ArticlePDF Available

Hematopoietic stem cell transplantation rescues the immunologic phenotype and prevents vasculopathy in patients with adenosine deaminase 2 deficiency

Authors:

Figures

Content may be subject to copyright.
Letter to the Editor
Hematopoietic stem cell transplantation res-
cues the immunologic phenotype and prevents
vasculopathy in patients with adenosine
deaminase 2 deficiency
To the Editor:
Recently, recessively inherited loss-of-function mutations in
CECR1 (cat eye syndrome chromosome region, candidate 1),
which encodes adenosine deaminase 2 (ADA2), were identified
in patients with a complex immunologic and vascular pheno-
type.
1,2
Possible mechanisms for this disorder are proinflamma-
tory polarization and disturbed endothelial integrity.
1,2
Zhou
et al
1
reported that aggressive systemic immunosuppressive
treatment was not effective in controlling inflammation but
hypothesized that hematopoietic stem cell transplantation
(HSCT) might be curative given that bone marrow–derived mono-
cytes and macrophages are the main source of secreted ADA2.
Here we report on 2 related patients with homozygous
p.Arg169Gln missense mutations in ADA2 located within the
putative receptor-binding domain.
3
Our observations in these sib-
lings demonstrate the clinical heterogeneity associated with
ADA2 deficiency and show that HSCT can be an effective
therapy. In the index patient the clinical course was dominated
by autoimmunity and lymphoproliferation with a combined
immunodeficiency–like phenotype, which prompted HSCT
from a healthy sibling. Despite early complications, transplanta-
tion was successful both in rescuing the immunologic phenotype
and in preventing vascular disease; at 5 years after HSCT, the
patient remains off treatment.
The index patient (P1) was the second child of a father of
Moroccan descent and a white mother. He was first admitted at
age 6 months for complicated human respiratory syncytial virus
infection. At this time, hypogammaglobulinemia was noted (see
Table E1 in this article’s Online Repository at www.jacionline.
org). At age 12 months, P1 presented with fever, lymphadenitis,
generalized lymphadenopathy, and hepatosplenomegaly. Staphy-
lococcus aureus was cultured from the lymph nodes, and fever
resolved within 24 hours of starting amoxicillin–clavulanic
acid treatment. Pancytopenia, hypogammaglobulinemia, and the
absence of specific antibodies were found (see Table E1). Results
of blood PCRs for EBV, cytomegalovirus, human herpesvirus
(HHV) 6, HHV-8, and adenovirus were negative. However,
adenovirus and norovirus were detected in the stool. Computed
tomographic scans confirmed generalized lymphoproliferation
with mediastinal and intra-abdominal lymphadenopathy and
splenomegaly. Lymphoma was suspected, but the results of lymph
node biopsy and bone marrow examination were normal. Macro-
phage activation syndrome as the cause of the pancytopenia and
lymphoproliferation was excluded based on serum markers
(including soluble IL-2 receptor) and the absence of hemo-
phagocytosis on bone marrow examination. A primary immune
deficiency (PID) with predominant lymphoproliferation and
autoimmunity was suspected, and known genetic causes
were excluded. Prednisone (2 mg/kg) led to resolution of the
thrombocytopenia and splenomegaly. However, attempts to taper
led to a relapse of thrombocytopenia. Despite the addition of
mycophenolate mofetil, sirolimus, tacrolimus, cyclosporine,
and mercaptopurine, the cytopenia and lymphoproliferation
persisted.
Because of growth failure secondary to chronic corticosteroid
treatment, HSCT was considered at the age of 3 years. The
patient’s HLA-identical healthy elder brother was chosen as the
donor. After conditioning with oral busulfan and cyclophospha-
mide, 7.5 310
6
CD34
1
bone marrow–derived hematopoietic
stem cells per kilogram were infused. Anti–graft-versus-host-
disease (GvHD) prophylaxis consisted of cyclosporine, whereas
steroids were slowly tapered. Antiviral prophylaxis consisting
of acyclovir and intravenous immunoglobulin (IVIG) administra-
tion and antifungal prophylaxis with fluconazole was added.
The transplantation was complicated by late engraftment of
neutrophils (day 26 <1.5 310
9
/L) and persistent severe
thrombocytopenia (<10 310
9
/L) refractory to transfusion,
although at day 28, whole blood chimerism was greater than
95%. At day 36, magnetic resonance imaging (MRI) of the brain,
which was performed because of severe sudden-onset headache,
identified a pineal gland hemorrhage (see Fig E1,A,in
this article’s Online Repository at www.jacionline.org). The
thrombocyte level was 2 310
9
/L but increased to greater
than 50 310
9
/L at day 40 after 2 infusions of rituximab.
Veno-occlusive disease (VOD) was diagnosed according to the
Seattle criteria at day 60 and was accompanied by a relapse of
thrombocytopenia. VOD responded well to fluid restriction.
Platelet levels of greater than 100 310
9
/L were reached at
day 111. Adenovirus reactivation was found at day 40, with
accompanying intestinal GvHD grade III, which responded to
corticosteroids. Cyclosporine was stopped at day 150. IVIG was
discontinued at day 180. Immunoreconstitution at day 360 was
excellent, including normal antibody levels, normal numbers
of B- and T-lymphocytes, and normal T-cell proliferation in
response to PHA. Moreover, response to polysaccharide vaccine
was normal (data not shown).
Five years after transplantation, P1 is clinically well and off all
medication. No more lymphoproliferation has occurred, and the
most recent MRI of the brain 5 years after HSCT did not show any
signs of acute or chronic small infarcts.
Two years after transplantation of P1, his younger brother (P2)
presented at age 5 months with profound Coombs (2) anemia
(hemoglobin, 2 g/dL), which was attributed to PCR-verified
HHV-6–associated erythroblastopenia. At this time, immuno-
logic analysis of P2 was normal. Several episodes of PCR-verified
facial herpes simplex virus infection followed. At age 23 months,
P2 was admitted with abdominal pain and ileus refractory to
conservative treatment. He had generalized lymphadenopathy
and hepatosplenomegaly, as well as hypogammaglobulinemia
and intermittent lymphopenia and neutropenia (see Table E1).
Results of blood polyomavirus PCR were positive. Bone marrow
examination was normal. Partial enterectomy was performed;
biopsy showed an atypical ulcerative bowel disease devoid of
plasma cells (see Fig E1,B), as can be seen in patients with
common variable immunodeficiency.
4
No cytomegalovirus,
Ó2014 The Authors. Published by Elsevier Inc. on behalf of the American Academy of
Allergy, Asthma & Immunology. This is an open access article under the CC BY-NC-
ND license (http://creativecommons.org/licenses/by-nc-nd/3.0/).
1
EBV, herpes simplex virus, HHV-6, polyomavirus, or adenovirus
could be detected in the biopsy specimen, and no signs of vascu-
litis could be observed in the entire surgical specimen.
Obstruction persisted despite aggressive systemic immunosup-
pressive treatment and was only relieved after treatment with
sirolimus. At this time, IVIG was started, and sirolimus was slowly
tapered without clinical relapse. Subsequently, P2 did not receive
any immunosuppressive treatment for a period of 13 months but
was solely treated with IVIG. At 3.5 years of age, P2 had
neurologic manifestations in the form of 2 episodes of acute loss
of balance in the absence of fever or signs of systemic
inflammation on blood analysis, Repeated MRI of the brain did
not reveal any lesions compatible with ischemic or hemorrhagic
stroke. A transient ischemic attack (TIA) was suspected, and
treatment with sirolimus was restarted.
