THE GENERATION of hematopoietic cells requires the coordinated response to a plethora of stimulatory and inhibitory signals that cells receive from their extracellular environment. The “positive” regulators are relatively well defined: the molecules that stimulate the proliferation, differentiation, and survival of hematopoietic cells have been extensively studied. These include the colony-stimulating factors (CSFs) and the majority of the interleukins (ILs). Several of these molecules are available for clinical use, and much is known about their structure, the consequences of their over-production, and the biological effects that result from their complete absence. The multi-protein receptor complexes that are used by these molecules at the cell surface have been characterized to reveal a complicated interplay of shared receptor components and unique receptor elements. More recently the intracellular pathways that are triggered by growth factor/receptor interactions have begun to be dissected, revealing an interdigitating network of signaling molecules that is striking both in terms of its complexity, and in terms of the recurring themes that are revealed in otherwise apparently divergent experimental systems.1 By comparison, the “negative” regulators of hematopoiesis have been relatively neglected. However, it has been clear for a number of years that molecules like transforming growth factor-β (TGF-β) are potent negative regulators of hematopoiesis and function as important molecules in determining hematopoietic responses.

The identification of the JAK/STAT signaling pathway provided an understanding of one mechanism by which stimulatory signals received at the cell surface are rapidly transmitted to the nucleus. The JAKs (or Janus kinases) are receptor-associated molecules that are phosphorylated on tyrosine in response to cytokine-receptor interactions. As a consequence, the STAT (for signal transducers and activators of transcription) molecules are recruited to the receptor and phosphorylated.2,3 This leads to their dimerization and translocation to the nucleus where they bind and activate transcription of target genes. The recruitment of particular JAK/STAT family members overlaps but differs for different cytokines. Thus, for example, IL-6 type cytokines use JAK1, JAK2, and the related molecule TYK2, and STAT1 and STAT3,4,5 although the critical molecules appear to be JAK1 and STAT3.6,7 In comparison, the IL-12 signaling pathway results in phosphorylation of JAK2 and TYK28,9 and requires STAT4 for IL-12–generated responses.10,11 

The mechanism by which “negative” regulators exert their inhibitory action is less clear. One possible mechanism involves the recruitment of protein tyrosine phosphatases that can then serve to inactivate JAK proteins. One such is SHP-1, the defective function of which results in the hyperproliferation and accumulation of several hematopoietic cell lineages and in the development of autoimmune disease as evidenced by the motheaten mouse.12Specific targeting of this molecule to the erythropoietin receptor can inhibit ligand-stimulated tyrosine phosphorylation and result in dephosphorylation of JAK2.13 The negative regulator TGF-β has been reported to activate protein tyrosine phosphatases14 and this may be one mechanism by which it exerts its inhibitory effect on the JAK/STAT pathway.15 

Another family of signaling molecules that are potentially important mediators of inhibitory signals have recently been described. There appear to be at least 8 of these “SOCS” proteins (for suppressors of cytokine signaling) including CIS, an early response gene, that encode SH2-domain containing proteins16-19 and 12 others that also share a C-terminal domain called the SOCS box.17These proteins were identified simultaneously using three different approaches. One involved a functional screen for cDNAs encoding proteins that blocked IL-6 function.20 A second approach involved a yeast two-hybrid strategy for proteins interacting with the kinase domain of JAK2.21 The third searched for proteins with antigen similarity to the SH2 domain of the STAT molecules.22 Cytokines induce the expression of the SOCS genes, and the SOCS proteins then serve to downregulate the JAK/STAT pathways and therefore curtail the biological response. Thus, for example, SOCS1 (also known as JAB and SSI-1) is induced within 20 minutes after stimulation by IL-6 and suppresses the cellular response to IL-6 via its interaction with activated JAK proteins through its SH2 domain.23 Levels of SOCS1 expression return to baseline within about 4 hours. The transcriptional activation of SOCS genes is mediated, at least in part by the STAT proteins; CIS expression is modulated by STAT5 and STAT3 is important for SOCS1 expression.24 SOCS1 also attenuates the biological response to a number of other cytokines including leukemia inhibitory factor, oncostatin-M, interferon-γ (IFN-γ), thrombopoietin, and growth hormone.20,25 Moreover, as well as acting to reduce phosphorylation of JAK kinases and STAT proteins, SOCS proteins may function as more general inhibitory regulators.26,27Therefore, these proteins are induced by cytokines, and act to suppress cytokine signaling, acting in a classical negative feedback loop.

