Está en la página 1de 5

The Role of Nuclear Macromolecules in Innate Immunity

David S. Pisetsky1
1
Durham VA Medical Center and Duke University Medical Center, Durham, North Carolina

Nuclear macromolecules, in addition to their intracellular role in THE IMMUNE PROPERTIES OF DNA IN SLE
regulating cell function, can translocate into the extracellular space
where they can activate innate immunity. This translocation can The recognition of DNA’s immune properties dates back half
occur in various settings and reflects the dynamic nature of nuclear a century to its discovery as a target antigen in systemic lupus
structure. Of nuclear molecules, DNA and the DNA-binding protein, erythematosus (SLE). This prototypic autoimmune disease is
HMGB1, display distinct patterns of immune activity. For DNA, im- characterized by the production of antinuclear antibodies, with
mune activity depends on sequence, base methylation, and context. antibodies to DNA (anti-DNA) serving as markers of diagnostic
While bacterial DNA is an immune activator, mammalian DNA is and prognostic significance. As shown in studies on patients with
either inert or inhibitory when free. In contrast, mammalian DNA SLE as well as murine models, these antibodies can also mediate
in the form of immune complexes can trigger immune cell activa- tissue injury, with DNA–anti-DNA immune complexes impor-
tion. As shown in in vivo and in vitro studies, DNA can exit cells tant inducers of glomerulonephritis (1–3).
during apoptotic as well as necrotic cell death in a process that As an antigen in SLE, DNA most likely functions in the form
may depend on the presence of macrophages. Like DNA, HMGB1 of the nucleosome, a highly organized structure comprised of
can exit cells and acquire immune properties. For HMGB1, the trans- DNA wrapped around a histone core. Consistent with the role
location occurs in macrophages that have been stimulated by Toll- of nucleosomes as the driving antigen for autoantibody induc-
like receptor (TLR) ligands as well as cytokines; HMGB1 release can tion, sera from patients with lupus contain antibodies to nucleo-
also occur with apoptotic as well as necrotic death. While HMGB1 somes, the DNA and histone components, and other structures
alone can display cytokine activity, it may also activate cells in con- comprised of DNA and histones. In this conceptualization, DNA
junction with other immune stimulators such as TLR ligands. For behaves as an epitope or surface of a larger structure that impacts
both DNA and HMGB1, the immune properties may therefore re- on the immune system; the use of the term DNA does not
flect the array of other endogenous as well as exogenous molecules imply that this molecule is free in solution and devoid of other
present. nucleosomal components (1, 3).
Because of the importance of DNA as antigen in SLE, efforts
Keywords: innate immunity; DNA; HMGB1; apoptosis; necrosis
to replicate this disease in induced animal models focused pri-
The nucleus is the central organelle in the cell because it is the marily on immunization with DNA. With a few possible excep-
location for replication, transcription, and the regulation of gene tions, these experiments failed, with DNA, even if attached to
expression. While often viewed as uniform in composition, the a carrier and presented in adjuvant, unable to induce an apprecia-
nucleus is a dynamic structure with a regional anatomy that can ble autoantibody response. In its immunologic inertness, DNA
appeared fundamentally different from proteins and carbohy-
vary during the cell cycle. Furthermore, the nucleus can serve
drates, the other major classes of macromolecule antigens (3).
as a repository for molecules that transit into and out of the
The failure of DNA to induce a specific autoimmune response
cytoplasm and even exit the cell entirely. Remarkably, once in
contrasts sharply with the situation of other autoimmune dis-
the extracellular space, some of these nuclear molecules can
eases, such as rheumatoid arthritis or multiple sclerosis, where
serve as immune mediators and trigger the innate immune sys-
animal disease models can be created in animals by immuniza-
tem in settings of injury and death. As such, the translocation
tion with self antigens such as collagen or myelin basic protein
of nuclear macromolecules from the inside to the outside of the
(4). Together, these considerations suggested that SLE reflects
cell is a central event in innate immunity.
a fundamental disturbance in the immune system that allows a
Among endogenous molecules stimulating innate immunity,
response to an essentially immunologic blank molecule. The
two nuclear molecules have attracted the most attention. Thus,
alternative explanation for the anti-DNA responses in SLE is
both DNA and the DNA-binding protein, HMGB1, can exit cells
the existence of a more immunogenic form of DNA that could
during death processes and, in the extracellular space, stimulate
induce responses under conditions in which experimental DNA–
responses via a variety of receptors to signal danger. While both
protein complexes fail.
