Vous êtes sur la page 1sur 10

ARTHRITIS & RHEUMATISM

Vol. 39, No. 12, December 1996, pp 1951-1960


0 1996, American College of Rheurnatology 1951

REVIEW

MOLECULAR THERAPEUTICS

Methotrexate and its Mechanism of Action

BRUCE N. CRONSTEIN

Since its reintroduction to the practice of rheu- tion of activated lymphocytes and other cells responsible
matology in the mid-l980s, methotrexate (MTX) has for tissue injury in RA-similar to its role in the
become the most widely used second-line agent for the treatment of cancer. Clearly, the side effects of low-dose
treatment of rheumatoid arthritis (RA). To date, MTX MTX therapy, stomatitis and marrow suppression, are
has been used to treat patients who have RA or other due to inhibition of cell replication; however, there is no
inflammatory diseases without a clear understanding of evidence that MTX specifically depletes specific “auto-
either its mechanism of action or the molecular basis for immune” cells.
its toxicity. A thorough understanding of the biochemi- In clinical use, the development of marrow sup-
cal mechanism(s) of action of MTX as well as the events pression is seen most often in patients who are relatively
that lead to the toxic effects of MTX may result in the folate-deficient at the start of therapy (2,3), and its
development of new, more effective and less toxic agents occurrence is usually a signal to either reduce the dosage
for the treatment of RA and other inflammatory dis- of MTX or discontinue the drug. Prevention of marrow
eases. In this review, the effects of MTX treatment on suppression, stomatitis, and other more common toxi-
those factors thought to be most important in the cities of MTX without diminution of the therapeutic
pathogenesis of chronic inflammatory arthritis will be effect in RA patients by folk acid or folinic acid
discussed and the evidence regarding the molecular supplements (4-6) would suggest that inhibition of
mechanism of MTX will be examined. cellular proliferation (or complete suppression of other
folate-dependent cellular reactions) by MTX is not
MTX is a cytotoxic agent critical to its therapeutic effect (7).
The first report on the efficacy of MTX in the
treatment of RA appeared in 1951 (1). MTX had MTX modulates humoral immunity
recently been introduced as a cytotoxic agent for the
One of the hallmarks of RA is the presence of
treatment of malignancies, and it was thought that the
serum antibodies directed against the Fc portion of IgG
potent inhibition of purine and pyrimidine synthesis
rheumatoid factor (RF), although its role in the patho-
mediated by MTX might lead to similar suppression of a
genesis of RA is debatable. Several groups of investiga-
malignant immune response. When reintroduced to tors have reported that MTX treatment does not affect
clinical rheumatology in the 1980s, it was thought to act
circulating levels of IgM-RF (8-10). In contrast, Alarcdn
as a chemotherapeutic agent for inhibiting the replica-
and colleagues (11) used a sensitive enzyme-linked
immunosorbent assay to study the concentrations of IgM
Supported by grants from the USPHS (AR-AI-41911, AR- and IgA RFs in patients involved in a blinded study of
11949, and HL-19721), the GCRC (MOl-RR-00096), and the Kaplan the efficacy of MTX in the treatment of RA; those
Cancer Center (CA-16087). authors observed a significant drop in R F levels in
Bruce N. Cronstein, MD: New York University Medical
Center, New York. patients who improved. When the data were analyzed,
Address reprint requests to Bruce N. Cronstein, MD, Profes- there was a strong statistical association between MTX
sor of Medicine and Pathology, Division of Rheumatology, New York treatment and a reduction in R F levels and a less
University Medical Center, 550 First Avenue, New York, NY 1UO16.
Submitted for publication June 18, 1996; accepted in revised impressive association between improvement in re-
form August 21, 1996. sponse to MTX and reduction in R F levels. Other
1952 CRONSTEIN

investigators (12) have reported that R F levels de- cell responses. In general, though, there are few data to
creased when inflammation was controlled by either support the contention that modulation of T cell num-
MTX or gold salts. bers or functions by MTX is responsible for the thera-
Olsen and coworkers (13) reported that treat- peutic effects of MTX.
ment of RA patients with MTX is associated with
diminished RF production by lymphocytes cultured ex
MTX diminishes phlogiston production and responses
vivo, although similar effects on R F production were
reported for gold salts as well. Moreover, MTX treat- A large and increasing number of soluble agents
ment of cultured peripheral blood mononuclear cells have been reported to play a role in the development of
(PBMC) has been shown to result in diminished RF synovial inflammation. These factors include lipid deriv-
production in vitro (14,15). In some of these studies, the atives (prostaglandins, leukotrienes, platelet-activating
investigators were able to reverse the effects of MTX on factor), complement activation products, cytokines, and
RF production by treating the cells with folinic acid. chemokines. Although MTX has not been reported to
Since reductions in the R F level may accompany affect the generation of complement activation products
successful therapy with other second-line agents (e.g., or prostaglandins, it may affect either the generation of
gold salts and penicillamine) and since the role of R F in or response to leukotrienes, cytokines, and chemokines.
the pathogenesis of RA is unclear, it is difficult to Leukotrienes, most notably leukotriene B4
conclude that the effects of MTX therapy on R F levels (LTB,), are potent lipid-derived stimuli for leukocytes.
in vivo or on RF production in vitro are causally related to The effect of MTX therapy on the generation of leuko-
the beneficial effects of this drug in the therapy of RA. trienes remains somewhat controversial. Sperling and
coworkers (21,22) have reported that neutrophils from
MTX modulates cellular immunity RA patients or normal controls treated with MTX
produce less LTB, than do cells from individuals not
The central role of cellular immune reactions and
treated with MTX (21,22). Similarly, Leroux and co-
T cells in the development and pathogenesis of RA has
workers (23) reported that MTX treatment (a single
been generally accepted for a number of years. The
dose) inhibited the generation of 5- and 12-lipoxygenase
strong HLA-DR-associated risk of developing RA and
products. In contrast, Hawkes and colleagues (24) ob-
the presence of large numbers of T cells in the affected
served that MTX administered in vivo did not inhibit the
synovium support the contention that T cells are impor-
production of lipoxygenase products by neutrophils
tant determining elements in the pathogenesis of joint
inflammation and destruction in RA. Thus, one poten- stimulated ex vivo. Methodologic differences may ac-
tial explanation for the therapeutic effects of MTX in count for the differing results. Nonetheless, the resulting
RA is diminution in the number or reactivity of T cells diminution in LTB, synthesis and synovial fluid concen-
involved in the pathogenesis of inflammation (16). How- tration is probably not responsible for the therapeutic
ever, there is little evidence to support the hypothesis effects of MTX, since the magnitude of the decrement in
that MTX affects either the number or function of T synovial fluid LTB, induced by substitution of dietary
cells involved in the pathogenesis of RA. Moreover, lipid with fish oil is similar to that induced by MTX
recent controlled studies of monoclonal antibodies di- therapy, but does not lead to as great an improvement in
rected against CD4+ T cells have provided little evi- arthritis (25-27).
dence that the elimination or modulation of T cells Cytokines are low molecular weight proteins that
significantly alters the course of RA (17,18). are secreted by various immune and inflammatory cells.
Although a reduction in T cell number or func- Based on studies in animal models of RA and, more
tion might be the predicted result of MTX treatment, recently, in RA patients, it is increasingly clear that
Weinblatt and colleagues (19) reported that, in fact, cytokines play a critical role in the development of the
long-term therapy with MTX leads to an increase in the clinical and laboratory manifestations of RA. Although
percentage of CD3 and CD4 cells in the peripheral it is currently thought that tumor necrosis factor a (TNFa)
blood. In contrast, Wascher et a1 (20) reported that plays the central role in the cytokine cascades responsi-
short-term therapy (12 weeks) diminishes the number of ble for synovial inflammation in RA, interleukin-1 (IL-1)
circulating T and B cells. One caveat in interpreting and IL-6, among others, are responsible for many of the
these studies is that the peripheral blood T cell number, manifestations of RA (28). The effects of MTX therapy
subsets, or function may not accurately reflect synovial T on both the response to and the production of these
MECHANISM OF ACTION OF MTX 1953

