• Perforin deficiency causes immune dysregulation and hemophagocytic lymphohistiocytosis, which requires allogeneic HCT for long-term cure.

  • In prf−/− mice, wild-type hematopoietic or CD8+ T-cell engraftment of only 10-20% is sufficient to reestablish normal immune regulation.

Defects in perforin and related genes lead to a loss of normal immune regulation and underlie hemophagocytic lymphohistiocytosis (HLH), which requires hematopoietic cell transplantation for long-term cure. However, transplantation may be complicated by the development of mixed chimerism and uncertainty regarding the risk of HLH recurrence. To help clarify this risk and investigate how perforin influences immune activation, we studied perforin-mediated immune regulation in the context of mixed chimerism using a murine model of HLH. We found that there is a distinct threshold of ∼10% to 20% perforin expression with either mixed hematopoietic or CD8+ T cell chimerism, above which immune regulation was reestablished. These findings demonstrate that perforin-mediated immunoregulation functions in trans and are consistent with a feedback model in which cytotoxic T cells control immune activation by killing dendritic cells. These findings also suggest rational targets for maintenance of minimal posttransplant chimerism and for therapeutic strategies involving gene correction.

Hemophagocytic lymphohistiocytosis (HLH) is a unique immunoregulatory disorder in which defects in perforin-dependent cytotoxicity lead to a syndrome of fatal immune activation.1,2  Short-term treatment of HLH involves immunosuppressive therapies, while long-term treatment requires allogeneic hematopoietic cell transplantation (HCT) to prevent disease recurrence. Historically, transplant-related mortality has been unusually high in this patient population.3,4  Use of reduced-intensity conditioning regimens is associated with improved survival, but this approach has been complicated by a high incidence of mixed chimerism and secondary graft loss in patients with HLH.5,6  Anecdotal evidence has suggested that stable mixed chimerism is sufficient for long-term cure.7,8  However, because clinical data are extremely limited, it remains uncertain whether there is a threshold of donor chimerism above which patients are protected from HLH recurrence.

To better understand how perforin regulates the immune response and how HCT may restore this regulation, we studied the immune reactivity of perforin deficient (prf−/−) mice engrafted with varying amounts of wild-type (WT) hematopoietic or T cells and challenged with lymphocytic choriomeningitis virus (LCMV) infection. We observed that there was a distinct threshold, 10% to 20% engraftment of WT cells, that reestablished perforin-dependent immunoregulation. Furthermore, our studies also demonstrated that perforin expression in CD8+ T cells was necessary and sufficient to restore normal immune regulation.

For transplantation, animals were lethally irradiated and injected with 107 marrow cells, varying proportions of WT, and prf−/− marrow. WT cells were marked with green fluorescent protein (GFP) or CD45.1. For recombination activating gene (RAG) complementation studies, animals were administered 9 × 106 RAG−/− (or RAG/prf−/−) marrow cells along with a mixture of WT and prf−/− marrow (1 × 106). After waiting 12 to 16 weeks, animals were challenged with LCMV-WE infection (200 plaque-forming units intraperitoneally). For adoptive transfer, magnetically purified (Miltenyi) CD8+ or CD4+ T cells from B6.SJL/BoyJ mice (prf+/+ or prf−/−) were transferred 2 days after administering cyclophosphamide (100 mg/kg intraperitoneally). Assessment of serum interferon (IFN)-γ levels, ex vivo dendritic cell antigen presentation assays, and in vivo T-cell IFN-γ measurement was as described.2,9  All studies were repeated at least 3 times with consistent results. All studies were performed on an Institutional Animal Care and Use Committee-approved protocol.