DNA and HMGB1 reside primarily in the nucleus and, indeed,
may interact, they differ in intranuclear and intracellular mobility
THE INDUCTION OF IMMUNE RESPONSES TO DNA
as well as physical state during death processes. This review will
therefore consider the roles of DNA and HMGB1 in innate The mystery of DNA’s immune activity in SLE is now clearing
immunity and propose an integrated picture for their movement because of studies in two seemingly disparate areas that have
and function during death processes. converged to provide a picture of the triggering of innate immu-
nity by nuclear molecules. The first area of this research concerns
the immune properties of bacterial DNA or, as it is often called,
CpG DNA. As shown in studies conducted over the last 20
years, bacterial DNA, unlike mammalian DNA, is immunologi-
(Received in original form January 23, 2007; accepted in final form March 1, 2007 )
cally active and can induce cytokine production and B cell mito-
Supported by VA Medical Research Service and the Lupus Research Institute.
genesis. As demonstrated in elegant molecular studies, these
Correspondence and requests for reprints should be addressed to David S. Pisetsky, responses result from stimulation of the TLR9 receptor which,
M.D., Ph.D., Durham VA Medical Center, Box 151G, 508 Fulton St., Durham,
in contrast to some other Toll-like receptors (TLRs), resides on
NC 27705. E-mail: dpiset@acpub.duke.edu
the inside of cells as opposed to the membrane (5, 6).
Proc Am Thorac Soc Vol 4. pp 258–262, 2007
DOI: 10.1513/pats.200701-027AW The induction of immune responses by bacterial DNA reflects
Internet address: www.atsjournals.org structural microheterogeneity and its content of sequence motifs
Pisetsky: Nuclear Molecules in Immunity 259

that center on unmethylated CpG dinucleotides. These se- regard, immune complexes with ribonucleoproteins can also
quences occur much more commonly in bacterial than mamma- stimulate IFN-␣, perhaps related to their content of RNA mole-
lian DNA because of differences in the patterns of base methyla- cules which can trigger TLR3 or TLR7 (14).
tion as well as a phenomenon known as CpG suppression. The activity of DNA in immune complexes contrasts with
Because of the differential display of the CpG motifs, bacterial the activity of free mammalian DNA which is either inert or
DNA, like LPS, can act as a pathogen-associated molecular inhibitory. As shown in vitro studies, free mammalian DNA can
pattern (PAMP). Importantly, in the studies on the structure– inhibit the activity of bacterial DNA, raising the possibility that
function relationships of immune stimulation by DNA, intact high concentrations of extracellular DNA can attenuate stimula-
mammalian DNA has been consistently inactive in in vitro or tion by bacterial DNA and possibly down-regulate innate immu-
in vivo systems. These observations point to the exquisite speci- nity. This inhibitory activity can be termed “safety” in contrast
ficity for base recognition in the triggering of innate immunity to “danger” elicited by TLR ligands (17). Thus, as these experi-
via TLR9. ments indicate, the formation of immune complexes can radically
The demonstration of the intrinsic immune activity of bacte- transform the activity of mammalian DNA to allow stimulation,
rial DNA is notable since it joins DNA with proteins and carbo- rather than inhibition, of innate immunity.