cytokines have been studied extensively in animal mod- (for review, see ref. 28). In general, in vitro studies of
els, in RA patients, and in their isolated cells. MTX using either animal or human cells have shown
IL-1 was among the first cytokines to be discov- that there is little effect of MTX on TNFa production,
ered. Because of the availability of reagents with which although liposomal preparations of MTX dramatically
I L 1 production or the response to IL-1 can be mea- inhibit TNFa production in vitro, most likely because of
sured, the levels of this cytokine in both the circulation improved uptake of liposomal MTX by target cells
and the synovial fluid of animals and patients with RA (38-41). Studies in the adjuvant arthritis model of RA
has been studied in great detail. In two studies, it was have demonstrated that MTX treatment leads to a
shown that MTX treatment of animals with inflamma- profound decrease in the synovial fluid TNFa concen-
tory arthritis decreased the production of IL-1 in vivo as tration (42), and treatment with liposomal MTX results
well as ex vivo (29,30). Subsequent studies produced in diminished TNFa production ex vivo in this model
more ambiguous results; MTX treatment of animals or (39). No such study in humans has yet been reported.
patients or their cells did not inhibit IL-1 production, but The studies reported to date do not demonstrate any
significantly diminished the ability of their cells to significant effect of MTX therapy on TNFa concentra-
respond to IL-1, an effect which could be overcome in tions in the peripheral blood of patients with RA
some studies by treatment with folinic acid (31-33). (43,44). As with IL-1 (see above), there may be a
One potential explanation for the effects of MTX disparity between the effects of MTX on the synovial
treatment on cellular responses to IL-1 was reported by production of TNFa and total circulating levels of
Brody and colleagues (34) who observed that, when TNFa, and the utility of measuring circulating TNFa
studied in vitro, MTX markedly inhibited the binding of concentrations as an indicator of response to therapy
IL-1p to its receptor on peripheral blood cells, an effect remains unclear. Thus, although MTX therapy leads to a
that could be overcome by an excess of IL-1p. Although decrease in TNFa production locally, the role of this
those authors' findings may explain the in vitro effects of reduction in the mechanism of action of MTX is not
MTX, it is difficult to understand how reversible inhibi- settled.
tion of IL-1 binding can be relevant to the mechanism of A variety of other pro- and antiinflammatory
action of a drug which is administered on a weekly basis proteins have been measured in the serum of RA
and which is present as free drug in biologic fluids for patients treated with MTX. Among these, the one
only a few hours after the dose is administered (35). cytokine that is most consistently diminished in the
Later studies reported that MTX treatment of patients serum is IL-6, a cytokine most tightly linked to produc-
leads to diminished I G 1 concentrations in the synovial tion of acute-phase reactants by the liver. Serum con-
fluid of patients who achieved a response to the drug; centrations of IL-6 were found to decline during MTX
however, no reduction in serum IL-1 concentrations was therapy in 4 studies (44-47) but did not change in
noted (36,37). These observations suggested that a local another (48). Although MTX therapy leads to an in-
effect of MTX treatment on IL-1 production might crease in the cellular production of IL-1 receptor antag-
contribute to the therapeutic effects of low-dose MTX. onist and soluble TNF receptors (43), these changes are
Although none of these studies is definitive, they are all not reflected by alterations in the circulating levels of
consistent with the hypothesis that interference with these antiinflammatory proteins. The chemokine IL-8 is
either IL-1 production or the effects of IL-1 is related to a potent chemoattractant for neutrophils and an angio-
the reduction in symptomatic synovial inflammation genic factor produced by mononuclear cells, among
observed in patients with RA who are being treated with others; in patients with RA, MTX therapy was shown to
MTX. With the possible exception of the study by Brody inhibit the spontaneous production of IL-8 by PBMC
et a1 (34), none of the studies summarized here have but not synoviocytes (49,50). As with the other cytokines
determined how MTX decreases IL-1 secretion or the studied, the mechanistic role of IL-8 inhibition by MTX
response to IL-1. therapy in the antiinflammatory effects of MTX remains
Recently, investigators have concentrated their uncertain.
attention on the central role of TNFa in the pathogen- The studies summarized above suggest that at
esis of RA. This focus has led to the development of least one of the mechanisms by which MTX inhibits
potentially useful therapeutic agents (monoclonal anti- inflammation in RA is by modulating the production of
TNFa antibodies and soluble TNF receptors). Adminis- cytokines in the synovium. Although the production of
tration of anti-TNFa antibodies to patients with RA has some of these factors was clearly inhibited by MTX
resulted in dramatic, albeit temporary, improvements treatment in vitro or ex vivo, other agents also affected
1954 CRONSTEIN

P oly am in e s
putrescin (S permine, sp ermidine)

M e t h o;t e x a t e SAM
Methionine
D H F -**TH F- N 5-C H ,-TH F 1 Methylation of
Homocysteine p h o sp h olipid s ,
proteins, R N A ,
Adenosine DNA, etc

Figure 1. Inhibition of transmethylation reactions and polyamine formation by methotrexate.


