coxsackievirus, Clostridial, and Streptococcal antibodies with T1DM and AM antigens. These data provide the basis for developing novel animal models of AM and T1DM and may provide a generalizable method for revealing the etiologies of other autoimmune diseases. Theories to explain these results are explored.
Chapter
There are over 80 defined autoimmune diseases, characterized by the activation of the immune system and tissue damage in the absence of an external threat to the organism, which affect 5 percent-7 percent of the population. Diseases considered to be autoimmune in origin can be characterized by activation of the adaptive immune response with B and T lymphocytes responding to self-antigens in the absence of any detectable tumor invasion or microbial assault; or as those that display the activation of the innate immune system and an excess of inflammatory mediators, but no evidence of an antigen-specific immune response, the former constituting the vast majority of such disease entities. Here we will discuss the immune cells and immune responses involved in autoimmune reaction, initiation and facilitation of autoimmunity and then will elaborate on human multisystem autoimmune diseases and some more common system-specific autoimmune diseases.
Chapter
Autoimmune diseases are common diseases in which loss of tolerance within the immune system results in pathologic immune responses that target either cellular or organ-specific self-antigens. There is a genetic tendency toward autoreactivity in affected individuals, and both innate and adaptive immune activation may contribute to disease. An important recent advance is the identification of genetic polymorphisms that contribute to risk in most autoimmune diseases and are associated with a variety of immune activation pathways; these associations have been informative about disease-specific pathogenesis and have led to the development of successful therapies for some diseases. A deeper understanding of the components of the innate and adaptive immune system has led to highly effective therapeutic targeting of cytokines, cell surface molecules, and intracellular signaling molecules with marked improvements in outcome in several autoimmune diseases. Fertile new avenues for research include mechanisms of regulation by regulatory immune cells, metabolic regulation of immune cell function, and immune activation by commensal bacteria that colonize humans. The goal of regulating autoimmunity without causing excessive immunosuppression remains elusive but is the holy grail of current research efforts.
Article
Full-text available
Objective: to identify the factors associated with mortality in mitral valve reoperation, to create a predictive model of mortality and to evaluate the EuroSCORE. Methods: a total of 65 patients were evaluated from January 2008 to December 2017. It was verified the association of variables with death and a multiple logistic regression model was used to stratify patients. Results: hospital mortality was 13.8% and in the Death Group: EuroSCORE was 12.33±8.87 (p=0.017), the left ventricular ejection fraction (LVEF) was 45.33±5.10 (p=0.000), the creatinine was 1.56±0.29 (p=0.002), the prothrombin time (TAP) was 1.64 (p=0.001), pulmonary artery systolic pressure (PSAP): 66.1±13.6 (p=0.002), female: 88% (p=0.000), malnutrition: 77.7% (p=0.007), associated tricuspid disease: 44,4% (p=0.048), presence of ventricular arrhythmia: 77.7% (p=0.005), implantation of a biological prosthesis: 55.5% (p=0.034), bronchopneumonia and sepsis: 33,3% (p=0.048), systemic inflammatory response syndrome (SIRS): 55.5% (p=0.001), low cardiac output syndrome (LCOS): 88.8% (p=0.000). Conclusion: the factors associated with mortality were: EuroSCORE, LVEF, creatinine, TAP, PSAP, female, malnutrition, tricuspid disease, ventricular arrhythmia, implantation of biological prosthesis, SIRS, SBDC, bronchopneumonia and sepsis. The explanatory variables of death of the model were: EuroSCORE, creatinine, TAP, LVEF, length of stay in the intensive care unit (ICU), interval between surgeries and presence of ventricular arrhythmia. The high EuroSCORE is related to higher mortality.
Article
Full-text available
M proteins that define the serotypes of group A streptococci are powerful blastogens for human T lymphocytes. The mechanism by which they activate T cells was investigated and compared with the conventional T cell mitogen phytohemagglutinin, and the known superantigen staphylococcal enterotoxin B. Although major histocompatibility complex (MHC) class II molecules are required for presentation, there is no MHC restriction, since allogeneic class II molecules presented the bacterial protein to human T cells. Type 5 M protein appears to bind class II molecules on the antigen-presenting cells and stimulate T cells bearing V beta 8 sequences. Our results indicate that this streptococcal M protein is a superantigen and suggest a possible mechanism of its role in the pathogenesis of the postinfectious autoimmune sequelae.