hydrates in the family of immune active macromolecules. Studies Since even high concentrations of mammalian DNA are inac-
on the antibody response to mammalian and bacterial DNA tive, it appears unlikely that the role of the complex is solely to
in humans confirmed and extended this role. As this serologic deliver DNA to TLR9. Even if mammalian DNA contains some
analysis showed, the sera of normal human subjects (NHS) have CpG motifs, the presence of nonstimulatory or inhibitory motifs
significant levels of antibody directed to bacterial DNA. These appears predominant. An interaction of active motifs within
antibodies bind with high avidity and specificity to some, but not mammalian DNA may nevertheless cause activation of TLR9,
all, bacterial DNA antigens. While the antibodies in NHS differ although the triggering of other internal receptors appears more
in isotype from those in SLE, their presence nevertheless sug- plausible. The operation of such receptors can be inferred from
gests that, during the ordinary encounter with bacteria, bacterial the activity of mammalian DNA in complexes with transfection
DNA triggers responses and can drive responses to sequential reagents known as cytofectins (18). These reagents promote
DNA epitopes (7–9). DNA internalization and, while the mode of action of cytofectins
Together, these observations raise the possibility that bacte- and immune complexes may differ, the potency of DNA in
rial DNA can initiate or sustain anti-DNA production in SLE cytofectin complexes provides further evidence that the context
because of its intrinsic immunologic activity as well as display of DNA determines its activity on innate immunity (18–20).
of the double-stranded B DNA antigen conformation. According The immunostimulatory activity of DNA-containing immune
to this model, the lesion in SLE would reflect antigen recognition, complexes has attracted great interest as a mechanism for pro-
not responsiveness, with abnormalities in the immune repertoire moting nonspecific immune activation in the pathogenesis of
in SLE providing an array of B cell precursors that can be SLE. While such complexes can sustain or intensify immune
triggered by bacterial DNA antigen to produce autoantibodies activation, they do not explain the initial generation of the anti-
(10). This possibility is supported by studies showing that bacte- DNA antibody which is critical to the activation by DNA. Fur-
rial DNA can induce an autoantibody response by immunization thermore, the necessity for complex formation suggests that for
of pre-autoimmune NZB/NZW mice under conditions in which endogenous DNA to activate innate immunity, it must exist in
mammalian DNA is inactive (11). While supporting the ability association with another moiety or structure to allow access to
of bacterial DNA to stimulate responses, these studies attest to internal DNA receptors. These considerations (Table 1) thus
the paucity of immunologic activity in mammalian DNA. focus attention on the mechanisms by which DNA is released
from cells and the molecules to which it is attached during this
THE ACTIVITY OF IMMUNE COMPLEXES process.
The second line of research on the immune activity DNA devel-
oped originally in efforts to characterize a factor in the sera of RELEASE OF DNA FROM DEAD AND DYING CELLS
patients with lupus that could stimulate the in vitro production As shown using a variety of assays, DNA appears at high levels
of interferon-␣ (IFN-␣). As shown in seminal experiments, this in the blood in many conditions including SLE, pulmonary in-
factor is an immune complex comprised of DNA and anti-DNA. farction, malignancy, and trauma among others. These condi-
While either component alone is devoid of activity, the complex tions are all associated with cell activation or cell death, although
can potently stimulate IFN-␣ production by plasmacytoid den- death has been considered the most likely source of blood DNA
dritic cells. Furthermore, this stimulatory activity can be mim- (21, 22). As now conceptualized, cell death can be divided into
icked by mixing antibody preparations with supernatants of apo- mechanistically distinct processes, with apoptosis and necrosis
ptotic cells. Studies on the mechanisms of B cell activation in a representing the prototype pathways. Apoptosis, or programmed
rheumatoid factor transgenic system in the mouse also showed cell death, is a highly regulated process characterized by cellular
that immune complexes with DNA have activity not present collapse and cleavage of nuclear molecules. In contrast, necrosis
with DNA alone (12–14).
Subsequent studies on the stimulation by plasmacytoid DCs
by DNA immune complexes established a role for both Fc recep-
tors and TLR9 receptor; non–TLR receptor mechanisms may TABLE 1. ROLE OF DNA IN INNATE IMMUNITY
also operate, however. Since TLR9 resides on the inside of the
References
cells in an endosomal compartment, immune complexes may
allow DNA internalization, where interaction with TLR9 may Stimulation of B cells, macrophages, and dendritic cells 3, 5
occur. The interaction with the Fc receptor could lead to addi- Vary with sequence, base methylation, and backbone structure 3, 5
tional signaling or provide an alternative mechanism for DNA Vary with context and mode of cell entry 18
Stimulation of TLR and non-TLR receptors 19, 20
entry into the cell (15, 16). While the identity of the DNA in Induction of specific autoantibodies in normal humans 3, 7–10
immune complexes from sera has not been characterized exten-
sively, endogenous human DNA is the likely source. In this Definition of abbreviations: TLR ⫽ Toll-like receptor.
