ArticlePDF Available

Impaired osteoblast and osteoclast function characterize the osteoporosis of Snyder - Robinson syndrome

Authors:

Abstract and Figures

Snyder-Robinson Syndrome (SRS) is an X-linked intellectual disability disorder also characterized by osteoporosis, scoliosis, and dysmorphic facial features. It is caused by mutations in SMS, a ubiquitously expressed gene encoding the polyamine biosynthetic enzyme spermine synthase. We hypothesized that the tissue specificity of SRS arises from differential sensitivity to spermidine toxicity or spermine deficiency. We performed detailed clinical, endocrine, histopathologic, and morphometric studies on two affected brothers with a spermine synthase loss of function mutation (NM_004595.4:c.443A > G, p.Gln148Arg). We also measured spermine and spermidine levels in cultured human bone marrow stromal cells (hBMSCs) and fibroblasts using the Biochrom 30 polyamine protocol and assessed the osteogenic potential of hBMSCs. In addition to the known tissue-specific features of SRS, the propositi manifested retinal pigmentary changes, recurrent episodes of hyper- and hypoglycemia, nephrocalcinosis, renal cysts, and frequent respiratory infections. Bone histopathology and morphometry identified a profound depletion of osteoblasts and osteoclasts, absence of a trabecular meshwork, a low bone volume and a thin cortex. Comparison of cultured fibroblasts from affected and unaffected individuals showed relatively small changes in polyamine content, whereas comparison of cultured osteoblasts identified marked differences in spermidine and spermine content. Osteogenic differentiation of the SRS-derived hBMSCs identified a severe deficiency of calcium phosphate mineralization. Our findings support the hypothesis that cell specific alterations in polyamine metabolism contribute to the tissue specificity of SRS features, and that the low bone density arises from a failure of mineralization.
Content may be subject to copyright.
R E S E A R C H Open Access
Impaired osteoblast and osteoclast function
characterize the osteoporosis of Snyder - Robinson
syndrome
Jessica S Albert
1,2
, Nisan Bhattacharyya
3
, Lynne A Wolfe
1,2
, William P Bone
1
, Valerie Maduro
1
, John Accardi
1
,
David R Adams
1,2
, Charles E Schwartz
4
, Joy Norris
3
, Tim Wood
4
, Rachel I Gafni
3
, Michael T Collins
3
, Laura L Tosi
5,6
,
Thomas C Markello
1,2
, William A Gahl
1,2*
and Cornelius F Boerkoel
1
Abstract
Background: Snyder-Robinson Syndrome (SRS) is an X-linked intellectual disability disorder also characterized by
osteoporosis, scoliosis, and dysmorphic facial features. It is caused by mutations in SMS, a ubiquitously expressed
gene encoding the polyamine biosynthetic enzyme spermine synthase. We hypothesized that the tissue specificity
of SRS arises from differential sensitivity to spermidine toxicity or spermine deficiency.
Methods: We performed detailed clinical, endocrine, histopathologic, and morphometric studies on two affected
brothers with a spermine synthase loss of function mutation (NM_004595.4:c.443A > G, p.Gln148Arg). We also
measured spermine and spermidine levels in cultured human bone marrow stromal cells (hBMSCs) and fibroblasts
using the Biochrom 30 polyamine protocol and assessed the osteogenic potential of hBMSCs.
Results: In addition to the known tissue-specific features of SRS, the propositi manifested retinal pigmentary
changes, recurrent episodes of hyper- and hypoglycemia, nephrocalcinosis, renal cysts, and frequent respiratory
infections. Bone histopathology and morphometry identified a profound depletion of osteoblasts and osteoclasts,
absence of a trabecular meshwork, a low bone volume and a thin cortex. Comparison of cultured fibroblasts from
affected and unaffected individuals showed relatively small changes in polyamine content, whereas comparison of
cultured osteoblasts identified marked differences in spermidine and spermine content. Osteogenic differentiation
of the SRS-derived hBMSCs identified a severe deficiency of calcium phosphate mineralization.
Conclusions: Our findings support the hypothesis that cell specific alterations in polyamine metabolism contribute
to the tissue specificity of SRS features, and that the low bone density arises from a failure of mineralization.