Whole-exome sequencing was performed on the untreated
patient (P2), the parents, and the healthy sibling (for details, see
the Methods section in this article’s Online Repository at www.
jacionline.org). We hypothesized a recessive model of inheri-
tance. After filtering out common polymorphisms, we identified
a homozygous c.G506A variant in CECR1, resulting in a
p.Arg169Gln missense mutation in ADA2. Sanger sequencing
on DNA obtained from the cheek swab of the patient who under-
went transplantation confirmed that he was also homozygous for
this variant. Both parents were carriers, whereas the sibling donor
was homozygous for the wild-type form of CECR1 (see Fig E2 in
this article’s Online Repository at www.jacionline.org).
ADA2 enzyme activity in plasma (Table I) was essentially
absent in P2, the patient who did not undergo transplantation,
whereas in post-HSCT plasma from P1, ADA2 activity was
comparable with that of his healthy donor and in the range for
healthy control subjects. Both parents have intermediate plasma
ADA2 activity. Of note, neither adenosine nor deoxyadenosine
levels were increased (<0.4 mmol/L) in plasma of P2 (these levels
have not been measured in previous patients). Both P1 and P2 had
normal ADA1 activity in dried blood spots, and deoxyadenosine
nucleotides were undetectable.
Although it has been speculated that the clinical consequences
of ADA2 deficiency might be due to increased extracellular
adenosine, our findings suggest this is not the case and that ADA2
actually has a minimal role compared with ADA1 in adenosine
metabolism in vivo, which is consistent with the very different
substrate affinities of the 2 ADA enzymes (see the Methods
section in this article’s Online Repository).
Because of the observed immunodeficiency, we performed
extensive profiling of peripheral immune cells of P2 (for details,
see the Methods section in this article’s Online Repository). Of
the major mononuclear leukocyte cell types surveyed, CD4
1
T-cell numbers were increased and CD8
1
T-cell numbers were
reduced in P2 compared with those in healthy age-matched con-
trol subjects. B-cell, natural killer cell, and dendritic cell numbers
were within 1 SD of the mean of the healthy control subjects
(Fig 1,A). Within the T-lymphocyte population, we found
defective T-cell activation, with increased naive and low effector
and memory subsets (Fig 1,Band D). Within the T
H
cell
population, numbers of regulatory T cells were increased,
whereas T
H
1, T
H
2, and follicular helper CD4
1
T-cell numbers
were low (Fig 1,C). T-cell proliferation in response to
Candida species, tetanus, and PHA was normal (data not shown).
Within the B-lymphocyte population, naive B-cell numbers were
increased at the expense of memory and plasmablasts (Fig 1,E),
which is suggestive of a defect in B-lymphocyte differentiation or
T-cell provision of help. Limited immunoprofiling performed
before HSCT showed similar findings in P1 (see Table E2 in
this article’s Online Repository at www.jacionline.org).
Because ofthe presence of severe inflammation inP1, serum IL-6
levels were measured from initial evaluation to last follow-up
(Fig 1,F). IL-6 levels were persistently high before HSCT and
before engraftment, but after HSCT IL-6 levels slowly decreased
and were undetectable at 3 years post-HSCT. In P2 serum IL-6
levels were extremely increased, despite the absence of clinical
signs of inflammation, with levels peaking at the time of bowel
obstruction and at the time of the suspected TIAs. IL-6 was unde-
tectable inthe healthy sibling and in healthy control subjects. More-
over, TNF-awas not detectable in the serum of P1 andP2 at the time
of the highest IL-6 levels. The immune profile of the other family
members wasnormal (data not shown). Together, these data demon-
strate a profound defect in T cell–dependent antibody-mediated
responses and a failure to regulate normal inflammatory cytokine
production in ADA2-deficient patients, adding to the previously
identified function of ADA2 in in vitro stimulation of T
H
cells.
5
PIDs with autoimmunity and lymphoproliferation dominated
the clinical image in our patients. The index patient P1 presented
with persistent autoimmune pancytopenia and lymphoprolifera-
tion, whereas P2 had an episode of lymphoproliferation, bowel
involvement, and 2 possible TIAs. Both patients only had fever
during infectious episodes, and unlike previously reported
patients, neither showed skin involvement or clear signs of
vasculitis. P1 had a stroke as an apparent early complication of
HSCT in the context of prolonged and severe thrombocytopenia.
Only 3 years after initial presentation, P2 presented with 2
potential TIAs, although transient labyrinthitis caused by a viral
infection could not be excluded. Therefore in retrospect
vasculitis and inflammation might have been present at
a subclinical level in both patients, but vasculopathy and
inflammation did not dominate the clinical presentation, as is
the case in the patients reported by Zhou et al
1
and Elkan et al.
2
Interestingly, serum IL-6 levels were increased in both patients
in the absence of clinical and (routine) biochemical signs of
inflammation. This suggests that ADA2 deficiency might lead
to a subclinical state of inflammation. This phenotypic
discrepancy cannot be explained entirely by CECR1 genotype
because the p.Arg169Gln mutation was previously observed in
hemizygous and homozygous form.
1,2
The ADA2-deficient
patients previously described had decreased serum immuno-
globulin levels and enhanced B-cell apoptosis in vitro.
1
By
TABLE I. Plasma ADA2 activity in the affected pedigree
Sample Age (y)
Plasma ADA2 activity
(mU/mL)
Patient 1 after HSCT 8 22.07
Patient 2 3 0.11
Healthy sibling (5HSCT donor) 10 19.14
Father 43 7.20
Mother 40 2.91
Reference values for plasma ADA2 activity (mU/mL), mean 6SD
(min-max)
ADA2 deficient (n 54) 1.0 60.4 (0.6-1.4)
ADA2 carriers (n 54) 4.9 60.3 (4.6-5.3)
Control subjects (n 551pooled human plasma) 14.0 66.1 (4.8-21.3)
J ALLERGY CLIN IMMUNOL
nnn 2014
2LETTER TO THE EDITOR
FIG 1. Serum IL-6 levels and immunoprofiling in ADA2-deficient patients. A, Major blood leukocyte subsets.
B, CD4
1
T-lymphocyte subsets. C, T
H
cell lineages. D, CD8
1
T-lymphocyte subsets. E, B-cell subsets. P2’s
values are shown as filled circles, and values of healthy age-matched control subjects are indicated by
open circles. Means and SDs (error bars) shown exclude values for the patient. F, IL-6 levels in sera of P1
and P2. The vertical line indicates the moment of HSCT followed by pineal stroke in P1. The gray shading
indicates the periods in which P2 was treated with sirolimus. DC, Dendritic cell; mDC, myeloid dendritic
cell; NK, natural killer cell; NKT, natural killer T cell; pDC, plasmacytoid dendritic cell; RTE, recent thymic
emigrant; TCM, central memory T cell; TEM, effector memory T cell; TEMRA, CD45RA-expressing effector
memory T cell; Tfh, follicular T cell; Th17, IL-17–expressing helper T cell; Treg, regulatory T cell.
J ALLERGY CLIN IMMUNOL
VOLUME nnn, NUMBER nn
LETTER TO THE EDITOR 3
contrast, our patients had abnormalities suggesting an in vivo
defect in T-cell activation and proliferation, corresponding to
their increased susceptibility to viral infections and combined
immunodeficiency–like phenotype. Taken together, these
observations suggest that ADA2 deficiency has a more varied
clinical phenotype than initially reported and that the diagnosis
should be considered in cases of undiagnosed PID characterized
by lymphoproliferation and autoimmunity, even in the absence
of overt vasculopathy or inflammation.
As reported by Zhou et al,
1
we found that treatment with a
variety of immunosuppressive medications resulted in poor
disease control in P1. However, both at the time of bowel
obstruction and at the time of potential TIA, P2 seemed to
respond well to sirolimus treatment. Sirolimus reduces M1
macrophage differentiation and IL-6 production.