DHF = dihydrofolate; THF = tetrahydrofolate; N"-CH,-THF = N"-methyl-tetrahydrofolate;
ATP = adenosine triphosphate; THF = tetrahydrofolate; SAM = S-adenosyl-rnethionine; SAH =
S-adenosyl-hornocysteine.

cytokine release (e.g., corticosteroids), indicating that the treatment of RA have been ascribed to two distinct,
diminution of cytokine release at sites of inflammation is although not mutually exclusive, biochemical mecha-
not a specific property of MTX. Finally, none of the nisms, as follows.
studies cited addressed the question of the molecular
mechanism by which MTX treatment modulates cyto- The biochemical mechanism of action of MTX:
kine release, although a number of hypotheses have two hypotheses
been suggested.
MTX inhibits methylation reactions. MTX and
its polyglutamates inhibit a number of intracellular
The molecular mechanism(s) of MTX
folate-dependent reactions, among which is the methyl-
One potential explanation for the effects of MTX ation of homocysteine to methionine. The conversion of
therapy on cellular and humoral immunity and cytokine homocysteine to methionine is required for the genera-
secretion is that MTX, via its effects on the purine and tion of S-adenosyl-methionine ( S A M ) , which acts as the
pyrimidine synthesis required for cell division, is cyto- proximal methyl donor to RNA, DNA, amino acids,
toxic for the cells that generate cytokines or incite other proteins, and phospholipids, as well as in the synthesis of
cells to generate cytokines. As discussed above, there is polyamines such as spermidine and spermine (Figure 1).
no evidence that a specific subset of immune or other Methylation of all of these biologically active molecules
cells is depleted in patients undergoing MTX therapy, is required for cellular survival and function, particularly
although the cytotoxic effects of MTX probably play a in the immune system. After donation of its methyl
role in the toxicity of MTX (stomatitis, cytopenias). group, SAM is converted to S-adenosyl-homocysteine
Alternatively, MTX treatment may induce a biochemical ( S A H ) which, in turn, is metabolized to adenosine and
change in the cells of the inflamed synovium that leads homocysteine, which can be recycled to form SAM
to a decrease in the secretion of soluble mediators of (Figure 1).
inflammation. During the late 1970s and early 1980s, investiga-
After absorption from the gut (or after systemic tions into the pathogenesis of severe combined immu-
administration), MTX is present in the circulation as nodeficiency associated with adenosine deaminase
either the native compound or its active metabolite (ADA) deficiency demonstrated that SAH accumulates
7-hydroxy-MTX for a relatively short period of time. in the ADA-deficient cells and that the accumulation of
Both of these compounds are taken up by cells and SAH leads, via competitive inhibition of SAM-
converted intracellularly to their polyglutamates, which dependent methyl-transfer reactions, to marked inhibi-
are both active and quite long-lived. It is likely that the tion of transmethylation reactions (51-54). This obser-
accumulation of MTX polyglutamates is responsible for vation led to the hypothesis that the inhibition of
both the therapeutic effects and the toxic effects of the transmethylation reactions was an important factor in
drug (35). The therapeutic effects of low-dose MTX in the development of severe combined immunodeficiency.
MECHANISM OF ACTION OF MTX 1955

In othcr studics, investigators induced a marked increase reactions. Again, SAM and folinic acid (but not spcrmi-
in intracellular SAH concentration by incubating cells dine) rcversed the cffect of MTX on monocytc function
with large cxcesses of adcnosine and homocystcine in (65). Although the concentrations of MTX that were
the prcsence of an ADA inhibitor in order to inhibit required to inhibit cellular function in these two studies
transmethylation rcactions; they found that this treat- wcrc high, the results rcportcd are consistent with the
ment markedly inhibited lymphocyte and mononuclear hypothesis that inhibition of transmethylation reactions
cell function (55-57). The immunologic cffects of inhib- contributcs to the antiinflammatory cffects of MTX.
iting transmethylation reactions were thought to be of A further test of the hypothesis that inhibition of
potcntial use in the treatment of RA or othcr forms of transmethylation reactions is responsible for the antiin-
inflammatory disease in which cellular and humoral flainmatory effects of MTX would be to determine
immunity play a role. whether other inhibitors of transmethylation reactions
Evidencc that polyamincs, the synthesis of which are also antiinflammatory. Because of the interest in
is dependent upon SAM, might be important in the transmethylation reactions that followed from thc stud-
pathogenesis of KA was provided in later studies that ies of the mechanism of immunodeficiency in childrcn
reported an increase in polyaminc levels in urinc, syno- lacking ADA, a potent inhibitor of S A H hydrolase that
vial fluid, synovial tissuc, and PBMC from paticnts with thereby inhibited transmethylation reactions,
KA (58-61). Indeed, Fleschcr and coworkcrs (60,61) 3-deazaadenosine, was developed. In in vitro and in vivo
reported that the defect in IL-2 secretion observed whcn studies, this compound was found to be an active
peripheral blood T cells from patients with RA were antiinflammatory agcnt (66-71). Moreover, the drug was
cultured ex vivo could be rcversed by inhibitors of studied in clinical trials of paticnts with RA, although its
polyamines. Thosc investigators also suggcsted that the efficacy has never been reported (72). Intercstingly,
ammonia, H,O,, or other toxic substances generated although most of its effects were initially ascribed to its
from the oxidation of polyamines by mononuclear cells capacity to inhibit transmethylation reactions, some
(but not lymphocytes) in these cultures was responsiblc antiinflammatory cffects of 3-deazaadenosine wcre later
for the inhibition of IL-2 synthesis. The association of shown to be independent of its ability to inhibit trans-
polyamine accumulation with this specific defect in methylation rcactions (73-75). Thus, the evidence to
immune regulation has only bcen documented in KA. date suggcsts that the inhibition of transmethylation
Although similar defects in IL-2 production have bcen reactions by MTX may inhibit the cellular synthesis of
documented for the lymphocytcs of patients with sys- polyamines, which accumulate in the synovium of pa-
temic lupus erythcmatosus (see ref. 62) and Sjogrcn’s tients with RA and which may contribute to joint
syndrome (see ref. 63), the role of polyamines in the destruction in this disease.
pathogenesis of these alterations has not been explored. MTX promotes adenosine release. As discussed
Intracellular polyaminc concentrations and IL-2 re- above, MTX is taken up by cells and convcrted to
sponses in inflammatory cells, tissucs, or fluids of pa- long-livcd polyglutamatcs (76). Rcsults of studies per-
tients with other forms of inflammatory arthritis that formed by Nlcgra et a1 (77) and, subsequently, Baggott
respond to MTX have also not been studied. and coworkcrs (78) demonstrated that MTX polygluta-
In 1990, Neshcr and Moore (64) noted that M‘IX matcs were as potent, if not more potent, inhibitors of a
might also inhibit transmethylation reactions, and first varicty of folate-dependent enzymcs as thc native mol-
tested the hypothcsis that inhibition of these transmeth- ecule. Moreovcr, the enzyme inhibited most effectively
ylation reactions was responsiblc for thc beneficial ef- by MTX polyglutamatcs was 5-aminoimidazole-4-
fects of MTX treatment in RA. In their initial studies, carboxarnide ribonucleotide (AICAR) transformylase
they found that MTX inhibited the production of (77,78). If the synthesis of AICAR is less inhibited than
IgM-RF by cultured PBMC. Trcatrnent of the cells with its metabolism, as predicted by the rclative scnsitivitics
folinic acid reversed the cffect of MTX treatment on thc of the enzymatic steps to inhibition by MTX polygluta-
measured function. Morc telling, howevcr, was the ca- mates, then the inhibition of AICAR transformylase by
pacity of methionine or spermidine (a polyamine whose MTX polyglutamates would result in intracellular accu-
synthesis is dependent upon SAM) to revcrse thc effects mulation of AICAR (Figure 2). Since AICARibosidc
of MTX on mononuclear cell function (64). In a subse- directly inhibits ADA and AICAR inhibits AMP deami-
quent study, this same laboratory rcported further in nasc, the intracellular accumulation of AICAR could
vitro evidence that MTX inhibits cellular, in this case lead to the respective release of adenosine or AMP
monocytc, function via inhibition of transmethylation (which is convcrted by 5’-nucleotidase to adenosine)
1956 CRONSTEIN