One situation in which a complex array of competing “positive” and “negative” regulators is clearly evident, is in the generation of cells important in antigen-specific or cell-mediated immune responses. Studies of T-lymphocyte clones have identified two major subpopulations of CD4+ cells, T-helper lymphocyte (TH) 1 induced by IL-12 and IFN-γ and TH2 cells induced by IL-4.28-30 The development of the TH1 versus TH2 phenotype is dependent on cell-intrinsic as well as extrinsic stimuli.31 These cells differ in the cytokines that they produce after activation. Thus, a cellular immune response dominated by a TH1 response is characterized by cells that produce IFN-γ, IL-2, and TNF-β. Conversely, a TH2 response is typified by the production of IL-4, IL-5, IL-6, IL-13, and IL-10 (Fig 1). Both TH1 and TH2 cells produce granulocyte-macrophage CSF (GM-CSF), tumor necrosis factor-α (TNF-α), and IL-3; however, the relative levels may vary. These patterns of cytokine expression can be used to categorize most murine CD4+ T-lymphocyte clones, and as such, allow classification in terms of the principal immune response induced in experimental systems.32 This may also be relevant to some human disease states.33 As expected from the differing cytokine expression patterns, these subsets of cells also differ in their functional properties, with TH1 cells being predominant in assisting microbiocidal macrophage responses (via IFN-γ and TNF-β) while TH2 cytokines augment B-cell responses. This divergence in function is particularly evident during infection with the protozoa, Leishmania, where macrophage activation is required for protection from this intracellular parasite.34 In this system TH1-responses are protective while TH2 responses fail to offer protection and lead to a nonhealing of disease. In BALB/c mice, a bias toward a TH2 response, with production of IL-4 and the requirement for STAT6-mediated signaling,35 results in the genetic susceptibility toLeishmania infection.

Fig. 1.

(A) Cytokines controlling the development of cell-mediated (TH1) and humoral (TH2) responses. Certain infections such as viruses induce the production of IL-12 and IFN-γ by antigen presenting cells (APC). These factors promote the differentiation of TH0 cells to the TH1 phenotype. Other infections such as helminths induce the production of IL-4 (by eosinophils) which induces differentiation to the TH2 phenotype. TH1 cytokines such as IFN-γ inhibit the production of IL-4 and IL-10 while TH2 cytokines such as IL-10 inhibit the production and action of IL-12. (Part B) Factors such as IL-12 and IFN-γ activate a cascade of intracellular signaling molecules including molecules in the JAK/STAT pathway. In contrast, inhibitory molecules may produce their inhibitory effects in part via the activation of molecules such as phosphatases in the case of TGF-β, and SOCS genes in the case of IL-10. These pathways would serve to attenuate a biological response.

Fig. 1.

(A) Cytokines controlling the development of cell-mediated (TH1) and humoral (TH2) responses. Certain infections such as viruses induce the production of IL-12 and IFN-γ by antigen presenting cells (APC). These factors promote the differentiation of TH0 cells to the TH1 phenotype. Other infections such as helminths induce the production of IL-4 (by eosinophils) which induces differentiation to the TH2 phenotype. TH1 cytokines such as IFN-γ inhibit the production of IL-4 and IL-10 while TH2 cytokines such as IL-10 inhibit the production and action of IL-12. (Part B) Factors such as IL-12 and IFN-γ activate a cascade of intracellular signaling molecules including molecules in the JAK/STAT pathway. In contrast, inhibitory molecules may produce their inhibitory effects in part via the activation of molecules such as phosphatases in the case of TGF-β, and SOCS genes in the case of IL-10. These pathways would serve to attenuate a biological response.