Keywords: Spermine, Snyder-Robinson syndrome, Osteoblast, Osteoclast, Osteoporosis
Background
Polyamines are ubiquitous, aliphatic, positively charged
molecules that interact with anionic compounds such as
DNA, RNA, and ATP [1,2]. Homeostasis of the poly-
amines putrescine, spermidine, and spermine is essential
to cell growth and survival [3]. By addition of a propyla-
mine moiety, spermidine synthase (SRM) converts pu-
trescine into spermidine, and spermine synthase (SMS)
converts spermidine into spermine [4]. The balance of
spermine and spermidine is crucial for ion channel regu-
lation, transcription and translation [5-9].
Mutations of SMS, the gene encoding spermine synthase,
cause Snyder-Robinson syndrome (SRS), an X-linked
disorder first reported in 1969 [10]. The clinical features
of SRS include intellectual disability, dysmorphic facies,
speech and gait abnormalities, seizures, muscle hypopla-
sia, kyphoscoliosis, and osteoporosis [11-17]. All affected
males have hemizygous mutations in SMS that result in
reduced SMS activity and a decreased spermine:spermi-
dine ratio.
* Correspondence: gahlw@helix.nih.gov
1
Undiagnosed Diseases Program, Common Fund, Office of the Director,
National Institutes of Health, Bethesda, MD 20814, USA
2
Medical Genetics Branch, National Human Genome Research Institute,
Bethesda, MD, USA
Full list of author information is available at the end of the article
© 2015 Albert et al.; licensee BioMed Central. This is an Open Access article distributed under the terms of the Creative
Commons Attribution License (http://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and
reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain
Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article,
unless otherwise stated.
Albert et al. Orphanet Journal of Rare Diseases (2015) 10:27
DOI 10.1186/s13023-015-0235-8
Atraumatic osteoporotic fractures commonly occur in
individuals with SRS, leading to significantly impaired
quality of life. Osteoporosis arises from disruption of the
equilibrium between osteoclastic bone resorption and
osteoblastic bone formation [18], which is regulated by
mechanical and endocrine stimuli [19]. This general un-
derstanding of osteoporosis has led to established thera-
peutic interventions, but further insights are required to
address the osteoporosis of SRS in a disease-specific
manner.
Here we define the osteoporotic disease of SRS in two
brothers with a missense mutation in SMS [20] and report
depletion of osteoblasts and osteoclasts, reduced cancel-
lous and cortical bone, reduced calcium-phosphate
mineralization in vitro, and markedly abnormal polyamine
content in human bone marrow stromal cells (hBMSCs).
These data offer new insights into the role of polyamines
in bone formation.
Clinical reports
Patient II-1
The propositus (II-1, Figure 1) is the 18-year-old son of
non-consanguineous healthy parents with no family his-
tory of intellectual disability or skeletal problems.
He was born by cesarean section at 40 weeks following
a gestation complicated by poor maternal weight gain and,
at 8 months, atypical fetal movements suggestive of in
utero seizures. His birth weight, length and occipitofrontal
circumference (OFC) were 2.38 kg (3%), 47 cm (13%) and
34.5 cm (26%), respectively. Apgar scores were 8 and 9 at
one and five minutes. He had saggy skin but no other
dysmorphic features. Following a perinatal intraven-
tricular hemorrhage associated with thrombocytopenia
(7×10
3
cells/μl) that corrected after 3 platelet transfusions,
he developed seizures, apnea with cyanosis, temperature
instability and hypoglycemia. His recurrent episodes of
hyperglycemia and hypoglycemia resolved with age and
the placement of a gastric tube that allowed more frequent
feedings.
Patient II-1 had tracheomalacia, upper airway obstruc-
tion, increased respiratory secretions, frequent aspira-
tions, and pulmonary infections from infancy. By age 7
years, he had chronic Pseudomonas aeruginosa respira-
tory infection. At 6 years, he developed proximal renal
tubular acidosis (RTA), nephrocalcinosis and nephro-
lithiasis. His renal stones were composed of carbonate
apatite and calcium oxalate. His RTA has been managed
with fluid and electrolyte replacement, spironolactone
and hydrochlorothiazide.
Although initially controlled with phenobarbital, his
seizures progressed to infantile spasms by 15 months.