Article
Group A streptococci are model extracellular gram-positive pathogens responsible for pharyngitis, impetigo, rheumatic fever, and acute glomerulonephritis. A resurgence of invasive streptococcal diseases and rheumatic fever has appeared in outbreaks over the past 10 years, with a predominant M1 serotype as well as others identified with the outbreaks. emm (M protein) gene sequencing has changed serotyping, and new virulence genes and new virulence regulatory networks have been defined. The emm gene superfamily has expanded to include antiphagocytic molecules and immunoglobulin-binding proteins with common structural features. At least nine superantigens have been characterized, all of which may contribute to toxic streptococcal syndrome. An emerging theme is the dichotomy between skin and throat strains in their epidemiology and genetic makeup. Eleven adhesins have been reported, and surface plasmin-binding proteins have been defined. The strong resistance of the group A streptococcus to phagocytosis is related to factor H and fibrinogen binding by M protein and to disarming complement component C5a by the C5a peptidase. Molecular mimicry appears to play a role in autoimmune mechanisms involved in rheumatic fever, while nephritis strain-associated proteins may lead to immune-mediated acute glomerulonephritis. Vaccine strategies have focused on recombinant M protein and C5a peptidase vaccines, and mucosal vaccine delivery systems are under investigation.
Article
Excerpt Numerous observations have demonstrated the importance of genetic factors in the recognition of immunogenicity and in the ability to identify certain antigenic determinants, even though the antibodies produced are a heterogeneous population of molecules. (1.) There is a statistically significant relation between the ability of parents and offspring to respond to some antigens (Carlinfanti, 1948; Fjord-Scheibel, 1943; Sang and Sobey, 1954) and pure strains of mice differ in the amounts of antibody which they produce to different antigens (Ibsen, 1959). (2.) More recently, Sobey, Magrath, and Reisner (1966) have observed that an occasional mouse from randomly bred stock is naturally unresponsive to bovine serum albumin (BSA) and that such mice, when bred, produce both responsive and unresponsive mice. The fraction of offspring which are unresponsive increases with successive generations, and after the 6th or 7th generation, approximately 90% of the offspring of such matings are unresponsive to BSA. These experiments...
Article
Although it is widely held that T cells are highly specific for a particular antigen, in this article Don Mason argues that it is essential that T cells recognize more than one antigen in order to ensure recognition of all possible antigens, and estimates the number of antigens to which each T cell responds.
Article
The distribution of CD3+, CD4+, CD8+, CD19+, CD16+, and CD25+ lymphocyte populations in peripheral blood as well as the plasma concentrations of interleukin-1 α (IL-1 α), and IL-2 and tumor necrosis factor α (TNF-α) were investigated in 25 children with acute rheumatic fever (ARF) at the time of admission and after 3 months and in 15 children with chronic rheumatic heart disease (CRHD) and in 15 children with streptoccocal pharyngitis (SP) in order to determine changes in lymphocyte subsets and cytokine concentrations occurring during different stages of the disease. The percentages and absolute counts of CD4+, CD16+, CD25+ cells, the ratio of CD4/CD8 and plasma concentrations of IL-1 α and IL-2 in patients with ARF were significantly higher at admission than 3 months later. These levels were also significantly higher than in patients with CRHD, SP, or normal controls. Production of IL-2 in ARF and CRHD patients directly correlated with the percentages of CD4+ and CD25+ cells. According to our results, the evidences of increased cellular immune response in ARF are increased percentages CD4+ and CD25+ cells, CD4/CD8 ratio, and increased plasma concentrations of IL-1 α and IL-2. Furthermore, activation of cellular immune response was not present throughout all stages of rheumatic heart disease and also in SP.
Article
Logic programming with negation has been given a declarative semantics by Clark's completed database (CDB), and one can consider the consequences of the CDB in either two-valued or three-valued logic. Logic programming also has a proof theory given by SLDNF derivations. Assuming the data-dependency condition of strictness, we prove that the two-valued and three-valued semantics are equivalent. Assuming allowedness (a condition on occurrences of variables), we prove that SLDNF is complete for the three-valued semantics. Putting these two results together, we have completeness of SLDNF deductions for strict and allowed databases and queries under the standard two-valued semantics. This improves a theorem of Cavedon and Lloyd, who obtained the same result under the additional assumption of stratifiability.
Article
A new class of genes that controls the formation of specific immune responses has been identified.