Close modal

In this context, IL-12 and IFN-γ are important, proinflammatory cytokines that act as a powerful inducers of a TH1 response.36,37 IL-12 is also active on natural killer (NK) cells. IL-12 is produced by antigen-presenting cells (APC) as a result of an interaction, via CD40 and its ligand, between the APC and activated T lymphocytes.38,39 In this incestuous relationship, the production of IL-12 by APC is further stimulated by the TH1 cells and their product IFN-γ and vice versa.40,41 Conversely, a defect in the production of IL-12 and IFN-γ may be important in impaired TH1-mediated immune responses,42,43 with evidence that IL-12 and IFN-γ production, and the IL-12 responsiveness of TH1 cells, can be antagonized by TH2 cells and the cytokines that they produce (IL-10, IL-4, IL-13).44,45 

The target cell for the action of IL-12, the activated T cell, is also subject to additional opposing, inhibitory cytokine influences. TGF-β is an immunosuppressive molecule that, in addition to its inhibitory action on lymphoid cells, inhibits proliferation, differentiation, and functional activity of myeloid cells and NK cells. TGF-β can interfere with IL-2–mediated tyrosine phosphorylation and activation of JAK1 and STAT515 and with multiple components of the IL-5 signaling pathway including an inhibitory effect on JAK2 and STAT1.46 In the report in this issue by Pardoux et al,47 the mechanism by which the inhibitory effect of TGF-β is able to oppose the stimulatory action of IL-12 on activated T cells is addressed: how does an activated T-cell resolve these conflicting signals?

As outlined above, IL-10 (along with IL-4 and IL-13) is an anti-inflammatory cytokine produced by TH2 cells.48,49 It serves to maintain a TH2-type immune response in part by preventing the development of TH1 cells, thereby preventing production of the macrophage activating molecules such as IFN-γ. IL-10 is also produced by monocytes in response to lipopolysaccharides. In addition to its action on lymphoid cells, IL-10 acts as a direct inhibitor of macrophages, inhibiting gene transcription and inhibiting production of inflammatory cytokines.50-52 Thus, as well as being a product of TH2 cells, IL-10 has an auto-regulatory role in monocytes53 and its production by monocytes is inhibited by the proinflammatory molecule IFN-γ.54 What are the mechanisms by which the opposing influences of IL-10 and IFN-γ are reconciled within the monocyte? In the accompanying report by Ito et al55 these authors examine the mechanisms by which the action of IL-10 and IFNs are balanced.

Pardoux et al have taken advantage of their recent demonstration that TGF-β inhibited the development of cytotoxic and proliferative allogeneic (TH1) responses by a mechanism that involved decreased production of IL-12.56 They further observed that TGF-β–mediated inhibition of alloreactive T cells was not overcome by the addition of exogenous IL-12. This was confirmed in their report in this issue by the demonstration that addition of IL-12 was not able to completely overcome the inhibitory effect of TGF-β on IFN-γ production by these cells. They reasoned that this failure implied that TGF-β also interfered either with IL-12 receptor expression on the activated T cell, or with the signal transduction pathway initiated by IL-12 receptor. They showed that the former was not the case; there was no consistent difference in IL-12 receptor expression in response to TGF-β. The addition of TGF-β did not alter the levels of JAK2 and Tyk2, two intracellular signaling molecules normally phosphorylated in response to IL-12; however, there was a significant decrease in the tyrosine phosphorylation of both molecules within 20 minutes of incubation with TGF-β. A consequence of activation of JAKs is the phosphorylation of STAT molecules, with STAT4 being activated in response to IL-12 signaling. Again there was no decrease in the level of STAT4 protein but tyrosine phosphorylation and DNA binding activity of STAT4 were decreased in response to TGF-β within 20 minutes. Thus, these results suggest that TGF-β directly interferes with intracellular signaling by IL-12 and the authors suggest that this may be mediated by TGF-β–induced tyrosine phosphatases (Fig 1).

The results presented by Ito et al55 are remarkably similar, but they provided additional evidence that may further explain the mechanism by which interference at the level of intracellular signaling occurs. The authors showed that IL-10 inhibited the transcription of some genes normally induced in human monocytes in response to stimulation with IFN-γ and IFN-α. Consistent with the studies by Pardoux et al, there was no change in number or affinity of IFN-γ receptors in response to treatment with IL-10. Similarly, IL-10 did not alter the levels of STAT1, the principal target for IFN-γ signaling in these cells, but decreased the phosphorylation of STAT1 and decreased DNA binding activity. These inhibitory effects of IL-10 were more evident when lower concentrations of IFN were examined. There was no evidence that this effect of IL-10 was due to induction of phosphatase activity. However, this study presents a result that potentially could explain the observations reported in both papers. These authors showed that treatment of monocytes with IL-10 induced the expression of the SOCS3 gene within 30 minutes and maintained its expression for at least 120 minutes. These kinetics are consistent with the observed action of IL-10 to inhibit IFN-induced STAT1 activation and IFN-stimulated gene expression. Although studies have shown that in myeloid and fibroblast cell lines SOCS1 but not SOCS3 was an inhibitor of IFN-γ signaling,57,58 more recent results have also implicated SOCS3 as an inhibitor of IFN signaling.59 It will be important to determine whether other SOCS proteins are induced by IL-10.