WT and prf−/− mice display divergent immune responses to LCMV infection.10  Though WT animals mount a vigorous response, prf−/− animals display unusually strong immune and T-cell activation, which lead to fatal immunopathology.9  This immune-mediated pathology is widely recognized as an experimental model of HLH.10-15  A critical aspect of this pathologic response is prolonged and heightened production of IFN-γ. To study the effects of marrow transplantation in this model system, we lethally irradiated and reconstituted prf−/− mice with either prf−/− or WT bone marrow. After waiting for immune reconstitution, we challenged these transplanted animals with LCMV infection. As displayed in Figure 1A, we found that transplantation with WT (perforin-expressing) marrow led to an IFN-γ response pattern in these animals that was very similar to that seen in intact WT animals.10  Thus, similar to what is clinically seen, HCT with perforin-expressing marrow restores perforin-mediated immunoregulation in experimental animals. Next, we generated a series of mixed hematopoietic chimeric mice using WT and prf−/− marrow. For these studies, we transplanted a fixed amount of total marrow but varied the proportions coming from each donor. When we plotted the post-LCMV day 8 IFN-γ level against the percent of WT chimerism, we observed a distinctive relationship between these 2 parameters (Figure 1B). At low levels of WT chimerism, IFN-γ levels were very elevated and similar to those seen in animals with complete prf−/− hematopoiesis. At WT chimerism levels >10% to 20%, cytokine levels were similar to those observed in animals reconstituted entirely with WT marrow. When we assessed mixed chimeric animals for the development of HLH, we found that those with WT chimerism >20% were protected from the development of severe anemia and death after LCMV infection (supplemental Figure 1, available on the Blood Web site).

Figure 1

Restoration of normal perforin expression in a fraction of hematopoietic cells restores perforin-dependent immune regulation in vivo. (A) Prf−/− mice were transplanted with WT (GFP marked) or prf−/− bone marrow. Twelve weeks later, they were infected with LCMV and serum IFN-γ levels were measured. N > 8 mice/point. (B) Mixed hematopoietic chimeric mice (prf−/− mice, reconstituted with mixtures of WT and prf−/− marrow) were challenged with LCMV, and day 8 serum IFN-γ levels were measured. (C) Six days after LCMV infection, splenic dendritic cells were sorted from groups of mixed chimeric mice (N = 3/group) and plated with LCMV-specific T cells. After overnight culture, IFN-γ production was assessed as a measure of antigen presentation. *P < .01 (comparing WT and prf curves in panel A; comparing the 2 upper curves with the lower 2 curves in panel C).

Figure 1

Restoration of normal perforin expression in a fraction of hematopoietic cells restores perforin-dependent immune regulation in vivo. (A) Prf−/− mice were transplanted with WT (GFP marked) or prf−/− bone marrow. Twelve weeks later, they were infected with LCMV and serum IFN-γ levels were measured. N > 8 mice/point. (B) Mixed hematopoietic chimeric mice (prf−/− mice, reconstituted with mixtures of WT and prf−/− marrow) were challenged with LCMV, and day 8 serum IFN-γ levels were measured. (C) Six days after LCMV infection, splenic dendritic cells were sorted from groups of mixed chimeric mice (N = 3/group) and plated with LCMV-specific T cells. After overnight culture, IFN-γ production was assessed as a measure of antigen presentation. *P < .01 (comparing WT and prf curves in panel A; comparing the 2 upper curves with the lower 2 curves in panel C).

Close modal

We have recently shown that perforin-dependent immunoregulation involves the suppression of antigen presentation and loss of antigen-presenting dendritic cells.2,9  To test whether this process was being restored, we sorted and assessed antigen presentation by splenic dendritic cells from LCMV-infected chimeric animals. When we cultured these dendritic cells with transgenic, LCMV-specific T cells, we found that presentation of endogenously acquired viral antigen was suppressed in animals with >10% WT chimerism to levels at or below those seen in intact WT animals (Figure 1C).2  Thus, a distinct threshold exists (>10% to 20% prf+/+ hematopoietic chimerism), above which perforin-dependent immune regulation is reestablished.

