ArticlePDF Available

The Nature of the Principal Type 1 Interferon-Producing Cells in Human Blood

Authors:
  • St. Vincent Hospital NYC Closed

Abstract and Figures

Interferons (IFNs) are the most important cytokines in antiviral immune responses. “Natural IFN-producing cells” (IPCs) in human blood express CD4 and major histocompatibility complex class II proteins, but have not been isolated and further characterized because of their rarity, rapid apoptosis, and lack of lineage markers. Purified IPCs are here shown to be the CD4+CD11c− type 2 dendritic cell precursors (pDC2s), which produce 200 to 1000 times more IFN than other blood cells after microbial challenge. pDC2s are thus an effector cell type of the immune system, critical for antiviral and antitumor immune responses.
Content may be subject to copyright.
9. Fuel load quantities, divided into standard time-lag
size classes (1 hour, 10 hour, 100 hour, and 1000
hour; corresponding to 0 to 0.6, .0.6 to 2.5, .2.5 to
7.6, and .7.6 cm in diameter), fuel height, and leaf
litter depth were measured along randomly directed
10-m transects at three points within each plot (125,
250, and 375 m). Additional fuel load measurements
were made in the vicinity of all observed fires.
10. At each observation site (n544), roughly 10 m of
fireline was observed every 30 s for several minutes
(average observation time, 7.4 min), and the average
minimum and maximum of each flame characteristic
were recorded. Flareups of short duration or small
area were noted separately.
11. J. K. Agee, Fire Ecology of Pacific Northwest Forests
(Island Press, Washington, DC, 1993).
12. R. C. Rothermel, How to Predict the Spread and
Intensity of Forest and Range Fires (General Technical
Report INT-143, U.S. Forest Service, Ogden, UT,
1983).
13. S. J. Pyne, Introduction to Wildland Fire (Wiley, New
York, 1984).
14. Leaf fall from damaged trees begins within 2 days of
a burn. We have observed contiguous layers of leaf
litter in recently burned forests in close proximity to
smoldering logs. Frequent observations by local res-
idents that forest stands often burn more than once
in a season may be explained by smoldering logs
reigniting fires once litter depths become sufficient
to carry fire through the stand.
15. The mortality of trees (.10 cm dbh) in previously
unburned forests that burned in 1995 was 38% 1
year after the fire and 68% at the end of the second
year. Annual mortality in unburned forest during this
time period was ,1%.
16. D. L. Peterson and K. C. Ryan, Environ. Manag.10, 797
(1986).
17. In forest subjected to fire, vines frequently form a
dense mat at 30 to 200 cm above the ground, and
grasses can form up to 70% of the ground cover.
18. Field studies of forests in 1996 revealed a repre-
sentative plot with only 18 live trees per hectare
(with 302 trees standing dead per hectare). The
fires of 1997 had left only three live trees in this
plot, which was threatened by an oncoming fire,
when recensused.
19. E. Silva, thesis, Pennsylvania State University (1996).
20. M. A. Cochrane and C. M. Souza Jr., Int. J. Remote
Sens.19, 3433 (1998).
21. C. E. Van Wagner, Can. J. For. Res.8, 220 (1978).
22. D. Skole and C. Tucker, Science 260, 1905 (1993).
23. INPE (Instituto Nacional de Pesquisas Espaciais) Des-
florestamento 1995–1997 (Sa˜o Jose´ dos Campos, Sa˜o
Paulo, Brazil, 1997).
24. INPE Desflorestamento, 1993–1994 (Sa˜o Jose´ dos
Campos, Sa˜o Paulo, Brazil, 1996).
25. W. D. Jackson, Proc. Ecol. Soc. Aust.3(1968).
26. M. Mueller-Dombois, in Proceeding of the Confer-
ence—Fire Regimes and Ecosystem Properties (Gen-
eral Technical Report WO-26, U.S. Forest Service,
Honolulu, HI, 1981), pp. 137–176.
27. J. Shukla et al.,Science 247, 1322 (1990).
28. C. E. Van Wagner, Can. J. For. Res.3, 373 (1973).
29. In Paragominas, burns detected in the imagery were
compared with data from landowner questionnaires
(n575) that described fire history from 1982 to
1995. Questionnaire data included 51.4% of the
study region and showed 100% detection of reported
fires that occurred within 1 year of the image date.
Comparisons between the area reported burned by
landowners with data from the imagery classifica-
tions showed that the area burned was systematical-
ly underreported (P,0.001; sign test) by an average
of 43%. Only small fires (,50 ha) were overestimat-
ed by landowners.
30. We thank C. Uhl, A. Taylor, G. P. Patil, F. Williams, and
four anonymous reviewers for comments on earlier
versions of the manuscript. This research was funded
by a grant from the PPG7 “Programa de Pesquisa
Dirigida” (MMA/MCT/FINEP). Additional financial
support was provided by NSF, NASA, and the U.S.
Agency for International Development.
16 February 1999; accepted 16 April 1999
The Nature of the Principal
Type 1 Interferon–Producing
Cells in Human Blood
Frederick P. Siegal,
1*
Norimitsu Kadowaki,
2
Michael Shodell,
3
Patricia A. Fitzgerald-Bocarsly,
4
Kokila Shah,
1
Stephen Ho,
2
Svetlana Antonenko,
2
Yong-Jun Liu
2*
Interferons (IFNs) are the most important cytokines in antiviral immune re-
sponses. “Natural IFN-producing cells” (IPCs) in human blood express CD4 and
major histocompatibility complex class II proteins, but have not been isolated
and further characterized because of their rarity, rapid apoptosis, and lack of
lineage markers. Purified IPCs are here shown to be the CD4
1
CD11c
2
type 2
dendritic cell precursors (pDC2s), which produce 200 to 1000 times more
IFN than other blood cells after microbial challenge. pDC2s are thus an
effector cell type of the immune system, critical for antiviral and antitumor
immune responses.