Together, these results suggest that one mechanism by which inhibitory cytokines exert their effect is via the direct activation of inhibitory signaling pathways like phosphatases and SOCS proteins. The SOCS pathway is a potential candidate for mediating this type of response because SOCS proteins are normally activated in response to positive cytokine signals, thus ensuring that a biological response is transient. Direct access to these same signaling molecules by inhibitory cytokines provides an attractive and efficient mechanism whereby competing signals are resolved to redirect a cellular response. It seems possible that this might prove a common mechanism for cells faced with alternate choices and competing extracellular demands.

1
Nicola
NA
Hilton
DJ
General classes and functions of four-helix bundle cytokines.
Adv Pr Chem
52
1998
2
2
Leonard
WJ
O’Shea
JJ
Jaks and STATs: Biological implications.
Annu Rev Immunol
16
1998
293
3
Liu
KD
Gaffen
SL
Goldsmith
MA
JAK/STAT signaling by cytokine receptors.
Curr Opin Immunol
10
1998
271
4
Heinrich
PC
Behrmann
I
Muller-Newen
G
Schaper
F
Graeve
L
Interleukin-6-type cytokine signalling through the gp130/Jak/STAT pathway.
Biochem J
334
1998
297
5
Nakashima
K
Taga
T
gp130 and the IL-6 family of cytokines: Signaling mechanisms and thrombopoietic activities.
Semin Hematol
35
1998
210
6
Minami
M
Inoue
M
Wei
S
Takeda
K
Matsumoto
M
Kishimoto
T
Akira
S
STAT3 activation is a critical step in gp130-mediated terminal differentiation and growth arrest of a myeloid cell line.
Proc Natl Acad Sci USA
93
1996
3963
7
Rodic
SJ
Meraz
MA
White
JM
Lampe
PA
Riley
JK
Arthur
CD
King
KL
Sheehan
KC
Yin
L
Pennica
D
Johnson
EM
Schreiber
RD
Disruption of the Jak1 gene demonstrates obligatory and nonredundant roles of the Jaks in cytokine-induced biologic responses.
Cell
93
1998
373
8
Bacon
CM
McVicar
DW
Ortaldo
JR
Rees
RC
O’Shea
JJ
Johnston
JA
Interleukin 12 (IL-12) induces tyrosine phosphorylation of JAK2 and TYK2: Differential use of Janus family tyrosine kinases by IL-2 and IL-12.
J Exp Med
181
1995
399
9
Zou
J
Presky
DH
Wu
CY
Gubler
U
Differential associations between the cytoplasmic regions of the interleukin-12 receptor subunits beta1 and beta2 and JAK kinases.
J Biol Chem
272
1997
6073
10
Thierfelder
WE
van Deursen
JM
Yamamoto
K
Tripp
RA
Sarawar
SR
Carson
RT
Sangster
MY
Vignali
DA
Doherty
PC
Grosveld
GC
Ihle
JN
Requirement for Stat4 in interleukin-12–mediated responses of natural killer and T cells.
Nature
382
1996
171
11
Kaplan
MH
Sun
YL
Hoey
T
Grusby
MJ
Impaired IL-12 responses and enhanced development of Th2 cells in Stat4-deficient mice.
Nature
382
1996
174
12
Schultz
LD
Rajan
TV
Greiner
DL
Severe defects in immunity and hematopoiesis caused by SHP-1 protein-tyrosine-phosphatase deficiency.
Trends Biotechnol
15
1997
302
13
Klingmuller
U
Lorenz
U
Cantley
LC
Neel
BG
Lodish
HF
Specific recruitment of SH-PTP1 to the erythropoietin receptor causes inactivation of JAK2 and termination of proliferative signals.
Cell
80
1995
729
14
Gruppuso
PA
Mikumo
R
Brautigan