Our prior studies and anecdotal clinical data have suggested that T cells play an important role in perforin-mediated immunoregulation.8,9,16  To assess the role of T cells, we generated mice with variable perforin expression in the T-cell and non–T-cell compartments. We reconstituted prf−/− mice with 90% RAG−/−/prf+/+ or RAG−/−/prf−/− marrow, along with 10% of a varying mixture of WT and prf−/− marrow. The resulting 3-way chimeric mice developed a T-cell compartment with variable proportions of prf+/+ genotype and 90% to 100% of non–T/B cells (myeloid, natural killer, etc) that were either prf+/+ or prf−/−. When challenged with LCMV, these animals demonstrated that perforin expression in only T cells was sufficient to control cytokine production (Figure 2A). Contribution of WT marrow to non–T (or B) cells ranged from 0% to 3% in most recipients (data not shown). Furthermore, when prf+/+/RAG−/− donors were used, giving normal perforin expression in the non–T-cell compartment (>90%), there was no additional effect on IFN-γ levels (Figure 2B). Thus, perforin expression in the T-cell compartment is uniquely necessary and sufficient for establishing perforin-dependent immune regulation. To assess the specific role of Foxp3+ regulatory T cells and CD1-restricted natural killer T cells, we performed studies using donor marrow or T cells from scurfy, Jalpha281−/−, and CD1−/− mice and observed similar results as with WT donor marrow, suggesting that neither of these T-cell subsets was critical (supplemental Figure 2).

Figure 2

Restoration of normal perforin expression in CD8+ T cells is necessary and sufficient to restore perforin-dependent immune regulation. (A) Prf−/− mice were reconstituted with 90% RAG/prf−/− (DKO) marrow and 10% mixed marrow (variable amounts of WT [GFP+] and prf−/−) to give animals with variable perforin expression within the T-cell compartment and perforin expression in <10% of non–T cells. After engraftment, animals were challenged with LCMV as in Figure 1, and day 8 IFN-γ levels are plotted against WT chimerism within the T-cell compartment. (B) Prf−/− mice were reconstituted with 90% RAG−/− marrow and 10% mixed marrow (WT [GFP+] and prf−/−) to give animals with variable perforin expression within the T-cell compartment and normal perforin expression in 90% to 100% of non–T cells. IFN-γ levels and chimerism are plotted as in panel A. (C) Purified naïve WT (CD45.1+) or prf−/− CD4+ T cells were transferred into prf−/− mice (after conditioning with cyclophosphamide and waiting 3 weeks for recovery/stable engraftment). Animals were challenged with LCMV; IFN-γ levels and WT CD4+ T cell chimerism were assessed. (D) Purified naïve WT (CD45.1+) CD8+ T cells were transferred into prf−/− mice (as in panel C). Animals were challenged with LCMV; IFN-γ levels and WT CD8+ T-cell chimerism were assessed. In vivo IFN-γ production by endogenous (prf−/−) T cells was assessed on day 8 after LCMV in animals receiving CD8+ T-cell transfers, as previously described.9  *P < .01.

Figure 2

Restoration of normal perforin expression in CD8+ T cells is necessary and sufficient to restore perforin-dependent immune regulation. (A) Prf−/− mice were reconstituted with 90% RAG/prf−/− (DKO) marrow and 10% mixed marrow (variable amounts of WT [GFP+] and prf−/−) to give animals with variable perforin expression within the T-cell compartment and perforin expression in <10% of non–T cells. After engraftment, animals were challenged with LCMV as in Figure 1, and day 8 IFN-γ levels are plotted against WT chimerism within the T-cell compartment. (B) Prf−/− mice were reconstituted with 90% RAG−/− marrow and 10% mixed marrow (WT [GFP+] and prf−/−) to give animals with variable perforin expression within the T-cell compartment and normal perforin expression in 90% to 100% of non–T cells. IFN-γ levels and chimerism are plotted as in panel A. (C) Purified naïve WT (CD45.1+) or prf−/− CD4+ T cells were transferred into prf−/− mice (after conditioning with cyclophosphamide and waiting 3 weeks for recovery/stable engraftment). Animals were challenged with LCMV; IFN-γ levels and WT CD4+ T cell chimerism were assessed. (D) Purified naïve WT (CD45.1+) CD8+ T cells were transferred into prf−/− mice (as in panel C). Animals were challenged with LCMV; IFN-γ levels and WT CD8+ T-cell chimerism were assessed. In vivo IFN-γ production by endogenous (prf−/−) T cells was assessed on day 8 after LCMV in animals receiving CD8+ T-cell transfers, as previously described.9  *P < .01.