260 PROCEEDINGS OF THE AMERICAN THORACIC SOCIETY VOL 4 2007

is a random process, provoked by physical or chemical trauma immune system, the distinction between apoptosis and necrosis
that culminates in cell lysis (23). According to current paradigms, may not be as stark as sometimes portrayed. Thus, in the intact
necrosis is pro-inflammatory while apoptosis is anti-inflamma- animal or in mixed cell cultures, both apoptotic and necrotic
tory, perhaps related to release or spillage of internal molecules. cells can release DNA that is similar in properties as measured
To investigate nuclear molecule dynamics during death, our by size distribution. These considerations do not imply that the
laboratory has used in vitro and in vivo system to assess the extracellular DNA from apoptotic and necrotic cells are equiva-
amount and properties of extracellular DNA released from dying lent immunologically, since the immune activity of DNA de-
cells. In addition, we have investigated the influence of macro- pends on context and the presence of other molecules that may
phages on the release of DNA, since macrophages can scavenge affect its intracellular trafficking. It is important therefore to
dead cells by phagocytosis and therefore potentially modify the consider other molecules that may be released in death settings.
amount of DNA arising from such cells. In these experiments,
we have measured DNA by both an ELISA for nucleosomes as THE IMMUNE ACTIVITY OF HMGB1
well as a direct chemical measurement using the dye PicoGreen.
This dye binds specifically to double-stranded DNA and allows HMGB1 is a nonhistone nuclear protein that has also been
sensitive detection by a fluorometric assay. implicated in the stimulation of innate immunity during death
Results of these studies have provided a novel perspective processes. Structurally, HMGB1 is 214 amino acids long and can
on DNA release during cell death as well as highlighted differ- be divided into an A box, B box, and a C-terminal tail domain,
ences in the in vitro and in vivo settings. Thus, during in vitro with the A and B boxes responsible for DNA interaction. This
culture, Jurkat leukemia T cells made apoptotic by chemical protein can bind DNA in a non–sequence-specific manner to
agents release DNA in a time-dependent process. This extracel- bend DNA, although it binds preferentially to distorted DNA
lular DNA shows laddering by gel electrophoresis, indicating structures such as strand junctions. In addition to binding DNA,
cleavage into nucleosomal fragments. In contrast to cells under- HMGB1 can interact with nuclear proteins. While the precise
going apoptosis, cells made necrotic by treatment with heat or function of HMGB1 is not known, this protein can regulate
ethanol fail to release DNA even after prolonged culture and transcription, perhaps by altering the architecture of DNA to
despite changes in cellular permeability, as demonstrated by promote interactions with other factors. Depending on cell type,
propidium iodide staining (24). HMGB1 can also appear in the cytoplasm (29, 30).
As our experiments showed, in the in vitro setting, macro- Like DNA, HMGB1 can leave the cell where, in the extracel-
phages can display important but divergent effects on DNA lular space, it can serve as an alarmin and promote immune
release. With apoptotic cells, the presence of macrophages (ei- activation. An alarmin is an intracellular molecule that, when
ther the RAW264.7 cell line or murine bone marrow–derived released from cells during death processes, can stimulate innate
macrophages) can reduce the amount of DNA released, whereas immune processes (31). In another terminology, an alarmin is
macrophages can increase the amount of DNA released by ne- a DAMP or damage (or death)-associated molecular pattern.
crotic cells. For necrotic cells co-cultured with macrophages, the In the case of HMGB1, the release processes was first detected
DNA present in the medium showed laddering suggestive of in the setting of macrophage activation rather than death. In
nuclease digestion. While indicating the importance of macro- these experiments, supernatants of cultures stimulated with LPS
phages in mediating DNA release, these findings also showed were characterized to identify other molecules that could medi-
that the size of the DNA in the extracellular space is not itself ate septic shock and therefore represent novel targets of therapy.