Interferons were discovered in the 1950s as
factors rapidly produced by virus-infected cells
that enable neighboring cells to resist virus
infection (1). IFN-a(leukocyte IFN) and IFN-b
(fibroblast IFN), the two type 1 antiviral IFNs,
are distinct from type 2 IFN-gproduced by
effector T cells. Specialized leukocytes, the
“natural IFN-producing cells” (IPCs), were
shown to be the chief IFN-aproducers in re-
sponse to enveloped viruses, bacteria, and tu-
mor cells (2–14). IPCs express CD4 and major
histocompatibility complex (MHC) class II, but
lack hematopoietic-lineage markers (2–14).
The nature of IPCs—whether they represent
dendritic cells (6,12,14) or cells of a distinct
lineage (7,9)—has been controversial. There is
a progressive loss of CD4
1
T lymphocytes and
functional IPCs during human immunodefi-
ciency virus (HIV) infection (15,16). Preser-
vation of IPCs is associated with protection
from opportunistic infections, suggesting the
importance of IPCs in the host defense (16).
A plasmacytoid cell type from human ton-
sils and blood that lacks lineage markers also
expresses CD4 and MHC class II (17–21).
These cells differentiate into type 2 dendritic
cells (DC2s) when cultured with interleukin-3
(IL-3) and CD40 ligand (19,21). Unlike mono-
cyte-derived type 1 dendritic cells (DC1s) that
induced type 1 T helper cell (T
H
1) differentia-
tion, DC2s induced type 2 T helper cell (T
H
2)
differentiation (21). Here we investigated
whether DC2 precursors (pDC2s) represent
IPCs. Human peripheral blood cells were sep-
arated into the following populations (19,21):
(i) monocytes (over 90% purity), obtained by
centrifugation through 52% Percoll, then
magnetic bead depletion of B, T, and natural
killer (NK) cells; (ii) CD4
1
CD3
2
CD11c
1
immature DCs (99% purity) and (iii) CD4
1
CD3
2
CD11c
2
pDC2s (99% purity), ob-
tained by magnetic bead depletion of B, T,
NK cells, and monocytes, followed by fluo-
rescence-activated cell sorting (FACS) (Fig.
1, A and B); (iv) pDC2-depleted blood mono-
1
Saint Vincents Hospital and Medical Center, New
York, NY 10011, and Long Island Jewish Medical
Center–Albert Einstein College of Medicine, New
Hyde Park, NY 11040, USA.
2
DNAX Research Insti-
tute, Palo Alto, CA 94304, USA.
3
The CW Post Cam-
pus of Long Island University, Brookville, NY 11548,
USA.
4
University of Medicine and Dentistry, New
Jersey Medical School, Newark, NJ 07103, USA.
*To whom correspondence should be addressed.
Table 1. Precursor DC2 cells are the natural IFN-producing cells. Cells (2 310
5
) were cultured for 24
hours with HSV. Without HSV, IFN activity from different cell types was less than 12.5 U/ml (23). PBMC:
total blood mononuclear cells; pDC2-dep: blood mononuclear cells positively selected for expressing CD3,
CD11c, CD19, CD14, and CD56; pDC2-enrich: blood mononuclear cells that were depleted of cells
expressing CD3, CD19, CD14, and CD56; pDC2: FACS-sorted CD4
1
CD11c
2
lin
2
cells; CD11c
1
DC:
FACS-sorted CD11c
1
lin
2
immature DCs; Mo: monocytes; DC1: monocyte-derived DCs after 6 days of
culture with either granulocyte-macrophage colony-stimulating factor (GM-CSF) 1IL-4 or GM-CSF 1
IL-4 1CD40 ligand (21); DC2: pDC2-derived DCs after 6 days of culture with IL-3 or IL-3 1CD40 ligand
(23). ND, not determined.
IFN (U/ml)
PBMC pDC2-dep pDC2-enrich pDC2 CD11c
1
DC Mo DC1 DC2
Exp. 1 500 ND 2,800 89,800 120 ND ,12.5 1,100
Exp. 2 40 ,12.5 180 20,000 ,12.5 350 ,12.5 ,12.5
Exp. 3 700 40 2,800 638,000 70 90 ND ND
REPORTS
www.sciencemag.org SCIENCE VOL 284 11 JUNE 1999 1835
nuclear cells; and (v) pDC2-enriched blood
mononuclear cells (19). pDC2s have a plas-
macytoid morphology, with rough endoplas-
mic reticulum and Golgi apparatus (Fig. 1, C
and D). The CD11c
1
blood immature DCs
display short dendrites (Fig. 1, E and F). The
frequency of pDC2s in human blood mono-
nuclear cells is less than 0.5% and increased
to 3 to 10% after magnetic bead depletion of
lineage-positive cells. The pDC2-depleted
population contains B, T, NK cells, mono-
cytes, and DCs. These cell populations were
exposed to ultraviolet (UV)-irradiated herpes
simplex virus (HSV) for 24 hours (22), and
IFN in the culture supernatant was measured
by a bioassay (23).
IFN production by total peripheral blood
mononuclear cells (PBMCs) from three donors
was 40, 500, and 700 international units (IU)
per 2 310
5
cells (Table 1). There was a four to
six times increase in IFN production from
pDC2-enriched blood mononuclear cells (180
to 2800 IU per 10
5
cells) and a 180 to 911 times
increase in IFN generation from purified
pDC2s (20,000 to 638,000 IU per 10
5
cells).
pDC2-depleted PBMCs, immature CD11c
1
DCs, monocytes, and monocyte-derived
DC1s produced little or low levels of IFN.
The ability of pDC2s to produce IFN was
decreased after maturation into DC2s by cul-
ture with IL-3 or IL-3 plus CD40L for 6 days
(Table 1). The geometric mean IFN-agener-
ation by pDC2s was ;1 IU per cell, similar to
previous estimates (2). Immunoperoxidase
staining for human IFN-aconfirmed that
most pDC2s contained IFN-aprotein after
6 hours of exposure to HSV (Fig. 2) (22).