DL
Braun
L
Growth arrest induced by transforming growth factor beta 1 is accompanied by protein phosphatase activation in human keratinocytes.
J Biol Chem
266
1991
3444
15
Bright
JJ
Kerr
LD
Sriram
S
TGF-beta inhibits IL-2-induced tyrosine phosphorylation and activation of Jak-1 and Stat 5 in T lymphocytes.
J Immunol
159
1997
175
16
Yoshimura
A
Ohkubo
T
Kiguchi
T
Jenkins
NA
Gilbert
DJ
Copeland
NG
Hara
T
Miyajima
A
A novel cytokine-inducible gene CIS encodes an SH2-containing protein that binds to tyrosine-phosphorylated interleukin 3 and erythropoietin receptors.
EMBO J
14
1995
2816
17
Hilton
DJ
Richardson
RT
Alexander
WS
Viney
EM
Willson
TA
Sprigg
NS
Starr
R
Nicholson
SE
Metcalf
D
Nicola
NA
Twenty proteins containing a C-terminal SOCS box form five structural classes.
Proc Natl Acad Sci USA
95
1998
114
18
Masuhara
M
Sakamoto
H
Matsumoto
A
Suzuki
R
Yasukawa
H
Mitsui
K
Wakioka
T
Tanimura
S
Sasaki
A
Misawa
H
Yokouchi
M
Ohtsubo
M
Yoshimura
A
Cloning and characterization of novel CIS family genes.
Biochem Biophys Res Commun
239
1997
439
19
Minamoto
S
Ikegame
K
Ueno
K
Narazaki
M
Naka
T
Yamamoto
H
Matsumoto
T
Saito
H
Hosoe
S
Kishimoto
T
Cloning and functional analysis of new members of STAT induced inhibitor (SSI) family: SSI-2 and SSI-3.
Biochem Biophys Res Commun
237
1997
79
20
Starr
R
Willson
TA
Viney
EM
Murray
LJ
Rayner
JR
Jenkins
BJ
Gonda
TJ
Alexander
WS
Metcalf
D
Nicola
NA
Hilton
DJ
A family of cytokine-inducible inhibitors of signalling.
Nature
387
1997
917
21
Endo
TA
Masuhara
M
Yokouchi
M
Suzuki
R
Sakamoto
H
Mitsui
K
Matsumoto
A
Tanimura
S
Ohtsubo
M
Misawa
H
Miyazaki
T
Leonor
N
Taniguchi
T
Fujita
T
Kanakura
Y
Komiya
S
Yoshimura
A
A new protein containing an SH2 domain that inhibits JAK kinases.
Nature
387
1997
921
22
Naka
T
Narazaki
M
Hirata
M
Matsumoto
T
Minamoto
S
Aono
A
Nishimoto
N
Kajita
T
Taga
T
Yoshizaki
K
Akira
S
Kishimoto
T
Structure and function of a new STAT-induced STAT inhibitor.
Nature
387
1997
924
23
Narazaki
M
Fujimoto
M
Matsumoto
T
Morita
Y
Saito
H
Kajita
T
Yoshizaki
K
Naka
T
Kishimoto
T
Three distinct domains of SSI-1/SOCS-1/JAB protein are required for its suppression of interleukin 6 signaling.
Proc Natl Acad Sci USA
95
1998
13130
24
Matsumoto
A
Masuhara
M
Mitsui
K
Yokouchi
M
Ohtsubo
M
Misawa
H
Miyajima
A
Yoshimura
A
CIS, a cytokine inducible SH2 protein, is a target of the JAK-STAT5 pathway and modulates STAT5 activation.
Blood
89
1997
3148
25
Adams
TE
Hansen
JA
Starr
R
Nicola
NA
Hilton
DJ
Billestrup
N
Growth hormone preferentially induces the rapid, transient expression of SOCS-3, a novel inhibitor of cytokine receptor signaling.
J Biol Chem
273
1998
1285
26
Ohya
Ki
Kajigaya
S
Yamashita
Y
Miyazato
A
Hatake
K
Miura
Y
Ikeda
U
Shimada
K
Ozawa
K
Mano
H
SOCS-1/JAB/SSI-1 can bind to and suppress Tec protein-tyrosine kinase.
J Biol Chem
272
1997
27178
27
Matuoka
K
Miki
H
Takahashi
K
Takenawa
T
A novel ligand for an SH3 domain of the adaptor protein Nck bears an SH2 domain and nuclear signaling motifs.
Biochem Biophys Res Commun
239
1997
488
28
Morel
PA
Oriss
TB