6
Because
ADA2 deficiency drives macrophages toward a more
proinflammatory M1 profile,
1
we present sirolimus as a
potential therapeutic option to at least temporarily control
inflammatory complications in ADA2-deficient patients. TNF-a
was undetectable in the serum of our patients. However, this
finding does not at all exclude a role for this cytokine in disease
pathogenesis. Indeed, etanercept led to a significant response in
all patients reported by Elkan et al
2
and should therefore be
considered as a potential treatment.
In the index patient P1 we successfully performed an
allogeneic HSCT. At 5 years after HSCT, consecutive clinical
and biochemical investigations in P1 have shown no signs of
immunologic disorder and no additional strokes. This result
supports the potential of HSCT as a long-term treatment strategy
for ADA2 deficiency. However, caution is warranted because the
HSCT procedure in P1 was characterized by severe early
complications. Indeed, ADA2-deficient patients might present
as high-risk candidates for HSCT. First, the inflammatory
response associated with conditioning is superimposed on the
inflammatory state intrinsic to ADA2 deficiency, which might
negatively affect engraftment. Second, the compromised
endothelial integrity observed in patients with ADA2 deficiency
could predispose to development of VOD, a potentially fatal
complication of HSCT. This combination of inflammation
and endothelial injury might further increase the risk of
stroke in the pre-engraftment and early postengraftment phases,
7
as observed in P1. It is reasonable to hypothesize that
ADA2-deficient patients might benefit from VOD prophylaxis
with defibrotide, as well as from pretreatment with anti–IL-6
mAbs, rituximab, or both. Moreover, treatment with etanercept
peri-HSCT could be considered in the context of ADA2
deficiency, especially given its usefulness in preventing and
treating acute GvHD. However, given the underlying
immunodeficiency, the risk of infection needs to be carefully
balanced when using anti–IL-6 and anti–TNF-amAbs.
Allogeneic HSCT restored normal plasma ADA2 activity in P1,
which is consistent with bone marrow–derived monocytes and
macrophages being the main sources of secreted ADA2. Whether
ADA2 plays a role in other tissues and the effect of this on
long-term prognosis remains unclear. A recent report on HSCT in a
patient with ADA2 deficiency with a 9-year follow-up is promising
and supports our findings.
8
However, it is plausible that the
benefit from HSCT to our patient is entirely due to restoration
of normal plasma ADA2 levels. If true, future treatment with
exogenous ADA2 might provide an alternative therapy
for ADA2 deficiency in patients in whom allogeneic HSCT is
contraindicated.
Lien Van Eyck, Jr, MD
a
Michael S. Hershfield, MD
b
Diana Pombal, MSc
a
Susan J. Kelly, PhD
b
Nancy J. Ganson, PhD
b
Leen Moens, PhD
c
Glynis Frans, MPharm
c
Heidi Schaballie, MD
d
Gert De Hertogh, MD, PhD
e
James Dooley, MSc
a
Xavier Bossuyt, MD, PhD
c
Carine Wouters, MD, PhD
d
Adrian Liston, PhD
a
*
Isabelle Meyts, MD, PhD
d
*
From
a
the Department of Immunology and Microbiology, Autoimmune Genetics Labo-
ratory, VIB and University of Leuven, Leuven, Belgium;
b
Duke University Medical
Center, Durham, NC;
c
the Department of Immunology and Microbiology, Experi-
mental Laboratory Immunology, University of Leuven, Leuven, Belgium;
d
the
Department of Immunology and Microbiology, Childhood Immunology, Department
of Pediatrics, University Hospitals Leuven and University of Leuven, Leuven,
Belgium; and
e
the Department of Pathology, University of Leuven, Leuven, Belgium.
E-mail: Isabelle.Meyts@uzleuven.be.
*These authors equally contributed to this work as senior authors.
Supported by the Research Foundation Flanders (FWO), the VIB, and the European
Research Council grant IMMUNO. I.M. is supported by a KOF mandate of the KU
Leuven, Belgium, and by the Jeffrey Modell Foundation.
Disclosure of potential conflict of interest: L. Van Eyck has received research support
from Research Foundation Flanders (FWO) and is employed by Un iversity Hospital
Leuven. M. S. Hershfield has consultant arrangements with and has received research
support from Sigma-Tau Pharmaceuticals and has a patent with and receives royalties
from Creata Pharmaceuticals. D. Pombal is employed by VIB. G. Frans has received a
GOA grant from the Catholic University of Leuven, Belgium. H. Schaballie has
received research support from Research Foundation Flanders (FWO). J. Dooley
has received research support from the European Research Council. X. Bossuyt has
received research support from the Research Council of Catholic University Leuven.
A. Liston has received research support from the European Research Council,
Research Foundation Flanders (FWO), and VIB. The rest of the authors declare
that they have no relevant conflicts of interest.
REFERENCES
1. Zhou Q, Yang D, Ombrello AK, Zavialov AV, Toro C, Zavialov AV, et al.
Early-Onset Stroke and Vasculopathy Associated with Mutations in ADA2.
N Engl J Med 2014;370:911-20.
2. Elkan PN, Pierce SB, Segel R, Walsh T, Barash J, Padeh S, et al. Mutant adenosine
deaminase 2 in a polyarteritis nodosa vasculopathy. N Engl J Med 2014;370:921-31.
3. Zavialov AV, Yu X, Spillmann D, Lauvau G, Zavialov AV. Structural basis for the
growth factor activity of human adenosine deaminase ADA2. J Biol Chem 2010;
285:12367-77.
4. Malamut G, Verkarre V, Suarez F, Viallard JF, Lascaux AS, Cosnes J, et al. The
enteropathy associated with common variable immunodeficiency: the delineated
frontiers with celiac disease. Am J Gastroenterol 2010;105:2262-75.
5. Zavialov AV, Gracia E, Glaichenhaus N, Franco R, Zavialov AV, Lauvau G.
Human adenosine deaminase 2 induces differentiation of monocytes into
macrophages and stimulates proliferation of T helper cells and macrophages.
J Leukoc Biol 2010;88:279-90.
6. Mercalli A, Calavita I, Dugnani E, Citro A, Cantarelli E, Nano R, et al. Rapamycin
unbalances the polarization of human macrophages to M1. Immunology 2013;140:
179-90.
7. DiCarlo J, Agarwal-Hashmi R, Shah A, Kim P, Craveiro L, Killen R, et al.
Cytokine and chemokine patterns across 100 days after hematopoietic stem cell
transplantation in children. Biol Blood Marrow Transplant 2014;20:361-9.
8. Van Montfrans J, Zavialov A, Zhou Q. Mutant ADA2 in vasculopathies. N Engl J
Med 2014;371:481.
http://dx.doi.org/10.1016/j.jaci.2014.10.010
J ALLERGY CLIN IMMUNOL
nnn 2014
4LETTER TO THE EDITOR
METHODS
The study was performed in accordance with the modified version of the
Declaration of Helsinki. The study was approved by the Ethics Committee of
UZ Leuven. Written informed consent was obtained before DNA isolation
from blood of all family members and from cheek epithelium of the
transplanted patient.
Functional assays
PBMCs were isolated from heparinized blood of patients, family members,
and control subjects and analyzed by using flow cytometry, as previously
described.
E1
Serum IL-6 levels were measured by means of ELISA, according
to the manufacturer’s instructions (BD Bioscience, San Jose, Calif).
Whole-exome sequencing
We performed whole-exome sequencing on the untreated patient and on the
unaffected parents and sibling. Genomic DNA samples for whole-exome
sequencing were prepared from heparinized peripheral blood by using the
QIAamp DNA Blood Midi Kit (QIAGEN, Hilden, Germany). Exome
sequence libraries were prepared with a SeqCap EZ Human Exome Library
v3.0 kit (Roche NimbleGen, Madison, Wis). Paired-end sequencing was
performed on the Illumina HiSeq2000 (Genomics Core Facility, University of
Leuven, Leuven, Belgium). BWA software was used to align the sequence
reads to the Human Reference Genome Build hg19. The GATK Unified
Genotyper was used to identify single nucleotide variants and insertions/
deletions. ANNOVAR was used for annotation.