Close modal

Finally, we tested the therapeutic effects of adoptive T-cell transfer into intact (nontransplanted) prf−/− mice. To achieve sufficient levels of donor T-cell engraftment, we lympho-depleted recipients with cyclophosphamide before transferring varying numbers of purified WT or prf−/− T cells and then waited 3 weeks for animals to recover before challenging with LCMV. When we transferred WT CD4+ T cells, we did not observe a clear trend for rescue (Figure 2C). However, when we transferred purified WT CD8+ T cells (but not prf−/− T cells) (supplemental Figure 2), we observed robust rescue when donor T-cell chimerism exceeded 10% to 20% (Figure 2D). Furthermore, if we examined in vivo IFN-γ production by endogenous (host) CD8+ T cells in recipients, we observed clear suppression in recipients with engraftment of >5% to 10% WT T cells (Figure 2D). Thus, perforin-mediated immune regulation functions in trans; perforin-sufficient cells are able to control the immune hyperactivation observed in perforin-deficient T cells.

We conclude that normal perforin expression in only 10% to 20% of hematopoietic cells, or within the conventional CD8+ T-cell compartment alone, is necessary and sufficient to reestablish normal immune regulation in mice. This finding is an important addition to limited long-term clinical data, consisting of only a few patients with stable donor chimerism in this range.7,8,17  However, additional clinical data are still needed to guide decision making. Our findings also suggest that perforin deficiency would be amenable to gene correction strategies and provide a target level for future studies.

The online version of this article contains a data supplement.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

This work was supported by National Institutes of Health: National Heart, Lung, and Blood Institute (RO1-HL091769) and a grant from the Histiocytosis Association.

Contribution: C.E.T. designed and performed experiments and analyzed data; and M.B.J. designed and performed experiments, analyzed data, and wrote the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Michael B. Jordan, Divisions of Cellular and Molecular Immunology, and Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital, 3333 Burnet Ave, ML 7038, Cincinnati, OH 45229; e-mail: michael.jordan@cchmc.org.

1
Jordan
 
MB
Allen
 
CE
Weitzman
 
S
Filipovich
 
AH
McClain
 
KL
How I treat hemophagocytic lymphohistiocytosis.
Blood
2011
, vol. 
118
 
15
(pg. 
4041
-
4052
)
2
Terrell
 
CE
Jordan
 
MB
Perforin deficiency impairs a critical immunoregulatory loop involving murine CD8(+) T cells and dendritic cells.
Blood
2013
, vol. 
121
 
26
(pg. 
5184
-
5191
)
3
Baker
 
KS
Filipovich
 
AH
Gross
 
TG
, et al. 
Unrelated donor hematopoietic cell transplantation for hemophagocytic lymphohistiocytosis.
Bone Marrow Transplant
2008
, vol. 
42
 
3
(pg. 
175
-
180
)
4
Trottestam
 
H
Horne
 
A
Aricò
 
M
, et al. 
Histiocyte Society
Chemoimmunotherapy for hemophagocytic lymphohistiocytosis: long-term results of the HLH-94 treatment protocol.
Blood
2011
, vol. 
118
 
17
(pg. 
4577
-
4584
)
5
Cooper
 
N
Rao
 
K
Goulden
 
N
Webb
 
D
Amrolia
 
P
Veys
 
P
The use of reduced-intensity stem cell transplantation in haemophagocytic lymphohistiocytosis and Langerhans cell histiocytosis.
Bone Marrow Transplant
2008
, vol. 
42
 