Crossregulation between Th1 and Th2 cells.
Crit Rev Immunol
18
1998
275
29
Muraille
E
Leo
O
Revisiting the Th1/Th2 paradigm.
Scand J Immunol
47
1998
1
30
O’Garra
A
Steinman
L
Gijbels
K
CD4+ T-cell subsets in autoimmunity.
Curr Opin Immunol
9
1997
872
31
Ouyang
W
Ranganath
SH
Weindel
K
Bhattacharya
D
Murphy
TL
Sha
WC
Murphy
KM
Inhibition of Th1 development mediated by GATA-3 through an IL-4-independent mechanism.
Immunity
9
1998
745
32
Etges
R
Muller
I
Progressive disease or protective immunity to Leishmania major infection: The result of a network of stimulatory and inhibitory interactions.
J Mol Med
76
1998
372
33
Del Prete
G
The concept of type-1 and type-2 helper T cells and their cytokines in humans.
Int Rev Immunol
16
1998
427
34
Reiner
SL
Locksley
RM
The regulation of immunity to Leishmania major.
Annu Rev Immunol
13
1995
151
35
Stamm
LM
Raisanen-Sokolowski
A
Okano
M
Russell
ME
David
JR
Satoskar
AR
Mice with STAT6-targeted gene disruption develop a Th1 response and control cutaneous leishmaniasis.
J Immunol
161
1998
6180
36
Chehimi
J
Trinchieri
G
Interleukin-12: A bridge between innate resistance and adaptive immunity with a role in infection and acquired immunodeficiency.
J Clin Immunol
14
1994
149
37
von Stebut
E
Belkaid
Y
Jakob
T
Sacks
DL
Udey
MC
Uptake of Leishmania major amastigotes results in activation and interleukin 12 release from murine skin-derived dendritic cells: Implications for the initiation of anti-Leishmania immunity.
J Exp Med
188
1998
1547
38
Snijders
A
Kalinski
P
Hilkens
CM
Kapsenberg
ML
High-level IL-12 production by human dendritic cells requires two signals.
Int Immunol
10
1998
1593
39
Mackey
MF
Gunn
JR
Maliszewsky
C
Kikutani
H
Noelle
RJ
Barth
RJ
Jr
Dendritic cells require maturation via CD40 to generate protective antitumor immunity.
J Immunol
161
1998
2094
40
Hilkens
CM
Kalinski
P
de Boer
M
Kapsenberg
ML
Human dendritic cells require exogenous interleukin-12-inducing factors to direct the development of naive T-helper cells toward the Th1 phenotype.
Blood
90
1997
1920
41
McRae
BL
Semnani
RT
Hayes
MP
van Seventer
GA
Type I IFNs inhibit human dendritic cell IL-12 production and Th1 cell development.
J Immunol
160
1998
4298
42
Clerici
M
Lucey
DR
Berzofsky
JA
Pinto
LA
Wynn
TA
Blatt
SP
Dolan
MJ
Hendrix
CW
Wolf
SF
Shearer
GM
Restoration of HIV-specific cell-mediated immune responses by interleukin-12 in vitro.
Science
262
1993
5140
43
Chehimi
J
Starr
SE
Frank
I
D’Andrea
A
Ma
X
MacGregor
RR
Sennelier
J
Trinchieri
G
Impaired interleukin 12 production in human immunodeficiency virus-infected patients.
J Exp Med
179
1994
1361
44
Ria
F
Penna
G
Adorini
L
Th1 cells induce and Th2 inhibit antigen-dependent IL-12 secretion by dendritic cells.
Eur J Immunol
28
1998
2003
45
Himmelrich
H
Parra-Lopez
C
Tacchini-Cottier
F
Louis
JA
Launois
P
The IL-4 rapidly produced in BALB/c mice after infection with Leishmania major down-regulates IL-12 receptor beta2-chain expression on CD4+ T cells resulting in a state of unresponsiveness to IL-12.
J Immunol
161
1998
6156
46
Pazdrak