a measure of whether death occurs by apoptosis or necrosis (25). As these experiments showed, HMGB1 appears in high concen-
In vivo systems can also help dissect the DNA release process. trations in the medium of macrophages activated by LPS as well
Thus, in normal mice, the intraperitoneal administration of apo- as pro-inflammatory mediators such as TNF-␣, IFN-␣, and nitric
ptotic or necrotic Jurkat cells leads to a prompt rise in the amount oxide. Furthermore, HMGB1 levels are high in the serum of
of DNA in the blood that returns to baseline after 24 h. With patients with shock and antibodies to HMGB1 can block shock
both types of dying cells, DNA shows laddering with a similar in mice (32). A series of insightful experiments established a
size distribution by gel electrophoresis. This result is similar to broad range of immunostimulatory activities of HMGB1, consis-
that obtained in vitro using mixed cultures of Jurkat cells and tent with its role as a late mediator of LPS and its function as
macrophages. Other studies with this model showed that the a cytokine. This stimulation appears dependent on the RAGE
extent of DNA release from transferred dead cells can be modi- receptor, although activation of TLR2 and TLR4 may also con-
fied by treatment of recipient mice with clodronate (which elimi-
tribute to stimulation (33–37).
nates macrophages), dexamethasone, or the induction of perito-
As shown subsequently, death processes, in addition to mac-
neal exudates (26–28).
rophage activation, can lead to HMGB1 release, with this protein
Together, these studies provide evidence that macromolecule
posited as an important mediator of inflammation induced by
release from dead cells is a modifiable process that may depend
necrotic cells. As shown in vitro, cells induced to undergo necro-
on the presence of more than one cell population, especially
sis by freeze-thawing release large concentrations of HMGB1.
macrophages (Table 2). Furthermore, they suggest that, to the
This release process differs from that occurring during activation,
extent that extracellular DNA can be a player in the innate
where acetylation and phosphorylation alter the charge of
HMGB1. These modifications affect the intracellular trafficking
of HMGB1, with the modified molecule transiting to the cyto-
TABLE 2. ORIGIN AND PROPERTIES OF EXTRACELLULAR DNA plasm into vesicles for secretion (38, 39). In contrast, for HMGB1
release during necrosis, the process appears passive and results
References from the diffusion of this protein away from chromatin and out
Increased blood levels in settings of cell death 21 of the cell. Reflecting its nuclear function, HMGB1 is only weakly
DNA laddering 21, 22 adherent to chromatin, differing markedly from histones in the
In vitro release by apoptotic cells 24 strength of its DNA interaction. Thus, unanchored to the nu-
In vivo release with apoptosis and necrosis 26
cleus, HMGB1 can leave readily when the permeability barriers
Role of macrophages 25, 27
break down during death (38).
Pisetsky: Nuclear Molecules in Immunity 261

While the release of HMGB1 during necrosis is consistent HMGB1 can enhance DNA transfection, suggesting its ability
with its biophysical properties, its behavior during apoptosis has to shuttle nucleic acid into cells (43).
been more uncertain. Studies by Scaffidi and coworkers indicated Together, these considerations suggest that immune activity
that apoptotic cells do not release HMGB1, with studies by FLIP of nuclear molecules may result from the ensemble of species
(fluorescence loss of photobleaching) in fact demonstrating that present, their physical interaction, and their ability to trigger
its nuclear diffusion decreases dramatically during apoptosis, more than one receptor type simultaneously. Whether DNA
implying greater adherence to chromatin (40). Since apoptotic promotes HMGB1 stimulation or vice versa is a matter of seman-
cells are generally considered anti-inflammatory, a retention of tics, as both components may have to be present simultaneously
HMGB1 in the nucleus could limit immune activation during in a complex to induce cell activation. The rules by which com-
this death process. These results are notable since they suggest plexes stimulate immunity are not known, although stimulation
that, while apoptotic cells release DNA, they do not release a by organelle fragments or particulates may be more analogous
companion DNA-binding protein (i.e., HMGB1). to a cell–cell interaction (where multiple molecular interactions
To resolve this seeming contradiction, we reevaluated occur) than the stimulation by a cytokine, a hormone, or other
HMGB1 release during death processes, using Jurkat cells as small molecular mediator (e.g., prostanoid), which may act in a
models. In these experiments, apoptosis was induced by chemical unitary manner.