INTRACELLULAR EXTRACELLULAR

---+.l
/
/
/ AIC#R ATP
METHOTREXATEdy 11
FAICAh
1
,,,q=+..
O0
ADP
tl
1 . AMP
tl
l n o s i n e m Adenosine,
b

~’N1
T
AMP
* ADENOSINE
1
I
1
Uric Acic) *Uric Acid

into the extracellular space (78,79). That MTX treat- leukocyte accumulation in this model of inflammation
ment may lead to the accumulation of AICAR was were due to the excess adenosine released into the
supported by the finding that humans treated with high inflammatory exudate, since elimination of extracellular
doses of MTX excrete increased concentrations of amino- adenosine by the addition of ADA to the exudate
imidazole carboxamide in their urine (80). reversed the effects of MTX treatment (82). Similar
The observations described above suggested the findings in a different animal model were reported by
hypothesis that adenosine, which is present in increased Asako and colleagues (83,84).
concentrations in the extracellular space, mediates at As with inhibition of transmethylation reactions,
least some of the antiinflammatory effects of low-dose a further test of the hypothesis that adenosine mediates
MTX therapy. This hypothesis was first tested in a series the antiinflammatory effects of MTX would be the
of in vitro experiments. Treatment of cultured human demonstration that other agents that induce adenosine
fibroblasts and endothelial cells with MTX led to in- release are antiinflammatory as well. Both adenosine
creased adenosine release but only after prolonged kinase inhibitors (85-88) and sulfasalazine (89), an
incubations, as might be predicted if the increase in antiinflammatory agent that directly inhibits AICAR
adenosine release resulted from intracellular accumula- transformylase (90), were also antiinflammatory by an
tion of AICAR (81). Moreover, MTX-induced adeno- adenosine-dependent mechanism in in vitro and in vivo
sine release was enhanced when the MTX-treated cells models of inflammation. This provides further evidence
were subject to exposure to a stressful agent, stimulated that the antiinflammatory effects of MTX are mediated,
neutrophils. The adenosine that was released from the at least in part, by adenosine. More support for the
MTX-treated cells was both necessary and sufficient to hypothesis that MTX enhances adenosine release in
diminish the adhesion of stimulated neutrophils, a sur- humans was provided by Bernini et a1 (91), who reported
rogate antiinflammatory effect (81). that high-dose MTX increases adenosine concentrations
Subsequent studies on the effects of MTX treat- in the cerebrospinal fluid of children treated for child-
ment in a murine model of acute inflammation revealed hood leukemia. Thus, MTX treatment does lead to local
that treatment of mice with pharmacologically relevant increases in adenosine concentration in both animals
doses of MTX at weekly intervals leads to intracellular and humans, and the evidence is consistent with the
AICAR accumulation and increased concentrations of notion that agents that promote adenosine release by
adenosine in inflammatory exudates. Moreover, the alternative mechanisms are also antiinflammatory in in
effects of low-dose, intermittent MTX treatment on vitro and animal models. Further proof of the hypothesis
MECHANISM OF ACTION OF MTX 1957

that agents that induce adenosine release are antiinflam- osine Al receptor agonist (109). Thus, enhanced aden-
matory will require trials in patients with RA or other osine release in extraarticular tissues may promote nod-
inflammatory diseases. ule formation in susceptible patients.
The antiinflammatory and immunologic effects Central nervous system (CNS) toxicity (head-
of adenosine result from the interaction of adenosine aches, light-headedness, and cognitive dysfunction) is
with specific receptors on the cell surface. These aden- not an uncommon problem for patients with rheumatic
osine receptors, of which 4 subtypes have been char- diseases treated with MTX. Similar toxic reactions are
acterized at the pharmacologic and molecular levels seen in children treated with higher doses of MTX for
(Al, A, AJ, are all members of the family of 7 leukemia. Bernini and colleagues (91) showed that the
transmembrane-spanning receptors related to the ad- severe CNS toxicity observed in MTX-treated leukemia
renergic receptors. Via interaction with an A,, receptor patients was associated with dramatic elevations in cere-
on stimulated neutrophils, adenosine inhibits stimulated brospinal fluid adenosine concentrations and was com-
release of toxic oxygen metabolites, adhesion to a variety pletely reversed by administration of an adenosine re-
of different cell types, and neutrophil-mediated injury ceptor antagonist (aminophylline). These observations
both in vitro and in vivo (92-96). Adenosine also inhibits suggest that the well-established CNS effects of adeno-
lymphocyte proliferation to mitogens and induces sup- sine are responsible for the CNS toxicity experienced by
pressor phenotype and function (97-102). Moreover, some patients.
adenosine, acting at one or another of its receptors on
monocyte/macrophages, inhibits the production of
TNFa, IL-6, and IL-8 (88,103-105) and, by a less clear Conclusion
mechanism, increases the secretion of the potent antiin- The introduction of MTX for the treatment of
flammatory cytokine IL-10 (106). One study demon- inflammatory diseases has transformed the way in which
strated that synovial cell synthesis of collagenase, but not RA is treated. Indeed, the standard of comparison for
tissue inhibitor of metalloprotease or stromelysin, is trials of new second-line agents is MTX. Although new
specifically inhibited in patients with RA who have been developments in the area of cytokine antagonists or gene
treated with MTX (107), and this highly selective effect therapy may lead to the development of potent, specific
of MTX therapy can be accounted for by an adenosine antiinflammatory drugs, the recognition that MTX may
receptor-mediated effect as well (108). The antiinflam-
act by two complementary biochemical mechanisms
matory effects of MTX treatment observed in a murine
should also permit the development of new agents that
model appeared to be mediated primarily by A, adeno-
are safer and more effective for the treatment of RA.
sine receptors, since an A, receptor antagonist com-
pletely reversed the antiinflammatory effects of MTX
(82). The role of A, receptors in the antiinflammatory ACKNOWLEDGMENT
effects of adenosine has not yet been explored in animal The author wishes to thank Dr. Susan M. Goodman for
models of inflammation. reviewing the manuscript.
Whether or not adenosine mediates the thera-
peutic effects of MTX, it is likely that some of the toxic
REFERENCES
effects of MTX may result from engagement of adeno-
sine receptors. One possible example of adenosine- 1. Gubner R, August S, Ginsberg V: Therapeutic suppression of
mediated MTX toxicity is accelerated rheumatoid nodu- tissue reactivity. 11. Effect of aminopterin in rheumatoid arthritis
and psoriasis. Am J Med Sci 221:176-182, 1951
losis, an increase in the number of rheumatoid nodules 2. Weinblatt ME, Fraser P: Elevated mean corpuscular volume as a
that occurs in MTX-treated patients with nodular RA, predictor of hematologic toxicity due to methotrexate therapy.
often despite good therapeutic responses. Merrill and Arthritis Rheum 32A.592-1596, 1989
3. Al-Awadhi A, Dale P, McKendry RJ: Pancytopenia associated
coworkers studied an in vitro model of rheumatoid with low dose methotrexate therapy: a regional survey. J Rheu-
nodule formation and found that MTX increases aden- matol 20:1121-1125, 1993
osine concentrations in supernatants of cultured mono- 4. Morgan SL, Baggott JE, Vaughn WH, Young PK, Austin JV,
Krumdieck CL, Alarc6n GS: The effect of folic acid supplemen-
nuclear cells (Merrill J, Schreibman D, Cronstein BN: tation on the toxicity of low-dose methotrexate in patients with
unpublished data) and enhances giant cell formation (a rheumatoid arthritis. Arthritis Rheum 33:9-18, 1990
surrogate for nodule formation in this in vitro model) by 5. Shiroky J, Neville C, Esdaile JM, Choquette D, Zummer M,
Hazeltine M, Bykerk V, Kanji M, St-Pierre A, Robidoux L,
a mechanism that is reversed by a specific A, adenosine Bourque L Low-dose methotrexate with leucovorin (folinic acid)
receptor antagonist and is mimicked by a specific aden- in the management of rheumatoid arthritis: results of a multi-
CRONSTEIN