Sanger sequencing
A somatic DNA sample of the patient undergoing transplantation was
obtained from a cheek swab by using the GenElute Mammalian Genomic
DNA Miniprep Kit (Sigma-Aldrich, St Louis, Mo). The region of interest in
exon 2 of CECR1 was sequenced with the primers 59-GTTTGTACCAAGG-
GAGACACCTACC-39and 59-CTGGCTGGTGAGGAATGTCAC-39. Sanger
sequencing was performed on an ABI 3730 XL Genetic Analyzer (Applied
Biosystems, Foster City, Calif) at the LGC Genomics Facility in Berlin, Ger-
many. Sequencing data were analyzed by using DNADynamo (Blue Tractor
Software, Llanfairfechan, United Kingdom).
Flow cytometry
PBMCs were isolated from heparinized blood of patients and control
subjects by using lymphocyte separation medium (MP Biomedicals, Solon,
Ohio) and frozen in 10% dimethyl sulfoxide (Sigma). Thawed cells were
stained with antibodies (from eBioscience [San Diego, Calif], unless stated
otherwise) against CD11c (3.9), CD3 (SK7), CD4 (RPA-T4), CD8a
(RPA-T8), CD19 (HIB19), CD45RA (HI100), CD56 (MEM188), HLA-DR
(LN3), forkhead box protein 3 (FOXP3; 206D; BioLegend, San Diego,
Calif), IFN-g(4S.B3 IL-17, eBio64DEC17), IL-2 (MQ1-17H12), CXCR5
(IgG23; R&D Systems, Minn eapolis, Minn), CD31 (WM-59), CCR7 (3D12),
IgM (MHM-88, BioLegend), CD27 (O323), IgE (IgE21), CD24 (eBioSN3,
SN3 A5-2 H10), CD38 (HIT2), gd T-cell receptor (B1.1), CD56 (MEM188),
CD14 (61D3), CD123 (6H6) , and IL-4 (8D4-8). For cytokine staining, T cells
were stimulated ex vivo for 5 hours in 50 ng/mL phorbol 12-myristate
13-acetate (Sigma) and 500 ng/mL ionomycin (Sigma) in the presence of
GolgiStop (BD Biosciences) before staining. Before intracellular staining,
cells were first surface stained as described, fixed, and permeabilized with
fixation/permeabilization buffer (eBioscience) for forkhead box protein 3
staining or Cytofix/Cytoperm (BD) for other intracellular stainings. All
data were acquired on BD FACSCanto II and analyzed with FlowJo (Tree
Star, Ashland, Ore).
ELISA for measurement of IL-6 levels in serum
An in-house validated ELISA was used based on a commercially available
antibody pair (BD Biosciences).
Measurements of ADA1 and ADA2 activity in
plasma
ADA2 activity in plasma was measured by using the HPLC method
described by Zhou et al.
E2
ADA1 activity and concentrations of total adeno-
sine and deoxyadenosine nucleotides in extracts of dried blood spots were
measured, as previously described.
E3,E4
The concentrations of adenosine
and deoxyadenosine in plasma were determined by means of HPLC analysis
of a neutralized perchloric acid extract of plasma. In brief, 200 mL of plasma
was acidified with 40 mL of 5 N perchloric acid and centrifuged, and the su-
pernatant was neutralized with 3 N KOH and 1 M KHCO
3
. After centrifuga-
tion, 100 mL of the supernatant was analyzed on a C18 mBondapak column
(Waters Corporation, Milford, Mass) by using 0.05 mol/L NH
4
H
2
PO
4
,8%
methanol, and 1% acetonitrile (pH 5.2; flow rate, 0.5 mL/min) as the mobile
phase and monitoring absorbance at 260 and 280 nm with a diode array detec-
tor. The lower limit of quantitation for adenosine and deoxyadenosine in this
assay was 0.8 mmol/L; the lower limit of detection was taken as half the lower
limit of quantitation or 0.4 mmol/L.
RESULTS
X-linked lymphoproliferative disease type I and II, Wiskott-
Aldrich syndrome, autoimmune lymphoproliferative syndrome,
ADA1 deficiency, purine nucleoside phosphorylase deficiency,
and immune dysregulation–polyendocrinopathy–enteropathy–X-
linked syndrome were excluded by means of functional and
genetic analyses.
REFERENCES
E1. Danso-Abeam D, Zhang J, Dooley J, Staats KA, Van Eyck L, Van Brussel T, et al.
Olmsted syndrome: exploration of the immunological phenotype. Orphanet J
Rare Dis 2013;8:79.
E2. Zhou Q, Yang D, Ombrello AK, Zavialov AV, Toro C, Zavialov AV, et al. Early-
Onset Stroke and Vasculopathy Associated with Mutations in ADA2. N Engl J
Med 2014;370:911-20.
E3. Hershfield MS, Fetter JE, Small WC, Small WC, Bagnara AS, Williams SR, et al.
Effects of mutational loss of adenosine kinase and deoxycytidine kinase on deox-
yATP accumulation and deoxyadenosine toxicity in cultured CEM human T-lym-
phoblastoid cells. J Biol Chem 1982;257:6380-6.
E4. Arredondo-Vega FX, Santisteban I, Richard E, Bali P, Koleilat M, Loubser M,
et al. Adenosine deaminase deficiency with mosaicism for a ‘‘second-site sup-
pressor’’ of a splicing mutation: decline in revertant T lymphocytes during
enzyme replacement therapy. Blood 2002;99:1005-13.
J ALLERGY CLIN IMMUNOL
VOLUME nnn, NUMBER nn
LETTER TO THE EDITOR 4.e1
FIG E1. Va sculopathology and immunopathology in patients with ADA2 deficiency. A, Sagittal T1-weighted
MRI of P1 showing pineal gland hemorrhage (arrow).B, Hematoxylin and eosin staining of jejunal ulcera-
tion in P2 showing chronic ulcer with predominant eosinophils (arrows), some neutrophils and lympho-
cytes, and very few plasma cells. Plasma cells stained by means of CD138 staining are indicated by
arrows in the inset.
J ALLERGY CLIN IMMUNOL
nnn 2014
4.e2 LETTER TO THE EDITOR
FIG E2. Familial inheritance of CECR1 mutation. The region of interest in
exon 2 of CECR1 was sequenced by means of Sanger sequencing. A-E,
Sequence reads for the father (Fig E2, A), mother (Fig E2, B), healthy sibling
(HSCT donor; Fig E2, C), patient 1 after HSCT (chimerism accounts for pres-
ence of a minor G peak; Fig E2, D), and patient 2 (Fig E2, E). F, Family tree of
the affected pedigree, indicating affected patients and CECR1 genotype.