Adrenocorticotropic hormone (ACTH) treatment transi-
ently reduced seizure frequency. His current anticonvul-
sant therapy includes rufinamide, felbamate, clonazepam,
and topiramate. He had delayed development with regres-
sion of several developmental milestones. He smiled and
laughed by 4 months, reached for toys by 6 months, had
vocalizations by 7 months, and could hold his head up
and roll onto his side by 8 months. He achieved his pincer
grasp at 18 months but lost it by 23 months. He never
walked. He lost many motor skills and all vocalization by
26 months of age. A cranial MRI at 14 years revealed cys-
tic encephalomalacia and a low parenchymal T2 signal in
the right temporal and right occipital lobes. These findings
were considered consequences of the perinatal intraven-
tricular hemorrhage.
When first evaluated at NIH at 15 years, Patient II-1
was awake but not interactive and could not sit inde-
pendently or hold up his head; he withdrew from nox-
ious stimuli. His height, weight and OFC were 129 cm
(<3%ile), 30.8kg (<3%ile) and 50.3 cm (<3%ile), respect-
ively. Facial dysmorphisms included a long, oval, asym-
metric face, midface hypoplasia, down-slanting palpebral
fissures, large, cupped ears, smooth philtrum, high-
arched palate and prognathia (Figure 1). His dental en-
amel was hypoplastic and secondary teeth 2, 6, 10, and
11 were absent. He had excessive drooling, sluggish
pupillary reflexes and left-sided hearing loss. He had fre-
quent seizures, severe hypotonia, decreased muscle bulk,
hypoactive deep tendon reflexes, kyphoscoliosis and flexion
contractures of most large and small joints (Figure 1). He
had one prepubertal (1-2 mL) testis that was undescended
but palpable in the inguinal canal and one undescended
testis, Tanner stage III pubic hair and a prepubertal
phallus. His skin was remarkable for excretions of carbon-
ate apatite, reflecting calcium/phosphate dysregulation.
His ophthalmologic exam revealed retinitis pigmentosa
and cortical blindness.
Previous skeletal problems included congenital bilateral
hip dislocation and fractures of his distal fibula (2 years),
right humerus (5 years) and spine (6 years). Kyphoscoliosis
developed between 6 and 12 years of age. A dual-energy
x-ray absorptiometry (DEXA) scan performed at age 15
years demonstrated a bone density of 0.341 gm/cm
2
BMD
(height adjusted Z-Score: -2.9 [21]) for the anteroposterior
Spine (L1-L4) and 0.342 gm/cm
2
bone mineral density
(height adjusted Z-Score: -6.5 [21]) for the right forearm.
Skeletal radiographs at 18 years revealed a 60° convex
right scoliosis, gracile bones with reduced mineral density,
and evidence of previous fractures (Figure 2E-J).
Patient II-3
Patient II-3, the brother of Patient II-1, was born at 37
weeks by cesarean section following an uncomplicated
pregnancy. His birth weight was 2.81 kg (25%). During
the immediate neonatal period his platelet count de-
creased from 90 to 45 k/μL, and he was admitted to
the neonatal intensive care unit for treatment with
Albert et al. Orphanet Journal of Rare Diseases (2015) 10:27 Page 2 of 13
Figure 1 Clinical and radiographic features. A. Face of patient II-1. B. Face of patient II-3. C. Palate of patient II-1. D. Hands of patient II-3. E-J.
Skeletal radiographs of Patient II-1 showing the left humerus (E), left forearm (F), left hand (G), pelvis (H), left femur (I) and left lower leg (J). Note
the gracile bones and undermineralization as well as the healing humeral fracture (E).
Albert et al. Orphanet Journal of Rare Diseases (2015) 10:27 Page 3 of 13
Figure 2 (See legend on next page.)
Albert et al. Orphanet Journal of Rare Diseases (2015) 10:27 Page 4 of 13
dexamethasone and a platelet transfusion. He also had
transient hypoglycemia, poor feeding and sensitivity to
light. By 4 days of life, his condition had stabilized and
he was discharged. Over the subsequent months, he
manifested moderate laryngomalacia, mild tracheobro-
chomalacia, severe torticollis and abnormally pigmen-
ted retinas. Like his brother, he has also had episodic
hyper- and hypoglycemia. He was diagnosed with
nephrocalcinosis at 1 year and RTA by 2 years. The
composition of his renal stones was carbonate apatite
and calcium oxalate.
