In this issue of Blood, Iriguchi et al report that T-lymphocyte–restricted overexpression of T-bet causes a maturational arrest in mononuclear phagocyte lineage cells and severe secondary pulmonary alveolar proteinosis (PAP).1 

Proposed mechanism by which constitutive, T-cell–restricted T-bet overexpression causes maturational arrest of mononuclear phagocyte lineage cells and secondary PAP. Transgenic mice overexpressing T-bet in T lymphocytes from the human CD2 promoter exhibit constitutive IFNγ expression and multiple primary and secondary downstream biological consequences. A critical primary effect (black arrow) is activation of CD4+ T cells and promotion of TH1 cell differentiation resulting in TH1 cell accumulation and activation. Secondary consequences (gray arrows) include lymphocytic infiltration of the lungs and tissues, marked accumulation of pulmonary alveolar macrophages, maturational arrest of mononuclear phagocytic lineage cells, and time-dependent accumulation of pulmonary surfactant in alveolar macrophages/alveoli (secondary PAP). Characteristics of the alveolar macrophages (large, foamy-appearing, CD11bHiCD11c+, reduced phagocytosis, reduced PPARγ, and ABCG1 messenger RNA [mRNA]) were similar to those of mice and humans with PAP caused by the disruption of GM-CSF signaling, yet GM-CSF mRNA was increased in the lungs of transgenic mice. Pulmonary MCP-1 was also increased (as it is in PAP, caused by the disruption of GM-CSF signaling) and likely contributed to mononuclear phagocyte recruitment (open arrows). Together, these results suggest that secondary PAP occurring in the context of increased expression of T-bet in T cells may be caused by an interruption of the GM-CSF-PU.1-PPARγ-ABCG1 axis, which is critical to surfactant clearance by alveolar macrophages but downstream of PU.1. However, the precise mechanism by which this signaling axis is disrupted in alveolar macrophages and the signaling molecule(s) responsible remain to be determined.

Proposed mechanism by which constitutive, T-cell–restricted T-bet overexpression causes maturational arrest of mononuclear phagocyte lineage cells and secondary PAP. Transgenic mice overexpressing T-bet in T lymphocytes from the human CD2 promoter exhibit constitutive IFNγ expression and multiple primary and secondary downstream biological consequences. A critical primary effect (black arrow) is activation of CD4+ T cells and promotion of TH1 cell differentiation resulting in TH1 cell accumulation and activation. Secondary consequences (gray arrows) include lymphocytic infiltration of the lungs and tissues, marked accumulation of pulmonary alveolar macrophages, maturational arrest of mononuclear phagocytic lineage cells, and time-dependent accumulation of pulmonary surfactant in alveolar macrophages/alveoli (secondary PAP). Characteristics of the alveolar macrophages (large, foamy-appearing, CD11bHiCD11c+, reduced phagocytosis, reduced PPARγ, and ABCG1 messenger RNA [mRNA]) were similar to those of mice and humans with PAP caused by the disruption of GM-CSF signaling, yet GM-CSF mRNA was increased in the lungs of transgenic mice. Pulmonary MCP-1 was also increased (as it is in PAP, caused by the disruption of GM-CSF signaling) and likely contributed to mononuclear phagocyte recruitment (open arrows). Together, these results suggest that secondary PAP occurring in the context of increased expression of T-bet in T cells may be caused by an interruption of the GM-CSF-PU.1-PPARγ-ABCG1 axis, which is critical to surfactant clearance by alveolar macrophages but downstream of PU.1. However, the precise mechanism by which this signaling axis is disrupted in alveolar macrophages and the signaling molecule(s) responsible remain to be determined.

Close modal

PAP is a rare syndrome characterized by pulmonary surfactant accumulation and hypoxemic respiratory failure for which the current treatment is whole lung lavage, an invasive and inefficient procedure to physically remove the excess pulmonary surfactant. It occurs in a heterogeneous group of diseases usefully subdivided into primary PAP, secondary PAP, and disorders of surfactant production.2  Surfactant is normally comprised of a thin phospholipid/protein layer that stabilizes alveoli by reducing alveolar wall surface tension and is maintained by balanced secretion by alveolar epithelial cells, and clearance by these cells and alveolar macrophages. In PAP, however, progressive surfactant accumulation eventually fills alveoli, thus displacing inhaled air and compromising gas exchange.

While significant research advances have elucidated the pathogenesis of primary PAP and led to the development of novel diagnostics and therapeutics,3,4  other than its association with myelodysplastic syndromes, the pathogenesis of secondary PAP remains obscure, its prognosis is poor, and therapeutic options are limited.5  In primary PAP, the disruption of granulocyte-macrophage colony-stimulating factor (GM-CSF) signaling causes alveolar macrophages to undergo maturational arrest, which impairs their ability to clear surfactant. From a mechanistic perspective, the disruption of the GM-CSF→PU.1→PPARγ signaling axis reduces the expression of a critical macrophage lipid exporter, ABCG1, which results in foamy, lipid-laden alveolar macrophages with impaired surfactant clearance capacity, intraalveolar surfactant accumulation, and PAP.6-9  The loss of GM-CSF signaling also increases pulmonary levels of monocyte chemoattractant protein-1 (MCP-1), a biomarker of primary PAP. It is thought that secondary PAP is caused by a reduction in either the functional capacity or absolute numbers of alveolar macrophages, but data supporting this hypothesis are limited.2 