J ALLERGY CLIN IMMUNOL
VOLUME nnn, NUMBER nn
LETTER TO THE EDITOR 4.e3
TABLE E1. Clinical presentation, laboratory values, and therapeutic history of ADA2-deficient patients
Patient 1 Patient 2
Clinical phenotype
Clinical presentation Hypogammaglobulinemia, pancytopenia,
lymphoproliferation
Hypogammaglobulinemia, (intermittent)
lymphopenia and neutropenia,
lymphoproliferation
Viral infections confirmed by means of PCR RSV, adenovirus, norovirus HHV-6, HSV, polyomavirus
Stroke Hemorrhage in the pineal gland None
Laboratory values*
White blood cell count (kU/mL) 2.02 9.08
Neutrophil count (kU/mL) 0.3 6.4
Lymphocyte count (kU/mL) 1.0 1.7
Hemoglobin (g/dL) 8.8 9.7
Thrombocytes (kU/mL) 25 300
ALT (5-38 U/L) 44 15
AST (0-41 U/L) 64 6
IgG (3.02-9.85 g/L) <1.00 2.77
IgA (0.13-1.08 g/L) <0.07 0.11
IgM (0.26-1.60 g/L) 0.09 0.27
IgE (0-91 IU/mL) <230
IgD (<10 U/mL) 0 0
ANA Negative Not determined
ANCA Negative Not determined
Thrombocyte autoantibodies Anti-gpIIB-IIIa antibody present Not determined
Erythrocyte autoantibodies Anti-MNS1 antibody present Not determined
Lymphocyte count (kU/mL) [at moment of
immunophenotyping]
0.39 3.0
Therapeutic history
Immunosuppressive medication Corticosteroids, sirolimus, mycophenolate mofetil,
tacrolimus, cyclosporine, mercaptopurine
Corticosteroids, azathioprine, sirolimus
Immunoglobulin substitution Yes (before HSCT) Yes
Allogeneic HSCT Yes No
ALT, Alanine aminotransferase; ANA, antinuclear antibody; ANCA, antineutrophil cytoplasmic antibody; AST, aspartate aminotransferase; HSV, herpes simplex virus; RSV,
respiratory syncytial virus.
*Values were obtained at initial clinical presentation unless stated otherwise.
J ALLERGY CLIN IMMUNOL
nnn 2014
4.e4 LETTER TO THE EDITOR
TABLE E2. Relative frequencies of peripheral blood leukocyte populations in P1 before HSCT compared with those in healthy
age-matched control subjects
Subset Defining surface markers Patients (%)
Healthy volunteers (%)
Range (minimum-maximum)
T cells CD3
1
80.6 52.9-65.2
CD4
1
T cells CD4
1
CD8
2
63.8 29.4-65.2
/Treg CD25
1
Foxp3
1
10.0
CD8
1
T cells CD4
2
CD8
1
0.83 17.6-23.2
B cells CD19
1
5.56 11.8-30.4
/Transitional CD38
high
CD24
high
0.2
/Naive CD27
2
IgD
1
97.8
/Immature CD27
1
IgD
1
0.1
/Switched memory CD27
1
IgD
2
0.3
CD3
1
, CD4
1
, and CD8
1
T cells and CD19
1
B cells are shown as percentages of total lymphocytes. Regulatory T (Treg) cells are shown as percentages of CD4
1
T cells. B-cell
subsets are shown as percentages of CD19
1
B cells.
Foxp3, Forkhead box protein 3.
J ALLERGY CLIN IMMUNOL
VOLUME nnn, NUMBER nn
LETTER TO THE EDITOR 4.e5
... early-onset stroke [1,2], the clinical phenotype was expanded [3][4][5]. An homozygous or compound heterozygous mutation causing a loss-of-functions in the gene of the Adenosine Deaminase 2 (ADA2), which encodes the homonymous enzyme, is the cause of this rare syndrome [1,2]. ...
... Hematopoietic stem cell transplantation (HSCT) has been attempted as rescue therapy, especially in patients with severe hematologic and immunologic disorders. This treatment is however burdened by well-known side effects [4,12,13]. ...
... After full text screening 90 papers have been used in our systematic review [1][2][3][4][5][6][7][8][9][10][11][12][13]: 8 papers were excluded for the publication type and 18 because they did not report the clinical phenotype or the confirmatory genetic mutations. Figure 1 represented the PRISMA flow diagram demonstrating the process of study selection. ...
Article
Full-text available
Introduction: Deficiency of adenosine deaminase 2 (DADA2) is a rare monogenic autoinflammatory disease, whose clinical phenotype was expanded since the first cases, originally described as mimicker of polyarteritis nodosa, with immunodeficiency and early-onset stroke. Methods: A systematic review according to PRISMA approach, including all articles published before the 31st of August 2021 in Pubmed and EMBASE database was performed. Results: The search identified 90 publications describing 378 unique patients (55.8% male). To date 95unique mutations have been reported. The mean age at disease onset was 92.15 months (range 0-720 months), 32 (8.5%) showed an onset of the first signs/symptoms after 18 years old and 96 (25.4%) after 10 years old. The most frequent clinical characteristics described were cutaneous (67.9%), haematological manifestations (56.3%), recurrent fever (51.3%), neurological as stroke and polyneuropathy (51%), immunological abnormalities (42.3%), arthralgia/arthritis (35.4%), splenomegaly (30.6%), abdominal involvement (29.8%), hepatomegaly (23.5%), recurrent infections (18.5%), myalgia (17.9%), kidney involvement (17.7%) etc. Patients with skin manifestations were older than the others (101.1 months SD ± 116.5, vs. 75.3 SD ± 88.2, p 0.041), while those with a haematological involvement (64.1 months SD ± 75.6 vs. 133.1 SD ± 133.1, p < 0.001) and immunological involvement (73.03 months SD ± 96.9 vs. 103.2 SD ± 112.9, p 0.05) are younger than the others. We observed different correlations among the different clinical manifestations. The use of anti-TNFα and hematopoietic cell stems transplantation (HCST) has improved the current history of the disease. Conclusion: Due to this highly variable phenotype and age of presentation, patients with DADA2 may present to several type of specialists. Given the important morbidity and mortality, early diagnosis and treatment are mandatory.
... Although immunomodulators and steroid are the mainstay of management, as a more definitive treatment, HSCT has been reported to control both the immunological, the haematological, and the vascular phenotype of DADA2. 11 In our case, a total of 4 samples were tested to rule out DADA2-the patient, her parents and an unrelated control sample were sent to a CLIA certified laboratory. We have a documented normal ADA2 enzyme activity with adequate control samples in our case, hence ruling out DADA2. ...
Article
Full-text available
Childhood Takayasu arteritis is one of the most common vasculitis in paediatric age group. It predominantly involves the greater vessels such as aorta and its branches, leading to granulomatous inflammation. The process of inflammation gives rise to the symptoms based on thrombotic or aneurysmal phenomena. This is a case report of a 11-year-old girl diagnosed in an urban tertiary care centre in India with childhood Takayasu arteritis. The said child had first presented with non-specific symptoms such as leg ache, back ache and fever. Over the course of illness, the child developed hypertension and had also suffered from PRES (posterior reversible encephalopathy syndrome). Clinical examination was consistent with Takayasu arteritis, however with predominant medium-sized vessel involvement on imaging. Since this is a relatively atypical presentation, it was mandatory to rule out other causes of medium-vessel arteritis, especially DADA2 (deficiency of ADA2).
... Patients at risk for infections benefit from antibiotic prophylaxis and/or immunoglobulin replacement therapy [2,3,7,8,15]. For patients with severe hematologic or immunologic manifestations, hematopoietic cell transplantation (HCT) has successfully been performed, resulting in full resolution of the vascular, immunologic and hematologic phenotype of DADA2 [3,15,18,19]. Therefore, HCT is particularly indicated in patients with refractory immunodeficiency or severe hematologic disease [3,4]. ...