Patient II-3 had global developmental delay, severe
hypotonia, and regression of milestones; he lost vocaliza-
tions and most motor skills by 15 months. An EEG at 6
months showed generalized slowing and disorganization;
at 18 months he manifested seizures and hypsarrhyth-
mia. An MRI performed at 2 years of age showed a mild
increase in ventricular size but no other abnormalities.
There was no evidence of hemorrhage or malformation.
Since age 5 years, he has required a vagal nerve stimula-
tor and bi-pap for adequate respiration. He also had re-
peated Pseudomonas aeruginosa pulmonary infections.
His infections are more frequent and severe than are
those of his brother.
Patient II-3 has had multiple atraumatic fractures in-
volving the clavicle, tibia, femur and humerus. He also
had congenital bilateral hip dislocation and, by 4 years
of age scoliosis.
Illness prevented Patient II-3 from traveling to the
NIH for evaluation, but review of his medical records
and photographs showed that he was alert and non-
ambulatory at age 10 years. He had facial features similar
to those of his brother (Figure 1), superficial skin excre-
tions, hypotonia, large joint contractures and muscle
atrophy.
Laboratory studies
The propositi had extensive laboratory testing. This
identified an elevated antibody titer to myelin basic pro-
tein and a mild intermittent anemia with low iron satur-
ation (7%; normal, 15-62) and ferritin (20 mcg/L; normal,
26 -388) for Patient II-1 and an elevated blood lactate level
of 7.8 mmol/L (0.5-2.2) and mild elevations of urine carni-
tine esters for Patient II-3. Testing for abnormalities in or-
ganic acids, amino acids, acylcarnitines, very long chain
fatty acids, lysosomal enzymes, biotinidase and copper
were unremarkable. Molecular testing of MECP2,mito-
chondrial DNA, a panel of lysosomal storage disease-
associated genes and the X-Linked Mental Retardation
9 Gene Panel (Greenwood Clinic, 2008), which did not
include SMS and FRMPD4, did not detect pathogenic
mutations. Patient II-1 had a normal karyotype (46,XY),
and clinical and research copy number variant analysis
did not detect any pathogenic variants (Additional file 1:
Table S 1 ) .
Methods
Patients
The propositi were accepted into the NIH Undiagnosed
Diseases Program (UDP) and enrolled in clinical proto-
col 76-HG-0238, approved by the Institutional Review
Board of the National Human Genome Research
Institute. Their parents gave written, informed consent.
CNV analysis
The NHGRI Genomics Core lab performed SNP determina-
tionsusingtheIlluminaBeadArrayPlatform(HumanOm-
niExpress, Illumina Corp., San Diego, CA, USA). Genome-
wide fluorescent intensities and genotype calls were analyzed
using Bead Studio and Genome Studio (Illumina Corp.).
Analysis of copy-number variations was performed using
PennCNV software, [22] and visual inspection using Gen-
ome Studio version 2010v3 build37/hg19 [23].
Exome sequence analysis
Genomic DNA was extracted from whole blood using
the Gentra Puregene Blood kit (Qiagen, Valencia, CA)
according to the manufacturers specifications. Exome
sequencing and analysis were performed as described
[24-26]. The potential pathogenicity of identified vari-
ants was predicted using CDPred, SIFT, PolyPhen2
[27-33] or the BLOSUM62 scoring matrix [34].
(See figure on previous page.)
Figure 2 Segregation, mutational analysis, and functional consequences of a novel SMS variant. A. Pedigree of the family of the propositi.
Affected males are shown by black squares. B. Sanger sequencing chromatograms showing the segregation of the SMS mutation NM_004595.4:c.443A
> G from the carrier mother to the affected boys. The unaffected father did not have this mutation. C. Conservation of the p.Gln148 (p.Q148) residue
across species. D. Drawing of the human SMS protein crystal complexed with spermidine and 5-methylthioadenosine. The mutated amino acid
(Gln148) is highlighted in yellow [Mac PyMOL [23]]. E-J. Immunofluorescent detection of SMS protein subcellular distribution in unaffected (E, F),
Patient II-1 (G, H) and Patient II-3 (I, J) skin fibroblasts. SMS protein is shown in red and the nucleus is shown in blue. K. Immunoblot of skin fibroblast
lysates showing reduced SMS protein levels in the patients (II-1, II-3) compared to an unaffected control (cnt). Tubulin is shown as a loading control. L.