agents and HMGB1 release measured by Western blotting. Con- While these issues require much further investigation, the
focal microscopy with staining with anti-HMGB1 was used to studies on DNA and HMGB1 nevertheless indicate the diversity
confirm translocation. Together, these experiments showed of intracellular molecules with immune activity and the impor-
clearly that, in apoptosis as well as necrosis, translocation of tance of context in the stimulation of innate immunity by nuclear
HMGB1 can occur, with the nucleus of cells showing dramati- macromolecules. Future studies will track the dynamics of nu-
cally reduced HMGB1 content in association with nuclear con- clear molecule trafficking during death processes, the role of
densation (41). macrophages in determining the extracellular release, and the
Since DNA release occurs during apoptosis, HMGB1 release signaling pathways stimulated by the complex mixtures of large
is not unexpected and suggests that permeability changes of molecules. Hopefully, this research will elucidate the pathogene-
nuclear and cytoplasmic membranes allow the diffusion of a sis of SLE and other autoimmune diseases as well as provide
variety of macromolecules into the extracellular space. The rea- new approaches to treat the broad range of immune-mediated
sons cells differ in their behavior during apoptosis is unknown, diseases.
although this process is not uniform among cells and may be Conflict of Interest Statement : D.S.P. does not have a financial relationship with
influenced by both the cell type as well as inducing agent. Fur- a commercial entity that has an interest in the subject of this manuscript.
thermore, comparisons between the amount of HMGB1 released
during apoptosis and necrosis can be misleading, since the extent References
of macromolecule release during necrosis may also vary de- 1. Tan EM. Antinuclear antibodies: diagnostic markers for autoimmune
pending upon the inducing agent or treatment. Thus, in our diseases and probes for cell biology. Adv Immunol 1989;44:93–151.
experience, the extent of HMGB1 released from cells undergo- 2. Kotzin BL. Systemic lupus erythematosus. Cell 1996;85:303–306.
ing freezing and thawing is much greater than cells that have 3. Pisetsky DS. Antibody responses to DNA in normal immunity and aber-
rant immunity. Clin Diagn Lab Immunol 1998;5:1–6.
been treated with ethanol or heat. The most relevant system for
4. Marrack P, Kappler J, Kotzin BL. Autoimmune disease: why and where
inducing necrosis is speculative, since necrosis is a random and it occurs. Nat Med 2001;7:899–905.
unregulated process that may follow a host of damaging agents. 5. Pisetsky DS. Immune activation by bacterial DNA: a new genetic code.
Table 3 summarizes the immune properties of HMGB1. Immunity 1996;5:303–310.
6. Wagner H. The immunobiology of the TLR9 subfamily. Trends Immunol
2004;25:381–386.
THE ACTIVITY OF EXTRACELLULAR 7. Wu ZQ, Drayton D, Pisetsky DS. Specificity and immunochemical prop-
NUCLEAR MATERIAL erties of antibodies to bacterial DNA in sera of normal human subjects
and patients with systemic lupus erythematosus (SLE). Clin Exp
While original studies suggested that HMGB1 has direct cyto- Immunol 1997;109:27–31.
kine activity, other studies have shown a more complicated situa- 8. Pisetsky DS, Drayton D, Wu ZQ. Specificity of antibodies to bacterial
tion. Thus, studies with purified mammalian HMGB1 show much DNA in the sera of healthy human subjects and patients with systemic
lower activity than molecularly cloned material, which may have lupus erythematosus (SLE). J Rheum 1999;26:1934–1938.
contamination by endotoxin or DNA. These considerations sug- 9. Hamilton KJ, Schett G, Reich CF III, Smolen J, Pisetsky DS. The binding
gest that HMGB1 may not be fully active as a cytokine but of sera of patients with SLE to bacterial and mammalian DNA. Clin
Immunol 2006;118:209–218.
rather may act in concert with other endogenous or exogenous 10. Pisetsky DS, Drayton DM. Deficient expression of antibodies for bacte-
molecules to trigger innate immunity (42). In this conceptualiza- rial DNA by patients with systemic lupus erythematosus. Proc Assoc
tion, HMGB1 may tune the system, serving a thermostatic func- Am Physicians 1997;109:237–244.
tion to allow other molecules to trigger responses. Furthermore, 11. Gilkeson GS, Ruiz P, Pippen AMM, Alexander AL, Lefkowitz JB,
HMGB1, like antibody or cytofectin, may serve as a carrier for Pisetsky DS. Modulation of renal disease in autoimmune NZB/NZW
mice by immunization with bacterial DNA. J Exp Med 1996;183:1389–
DNA to allow triggering of internal DNA receptors. Indeed,
1397.