Article
Full-text available
Deficiency of adenosine deaminase-2 (DADA2) is an autosomal recessive autoinflammatory disease with an extremely variable disease presentation. This paper provides a comprehensive overview of the Dutch DADA2 cohort. We performed a retrospective cohort study in 29 ADA2-deficient patients from 23 families with a median age at inclusion of 26 years. All patients had biallelic pathogenic variants in the ADA2 gene. The most common clinical findings included cutaneous involvement (79.3%), (hepato)splenomegaly (70.8%) and recurrent infections (58.6%). Stroke was observed in 41.4% of the patients. The main laboratory abnormalities were hypogammaglobulinemia and various cytopenias. Patients presented most often with a mixed phenotype involving vasculopathy, immunodeficiency and hematologic manifestations (62.1%). In this cohort, malignancies were reported in eight patients (27.6%), of whom five presented with a hematologic malignancy and two with a basal cell carcinoma. Four patients developed hemophagocytic lymphohistiocytosis (HLH) or an HLH-like episode, of whom three passed away during or shortly after the occurrence of HLH. TNF-inhibitors (TNFi) were effective in treating vasculopathy-associated symptoms and preventing stroke, but were hardly effective in the treatment of hematologic manifestations. Three patients underwent hematopoietic cell transplantation and two of them are doing well with complete resolution of DADA2-related symptoms. The overall mortality in this cohort was 17.2%. In conclusion, this cohort describes the clinical, genetic and laboratory findings of 29 Dutch DADA2 patients. We describe the occurrence of HLH as a life-threatening disease complication and report a relatively high incidence of malignancies and mortality. Supplementary Information The online version contains supplementary material available at 10.1007/s10875-023-01521-8.
... Systemic autoinflammatory manifestations often accompany cutaneous vasculitis and ischemic or hemorrhagic stroke [12]. However, several reports concerning patients with DADA2 have described the predominance of hematological and immunological manifestations, including hypogammaglobinemia, lymphopenia, neutropenia, thrombocytopenia, pure red cell aplasia (PRCA), and bone marrow failure [13][14][15]. The vasculitis phenotype is correlated with missense mutations and residual ADA2 protein function, while the hematological phenotype is correlated with insertion/deletion, nonsense or missense mutations, and loss of ADA2 activity [16]. ...
Article
Full-text available
Adenosine deaminase 2 deficiency (DADA2), a rare and potentially fatal systemic autoinflammatory disease, is characterized by low or lack of ADA2 activity due to ADA2 mutations. DADA2 symptoms are variable and include vasculitis, immunodeficiency, and cytopenia. Minimal data are available from Saudi Arabia. This retrospective study conducted at seven major tertiary medical centers examined the phenotypic and genotypic variabilities, clinical and diagnostic findings, and treatment outcomes among 20 Saudi patients with DADA2 from 14 families. The median age of the study cohort was 9.5 years (4–26 years). The clinical presentation was before the age of 5 months in 25% of patients. Homozygous c.1447-1451del mutation was the most frequent ADA2 alteration (40%), followed by c.882-2A:G (30%). All tested patients exhibited absent or near-absent ADA2 activity. Phenotypic manifestations included stroke (40%), hematological abnormalities (95%), lymphoproliferation (65%), and recurrent infection (45%). Five and three patients had extracranial vasculitis features and Hodgkin lymphoma, respectively. Atypical manifestations included growth retardation (30%) and transverse myelitis. Anti-tumor necrosis factor (anti-TNF) therapy was the main treatment. Some patients underwent blood transfusion, splenectomy, cyclosporine and colony-stimulating factor therapies, and hematopoietic stem cell transplantation due to anti-TNF therapy failure. Fulminant hepatitis and septic multiorgan failure caused mortality in three patients. Thus, this study revealed the variability in the molecular and clinical characteristics of DADA2 in the study cohort with predominant aberrant hematological and immunological characteristics. Consensus diagnostic criteria will facilitate early diagnosis and treatment. Additionally, disease registries or large prospective studies are needed for evaluating rare disease complications, such as cancer.
Article
Full-text available
Systemic autoinflammatory diseases (SAIDs) are disorders of innate immunity, which are characterized by unprovoked recurrent flares of systemic inflammation often characterized by fever associated with clinical manifestations mainly involving the musculoskeletal, mucocutaneous, gastrointestinal, and nervous systems. Several conditions also present with varied, sometimes prominent, involvement of the vascular system, with features of vasculitis characterized by variable target vessel involvement and organ damage. Here, we report a systematic review of vasculitis and vasculopathy associated with inborn errors of immunity.
Article
The deficiency of adenosine deaminase 2 (DADA2) is an autosomal recessive disease caused by loss-of-function (LOF) mutations in the ADA2 gene and was first described in 2014. Initially, it was described as vasculopathy/vasculitis that mostly affected infants and young children and closely resembled polyarteritis nodosa (PAN). Skin rash and ischemic/hemorrhagic stroke are predominant symptoms. However, the clinical spectrum of DADA2 has continued to expand since then. It has now been reported in adults as well. Besides vasculitis-related manifestations, hematological, immunological, and autoinflammatory manifestations are now well recognized. More than 100 disease-causing mutations have been described. The decrease in ADA2 enzyme leads to an increased extracellular adenosine level that, in turn, triggers a proinflammatory cascade. The disease is highly variable, and patients carrying same mutation may have different ages of presentation and clinical features. Anti-tumor necrosis factor (TNF) agents are mainstay of treatment of the vasculitis/vasculopathy phenotype. Hematopoietic stem cell transplant (HSCT) has been performed in patients with severe hematological manifestations. Recombinant ADA2 protein and gene therapy hold a promise for future.
Article
Full-text available
Importance: Deficiency of adenosine deaminase 2 (DADA2) is a recessively inherited disease characterized by systemic vasculitis, early-onset stroke, bone marrow failure, and/or immunodeficiency affecting both children and adults. DADA2 is among the more common monogenic autoinflammatory diseases, with an estimate of more than 35 000 cases worldwide, but currently, there are no guidelines for diagnostic evaluation or management. Objective: To review the available evidence and develop multidisciplinary consensus statements for the evaluation and management of DADA2. Evidence review: The DADA2 Consensus Committee developed research questions based on data collected from the International Meetings on DADA2 organized by the DADA2 Foundation in 2016, 2018, and 2020. A comprehensive literature review was performed for articles published prior to 2022. Thirty-two consensus statements were generated using a modified Delphi process, and evidence was graded using the Oxford Center for Evidence-Based Medicine Levels of Evidence. Findings: The DADA2 Consensus Committee, comprising 3 patient representatives and 35 international experts from 18 countries, developed consensus statements for (1) diagnostic testing, (2) screening, (3) clinical and laboratory evaluation, and (4) management of DADA2 based on disease phenotype. Additional consensus statements related to the evaluation and treatment of individuals with DADA2 who are presymptomatic and carriers were generated. Areas with insufficient evidence were identified, and questions for future research were outlined. Conclusions and relevance: DADA2 is a potentially fatal disease that requires early diagnosis and treatment. By summarizing key evidence and expert opinions, these consensus statements provide a framework to facilitate diagnostic evaluation and management of DADA2.
Article
Full-text available
Deficiency of adenosine deaminase 2 (DADA2) is an autosomal recessive that was first described in 2014. It is a monogenic disease that is caused by loss-of-function variants in the ADA2 gene. DADA2 involves small- and medium-sized vessels and its clinical presentations include polyarteritis nodosa (PAN)-like features such as livedoid rash, early-onset stroke, hypogammaglobulinemia, hematological abnormalities, and systemic inflammation. Early diagnosis and treatment of DADA2 are crucial as the clinical features could be potentially life-threatening but might be treatable. The first-line treatment of choice in DADA2 is tumor necrosis factor (TNF)-inhibitors. We aimed to give an overview of the known pathophysiology, clinical presentations, diagnosis, and treatment of DADA2. A clearer knowledge of DADA2 may help to better diagnose, manage, and improve the clinical outcome of DADA2 patients. However, further studies are required to investigate the genotype-phenotype associations and exact pathophysiology of DADA2.