Suppl 2
(pg. 
S47
-
S50
)
6
Marsh
 
RA
Jordan
 
MB
Filipovich
 
AH
Reduced-intensity conditioning haematopoietic cell transplantation for haemophagocytic lymphohistiocytosis: an important step forward.
Br J Haematol
2011
, vol. 
154
 
5
(pg. 
556
-
563
)
7
Ouachée-Chardin
 
M
Elie
 
C
de Saint Basile
 
G
, et al. 
Hematopoietic stem cell transplantation in hemophagocytic lymphohistiocytosis: a single-center report of 48 patients.
Pediatrics
2006
, vol. 
117
 
4
(pg. 
e743
-
e750
)
8
Cooper
 
N
Rao
 
K
Gilmour
 
K
, et al. 
Stem cell transplantation with reduced-intensity conditioning for hemophagocytic lymphohistiocytosis.
Blood
2006
, vol. 
107
 
3
(pg. 
1233
-
1236
)
9
Lykens
 
JE
Terrell
 
CE
Zoller
 
EE
Risma
 
K
Jordan
 
MB
Perforin is a critical physiologic regulator of T-cell activation.
Blood
2011
, vol. 
118
 
3
(pg. 
618
-
626
)
10
Jordan
 
MB
Hildeman
 
D
Kappler
 
J
Marrack
 
P
An animal model of hemophagocytic lymphohistiocytosis (HLH): CD8+ T cells and interferon gamma are essential for the disorder.
Blood
2004
, vol. 
104
 
3
(pg. 
735
-
743
)
11
Pachlopnik Schmid
 
J
Ho
 
CH
Chrétien
 
F
, et al. 
Neutralization of IFNgamma defeats haemophagocytosis in LCMV-infected perforin- and Rab27a-deficient mice.
EMBO Mol Med
2009
, vol. 
1
 
2
(pg. 
112
-
124
)
12
Jessen
 
B
Maul-Pavicic
 
A
Ufheil
 
H
, et al. 
Subtle differences in CTL cytotoxicity determine susceptibility to hemophagocytic lymphohistiocytosis in mice and humans with Chediak-Higashi syndrome.
Blood
2011
, vol. 
118
 
17
(pg. 
4620
-
4629
)
13
Pham
 
NL
Badovinac
 
VP
Harty
 
JT
Epitope specificity of memory CD8+ T cells dictates vaccination-induced mortality in LCMV-infected perforin-deficient mice.
Eur J Immunol
2012
, vol. 
42
 
6
(pg. 
1488
-
1499
)
14
Krebs
 
P
Crozat
 
K
Popkin
 
D
Oldstone
 
MB
Beutler
 
B
Disruption of MyD88 signaling suppresses hemophagocytic lymphohistiocytosis in mice.
Blood
2011
, vol. 
117
 
24
(pg. 
6582
-
6588
)
15
Sepulveda
 
FE
Debeurme
 
F
Ménasché
 
G
, et al. 
Distinct severity of HLH in both human and murine mutants with complete loss of cytotoxic effector PRF1, RAB27A, and STX11.
Blood
2013
, vol. 
121
 
4
(pg. 
595
-
603
)
16
Almousa
 
H
Ouachée-Chardin
 
M
Picard
 
C
, et al. 
Transient familial haemophagocytic lymphohistiocytosis reactivation post-CD34 haematopoietic stem cell transplantation.
Br J Haematol
2005
, vol. 
130
 
3
(pg. 
404
-
408
)
17
Marsh
 
RA
Vaughn
 
G
Kim
 
MO
, et al. 
Reduced-intensity conditioning significantly improves survival of patients with hemophagocytic lymphohistiocytosis undergoing allogeneic hematopoietic cell transplantation.
Blood
2010
, vol. 
116
 
26
(pg. 
5824
-
5831
)
Sign in via your Institution