Analysis of IFN-aand IFN-bmRNA by
polymerase chain reaction (PCR) showed that
among human blood cells, pDC2s were mak-
ing the most IFN-aand IFN-bmRNA (Fig.
3) (24). Thus, the blood cells responsible for
IFN generation in response to HSV, previ-
ously known as the IPCs, are actually the
DC2 precursors. These cells can be traced
and isolated by their expression of CD4 or
IL-3 receptor after depletion of cells express-
ing lineage markers and CD11c (19,21).
The purified IPCs also produced high levels
of IFN in response to Sendai virus and heat-
killed Staphylococcus aureus, confirming the
previous studies on PBMCs and partially puri-
fied IPCs (214). The ability of UV-irradiated
virus and heat-killed bacteria to induce IFN
production by IPCs suggests that viral infection
is not required for triggering IFN production.
The rapid production of IFN by IPCs in the
absence of other cells suggests that IPCs repre-
sent an effector cell type of the innate immune
system. We propose that the IPCs/pDC2s
should be included in the hematopoietic devel-
opmental chart as a distinct cell lineage. They
function as professional IFN-producing cells at
the precursor stages and as professional anti-
Fig. 1. Tracing and isolation of IPCs/pDC2s from human peripheral blood. CD3
1
T cells, CD19
1
B
cells, CD16
1
and CD56
1
NK cells, and CD14
1
monocytes were depleted from blood mononuclear
cells by immunomagnetic beads (Dynabeads M-450; Dynal, Oslo, Norway). The cells were stained
with anti-CD4-Tricolor (Immunotech, Marseille, France), anti-CD11c-PE (Becton Dickinson, San
Jose, California), and a mixture of fluorescein isothiocyanate–labeled antibodies to CD3, CD15,
CD16, CD20, CD57 (Becton Dickinson), CD14 (Coulter, Miami, Florida), and CD34 (Immunotech).
Within the lineage-negative population (A), CD4
1
CD11c
2
IPCs and CD11c
1
immature DCs were
isolated (B). IPCs are plasmacytoid by Giemsa staining (C) and contain rough endoplasmic
reticulum and Golgi apparatus under transmission electron microscopy (D). The CD11c
1
blood
immature DCs display dendrites (Eand F).
A
B
Fig. 2. Immunoperoxidase staining for IFN-a
(21,22). (A) Purified CD4
1
CD11c
2
lin
2
IPCs
were stimulated with HSV for 6 hours. (B) IPCs
cultured in medium for 6 hours. The isotype
controls for the primary antibody in both cell
preparations show no staining.
REPORTS
11 JUNE 1999 VOL 284 SCIENCE www.sciencemag.org1836
gen-presenting type 2 DCs upon terminal
differentiation.
Type 1 IFNs have pleiotropic effects on the
immune system, including up-regulation of
MHC class I on all cell types and activation of
macrophage and NK cells (2). IFNs are also
critical in the activation and survival of both
CD4
1
and CD8
1
T cells (25,26). Now with
the ability to trace and isolate IPCs, it should be
possible to directly study the interaction be-
tween IPCs and other cell types within the
immune system. IFN-ahas been widely used
for treating hepatitis B and C as well as various
cancers. A progressive loss of IPCs has been
observed during HIV infection, suggesting that
IPCs may represent targets for HIV-mediated
infection and deletion. The present study pro-
vides an approach to directly monitor the num-
ber and functional state of IPCs in these pa-
tients. The ability to purify and culture IPCs in
vitro will allow further studies on the molecular
mechanisms that control the survival and
growth of IPCs and their production of IFN,
which may lead to novel therapies for patients
with viral infections and cancer.
References and Notes
1. A. Isaacs and J. Lindemann, Proc. R. Soc. London 147,
258 (1957).
2. P. Fitzgerald-Bocarsly, Pharmacol. Ther. 60,39
(1993).
3. H. Kirchner et al.,Immunobiology 156, 65 (1979).
4. H. H. Peter et al.,Eur. J. Immunol. 10, 547 (1980).
5. J. Abb, H. Abb, F. Deinhart, Clin. Exp. Immunol. 52,
179 (1983).
6. B. Perussia, V. Fanning, G. Trinchieri, Nat. Immun. Cell
Growth Regul. 4, 120 (1985).
7. J. Chehimi et al.,Immunology 68, 488 (1989).
8. K. Sandberg et al., Scand. J. Immunol. 34, 565 (1991).
9. S. E. Starr et al.,Adv. Exp. Med. Biol. 329, 173 (1993).
10. H. Svensson et al., Scand. J. Immunol. 44, 164 (1996).
11. S. B. Feldman et al.,Virology 204, 1 (1994).
12. M. Feldman and P. Fitzgerald-Bocarsly, J. Interferon
Res. 10, 435 (1990).
13. J. J. Ferbas et al., J. Immunol. 152, 4649 (1994); S. B.
Feldman et al., J. Leukocyte Biol. 57, 214 (1995).
14. S. Ghanekar et al.,J. Immunol. 157, 4028 (1996).
15. C. Lopez, P. Fitzgerald, F. P. Siegal. J. Infect. Dis. 148,
962 (1983); D. M. Howell, S. B. Feldman, P. Kloser, P.
Fitzgerald-Bocarsly, Clin. Immunol. Immunopathol. 7,
223 (1994); J. Ferbas, J. Navratil, A. Logar, C. Rinaldo,
Clin. Diagn. Lab. Immunol. 2, 138 (1995).
16. F. P. Siegal et al.,J. Clin. Invest. 78, 115 (1986).
17. U. O’Doherty et al.,J. Exp. Med. 178, 1067 (1993).
18. U. O’Doherty et al.,Immunology 82, 487 (1994).
19. G. Grouard et al.,J. Exp. Med. 185, 1101 (1997).
20. J. Olweus et al.,Proc. Natl. Acad. Sci. U.S.A.94,
12551 (1997).