Chapter
Periodic fever syndromes (PFS) and other autoinflammatory diseases (AuID) are characterized by recurrent episodes of inflammation often accompanied by fever and without an apparent triggering factor. PFS have traditionally been categorized together due to the commonality of recurring febrile exacerbations with findings of systemic inflammation of the eyes, skin, joints, and serosal tissues. In these disorders, the predominant affectation is found in the inflammasomes, representing the collection of innate immune response mechanisms, which are tasked with controlling inflammatory responses. The remainder of the AuID discussed here are also linked by various pathways to overactivation of the innate immune system, leading to increased production of inflammatory cytokine pathways including IL-1β, IL-6, IL-18, and type 1 interferons. Some of these conditions have a focused cytokine effect, while others represent overlapping pathways of inflammatory responses. Diagnosis can be made with strong clinical suspicion, followed by confirmatory genetic testing in many cases. As we learn more about these monogenic syndromes, they can continue to inform our understanding of inflammation-related pathology, as well as help us to develop more precise therapies for better outcomes.
Article
Full-text available
Zhou et al. and Navon Elkan et al. (both in the March 6 issue) speculate that hematopoietic stem-cell transplantation (HSCT) or enzyme-replacement therapy may be beneficial in patients with adenosine deaminase 2 (ADA2) deficiency. We report the clinical course of two brothers with recently diagnosed ADA2 deficiency resulting from a homozygous mutation in CECR1 (p.R169Q). One sibling underwent HSCT in 2003. (CECR1 encodes the protein ADA2.) One brother presented in 1999, at 6 months of age, with livedo reticularis, hepatosplenomegaly, hypercoagulability, granulocytopenia, and complete red-cell aplasia. He underwent HSCT in 2003 for a presumed diagnosis of atypical Diamond–Blackfan anemia; the transplant was obtained from a matched unrelated donor after myeloablative conditioning. The patient showed rapid immune reconstitution, with resolution of cytopenias, skin lesions, hepatosplenomegaly, and hypercoagulability. He has had 100% donor chimerism in peripheral blood and has been not been taking medications for any of these conditions for 9 years. Serum ADA2 levels are now within the normal range for his age. A recent magnetic resonance image of the brain was negative for vasculopathic changes. HSCT corrected the CECR1 mutation in blood cells, but not in (somatic) buccal mucosa cells. This patient's younger brother presented in 2009, at 6 years of age, with hepatosplenomegaly, hypogammaglobulinemia, and profound lymphopenia. He also had unexplained fever, livedo reticularis, and stroke at the age of 10, which prompted us to sequence CECR1. Treatment with a tumor necrosis factor (TNF) inhibitor (etanercept) has stabilized his clinical condition, although he has persisting profound lymphopenia and low-grade inflammation. HSCT is being considered. The absence of vasculopathy and the resolution of hypercoagulability after HSCT in the older brother suggests that the correction of ADA2 blood levels reduces macrophage activation and endothelial disruption, both of which probably cause vasculitis and stroke in patients with ADA2 deficiency.
Article
Full-text available
Background: Polyarteritis nodosa is a systemic necrotizing vasculitis with a pathogenesis that is poorly understood. We identified six families with multiple cases of systemic and cutaneous polyarteritis nodosa, consistent with autosomal recessive inheritance. In most cases, onset of the disease occurred during childhood. Methods: We carried out exome sequencing in persons from multiply affected families of Georgian Jewish or German ancestry. We performed targeted sequencing in additional family members and in unrelated affected persons, 3 of Georgian Jewish ancestry and 14 of Turkish ancestry. Mutations were assessed by testing their effect on enzymatic activity in serum specimens from patients, analysis of protein structure, expression in mammalian cells, and biophysical analysis of purified protein. Results: In all the families, vasculitis was caused by recessive mutations in CECR1, the gene encoding adenosine deaminase 2 (ADA2). All the Georgian Jewish patients were homozygous for a mutation encoding a Gly47Arg substitution, the German patients were compound heterozygous for Arg169Gln and Pro251Leu mutations, and one Turkish patient was compound heterozygous for Gly47Val and Trp264Ser mutations. In the endogamous Georgian Jewish population, the Gly47Arg carrier frequency was 0.102, which is consistent with the high prevalence of disease. The other mutations either were found in only one family member or patient or were extremely rare. ADA2 activity was significantly reduced in serum specimens from patients. Expression in human embryonic kidney 293T cells revealed low amounts of mutant secreted protein. Conclusions: Recessive loss-of-function mutations of ADA2, a growth factor that is the major extracellular adenosine deaminase, can cause polyarteritis nodosa vasculopathy with highly varied clinical expression. (Funded by the Shaare Zedek Medical Center and others.).
Article
Full-text available
Background We observed a syndrome of intermittent fevers, early-onset lacunar strokes and other neurovascular manifestations, livedoid rash, hepatosplenomegaly, and systemic vasculopathy in three unrelated patients. We suspected a genetic cause because the disorder presented in early childhood. Methods We performed whole-exome sequencing in the initial three patients and their unaffected parents and candidate-gene sequencing in three patients with a similar phenotype, as well as two young siblings with polyarteritis nodosa and one patient with small-vessel vasculitis. Enzyme assays, immunoblotting, immunohistochemical testing, flow cytometry, and cytokine profiling were performed on samples from the patients. To study protein function, we used morpholino-mediated knockdowns in zebrafish and short hairpin RNA knockdowns in U937 cells cultured with human dermal endothelial cells. ResultsAll nine patients carried recessively inherited mutations in CECR1 (cat eye syndrome chromosome region, candidate 1), encoding adenosine deaminase 2 (ADA2), that were predicted to be deleterious; these mutations were rare or absent in healthy controls. Six patients were compound heterozygous for eight CECR1 mutations, whereas the three patients with polyarteritis nodosa or small-vessel vasculitis were homozygous for the p.Gly47Arg mutation. Patients had a marked reduction in the levels of ADA2 and ADA2-specific enzyme activity in the blood. Skin, liver, and brain biopsies revealed vasculopathic changes characterized by compromised endothelial integrity, endothelial cellular activation, and inflammation. Knockdown of a zebrafish ADA2 homologue caused intracranial hemorrhages and neutropenia phenotypes that were prevented by coinjection with nonmutated (but not with mutated) human CECR1. Monocytes from patients induced damage in cocultured endothelial-cell layers. Conclusions Loss-of-function mutations in CECR1 were associated with a spectrum of vascular and inflammatory phenotypes, ranging from early-onset recurrent stroke to systemic vasculopathy or vasculitis. (Funded by the National Institutes of Health Intramural Research Programs and others.) Adenosine deaminase 2 (ADA2) is an enzyme involved in purine metabolism and a growth factor that influences the development of endothelial cells and leukocytes. This study shows that defects in ADA2 cause recurrent fevers, vascular pathologic features, and mild immunodeficiency. Patients with autoinflammatory disease sometimes present with clinical findings that encompass multiple organ systems.(1) Three unrelated children presented to the National Institutes of Health (NIH) Clinical Center with intermittent fevers, recurrent lacunar strokes, elevated levels of acute-phase reactants, livedoid rash, hepatosplenomegaly, and hypogammaglobulinemia. Collectively, these findings do not easily fit with any of the known inherited autoinflammatory diseases. Hereditary or acquired vascular disorders can have protean manifestations yet be caused by mutations in a single gene. Diseases such as the Aicardi-Goutieres syndrome,(2),(3) polypoidal choroidal vasculopathy,(4) sickle cell anemia,(5) livedoid vasculopathy,(6) and the small-vessel vasculitides(7),(8) are examples of systemic ...