Graph showing steady state SMS protein levels in the patient and control fibroblasts relative to ß-tubulin levels. The data are based on 3 independent
experiments for each cell line. M. Graph quantifying immunoblot detected steady state SMS protein levels in the cytoplasm and nuclei of patient and
control fibroblasts. The cytoplasmic expression was normalized to β-tubulin expression and the nuclear expression to p84 expression. The data are
based on 2 independent experiments for each cell line. N. SMS enzyme activity (spermidine d8 peak per hour) in lymphoblasts of unaffected individuals
(Cnt), a cohort of 4 individuals with SRS (SRS) and patient II-1, * p < 0.05.
Albert et al. Orphanet Journal of Rare Diseases (2015) 10:27 Page 5 of 13
Confirmatory sequencing
For the SMS variant, Qiagen HotStarTaq master mix
(Qiagen, Valencia, CA) was used to amplify the putative
variant and 200 flanking nucleotides using the primers 5-
TGTGGCTTTCTTTTGCACAC-3and 5-TGCATCT
CAAAAACCAGCAG-3. Unincorporated primers and
nucleotides were removed using ExoSAP-IT reagent
(USB, Cleveland, OH, USA). Sanger capillary sequencing
was used to sequence the PCR products (Macrogen,
Rockville, MD), and the sequences were aligned and ana-
lyzed using Sequencher v.4.10.1 (Gene Codes, Ann Arbor,
MI, USA). Mutation interpretation was conducted using
Alamut 2.0 (Interactive Biosoftware, San Diego, CA, USA).
Bone biopsy and histomorphometry
Patient II-1 was given two courses of demeclocycline
prior to the biopsy so that dynamic histomorphometry,
including measurement of bone turnover, could be per-
formed [35]. A bicortical transiliac crest core biopsy was
performed and immediately split. The sample was placed
in 70% ethanol and sent to The Johns Hopkins School of
Medicine for histomorphometry, which was performed
as described [35].
Western blot analysis
Protein was extracted from fibroblast and osteoblast cell
lines with RIPA buffer (Thermo Scientific, Waltham, MA)
or used directly from Clontech human protein library (Clon-
tech Laboratories, Mountain View, CA). Lysates (30ug) were
electrophoresed on a 4-10% SDS-polyacrylamide gel and
transferred to a polyvinylidene fluoride (PVDF) membrane.
Using gentle agitation, the membrane was blocked overnight
at 4ºC with casein blocking buffer (Thermo Scientific,
Waltham, MA) and 10% horse serum. Anti-SMS (1:1000)
(Novus Biologicals, Littleton, CO), anti-β-tubulin (1:1000,
AbCam, Cambridge, UK), and anti-GAPDH (1:1000, Gene
Tex, Irvine, CA) were used as primary antibodies. Horsera-
dish peroxidase-conjugated secondary antibodies (1:10000,
Bio-rad Laboratories, Hurcules, CA) were used to detect the
primaryantibodies.Theantibody-enzyme complexes were
detected by chemiluminescence using Amersham ECL west-
ern blotting detection reagent (GE Life Sciences, Pittsburgh,
PA) or WesternSure Premium Chemiluminescent Substrate
(LI-COR, Lincoln, NE) according to the manufacturers
specifications. β-tubulin or GAPDH was detected as a load-
ing control.
Cellular fractionation
Cellular fractionation was performed using NE-PER Nuclear
and Cytoplasmic Extraction Reagents (Thermo Scientific,
Waltham, MA) per the manufacturers recommendations.
Immunoblotting was performed as described above. β-tubu-
lin and p84 (GeneTex, Irvine, CA) served as loading con-
trols for the cytoplasmic and nuclear fractions, respectively.
qRT-PCR analysis
RNA was extracted from cells using the RNeasy Mini Kit
(Qiagen, Valencia, CA) per the manufacturers specifica-
tions. RNA was converted to cDNA using the VILO cDNA
synthesis kit (Life Technologies, Grand Island, NY) per the
manufacturers protocol. 100ng of cDNA was amplified
using Sso Advanced SYBR Green supermix (Bio-Rad
Laboratories, Richmond, CA) per the manufacturersspeci-
fication. qRT-PCR for measuring SMS steady state mRNA
levels was performed on the Bio-Rad CFX96 Real-Time
system (Bio-Rad Laboratories, Richmond, CA) using
primers 5-gattggtgttgctggacctt-3and 5-tgactcaattcttt
cattctttcct-3. PCR was cycled 33 times and annealing
temperature was 58 degrees, melt curve was incremented
at 0.5 degrees from 65-95 degrees. mRNA levels were nor-
malized to GAPDH mRNA levels, a house-keeping gene.