21. M.-C. Rissoan et al.,Science 283, 1183 (1999).
22.
Cells were incubated with UV-irradiated HSV in
quadruplicate wells (2 310
5
cells in 200 ml of culture
medium per well with 2 310
4
plaque-forming units of
virus in 96-well culture plates) (23). IFN in supernatants
from 24-hour cultures with and without IL-3 were ana-
lyzed with cytopathic reduction in human foreskin fibro-
blast monolayers cultured with vesicular stomatitis virus
(sensitivity, 2 to 25 IU of IFN per milliliter) (23). Cytocen-
trifuge preparations of cells from 6-hour cultures were
prepared for IFN-aimmunostaining with mouse mono-
clonal antibody 7N4-1 (10 mg/ml; Schering-Plough Re-
search Institute, Kenilworth, NJ) (21). Cells from 6-hour
cultures were centrifuged and the sediment frozen for
IFN-aand IFN-bmRNA PCR analyses (24).
23. F. P. Siegal et al.,Leukemia 8, 1474 (1994).
24. Reverse transcriptase (RT )–PCR: RNA was isolated
with the acid guanidinium thiocyanate–phenol-chlo-
roform method [P. Chomczrski and N. Sacchi, Anal.
Biochem. 162, 156 (1987)]. DNA contamination was
removed by digestion with deoxyribonuclease I (5 U;
Boehringer-Mannheim, Indianapolis, IN) for 30 min at
37°C. Controls without RT were performed. Reverse
transcription was carried out with pd( T)12-18 (Phar-
macia, Alameda, CA) priming and Superscript II RT
(Life Technologies, Grand Island, NY ). PCR reaction
volume was 25 ml containing 100 ng of each primer,
40 nM of each deoxynucleoside triphosphate, 1 mlof
cDNA, and 1.25 U of AmpliTaq (Perkin-Elmer, Foster
City, CA). The primers used were as follows: IFN-a
(sense: 59-GATGGCCGTGCTGGTGCTCA-39; antisense:
59-TGATTTCTGCTCTGACAACCTCCC-39; probe: 59-
CTCAAGCCATCTCTGTCCTCC ATGAGATGA-39);
IFN-b(sense: 59-TTGAATGGGAGGCTTGAATA-39;
antisense: 59-CTATGGTCCAGGCACAGTGA-39; probe:
59-GGCTGGAATGAGACTATTGTTGAGAACCTC-39);
and human ribosomal protein S14 (sense: 59-GGCA-
GACCGAGATGAATCCTCA-39; antisense: 59-CAGG-
TCCAGGGGTCTTGGTCC-39). Each PCR amplification
contained primers for the ribosomal protein S14, to
verify the amounts of cDNA. A GeneAmp PCR Sys-
tem 9700 (Perkin-Elmer/Applied Biosystems) was
used with an initial denaturation step of 94°C for 5
min, followed by cycles of 94°C for 30 s, 60°C for
30 s, 72°C for 60 s, and a final elongation step of
72°C for 7 min. PCR products were separated on a
2% agarose gel, followed by DNA blotting and hy-
bridization with
32
P-labeled probes.
25. S. Sun et al., J. Exp. Med. 188, 2335 (1998).
26. P. Marrack et al., ibid. 189, 521 (1999).
27. We thank T. L. Nagabhushan, L. L. Lanier, H. Kanzler,
D. Imperato, C. Lopez, L. Filgueira, G. Grouard, and R.
Rai for continued support. F.P.S. is supported by a
grant from Pharmacia and Upjohn. DNAX Research
Institute of Molecular and Cellular Biology is support-
ed by Schering-Plough Corporation.
8 February 1999; accepted 3 May 1999
Math1: An Essential Gene for
the Generation of Inner Ear
Hair Cells
Nessan A. Bermingham,
1,2
Bassem A. Hassan,
1,2
Steven D. Price,
7
Melissa A. Vollrath,
5
Nissim Ben-Arie,
2
* Ruth Anne Eatock,
5,6
Hugo J. Bellen,
1,2,4,5
Anna Lysakowski,
7
Huda Y. Zoghbi
1,2,3,4,5
The mammalian inner ear contains the cochlea and vestibular organs, which are
responsible for hearing and balance, respectively. The epithelia of these sensory
organs contain hair cells that function as mechanoreceptors to transduce sound
and head motion. The molecular mechanisms underlying hair cell development
and differentiation are poorly understood. Math1, a mouse homolog of the
Drosophila proneural gene atonal, is expressed in inner ear sensory epithelia.
Embryonic Math1-null mice failed to generate cochlear and vestibular hair cells.
This gene is thus required for the genesis of hair cells.
The inner ear initially forms as a thickening
of the ectoderm, termed the otic placode, be-
tween rhombomeres 5 and 6 in the hindbrain.
The otic placode gives rise to neurons of the
VIIIth cranial nerve and invaginates to be-
come the otocyst, from which the inner ear
Fig. 3. RT-PCR amplification of IFN-aand IFN-b. PCR products amplified from each cell population
after 20, 25, 30, or 35 cycles were separated on a 2% agarose gel containing ethidium bromide.
Negative controls contained no cDNA. Marker is 1-kb DNA Ladder (Life Technologies, Grand Island,
New York). IFN-amRNA is apparent in PBMCs, is increased in the DC2 precursors (pDC2s) with
enrichment and purification, and is diminished in the monocyte fraction. IFN-bmRNA is visualized
only in the most highly purified DC2 precursors.