12. Vallin H, Perers A, Alm GV, Rönnblom L. Anti-double stranded DNA
antibodies and immunostimulatory plasmid DNA in combination
mimic the endogenous IFN-␣ inducer in systemic lupus erythematosus.
TABLE 3. ROLE OF HMGB1 IN INNATE IMMUNITY J Immunol 1999;163:6306–6313.
13. Viglianti GA, Lau CM, Hanley TM, Miko BA, Shlomchik MJ, Marshak-
References Rothstein A. Activation of autoreactive B cells by CpG dsDNA.
Induction of extracellular release by TLR ligands 32 Immunity 2003;19:837–847.
Stimulation of cytokine production 33, 35 14. Rönnblom L, Eloranta M-L, Alm GV. The Type I interferon system in
Interaction with RAGE 37 systemic lupus erythematosus. Arthritis Rheum 2006;54:408–420.
Role of acetylation and phosphorylation in translocation 38, 39 15. Båve U, Magnusson M, Eloranta M-L, Perers A, Alm GV, Rönnblom L.
Extracellular release with necrosis and apoptosis 40, 41 Fc␥RIIa is expressed on natural IFN-␣-producing cells (plasmacytoid
dendritic cells) and is required for the IFN-␣ production induced by
262 PROCEEDINGS OF THE AMERICAN THORACIC SOCIETY VOL 4 2007

apoptotic cells combined with lupus IgG1. J Immunol 2000;165:3519– 30. Thomas JO. HMG1 and 2: architectural DNA-binding proteins. Biochem
3526. Soc Trans 2001;29:395–401.
16. Means TK, Latz E, Hayashi F, Murali MR, Golenbock DT, Luster AD. 31. Erlandsson-Harris H, Raucci A. Alarmin(g) news about danger: work-
Human lupus autoantibody-DNA complexes activate DCs through shop on innate danger signals and HMGB1. EMBO Rep 2006:7;774–
cooperation of CD32 and TLR9. J Clin Invest 2005;115:407–417. 778.
17. Zhu FG, Reich CF III, Pisetsky DS. Inhibition of murine macrophage 32. Wang H, Bloom O, Zhang M, Vishnubhakat JM, Ombrellino M, Jiantu
nitric oxide production by synthetic oligonucleotides. J Leukoc Biol C, Frazier A, Yang H, Ivanova S, Borovikova L, et al. HMG-1 as a
2002;71:686–694. late mediator of endotoxin lethality in mice. Science 1999;285:248–251.
18. Jiang W, Reich CF III, Pisetsky DS. Mechanisms of activation of the 33. Andersson U, Wang H, Palmblad K, Aveberger A-C, Bloom O,
RAW264.7 macrophage cell line by transfected mammalian DNA. Erlandsson-Harris H, Janson A, Kokkola R, Zhang M, Yang H, et al.
Cell Immunol 2004;229:31–40. High mobility group 1 protein (HMG-1) stimulates proinflammatory
19. Okabe Y, Kawane K, Akira S, Taniguchi T, Nagata S. Toll-like receptor- cytokine synthesis in human monocytes. J Exp Med 2000;192:565–570.
independent gene induction program activated by mammalian DNA 34. Müller S, Scaffidi P, Degryse B, Bonaldi T, Ronfani L, Agresti A,
escaped from apoptotic DNA degradation. J Exp Med 2005;202:1333– Beltrame M, Bianchi ME. The double life of HMGB1 chromatin
1339. protein: architectural factor and extracellular signal. EMBO J
20. Ishii KJ, Coban C, Kato H, Takahashi K, Torii Y, Takeshita F, Ludwig 2001;20:4337–4340.
H, Sutter G, Suzuki K, Hemmi H, et al. takeuchi O, Akira S. A 35. Agnello D, Wang H, Yang H, Tracey KJ, Ghezzi P. HMGB-1, a DNA-
Toll-like receptor-independent antiviral response induced by double- binding protein with cytokine activity, induces brain TNF and IL-6
stranded B-form DNA. Nat Immunol 2006;7:40–48. production, and mediates anorexia and taste aversion. Cytokine 2002;
18:231–236.