Cell culture
Epstein-Barr virus (EBV)-transformed lymphoblast cells
were cultured as described previously [36]. Fibroblast cells
were cultured in Dulbeccos Modified Eagle Medium
(DMEM) supplemented with 4.5 g/L D-glucose, L-glutam-
ine, sodium pyruvate (Life Technologies, Grand Island,
NY), 10% fetal bovine serum (Life Technologies, Grand
Island, NY) and 1 X antibiotic (Life Technologies, Grand
Island, NY). Fibroblasts were grown at 37°C with 5% CO2.
Human bone marrow stromal cells (hBMSC) were iso-
lated and grown in culture as previously described [37].
Briefly, bone biopsy samples from patient(s) were used as
the starting material. Cells were grown in α-minimal es-
sential medium (α-MEM) containing 20% fetal bovine
serum (FBS, Atlanta Biologicals, Lawrenceville, GA, USA),
L-glutamine (Glutamax, GIBCO, Carlsbad, CA, USA), and
penicillin-streptomycin mix. When noted, cells were also
grown in the above medium containing Dexamethasone
(10-8 M dexamethasone and 10-4 M ascorbate). Cells
were cryopreserved in α-MEM containing 50% FBS and
5% dimethyl sulphoxide (DMSO). Control hBMSC were
obtained from the rib of a 51 year-old Caucasian male.
Osteogenic differentiation assay
Control and experimental BMSCs were plated (6x104/12-
well plate) in triplicate. Cells were kept either untreated (α-
MEM containing 20% FBS, L-glutamine, antibiotics) or
treated with an osteogenic differentiation media (α-MEM,
20% FBS, L-glutamine, antibiotics, 5x10-3M β-glycerophos-
phate, 1x10-4M Ascorbic Acid Phosphate and 1x10-8M
dexamethasone). Media was changed every 3-4 days. After
18 days, cells were rinsed with HBSS (HanksBalanced Salt
Solution, Invitrogen, Grand Island, NY) and were fixed at
room temperature using 4% paraformaldehyde solution in
1X phosphate buffered saline (PBS). Cells were washed
with distilled water, and were stained with Alizarin Red so-
lution at room temperature for 20 minutes. Finally, the cells
Albert et al. Orphanet Journal of Rare Diseases (2015) 10:27 Page 6 of 13
were washed with distilled water for 6 times and were
photographed.
Spermine synthase activity assay
To test SMS activity in lymphoblastoid cell lines, we
measured the production of deuterated spermine
(spermine d8) from deuterated spermidine (spermidine
d8). Briefly, cells were harvested by centrifugation and
washed 2X with PBS and suspended in 50mM sodium
phosphate buffer pH 7.2 with protease inhibitor (Sigma,
St. Louis, MO). Samples were frozen at -80ºC. Upon
thawing, the samples were subjected to 2 freeze-thaw cy-
cles using an ethanol and dry ice bath. After centrifuga-
tion, protein in the supernatant was quantified using the
Lowry assay. To test for SMS activity, 70 μg of protein
was incubated with 0.1M sodium phosphate buffer 7.5,
10μM spermidine d8 (Sigma, St. Louis, MO), protease
inhibitor (Sigma, St. Louis, MO), 100μM dcSAM, and
50uM 4-MCHA (Sigma, St. Louis, MO) in a total vol-
ume of 100 μL. Baseline reactions were stopped immedi-
ately with 100μL acetonitrile/0.1% formic acid; other
samples were incubated at 37ºC for 24 h and then
stopped by addition of acetonitrile/0.1% formic acid.
Spermine d8 was quantified by LC/MS/MS. Enzymatic
activity was represented as the area of the spermidine d8
peak per hour.