REPORTS
www.sciencemag.org SCIENCE VOL 284 11 JUNE 1999 1837
... The rarity of the cells challenged more specific identification, but further studies in the context of the AIDS epidemic advanced identification of the cells ultimately described as plasmacytoid dendritic cells (pDCs). [58][59][60][61] Linking pDCs, studied by Liu, 62 to the cells that produce IFN-I was facilitated by the insight of Siegal, a clinical immunologist working with patients with AIDS in New York City who noted that "pDCs really smelled like a cell type called the natural IFN-producing cells." 62 pDCs were defined as the major IFN-I-producing cells and demonstrated to lose ability to produce IFN-I after maturation, when they support adaptive immune responses. ...
... 62 pDCs were defined as the major IFN-I-producing cells and demonstrated to lose ability to produce IFN-I after maturation, when they support adaptive immune responses. 61 Identification of BDCA2, a specific marker for pDCs, accelerated study of those cells and their role as robust producers of IFN-α, and Dzionek et al, 63 Cisse et al, 64 and Soni et al 65 subsequently characterized the development of pDCs and their role in extrafollicular B cell responses supportive of autoimmunity. Toll-like receptor 7 (TLR7) and TLR9, endosomal nucleic acid sensors highly expressed in pDCs, were recognized as receptors not only for RNA and DNA viruses, respectively, but for endogenous RNA and DNA ligands, and IRF7 was identified as a critical transcription factor involved in IFN-α production by pDCs. ...
Article
Full-text available
The discovery of interferon in the 1950s represents much more than the identification of the first cytokine and the key mediator of antiviral host defense. Defining the molecular nature and complexity of the type I interferon family, as well as its inducers and molecular mechanisms of action, was the work of investigators working at the highest level and producing insights of great consequence. Current knowledge of receptor–ligand interactions, cell signaling, and transcriptional regulation derives from studies of type I interferon. It is on the shoulders of the giants who produced that knowledge that others stand and have revealed critical mechanisms of the pathogenesis of systemic lupus erythematosus and other autoimmune diseases. The design of novel therapeutics is informed by the advances in investigation of type I interferon, with the potential for important impact on patient management.
... In vitro, most cells can produce IFN-I. However, in vivo, most IFN-I-producing cells are myeloid APCs [30,32,[82][83][84][85]. Among APCs, iMOs, DCs, and plasmacytoid DCs (pDCs) can produce IFN-I in infected animals or humans. ...
Article
Full-text available
The interstitial fluids in tissues are constantly drained into the lymph nodes (LNs) as lymph through afferent lymphatic vessels and from LNs into the blood through efferent lymphatics. LNs are strategically positioned and have the appropriate cellular composition to serve as sites of adaptive immune initiation against invading pathogens. However, for lymph-borne viruses, which disseminate from the entry site to other tissues through the lymphatic system, immune cells in the draining LN (dLN) also play critical roles in curbing systemic viral dissemination during primary and secondary infections. Lymph-borne viruses in tissues can be transported to dLNs as free virions in the lymph or within infected cells. Regardless of the entry mechanism, infected myeloid antigen-presenting cells, including various subtypes of dendritic cells, inflammatory monocytes, and macrophages, play a critical role in initiating the innate immune response within the dLN. This innate immune response involves cellular crosstalk between infected and bystander innate immune cells that ultimately produce type I interferons (IFN-Is) and other cytokines and recruit inflammatory monocytes and natural killer (NK) cells. IFN-I and NK cell cytotoxicity can restrict systemic viral spread during primary infections and prevent serious disease. Additionally, the memory CD8 ⁺ T-cells that reside or rapidly migrate to the dLN can contribute to disease prevention during secondary viral infections. This review explores the intricate innate immune responses orchestrated within dLNs that contain primary viral infections and the role of memory CD8 ⁺ T-cells following secondary infection or CD8 ⁺ T-cell vaccination.
... En lo que respecta a las células dendríticas plasmocitoides, son las principales productoras de interferones tipo I (IFN), citocina que tienen un rol importante en cáncer al modular la respuesta inmune para eliminar el tumor (Reizis, 2019). Además, las pDC expresan moléculas de MHC II por lo cual, también contribuyen en la presentación de antígenos a las células T CD4 (Siegal et al., 1999). ...
Article
El cáncer es una de las principales causas de muerte en el mundo, por ello las investigaciones actuales se centran en evaluar los mecanismos implicados en el desarrollo de esta enfermedad. A lo largo de los años se ha demostrado que la respuesta inmune (tanto innata como adaptativa) juega un papel fundamental en la identificación y eliminación de células tumorales. Sin embargo, los tumores pueden evadir el reconocimiento inmunológico y seguir proliferando. En este artículo explicamos aspectos actuales sobre los mecanismos que utiliza el sistema inmune para eliminar las células tumorales, también hacemos una revisión de los mecanismos que utiliza el tumor para evadir la respuesta inmune.
... By analogy, GM-CSF probably has the potential for treatments for RV infection. IFN can also be produced by pDC in RV-induced immunoreactions (99). It appears that IFN-α and IFN-β couple to the same receptor, which is different from the receptor for IFN-λ. ...
Article
Full-text available
Rhinovirus is a widespread virus associated with several respiratory diseases, especially asthma exacerbation. Currently, there are no accurate therapies for rhinovirus. Encouragingly, it is found that during rhinovirus-induced immunoreactions the levels of certain cytokines in patients' serum will alter. These cytokines may have pivotal pro-inflammatory or anti-inflammatory effects via their specific mechanisms. Thus far, studies have shown that inhibitions of cytokines such as IL-1, IL-4, IL-5, IL-6, IL-13, IL-18, IL-25, and IL-33 may attenuate rhinovirus-induced immunoreactions, thereby relieving rhinovirus infection. Furthermore, such therapeutics for rhinovirus infection can be applied to viruses of other species, with certain practicability.
... Additionally, IFN-I also affects the adaptive immune response, augmenting antibody production and effector T cell responses (21). While the majority of cells are capable of producing IFN-I, pDCs are known to rapidly pro duce large quantities of IFN-I in response to viral infections, accounting for 95% of IFN-I production by mononuclear cells (22). IFN-I binds to a heterodimeric receptor composed of two subunits, Interferon Alpha and Beta Receptor Subunit 1 (IFNAR1) and Interferon Alpha and Beta Receptor Subunit 2 (IFNAR2), to form a ternary complex. ...