21. Sidransky D. Circulating DNA: what we know and what we need to
36. Park JS, Arcaroli J, Yum H-K, Yang H, Wang H, Yang K-Y, Choe K-H,
learn. Ann N Y Acad Sci 2000;906:1–4.
Strassheim D, Pitts TM, Tracey KJ, et al. Activation of gene expression
22. Lui YYN, Lo YMD. Circulating DNA in plasma and serum: biology
in human neutrophils by high mobility group box 1 protein. Am J
preanalytical issues and diagnostic applications. Clin Chem Lab Med
Physiol Cell Physiol 2003;284:C870–C879.
2002;40:962–968.
37. Kokkola R, Andersson Å, Mullins G, Östberg T, Treutigert C-J, Arnold
23. Fink SL, Cookson BT. Apoptosis, pyroptosis, and necrosis: mechanistic
B, Nawroth P, Andersson U, Harris RA, Harris HE. RAGE is the
description of dead and dying eukaryotic cells. Infect Immun
major receptor for the proinflammatory activity of HMGB1 in rodent
2005;73:1907–1916. macrophages. Scand J Immunol 2005;61:1–9.
24. Choi JJ, Reich CF III, Pisetsky DS. Release of DNA from dead and 38. Bonaldi T, Talamo F, Scaffidi P, Ferrera D, Porto A, Bachi A, Rubartelli
dying lymphocyte and monocytes cell lines in vitro. Scand J Immunol A, Agresti A, Bianchi ME. Monocytic cells hyperacetylate chromatin
2004;60:159–166. protein HMGB1 to redirect it towards secretion. EMBO J 2003;22:
25. Choi JJ, Reich CF III, Pisetsky DS. The role of macrophages in the in 5551–5560.
vitro generation of extracellular DNA from apoptotic and necrotic 39. Youn JH, Shin J-S. Nucleocytoplasmic shuttling of HMGB1 is regulated
cells. Immunology 2005;115:55–62. by phosphorylation that redirects it toward secretion. J Immunol 2006;
26. Jiang N, Reich CF III, Pisetsky DS. Role of macrophages in the genera- 177:7889–7897.
tion of circulating blood nucleosomes from dead and dying cells. Blood 40. Scaffidi P, Misteli T, Bianchi ME. Release of chromatin protein HMGB1
2003;102:2243–2250. by necrotic cells triggers inflammation. Nature 2002;418:191–195.
27. Jiang N, Pisetsky DS. The effect of dexamethasone on the generation of 41. Bell CW, Jiang W, Reich CF III, Pisetsky DS. The extracellular release
plasma DNA from dead and dying cells. Am J Pathol 2004;164:1751– of HMGB1 during apoptotic cell death. Am J Physiol Cell Physiol
1759. 2006;291:C1318–C1325.
28. Jiang N, Pisetsky DS. The effect of inflammation on the generation of 42. Rouhiainen A, Tumova S, Valmu L, Kalkkinen N, Rauvala H. Analysis
plasma DNA from dead and dying cells in the peritoneum. J Leukoc of proinflammatory activity of highly purified eukaryotic recombinant
Biol 2005;77:296–302. HMGB1 (Amphoterin). J Leukoc Biol 2007;81:49–58.
29. Bustin M. Regulation of DNA-dependent activities by the functional 43. Böttger M, Vogel F, Platzer M, Kiessling U, Grade K, Strauss M. Conden-
motifs of the high-mobility-group chromosomal proteins. Mol Cell sation of vector DNA by the chromosomal protein HMG1 results in
Biol 1999;19:5237–5246. efficient transfection. Biochim Biophys Acta 1988;950:221–228.

También podría gustarte