K
Justement
L
Alam
R
Mechanism of inhibition of eosinophil activation by transforming growth factor-beta. Inhibition of Lyn, MAP, Jak2 kinases and STAT1 nuclear factor.
J Immunol
155
1995
4454
47
Pardoux
C
Ma
X
Gobert
S
Pellegrini
S
Mayeux
P
Gay
F
Trinchieri
G
Chouaib
S
Downregulation of interleukin-12 (IL-12) responsiveness in human T cells by transforming growth factor-β: Relationship with IL-12 signaling.
Blood
93
1999
1448
48
Lalani
I
Bhol
K
Ahmed
AR
Interleukin-10: biology, role in inflammation and autoimmunity.
Ann Allergy Asthma Immunol
79
1997
469
49
Stordeur
P
Goldman
M
Interleukin-10 as a regulatory cytokine induced by cellular stress: molecular aspects.
Int Rev Immunol
16
1998
501
50
Hart
PH
Jones
CA
Finlay-Jones
JJ
Monocytes cultured in cytokine-defined environments differ from freshly isolated monocytes in their responses to IL-4 and IL-10.
J Leukoc Biol
57
1995
909
51
Wang
P
Wu
P
Siegel
MI
Egan
RW
Billah
MM
Interleukin (IL)-10 inhibits nuclear factor kappa B (NF kappa B) activation in human monocytes. IL-10 and IL-4 suppress cytokine synthesis by different mechanisms.
J Biol Chem
270
1995
9558
52
Tebo
JM
Kim
HS
Gao
J
Armstrong
DA
Hamilton
TA
Interleukin-10 suppresses IP-10 gene transcription by inhibiting the production of class I interferon.
Blood
92
1998
4742
53
de Waal Malefyt
R
Abrams
J
Bennett
B
Figdor
CG
de Vries
JE
Interleukin 10 (IL-10) inhibits cytokine synthesis by human monocytes: an autoregulatory role of IL-10 produced by monocytes.
J Exp Med
174
1991
1209
54
Chomarat
P
Rissoan
MC
Banchereau
J
Miossec
P
Interferon gamma inhibits interleukin 10 production by monocytes.
J Exp Med
177
1993
523
55
Ito
S
Ansari
P
Sakatsume
M
Dickensheets
H
Vazquez
N
Donnelly
RP
Larner
AC
Finbloom
DS
Interleukin-10 inhibits expres-sion of both interferon α- and interferon γ-induced genes by suppressing tyrosine phosphorylation of STAT1.
Blood
93
1999
1456
56
Pardoux
C
Asselin-Paturel
C
Chehimi
J
Gay
F
Mami-Chouaib
F
Chouaib
S
Functional interaction between TGF-beta and IL-12 in human primary allogeneic cytotoxicity and proliferative response.
J Immunol
158
1997
136
57
Sakamoto
H
Yasukawa
H
Masuhara
M
Tanimura
S
Sasaki
A
Yuge
K
Ohtsubo
M
Ohtsuka
A
Fujita
T
Ohta
T
Furuhawa
Y
Iwasa
S
Yamada
H
Yoshimura
A
A Janus kinase inhibitor, JAB, is an interferon-gamma-inducible gene and confers resistance to interferons.
Blood
92
1998
1668
58
Suzuki
R
Sakamoto
H
Yasukawa
H
Masuhara
M
Wakioka
T
Sasaki
A
Yuge
K
Komiya
S
Inoue
A
Yoshimura
A
CIS3 and JAB have different regulatory roles in interleukin-6 mediated differentiation and STAT3 activation in M1 leukemia cells.
Oncogene
17
1998
2271
59
Song
MM
Shuai
K
The suppressor of cytokine signaling (SOCS) 1 and SOCS3 but not SOCS2 proteins inhibit interferon-mediated antiviral and antiproliferative activities.
J Biol Chem
273
1998
35056

Author notes

Address reprint requests to C.G. Begley, MD, The Walter and Eliza Hall Institute of Medical Research, PO Royal Melbourne Hospital, Parkville, 3050 Victoria, Australia.

Sign in via your Institution