Article
Full-text available
Interferons (IFNs) are essential for defense against viral infections but also drive recruitment of inflammatory cells to sites of infection, a key feature of severe COVID-19. Here, we explore the complexity of the IFN response in COVID-19, examine the effects of manipulating IFN on SARS-CoV-2 viral replication and pathogenesis, and highlight pre-clinical and clinical studies evaluating the therapeutic efficacy of IFN in limiting COVID-19 severity.
... PDCs are not classical antigen-presenting cells but specialize in producing large quantities of type I IFNs in response to TLR7 and TLR9 ligation. 153,154 In addition, cytosolic PRRs, such as cGAS, RIG-I, and MDA5, are potent drivers of type I IFN production in many cell types including cDCs. CD8 + T cells in particular depend on interferon signaling for differentiation and function. ...
Article
Full-text available
Background Dermatomyositis (DM) manifests as an autoimmune and inflammatory condition, clinically characterized by subacute progressive proximal muscle weakness, rashes or both along with extramuscular manifestations. Literature indicates that DM shares common risk factors with atherosclerosis (AS), and they often co‐occur, yet the etiology and pathogenesis remain to be fully elucidated. This investigation aims to utilize bioinformatics methods to clarify the crucial genes and pathways that influence the pathophysiology of both DM and AS. Method Microarray datasets for DM (GSE128470, GSE1551, GSE143323) and AS (GSE100927, GSE28829, GSE43292) were retrieved from the Gene Expression Omnibus (GEO) database. The weighted gene co‐expression network analysis (WGCNA) was used to reveal their co‐expressed modules. Differentially expression genes (DEGs) were identified using the “limma” package in R software, and the functions of common DEGs were determined by functional enrichment analysis. A protein‐protein interaction (PPI) network was established using the STRING database, with central genes evaluated by the cytoHubba plugin, and validated through external datasets. Immune infiltration analysis of the hub genes was conducted using the CIBERSORT method, along with Gene Set Enrichment Analysis (GSEA). Finally, the NetworkAnalyst platform was employed to examine the transcription factors (TFs) responsible for regulating pivotal crosstalk genes. Results Utilizing WGCNA analysis, a total of 271 overlapping genes were pinpointed. Subsequent DEG analysis revealed 34 genes that are commonly found in both DM and AS, including 31 upregulated genes and 3 downregulated genes. The Degree Centrality algorithm was applied separately to the WGCNA and DEG collections to select the 15 genes with the highest connectivity, and crossing the two gene sets yielded 3 hub genes (PTPRC, TYROBP, CXCR4). Validation with external datasets showed their diagnostic value for DM and AS. Analysis of immune infiltration indicates that lymphocytes and macrophages are significantly associated with the pathogenesis of DM and AS. Moreover, GSEA analysis suggested that the shared genes are enriched in various receptor interactions and multiple cytokines and receptor signaling pathways. We coupled the 3 hub genes with their respective predicted genes, identifying a potential key TF, CBFB, which interacts with all 3 hub genes. Conclusion This research utilized comprehensive bioinformatics techniques to explore the shared pathogenesis of DM and AS. The three key genes, including PTPRC, TYROBP, and CXCR4, are related to the pathogenesis of DM and AS. The central genes and their correlations with immune cells may serve as potential diagnostic and therapeutic targets.
Article
People living with HIV have a 1.5- to 2-fold increased risk of developing cardiovascular disease. Despite treatment with highly effective antiretroviral therapy, people living with HIV have chronic inflammation that makes them susceptible to multiple comorbidities. Several factors, including the HIV reservoir, coinfections, clonal hematopoiesis of indeterminate potential (CHIP), microbial translocation, and antiretroviral therapy, may contribute to the chronic state of inflammation. Within the innate immune system, macrophages harbor latent HIV and are among the prominent immune cells present in atheroma during the progression of atherosclerosis. They secrete inflammatory cytokines such as IL (interleukin)-6 and tumor necrosis-α that stimulate the expression of adhesion molecules on the endothelium. This leads to the recruitment of other immune cells, including cluster of differentiation (CD)8 ⁺ and CD4 ⁺ T cells, also present in early and late atheroma. As such, cells of the innate and adaptive immune systems contribute to both systemic inflammation and vascular inflammation. On a molecular level, HIV-1 primes the NLRP3 (NLR family pyrin domain containing 3) inflammasome, leading to an increased expression of IL-1β, which is important for cardiovascular outcomes. Moreover, activation of TLRs (toll-like receptors) by HIV, gut microbes, and substance abuse further activates the NLRP3 inflammasome pathway. Finally, HIV proteins such as Nef (negative regulatory factor) can inhibit cholesterol efflux in monocytes and macrophages through direct action on the cholesterol transporter ABCA1 (ATP-binding cassette transporter A1), which promotes the formation of foam cells and the progression of atherosclerotic plaque. Here, we summarize the stages of atherosclerosis in the context of HIV, highlighting the effects of HIV, coinfections, and antiretroviral therapy on cells of the innate and adaptive immune system and describe current and future interventions to reduce residual inflammation and improve cardiovascular outcomes among people living with HIV.