center randomized. double-blind, placebo-controlled trial. arthritis beginning treatment with methotrexate. Arthritis Rheum
Arthritis Rheum 36:795-803, 1993 35:376-384, 1992
6. Morgan SL, Baggott JE, Vaughn WH, Austin JS, Veitch TA, Lee 23. Leroux JL, Damon M, Chavis C, Crastes de Paulet A, Blotman F:
JY, Koopman WJ, Krumdieck CL, Alarcon GS: Supplementation Effects of a single dose of methotrexate on 5- and 12-
with folic acid during methotrexate therapy for rheumatoid lipoxygenase products in patients with rheumatoid arthritis.
arthritis: a double-blind, placebo-controlled trial. Ann Intern J Rheumatol 19:863-866, 1992
Med 121333-841, 1994 24. Hawkes JS, Cleland LG, Proudman SM, James MJ: The effect of
7. Baggott JE, Morgan SL, Ha T, Alarcon GS, Koopman WJ, methotrexate on ex vivo lipoxygenase metabolism in neutrophils
Krumdieck C L Antifolates in rheumatoid arthritis: a hypotheti- from patients with rheumatoid arthritis. J Rheumatol 2155-58,
cal mechanism of action. Clin Eup Rheumatol 11 (suppl 8):SlOl- 1994
SlO5, 1993 25. Kremer JM, Lawrence DA, Jubiz W, DiGiacomo R, Rynes R,
8. Weinblatt ME, Coblyn JS, Fox DA, Fraser PA, Holdsworth DE, Bartholomew LE, Sherman M: Dietary fish oil and olive oil
Glass DN, Trentham D E Efficacy of low-dose methotrexate in supplementation in patients with rheumatoid arthritis: clinical
rheumatoid arthritis. N Engl J Med 312:818-822, 1985 and immunologic effects. Arthritis Rheum 33:810-820, 1990
9. Anderson PA, West SG, O’Dell JR, Via CS, Claypool RG, Kotzin 26. Kremer JM, Jubiz W, Michalek A, Rynes RI, Bartholomew LE,
BL: Weekly pulse methotrexate in rheumatoid arthritis: clinical Bigaouette J, Timchalk M, Beeler D, Lininger L Fish-oil fatty
and immunologic effects in a randomized double-blind study. acid supplementation in active rheumatoid arthritis: a double-
Ann Intern Med 103:489-496, 1985 blinded, controlled, crossover study. Ann Intern Med 106497-
10. Kremer JM, Phelps CT:Long-term prospective study of the use 503, 1987
of methotrexate in the treatment of rheumatoid arthritis: update 27. Kremer JM: The mechanism of action of methotrexate in rheu-
after a mean of 90 months. Arthritis Rheum 35:138-145, 1992 matoid arthritis: the search continues. J Rheumatol 21:l-5, 1994
11. Alarcon GS, Schrohenloher RE, Bartolucci AA, Ward JR, Wil- 28. Maini RN, Elliott MJ, Brennan FM, Williams RO, Chu CQ,
liams HJ, Koopman WJ: Suppression of rheumatoid factor Paleolog E, Charles PJ, Taylor PC, Feldmann M: Monoclonal
production by methotrexate in patients with rheumatoid arthritis: anti-TNFa antibody as a probe of pathogenesis and therapy of
evidence for differential influences of therapy and clinical status rheumatoid disease. Immunol Rev 144:195-223, 1995
on IgM and IgA rheumatoid factor expression. Arthritis Rheum 29. Hu SK, Mitcho YL, Oronsky AL, Kerwar SS: Studies on the
33:1155-1161, 1990 effect of methotrexate on macrophage function. J Rheumatol
12. Moore S, Ruska K, Peters L, Olsen NJ: Associations of IgA and 15:206-209, 1988
IgA-rheumatoid factor with disease features in patients with 30. Connolly KM, Stecher VJ, Dank E, Pruden DJ, LaBrie T:
rheumatoid arthritis. Immunol Invest 23:355-365, 1994 Alteration of interleukin-1 production and the acute phase
13. Olsen NJ, Teal GP, Brooks RH: IgM-rheumatoid factor and response following medication of adjuvant arthritic rats with
responses to second-line drugs in rheumatoid arthritis. Agents cyclosporin-A or methotrexate. Int J Immunopharmacol 10:717-
Actions 34:169-171. 1991 728, 1988
14, Olsen NJ, Murray LM: Antiproliferative effects of methotrexate 31. Chang DM, Baptiste P, Schur PH: The effect of antirheumatic
on peripheral blood mononuclear cells. Arthritis Rheum 32:378- drugs on interleukin 1 (IL-1) activity and IL-1 and IL-1 inhibitor
385, 1989 production by human monocytes. J Rheumatol 17:1148-1157,
15. Olsen NJ, Callahan LF, Pincus T: Immunologic studies of 1990
rheumatoid arthritis patients treated with methotrexate. Arthritis 32. Segal R, Mozes E, Yaron M, Tartakovsky B: The effects of
Rheum 30:481-488, 1987 methotrexate on the production and activity of interleukin-1.
16. Feldmann M, Brennan FM, Maini RN: Rheumatoid arthritis. Arthritis Rheum 32:370-377, 1989
Cell 85307-310, 1996 33. Meyer FA, Yaron I, Mashiah V, Yaron M: Methotrexate inhibits
17. Moreland LW, Pratt PW, Mayes MD, Postlethwaite A, Weisman proliferation but not interleukin 1 stimulated secretory activities
MH, Schnitzer T, Lightfoot R, Calabrese L, Zelinger DJ, Woody of cultured human synovial fibroblasts. J Rheumatol20:238-242,
JN, Koopman WJ: Double-blind, placebo-controlled multicenter 1993
trial using chimeric monoclonal anti-CD4 antibody, cM-T412, in 34. Brody M, Bohm I, Bauer R: Mechanism of action of methotrex-
rheumatoid arthritis patients receiving concomitant methotrex- ate: experimental evidence that methotrexate blocks the binding
of interleukin I p to the interleukin 1 receptor on target cells. Eur
ate. Arthritis Rheum 38:1581-1588, 1995
J Clin Chem Clin Biochem 31:667-674, 1993
18. Van der Lubbe PA, Dijkmans BAC, Markusse HM, Nassander
35. Furst DE: Practical clinical pharmacology and drug interactions
U, Breedveld FC: A randomized, double-blind, placebo-
of low-dose methotrexate therapy in rheumatoid arthritis. Br J
controlled study of CD4 monoclonal antibody therapy in early Rheumatol 34 (suppl 2):20-25, 1995
rheumatoid arthritis. Arthritis Rheum 38:1097-1106, 1995 36. Chang DM, Weinblatt ME, Schur PH: The effects of methotrex-
19. Weinblatt ME, Trentham DE, Fraser PA, Holdsworth DE, ate on interleukin 1 in patients with rheumatoid arthritis. J Rheu-
Falchuk KR, Weissman BN, Coblyn JS: Long-term prospective matol 19:1678-1682, 1992
trial of low-dose methotrexate in rheumatoid arthritis. Arthritis 37. Thomas R, Carroll GJ: Reduction of leukocyte and
Rheum 31:167-175, 1988 interleukin-lp concentrations in the synovial fluid of rheumatoid
20. Wascher TC, Hermann J, Brezinschek HP, Brezinschek R, arthritis patients treated with methotrexate. Arthritis Rheum
Wilders-Truschnig M, Rainer F, Krejs GJ: Cell-type specific 36: 1244-1252, 1993
response of peripheral blood lymphocytes to methotrexate in the 38. Williams AS, Punn YL, Amos N, Cooper AM, Williams BD: The
treatment of rheumatoid arthritis. Clin Invest 72535-540, 1994 effect of lipsomally conlugated methotrexate upon mediator
21. Sperling RI, Coblyn JS, Larkin JK, Benincaso A!, Austen KF, release from human peripheral blood monocytes. Br J Rheumn-
Weinblatt ME: Inhibition of leukotriene B, synthesis in neutro- to1 34:241-245, 1995
phils from patients with rheumatoid arthritis by a single oral dose 39. Williams AS. Camilleri JP, Topley N, Williams BD: Prostaglan-
of rnethotrexate. Arthritis Rheum 33:1149-1155, 1990 din and tumor necrosis factor secretion by peritoneal macro-
22. Sperling RI, Benincaso AI, Anderson RJ, Coblyn JS, Austen KF, phages isolated from normal and arthritic rats treated with
Weinblatt M E Acute and chronic suppression of leukotriene B, liposomal methotrexate. J Pharmacol Toxicol Methods 325368,
synthesis ex vivo in neutrophils from patients with rheumatoid 1994
MECHANISM OF ACTION OF MTX 1959