Article
Full-text available
We mapped the cytokine response to hematopoietic stem cell transplantation (HSCT) by assaying 51 cytokines and chemokines each week for 100 days in 51 children receiving allogeneic (n = 44) or autologous HSCT (n = 7). Assay values were reported as mean fluorescence intensity (MFI). Log transformation converted MFI to clinically relevant measures (ie, pg/mL). We searched for potential markers of transplant complications by using mixed treatment by subject analysis of variance. Global cytokine secretion in HSCT recipients was significantly lower than in concurrent control patients (n = 11). Coincident with the nadir in WBC count, the concentration of many cytokines declined further by the second and third week. All analytes (except monokine induced by gamma interferon [MIG]) subsequently rebounded by week 4 (coincident with engraftment and recovery of WBC count) but often still remained well below control levels. Concurrent with the collective nadir of multiple cytokines, monocyte chemoattractant protein 1 (MCP-1), growth-regulated oncogene alpha (GRO-a), and leptin surged during weeks 2 to 4. High levels of leptin persisted throughout the 100 post-transplant days. Also during weeks 2 to 4, hepatocyte growth factor (HGF) and IL-6 surged in children with complications but not in those without complications. The peak in HGF was more pronounced in veno-occlusive disease (VOD). HGF and IL-6 secretion rose at least 2 weeks before the clinical diagnosis of VOD or graft-versus-host disease (GVHD). From week 4 onward in all groups, the MFI of the cytokine resistin increased to 5 to 15 times above concurrent control. HGF has now emerged in 3 or more biomarker discovery efforts for GVHD (and in our population for VOD as well). HGF (with or without IL-6) should be investigated as a potential predictive biomarker of VOD or GVHD. Alternatively, the hyperinflammatory “signature” provided by a multicytokine assay may be predictive.
Article
Full-text available
Background Olmsted syndrome is a rare congenital skin disorder presenting with periorifical hyperkeratotic lesions and mutilating palmoplantar keratoderma, which is often associated with infections of the keratotic area. A recent study identified de novo mutations causing constitutive activation of TRPV3 as a cause of the keratotic manifestations of Olmsted syndrome. Methods Genetic, clinical and immunological profiling was performed on a case study patient with the clinical diagnosis of Olmsted syndrome. Results The patient was found to harbour a previously undescribed 1718G-C transversion in TRPV3, causing a G573A point mutation. In depth clinical and immunological analysis found multiple indicators of immune dysregulation, including frequent dermal infections, inflammatory infiltrate in the affected skin, hyper IgE production and elevated follicular T cells and eosinophils in the peripheral blood. Conclusions These results provide the first comprehensive assessment of the immunological features of Olmsted syndrome. The systemic phenotype of hyper IgE and persistent eosinophilia suggest a primary or secondary role of immunological processes in the pathogenesis of Olmsted syndrome, and have important clinical consequences with regard to the treatment of Olmsted syndrome patients.
Article
Full-text available
Discovery of the growth factor activity of ADA2. ADAs play a pivotal role in regulating the level of adenosine, a signaling molecule controlling a variety of cellular responses by binding to and activating four ADRs. Two enzymes, ADA1 and ADA2, are known to possess ADA activity in humans. Although the structure of ADA1 and its role in lymphocytic activation have been known for a long time, the structure and function of ADA2, a member of ADGF, remain enigmatic. Here, we found that ADA2 is secreted by monocytes undergoing differentiation into macrophages or DCs and that it binds to the cell surface via proteoglycans and ADRs. We demonstrate that ADA1 and ADA2 increase the rate of proliferation of monocyte-activated CD4+ T cells independently of their catalytic activity. We also show that ADA2 induces T cell-dependent differentiation of monocytes into macrophages and stimulates macrophage proliferation. Our discovery of the growth factor-like activity of ADA2 explains clinical observations and suggests that this enzyme could be used as a drug candidate to modulate the immune responses during inflammation and cancer.
Article
Four patients from 3 Saudi Arabian families had delayed onset of immune deficiency due to homozygosity for a novel intronic mutation, g.31701T>A, in the last splice acceptor site of the adenosine deaminase (ADA) gene. Aberrant splicing mutated the last 4 ADA amino acids and added a 43-residue “tail” that rendered the protein unstable. Mutant complementary DNA (cDNA) expressed inEscherichia coli yielded 1% of the ADA activity obtained with wild-type cDNA. The oldest patient, 16 years old at diagnosis, had greater residual immune function and less elevated erythrocyte deoxyadenosine nucleotides than his 4-year-old affected sister. His T cells and Epstein-Barr virus (EBV) B cell line had 75% of normal ADA activity and ADA protein of normal size. DNA from these cells and his whole blood possessed 2 mutant ADA alleles. Both carried g.31701T>A, but one had acquired a deletion of the 11 adjacent base pair, g.31702-12, which suppressed aberrant splicing and excised an unusual purine-rich tract from the wild-type intron 11/exon 12 junction. During ADA replacement therapy, ADA activity in T cells and abundance of the “second-site” revertant allele decreased markedly. This finding raises an important issue relevant to stem cell gene therapy.
Article
Plasticity is a hallmark of macrophages, and in response to environmental signals these cells undergo different forms of polarized activation, the extremes of which are called classic (M1) and alternative (M2). Rapamycin (RAPA) is crucial for survival and functions of myeloid phagocytes, but its effects on macrophage polarization are not yet studied. To address this issue, human macrophages obtained from six normal blood donors were polarized to M1 or M2 in vitro by LPS plus IFN-γ or IL-4, respectively. The presence of RAPA (10 ng/ml) induced macrophage apoptosis in M2 but not in M1. Beyond the impact on survival in M2 RAPA reduced CXCR4, CD206 and CD209 expression and SCGF-β, CCL18 and CCL13 release. On the other side, in M1 RAPA increased CD86 and CCR7 expression and IL-6, TNF-α and IL-1β1γ release while it reduced CD206 and CD209 expression and IL-10, VEGF and CCL18 release. In view of the in vitro data, we examined the in vivo effect of RAPA monotherapy (0.1 mg/kg/day) in 12 patients who were treated for at least 1 month prior to islet transplant. Cytokine release by TLR4-stimulated peripheral blood mononuclear cells showed a clear shift to an M1-like profile. Moreover, macrophage polarization 21 days after treatment showed a significant quantitative shift to M1. These results suggest a role of mTOR into the molecular mechanisms of macrophage polarization and propose new therapeutic strategies for human M2-related diseases through mTOR inhibitor treatment. This article is protected by copyright. All rights reserved.
Article
The enteropathy associated with common variable immunodeficiency (CVID) is poorly characterized, and its possible relationships with well-defined causes of enteropathy, such as celiac sprue (CS), remain debated. We aimed to assess the clinical and histopathological features of the enteropathy associated with CVID. The medical files of 50 CVID patients with gastrointestinal symptoms were analyzed retrospectively. Histological, phenotypic, and molecular analysis of intestinal endoscopic specimens was centrally performed. Chronic diarrhea was the most frequent gastrointestinal symptom (92%), and biological evidence of malabsorption was observed in 54% of patients. Chronic gastritis associated or not with pernicious anemia and microscopic colitis were the most frequently observed histopathological features in gastric and colonic mucosa, respectively. Small-bowel biopsies available in 41 patients showed moderate increase in intestinal intraepithelial lymphocytes in 31 patients (75.6%) and villous atrophy in 21 patients (51%). Distinctive features from CS were a profound depletion in plasma cells and follicular lymphoid hyperplasia. Presence of peripheral blood CD8+ hyperlymphocytosis was predictive of intestinal intraepithelial hyperlymphocytosis. Intravenous (i.v.) immunoglobulin (Ig) therapy had no effect on enteropathy-related symptoms. Gluten-free diet improved only two out of 12 patients with villous atrophy, whereas all patients (7/7) responded to steroid therapy. Several distinctive features differentiate CVID enteropathy from other causes of enteropathy including CS. Replacement i.v. Ig therapy is insufficient to improve gastrointestinal symptoms. Steroids are effective in reducing inflammation and restoring mucosal architecture.