Article
Full-text available
Type I and III interferons (IFNs) are essential for antiviral immunity and act through two different but complimentary pathways. First, IFNs activate intracellular antimicrobial programs by triggering the upregulation of a broad repertoire of viral restriction factors. Second, IFNs activate innate and adaptive immunity. Dysregulation of IFN production can lead to severe immune system dysfunction. It is thus crucial to identify and characterize the cellular sources of IFNs, their effects, and their regulation to promote their beneficial effects and limit their detrimental effects, which can depend on the nature of the infected or diseased tissues, as we will discuss. Plasmacytoid dendritic cells (pDCs) can produce large amounts of all IFN subtypes during viral infection. pDCs are resistant to infection by many different viruses, thus inhibiting the immune evasion mechanisms of viruses that target IFN production or their downstream responses. Therefore, pDCs are considered essential for the control of viral infections and the establishment of protective immunity. A thorough bibliographical survey showed that, in most viral infections, despite being major IFN producers, pDCs are actually dispensable for host resistance, which is achieved by multiple IFN sources depending on the tissue. Moreover, primary innate and adaptive antiviral immune responses are only transiently affected in the absence of pDCs. More surprisingly, pDCs and their IFNs can be detrimental in some viral infections or autoimmune diseases. This makes the conservation of pDCs during vertebrate evolution an enigma and thus raises outstanding questions about their role not only in viral infections but also in other diseases and under physiological conditions.
Article
Full-text available
Simple Summary IFNs are cytokines that play critical roles in the immune defense mechanisms that prevent virus spread. They also exhibit regulatory effects on the immune system and contribute to the elimination of malignant cells. These cytokines mediate their effects by binding on unique receptors on the surface of immune cells, initiating signaling cascades that trigger expression of IFN-stimulated genes (ISGs) that ultimately drive expression of specific proteins that mediate the various interferon responses and effects. In this review, we discuss the mechanisms by which interferons control different types of cells of the immune system, as well as malignant cells to mediate their important biological properties. Abstract This review addresses interferon (IFN) signaling in immune cells and the tumor microenvironment (TME) and examines how this affects cancer progression. The data reveal that IFNs exert dual roles in cancers, dependent on the TME, exhibiting both anti-tumor activity and promoting cancer progression. We discuss the abnormal IFN signaling induced by cancerous cells that alters immune responses to permit their survival and proliferation.
Article
Full-text available
A new method of total RNA isolation by a single extraction with an acid guanidinium thiocyanate-phenol-chloroform mixture is described. The method provides a pure preparation of undegraded RNA in high yield and can be completed within 4 h. It is particularly useful for processing large numbers of samples and for isolation of RNA from minute quantities of cells or tissue samples.
Article
Full-text available
A subset of CD4+CD11c−CD3− blood cells was recently shown to develop into dendritic cells when cultured with monocyte conditioned medium. Here, we demonstrate that CD4+ CD11c−CD3− cells, isolated from tonsils, correspond to the so-called plasmacytoid T cells, an obscure cell type that has long been observed by pathologists within secondary lymphoid tissues. They express CD45RA, but not markers specific for known lymphoid- or myeloid-derived cell types. They undergo rapid apoptosis in culture, unless rescued by IL-3. Further addition of CD40-ligand results in their differentiation into dendritic cells that express low levels of myeloid antigens CD13 and CD33.
Article
Full-text available
It is not known whether subsets of dendritic cells provide different cytokine microenvironments that determine the differentiation of either type-1 T helper (TH1) or TH2 cells. Human monocyte (pDC1)–derived dendritic cells (DC1) were found to induce TH1 differentiation, whereas dendritic cells (DC2) derived from CD4+CD3–CD11c–plasmacytoid cells (pDC2) induced TH2 differentiation by use of a mechanism unaffected by interleukin-4 (IL-4) or IL-12. The TH2 cytokine IL-4 enhanced DC1 maturation and killed pDC2, an effect potentiated by IL-10 but blocked by CD40 ligand and interferon-γ. Thus, a negative feedback loop from the mature T helper cells may selectively inhibit prolonged TH1 or TH2 responses by regulating survival of the appropriate dendritic cell subset.
Article
Full-text available
We evaluated the cellular immunity of 408 clinically stratified subjects at risk for acquired immune deficiency syndrome (AIDS), to define the role of interferon-alpha production deficits in the pathogenesis of opportunistic infections (OI). We followed 115 prospectively for up to 45 mo. Onset of OI was associated with, and predicted by, deficiency both of interferon-alpha generation in vitro, and of circulating Leu-3a+ cells. Interferon-alpha production is an index of the function of certain non-T, non-B, large granular lymphocytes (LGL) that are independent of T cell help. Leu-3a+ cell counts are a marker of T cell function. OI did not usually develop until both of these mutually independent immune functions were simultaneously critically depressed, leading to a synergistic interaction. These data suggest that the AIDS virus affects a subset of LGL, and that cytokine production by these cells is an important component of the host defense against intracellular pathogens that becomes crucial in the presence of severe T cell immunodeficiency.
Article
The producer cells of interferon were studied in human leucocyte cultures stimulated by a variety of stimulants, including phytohemagglutinin (PHA), pokeweed mitogen (PWM), Corynebacterium parvum (CP) and Herpes Simplex Virus (HSV). When the cells were fractionated by the use of neuraminidase-treated sheep red blood cells (SRBC), the T cell population responded with interferon production to PHA and PWM but not to CP or HSV. However, the non-T population showed a vigorous response to the latter two stimuli. In contrast, nylon column eluate cells enriched for T cells responded well to CP and HSV with production of interferon. To resolve these contradictory data, we have used combinations of techniques. Nylon column eluate cells were further separated by SRBC and it was found that the nylon non-adherent rosetting cell did not produce interferon in response to HSV or CP whereas the nylon-nonadherent non-rosetting cell did. In additional experiments more elaborate techniques were used. Leucocytes were treated by plastic adherence and iron filings, passed over a nylon column and subsequently over an Ig-anti-Ig column, and then rosetted with SRBC. Again only the non-rosetting population produced interferon. In parallel experiments the capacity of the different cell populations to lyse three types of target cells in a chromium release assay as a test for "natural killer" (NK) cell activity was investigated. There was some correlation between interferon production and NK cell activity. Thus, our data indicate that interferon is produced by non-T, non-B cells, possibly by cells related to NK cells.