40. Williams AS, Topley N, Amos N, Williams BD: Effect of three affinity and functional activity of chemotactic factor receptors on
lipophilic methotrexate derivatives upon mediator release by macrophages. Cell 28:107-114, 1982
lipopolysaccharide-stimulated rat peritoneal macrophages. 56. Pike MC, Kredich NM, Snyderman R: Requirement of
J Pharm Pharmacol 46:291-295, 1994 S-adenosyl-L-methionine-mediatedmethylation for human
41. Williams AS, Topley N, Williams BD: Effect of liposomally monocyte chemotaxis. Proc Natl Acad Sci U S A 75:3928-3932,
encapsulated MTX-DMPE conjugates upon TNFa and PGE, 1978
release by lipopolysaccharide stimulated rat peritoneal macro- 57. Johnston JM, Kredich NM: Inhibition of methylation by adeno-
phages. Biochim Biophys Acta 1225217-222, 1994 sine in adenosine deaminase-inhibited, phytohemagglutinin-
42. Smith-Oliver T, Noel LS, Stimpson SS, Yarnall DP, Connolly stimulated human lymphocytes. J Immunol 123:97-103, 1979
KM: Elevated levels of TNF in the joints of adjuvant arthritic 58. Furumitsu Y, Yukioka K, Kojima A, Yukioka M, Shichikawa K,
rats. Cytokine 5:298-304, 1993 Ochi T, Matsui-Yuasa I, Otani S, Nishizawa Y, Morii H: Levels
43. Seitz M, Loetscher P, Dewald B, Towbin H, Rordorf C, Gallati of urinary polyamines in patients with rheumatoid arthritis.
H, Baggiolini M, Gerber NJ: Methotrexate action in rheumatoid J Rheumatol 20:1661-1665, 1993
arthritis: stimulation of cytokine inhibitor and inhibition of 59. Yukioka K, Wakitani S, Yukioka M, Furumitsu Y, Shichikawa K,
chemokine production by peripheral blood mononuclear cells. Ochi T, Goto H, Matsui-Yuasa I, Otani S, Nishizawa Y, Morii H:
Br J Rheumatol 34:602-609, 1995 Polyamine levels in synovial tissues and synovial fluids of patients
44. Barrera P, Haagsma CJ,Boerbooms AMT, van Riel PLC, Born with rheumatoid arthritis. J Rheumatol 19:689-692, 1992
GF, van de Putte LBA, van der Meer JWM: Effect of metho- 60. Flescher E, Bowlin TL, Talal N: Regulation of IL-2 production by
trexate alone or in combination with sulphasalazine on the mononuclear cells from rheumatoid arthritis synovial fluids. Clin
production and circulating concentrations of cytokines and their Exp Immunol 87:435-437, 1992
antagonists: longitudinal evaluation in patients with rheumatoid 61. Flescher E, Bowlin TL, Ballester A, Houk R, Talal N: Increased
arthritis. Br J Rheumatol 34:747-755, 1995 polyamines may downregulate interleukin 2 production in rheu-
45. Barrera P, Boerbooms AMT, Janssen EM, Sauenvein RW, matoid arthritis. J Clin Invest 83:1356-1362, 1989
Gallati H, Mulder J, de Boo T, Demacker PNM, van de Putte 62. Miyasaka N, Nakamura T, Russell IJ, Talal N: Interleukin 2
LBA, van der Meer JWM: Circulating soluble tumor necrosis deficiencies in rheumatoid arthritis and systemic lupus erythem-
factor receptors, interleukin-2 receptors, tumor necrosis factor a, atosus. Clin Immunol Imrnunopathol 31:109-117, 1984
and interleukin-6 levels in rheumatoid arthritis: longitudinal 63. Miyasaka N, Murota N, Yamaoka K, Sat0 K, Yamada T, Nishido
evaluation during methotrexate and azathioprine therapy. Arthri- T, Okumura K Interleukin 2 defect in the peripheral blood and
tis Rheum 36:1070-1079, 1993 the lung in patients with Sjogren’s syndrome. Clin Exp Immunol
46. Crilly A, McInness IB, McDonald AG, Watson J, Capell HA, 65~497-505, 1986
Madhok R: Interleukin 6 (IL-6) and soluble IL-2 receptor levels 64. Nesher G. Moore T L The in vitro effects of methotrexate on
in patients with rheumatoid arthritis treated with low dose oral peripheral blood mononuclear cells: modulation by methyl do-
methotrexate. J Rheumatol 22:224-226, 1995 nors and spermidine. Arthritis Rheum 33:954-959, 1990