Although expression of T-bet, a “master” TH1 transcription factor, is increased in inflammatory, autoimmune, and hematologic disorders including myelodysplastic syndromes, its precise role in disease pathogenesis is unknown. To address this question and determine the contribution made by increased expression of T-bet to the pathogenesis of inflammatory diseases, Iriguchi et al studied wild-type (wt) and transgenic mice heterozygous or homozygous for a human CD2–T-bet transgene (tg) (wt/wt, tg/wt, or tg/tg mice, respectively). Unexpectedly, the results identified a novel mechanism by which increased expression of T-bet, exclusively in T lymphocytes, spontaneously drives the pathogenesis of both myelodysplasia and secondary PAP in a dose-dependent manner.1 

Prior studies with these mice had shown that T-bet overexpression increased interferon-γ (IFNγ) production in CD4+ cells, enhanced TH1 and suppressed TH2 antibody responses, and caused dermatitis.10  The present report shows that compared with wt/wt mice, tg/wt mice spontaneously developed maturational arrest in the mononuclear phagocytic lineage, while tg/tg mice also spontaneously developed severe lung inflammation including perivascular/peribronchiolar lymphocytic infiltration, secondary PAP, and had increased mortality correlating with the presence and severity of the lung disease.

These results identify a previously unsuspected mechanism in which T-bet expression in TH1 cells regulates myeloid lineage progression that, when activated constitutively, causes maturational arrest of mononuclear phagocytes, dose-dependent tissue inflammation, and secondary PAP. These findings are important because they provide a molecular explanation for the association between PAP and myelodysplastic syndromes, and confirm the leading hypothesis about the pathogenesis of secondary PAP (see figure). Nonetheless, the precise mechanism by which T-bet overexpression results in maturational arrest of myeloid lineage cells or the accumulation of functionally impaired alveolar macrophages remains to be determined. Future studies are needed to determine the pathogenic mechanism and to explore the clinical implications of these findings for myelodysplasia and secondary PAP.

Conflict-of-interest disclosure: The author declares no competing financial interests.

1
Iriguchi
 
S
Kikuchi
 
N
Kaneko
 
S
, et al. 
T-cell–restricted T-bet overexpression induces aberrant hematopoiesis of myeloid cells and impairs function of lung macrophages in the lung.
Blood
2015
 
125(2):370-382
2
Trapnell
 
BC
Whitsett
 
JA
Nakata
 
K
Pulmonary alveolar proteinosis.
N Engl J Med
2003
, vol. 
349
 
26
(pg. 
2527
-
2539
)
3
Suzuki
 
T
Sakagami
 
T
Young
 
LR
, et al. 
Hereditary pulmonary alveolar proteinosis: pathogenesis, presentation, diagnosis, and therapy.
Am J Respir Crit Care Med
2010
, vol. 
182
 
10
(pg. 
1292
-
1304
)
4
Suzuki
 
T
Arumugam
 
P
Sakagami
 
T
, et al. 
Pulmonary macrophage transplantation therapy.
Nature
2014
, vol. 
514
 
7523
(pg. 
450
-
454
)
5
Ishii
 
H
Tazawa
 
R
Kaneko
 
C
, et al. 
Clinical features of secondary pulmonary alveolar proteinosis: pre-mortem cases in Japan.
Eur Respir J
2011
, vol. 
37
 
2
(pg. 
465
-
468
)
6
Shibata
 
Y
Berclaz
 
PY
Chroneos
 
ZC
Yoshida
 
M
Whitsett
 
JA
Trapnell
 
BC
GM-CSF regulates alveolar macrophage differentiation and innate immunity in the lung through PU.1.
Immunity
2001
, vol. 
15
 
4
(pg. 
557
-
567
)
7
Bonfield
 
TL
Farver
 
CF
Barna
 
BP
, et al. 
Peroxisome proliferator-activated receptor-gamma is deficient in alveolar macrophages from patients with alveolar proteinosis.
Am J Respir Cell Mol Biol
2003
, vol. 
29
 
6
(pg. 
677
-
682
)
8
Thomassen
 
MJ
Barna
 
BP
Malur
 
AG
, et al. 
ABCG1 is deficient in alveolar macrophages of GM-CSF knockout mice and patients with pulmonary alveolar proteinosis.
J Lipid Res
2007
, vol. 
48
 
12
(pg. 
2762
-
2768
)
9
Malur
 
A
Baker
 
AD
McCoy
 
AJ
, et al. 
Restoration of PPARγ reverses lipid accumulation in alveolar macrophages of GM-CSF knockout mice.
Am J Physiol Lung Cell Mol Physiol
2011
, vol. 
300
 
1
(pg. 
L73
-
L80
)
10
Ishizaki
 
K
Yamada
 
A
Yoh
 
K
, et al. 
Th1 and type 1 cytotoxic T cells dominate responses in T-bet overexpression transgenic mice that develop contact dermatitis.
J Immunol
2007
, vol. 
178
 
1
(pg. 
605
-
612
)
Sign in via your Institution