Article
Herpes simplex virus-infected cells induce high interferon-alpha (IFN-alpha) production in infrequent cells among peripheral blood mononuclear cells (PBMC), designated natural IFN-alpha producing (NIP) cells. The properties of such NIP cells were compared with defined populations of leucocytes by means of flow cytometric analysis and sorting. The NIP cells are characterized as a discrete population of cells with high forward and low to intermediate orthogonal light scattering, similar to that of early progenitors of myeloid and lymphoid cells. However, they appear to lack the stem cell-associated molecule CD34. Furthermore, NIP cells cannot be localized to the myeloid line of cell differentiation, because they do not express the CD33, CD13, CD11b, CD15 or CD14 antigens. Neither do they express CD10 and CD19 antigens which are present in all stages of B-cell differentiation plasma cells excepted, nor CD7 antigens expressed on early T cells. In combination with previous results, our data support the view that the NIP cell is a unique and distinct cell type in peripheral blood, possibly with a physiological role in the defence against certain viral infections.
Article
Human HLA-DR+ peripheral mononuclear cells (PBMC) produce interferon-alpha (IFN-alpha) in response to herpes simplex virus type 1 (HSV-1) or HSV-1-infected fibroblasts (HSV-FS). We have developed a protocol, based partly on a technique known to enrich for dendritic cells, that allows for a greater than 125-fold enrichment of these IFN-alpha-producing cells. Nylon wood nonadherent PBMC (NWNA) were fractionated on a 48% Percoll gradient into low-density (LD) and high-density (HD) populations. The LD cells were 10- to 30-fold enriched for the production of IFN-alpha compared to PBMC when stimulated with HSV-FS. LD cells were treated further to deplete any contaminating monocytes, CD3+ T cells and CD56+ natural killer (NK) cells. The resulting population (CD3/CD56-depleted) produced approximately 30,000 IU/ml IFN-alpha compared to 3,000-10,000 IU/ml for the corresponding LD cells and 30-300 IU/ml for PBMC. Immunocytochemistry to detect cytoplasmic IFN-alpha indicated that PBMC, NWNA, HD, LD, and CD3/CD56-depleted populations contained an average of less than 0.1%, 0.3%, less than 0.1%, 3%, and 12% IFN-alpha-producing cells, respectively. The cells responsible for IFN-alpha production in response to HSV-1 were of medium to large diameter and possessed eccentric nuclei that were often indented, with lightly staining perinuclear areas. The CD3/CD56-depleted populations were fivefold enriched for HLA-DR+ cells. This enrichment procedure partially overcomes the barrier of low frequency that has contributed to the elusive identification of these cells.
Article
In this report, we characterize the major human peripheral blood nonadherent mononuclear cell subset that is responsible for the production in vitro of alpha-interferon (alpha IFN) in response to influenza, Sendai, and Newcastle disease viruses. Using a panel of anti-monocyte, anti-B, anti-T and anti-natural killer cell monoclonal antibodies to purify and recover both positive and negative cell populations, we show that the major alpha IFN-producing cells are HLA-DR(+) cells with no other surface markers characteristic of either lymphocytes or myelomonocytic cells. These cells copurify, on Percoll density gradients, with cells mediating NK activity, but the cell population responsible for alpha IFN production can be distinguished unambiguously from NK cells based on the former's reactivity with an anti-HLA-DR monoclonal antibody, the lack of reactivity with antibodies that detect the low-affinity receptor for IgG on human natural killer cells and granulocytes, and the inability to mediate spontaneous cytotoxicity. Double immunofluorescence assays indicate that cells of this subset, the lineage of which is as yet undetermined but which might be related to dendritic cells, constitute a minor proportion (approximately 1-1.5%) of the nonadherent mononuclear cells from healthy donors.
Article
Human blood lymphocytes, exposed for 6 to 24 h in vitro to tumor cells (K 562, IGR3, L1210), Herpes simplex virus type 1 (HSV) or Corynebacterium parvum (CP), produced high levels of anti-viral activity which was identified as type-1 interferon (IF). In mixed lymphocyte tumor cell cultures (MLTC), the generated type-1 IF was definitely shown to originate from the lymphocytes and not from the tumor cells. Supplementation of leukocyte cultures with 10% fetal calf serum instead 10% human AB serum had little influence on tumor cell-induced IF production, but strongly reduced CP-induced IF production. Lymphocyte fractionation procedures involving iron/plastic treatment, nylon wool columns, Ig-anti-Ig columns and rosette (E, EA) separation led to the identification of null cells as highly efficient producers of type-1 IF. T cells obtained by different ways (E-rosette sedimentation, passage through 1 nylon and 2 Ig-anti-Ig columns, or thoracic duct lymphocytes) were poor IF producers in response to tumor cells, HSV and CP, but secreted anti-viral activity when stimulated with phytohemagglutinin. In MLTC, the level of generated type-1 IF roughly stimulated with phytohemagglutinin. In MLTC, the level of generated type-1 IF roughly paralleled nautral killer (NK) cell activity. Evidence is presented that type-1 IF can be produced by an Fc receptor-negative null cell subset, whereas NK activity requires Fc receptor-positive cells. It is suggested that production of type-1 IF represents one of the earliest functions in the differentiation process of mononuclear phagocytes and is likely to develop before the appearance of Fc receptors, diffuse esterase staining and latex phagocytosis.
Article
Monoclonal antibodies with specificities for subsets of human leucocytes have been used for the characterization of alpha-interferon (alpha-IFN) producing cells. The production of alpha-IFN was demonstrated to be a function of Ia+ leucocytes. OKT3+ T lymphocytes, BA-1+ B lymphocytes and Leu 7+ natural killer (NK) cells did not contribute to the production of alpha-IFN. OKM1+ monocytes were essential for the production of alpha-IFN in response to bacterial products or leukaemia cells, but were not required for the synthesis of virus- or poly I:C-induced alpha-IFN. The results indicate that alpha-IFN producing cells represent a heterogenous population of cells of the myeloid lineage.