47. Lacki JK, Mama K, Mackiewicz SH, Mackiewicz U, Muller W: 65. Nesher G, Moore TL, Dorner RW: In vitro effects of methotrex-
Circulating interleukin-10 and interleukin-6 serum levels in rheu- ate on peripheral blood monocytes: modulation by folinic acid
matoid arthritis patients treated with methotrexate or gold salts: and S-adenosylmethionine. Ann Rheum Dis 50:637-641, 1991
preliminary report. Inflamm Res 44:24-26, 1995 66. Krenitsky TA, Rideout JL, Chao EY, Koszalka GW, Gurney F,
48. Wascher TC, Hermann J, Brezinschek R, Brezinschek HP, Crouch RC, Cohn NK, Wolberg G, Vinegar R: Imidazo[4,5-c]-
Wilders-Truschnig M, Rainer F, Krejs GJ: Serum levels of pyridines (3-deazapurines) and their nucleosides as immunosup-
interleukin-6 and tumour-necrosis-factor-alpha are not corre- pressive and antiinflammatory agents. J Med Chem 29:138-143,
lated to disease activity in patients with rheumatoid arthritis after 1986
treatment with low-dose methotrexate. Eur J Clin Invest 24:73- 67. Jurgensen CH, Huber BE, Zimmerman TP, Wolberg G:
75, 1994 3-deazaadenosine inhibits leukocyte adhesion and ICAM-1 bio-
49. Seitz M, Dewald B, Ceska M, Gerber N, Baggiolini M: synthesis in tumor necrosis factor-stimulated human endothelial
Interleukin-8 in inflammatory rheumatic diseases: synovial fluid cells. J Immunol 144:65>661, 1990
levels, relation to rheumatoid factors, production by mono- 68. Jurgensen CH, Wolberg G, Zimmerman T P Inhibition of neu-
nuclear cells, and effects of gold sodium thiomalate and metho- trophil adherence to endothelial cells by 3-deazaadenosine.
trexate. Rheumatol Int 12:159-164, 1992 Agents Actions 27:398-400, 1989
50. Loetscher P, Dewald B, Baggiolini M, Seitz M: Monocyte che- 69. Fantone JC, Duque RE, Davis BH, Phan SH: 3-deazaadenosine
moattractant protein 1 and interleukin 8 production by rheuma- inhibition of stimulus-response coupling in human polymorpho-
toid synoviocytes: effects of anti-rheumatic drugs. Cytokine nuclear leukocytes. J Leukoc Biol 45:121-128, 1989
6:162-170, 1994 70. Leonard W, Skeel A, Chiang PK, Cantoni G L The action of the
51. Hershfield MS, Krodich NM: S-adenosylhomocysteine hydrolase adenosylhomocysteine hydrolase inhibitor, 3-deazaadenosine, on
is an adenosine-binding protein: a target for adenosine toxicity. phagocytic function of mouse macrophages and human mono-
Science 202:757-760, 1978 cytes. Biochem Biophys Res Commun 84:102-109, 1978
52. Kredich NM, Martin DV Jr: Role of S-adenosylhomocysteine in 71. Zimmerman TP, Wolberg G, Duncan GS: Inhibition of
adenosine mediated toxicity in cultured mouse T lymphoma cells. lymphocyte-mediated cytolysis by 3-deazaadenosine: evidence for
Cell 12:931-938, 1977 a methylation reaction essential for cytolysis. Proc Natl Acad Sci
53. Hershfield MS, Kurtzberg J, Aiyar VN, Suh EJ, Schiff R: U S A 7516220-6224, 1978
Abnormalities in S-adenosylhomocysteine hydrolysis, ATP catab- 72. Smith DM, Johnson JA, Turner RA: Biochemical perturbations
olism, and lymphoid differentiation in adenosine deaminase of BW 91Y (3-deazaadenosine) on human neutrophil chemotac-
deficiency. Ann N Y Acad Sci 451:78-86, 1985 tic potential and lipid metabolism. Int J Tissue React 13:l-18,
54. Palella TD, Schatz RA, Wilens TE, Fox IH: S- 1991
Adenosylhomocysteine accumulation and selective cytotoxicity in 73. Sung SSJ, Silverstein SC: Inhibition of macrophage phagocytosis
cultured T- and B-lymphocytes. J Lab Clin Med 100:269-278, by methylation inhibitors: lack of correlation of protein car-
1982 boxymethylation and phospholipid methylation with phagocyto-
55. Pike MC, Snyderman R: Transmethylation reactions regulate sis. J Biol Chem 260:546-554, 1985
1960 CRONSTEIN

74. Stopford CR, Wolberg G, Prus KL, Reynolds-Vaughn R, Zim- R: Adenosine: an endogenous inhibitor of neutrophil-mediated
merman TP: 3-Deazaadenosine-induced disorganization of mac- I injury to endothelial cells. J Clin Invest 78:760-770, 1986
rophage microfilaments. Proc Natl Acad Sci U S A 82:4060-4064, 93. Cronstein BN, Kramer SB, Weissmann G, Hirschhorn R: Aden-
1985 osine: a physiological modulator of superoxide anion generation
75. Zimmerman TP, Iannone M, Wolberg G: 3-Deazaadenosine: by human neutrophils. J Exp Med 158:1160-1177, 1983
S-adenosylhomocysteine hydrolase-independent mechanism of 94. Cronstein BN, Duguma L, Nicholls D. Hutchison A, Williams M:
action in mouse lymphocytes. J Biol Chem 259:1122-1126, 1984 The adenosine/neutrophil paradox resolved: human neutrophils
76. Chabner BA, Allegra CJ, Curt G.4, Clendeninn NJ, Baram J, possess both A1 and A2 receptors which promote chemotaxis and
Koizumi S, Drake JC, Jolivet J: Polyglutamation of methotrexate: inhibit 0, generation, respectively. J Clin Invest 85:1150-1157,
is methotrexate a prodrug? J Clin Invest 76:907-912, 1985 1990
77. Allegra CJ, Drake JC, Jolivet J, Chabner BA: Inhibition of 95. Salmon JE, Cronstein BN: Fcy receptor-mediated functions in
phosphoribosylaminoimidazolecarboxamide transformylase by neutrophils are modulated by adenosine receptor occupancy: A1
methotrexate and dihydrofolic acid polyglutamates. Proc Natl receptors are stimulatory and A2 receptors are inhibitory. J Im-
Acad Sci U S A 82:4881-4885, 1985 munol 145:2235-2240, 1990
78. Baggott JE, Vaughn WH, Hudson BB: Inhibition of 5- 96. Cronstein BN, Levin RI, Philips MR, Hirschhorn R, Abramson
aminoimidazole-4-carboxamide ribotide transformylase, adeno- SB, Weissmann G: Neutrophil adherence to endothelium is
sine deaminase and 5'-adenylate deaminase by polyglutamates of enhanced via adenosine A1 receptors and inhibited via adenosine
A2 receptors. J Immunol 148:2201-2206, 1992
methotrexate and oxidized folates and by 5-aminoimidazole-4-
carboxamide riboside and ribotide. Biochem J 236:193-200, 1986 97. Kammer GM, Birch RE, Polmar SH: Impaired immunoregula-
tion in systemic lupus erythematosus: defective adenosine-
79. Ha T, Morgan SL, Vaughan WH, Baggott JE: Inhibition of
induced suppressor T lymphocyte generation. J Immunol 130:
adenosine deaminase and S-adenosyl homocysteine hydrolase by 1706-1712, 1983
5-aminoimidazole-4-carboxamide riboside. FASEB J 6:1210- 98. Schultz LA?Kammer GM, Rudolph SA: Characterization of the
1215, 1992 human T lymphocyte adenosine receptor: comparison of normal
80. Luhby AL, Cooperman JH: Aminoimidazole carboxamide excre- and systemic lupus erythematosus cells. FASEB J 2244-250,
tion in vitamin B12 and folk acid deficiencies. Lancet 2:1381- 1988
1382, 1962 99. Mandler R, Birch RE, Polmar SH, Kammer GM, Rudolph SA:
81. Cronstein BN, Eberle MA, Gruber HE, Levin RI: Methotrexate Abnormal adenosine-induced immunosuppression and CAMP
inhibits neutrophil function by stimulating adenosine release metabolism in T lymphocytes of patients with systemic lupus
from connective tissue cells. Proc Natl Acad Sci U S A 88:244- erythematosus. Proc Natl Acad Sci U S A 79:7542-7546, 1982
2445, 1991 100. Kammer GM, Rudolph SA: Regulation of human T lymphocyte
82. Cronstein BN, Naime D, Ostad E: The antiinflammatory mech- surface antigen mobility by purinergic receptors. J Immunol
anism of methotrexate: increased adenosine release at inflamed 133:3298-3302, 1984
sites diminishes leukocyte accumulation in an in vivo model of 101. Kammer GM, Smith JA, Mitchell R: Capping of human T-cell
inflammation. J Clin Invest 92:2675-2682, 1993 specific determinants: kinetics of capping and receptor re-
83. Asako H, Kubes P, Baethge B, Wolf R, Granger DN: Colchicine expression and regulation by the cytoskeleton. J Immunol 130:
and methotrexate reduce leukocyte adherence and emigration in 38-44, 1983
rat mesenteric venules. Inflammation 16:45-56, 1992 102. Dong RP; Kameoka J, Hegen M, Tanaka T, Xu Y, Schlossman
84. Asako H, Wolf RE, Granger DN: Leukocyte adherence in rat SF, Morimoto C: Characterization of adenosine deaminase bind-
mesenteric venules: effects of adenosine and methotrexate. Gas- ing to human CD26 on T cells and its biologic role in immune
troenterology 104:31-37, 1993 response. J Immunol 156:1349-1355, 1996
85. Firestein GS, Boyle D, Bullough DA, Gruber HE, Sajjadi FG, 103. Parmely MJ, Zhou W-W, Edwards CK 111, Borcherding DR,
Montag A, Sambol B, Mullane K Protective effect of an aden- Silverstein R, Morrison DC: Adenosine and a related carbocyclic
osine kinase inhibitor in septic shock. J Immunol 1525853-5859, nucleoside analogue selectively inhibit tumor necrosis factor-
1994 alpha production and protect mice against endotoxin challenge.
86. Firestein GS, Bullough DA, Erion MD, Jimenez R, Ramirez- J Immunol 151389-396, 1993
Weinhouse M, Barankiewicz J, Smith CW, Gruber HE, Mullane 104. Bouma MG, Stad RK, van den Wildenberg FAJM, Buurman
KM: Inhibition of neutrophil adhesion by adenosine and an WA: Differential regulatory effects of adenosine on cytokine
adenosine hnase inhibitor: the role of selectins. J Immunol release by activated human monocytes. J Immunol 153:4159-
154:326-334, 1995 4168, 1994
87. Rosengren S, Bong GW, Firestein GS: Anti-inflammatory effects 105. Sajjadi FG, Takabayashi K, Foster AC, Domingo RC, Firestein
of an adenosine kinase inhibitor: decreased neutrophil accumu- GS: Inhibition of TNFa expression by adenosine: role of A3
lation and vascular leakage. J Immunol 1545444-5451, 1995 adenosine receptors. J Immunol 156:3435-3442, 1996
88. Cronstein BN, Naime D, Firestein GS: The antiinflammatory 106. Le Moine 0, Stordeur P. SchandenC L, Marchant A, de Groote
effects of an adenosine kinase inhibitor are mediated by adeno- D, Goldman M, Devitre J: Adenosine enhances IL-10 secretion
sine. Arthritis Rheum 38:1040-1045, 1995 by human monocytes. J Immunol 156:4408-4414, 1996
89. Gadangi P, Longaker M, Naime D, Levin RI, Recht PA, Mon- 107. Firestein GS, Paine MM, Boyle DL: Mechanisms of methotrex-
tesinos MC, Buckley MT, Carlin G, Cronstein BN: The antiin- ate action in rheumatoid arthritis: selective decrease in synovial
flammatory mechanism of sulfasalazine is related to adenosine collagenase gene expression. Arthritis Rheum 37:193-200, 1994
release at inflamed sites. J Immunol 156:1937-1941, 1996 108. Boyle DL, Sajjadi FG, Firestein GS: Inhibition of synoviocyte
90. Baggott JE, Morgan SL, Ha T, Vaughn WH, Hine RJ: Inhibition collagenase gene expression by adenosine receptor stimulation.
of folate-dependent enzymes by non-steroidal anti-inflammatory Arthritis Rheum 39:92>930, 1996
drugs. Biochem J 282:197-202, 1992 109. Merrill JT, Coffey D, Shen C , Zakharenko 0, Zhang HW,Ldhita
91. Bernini JC, Fort DW, Griener JC, Kane BJ, Chappell WB, RG, Cronstein BN: Mechanisms of rheumatoid nodulosis:
Kamen BA: Arninophylline for methotrexate-induced neurotoxi- methotrexate-enhanced monocyte fusion requires protein synthe-
city. Lancet 345544547, 1995 sis and intact microtubules (abstract). Arthritis Rheum 38 (suppl
92. Cronstein BN, Levin RI, Belanoff J, Weissmann G, Hirschhorn 9):S157, 1995

Vous aimerez peut-être aussi