Measurement of polyamine content
The spermine/spermidine ratio was determined in the
lymphoblastoid cell lines using LC/MS/MS as described
[36]. Fibroblast and hBMSC polyamines were measured
using the Biochrom 30 polyamine protocol. Briefly, hu-
man fibroblasts and hBMSCs were cultured as described
above. Cells were harvested and pelleted and polyamines
were extracted with a volume of 10% PCA equivalent to
4-fold the weight of the cells in milligrams. After 1 h of
incubation on ice, cells were centrifuged at 13,000 rpm
for 10 min at 4 degrees. The polyamines were fraction-
ated and quantitated on the Biochrom 30 using an ion
exchange polyamine column, compatible with sodium
chemistry.
Immunofluorescent localization of SMS
Immunofluorescent detection of SMS was modified from a
previously described protocol [38]. Briefly, 2 x 104 cells were
grown overnight on a coverslip in a 6-well plate. The cells
were fixed with 3.7% paraformaldehyde (PFA) for 25 min at
room temperature and permeabilized with 0.1% Triton X-
100, 2mg/ml BSA and 1mM NaN3 for 5 min at room
temperature. All cells were blocked for 2 h with casein
blocking buffer with 10% horse serum and then incubated
at 4°C overnight with anti-SMS (1:100) (Sigma, St. Louis,
MO) diluted in blocking buffer. They were then gently
washed 3 times with PBS and incubated with secondary
antibodies conjugated with Alexa 488 or Alexa 555 (2ug/ml,
Life Technologies, Grand Island, NY) for 2 h at room
temperature. Cells were then washed 3 times with PBS and
mounted in Vectashield containing 4, 6-diamidino-2-phe-
nylindole (DAPI, Vector Laboratories, Burlington, ON,
Canada). Images were acquired using a 63X Zeiss plan-apo-
chromat oil, 1.4 NA, DIC objective lens on a Zeiss LSM 780
confocal microscope using Zen 2011 acquisition software.
Alkaline phosphatase activity
Upon reaching confluence, hBMSCs were plated in
6- well plates (100,000 cells/well). After 10 days of
growth in MEM Alpha with 20% FBS, the hBMSCs
were stimulated with 10mM β-glycerophosphate (Sigma
St. Louis, MO), 50μM ascorbic acid 2-phosphate
(Sigma, St. Louis, MO) and 100nM Dexamethasone
(Sigma, St. Louis, MO) for 5 days. Alkaline phosphatase
activity was quantified using StemTAG Alkaline Phos-
phatase Colorimetric Kit (Cell Biolabs, San Diego, CA).
Results
Exome sequencing identifies a novel SMS mutation
diagnostic of SRS
The propositi had a maternally inherited hemizygous
transition (NM_004595.4:c.443A > G) in SMS identified
by exome sequencing (Additional file 1: Table S2; full
VCF file is available on request) and confirmed by
Sanger sequencing (Figure 2A-B). This mutation en-
codes the missense mutation p.Gln148Arg (CDPred
score: -9; SIFT score: 0; PolyPhen2: probably damaging
(0.998, Sensitivity 0.27, Specificity 0.99)). The Gln148
residue is conserved to S. cerevisiae (Figure 2C) and re-
sides in the central β-strand domain that functions as a
cap for the carboxyl terminal catalytic domain. It is one
of 8 residues involved in the binding of 5-methylthioa-
denosine (MTA) (Figure 2D) [39,40], which functions as
an amine acceptor.
Similar to other SRS-associated SMS mutations [11-17],
the p.Gln148Arg variant decreased the steady state level of
SMS protein detectable in cultured fibroblasts by immuno-
fluorescence and immunoblotting (Figure 2E-K). It reduced
total SMS steady state levels 2.6-fold (Figure 2L); nuclear
SMS was reduced 5.8-fold and soluble cytosolic SMS 2.7-
fold (Figure 2M).
Spermine d8 generation by lymphoblastoid lysates ex-
pressing p.Gln148Arg SMS was 37- fold less than that
for unaffected controls (Figure 2N). Additionally, as
measured by LC/MS/MS, the ratio of spermine: spermi-
dine in the lymphoblastoid lysates was reduced 10-fold
compared to unaffected controls.
The propositi had features previously not reported with
SRS, including more profound intellectual disability, retinal
pigmentary changes, renal dysfunction, frequent pulmonary
infections and hyper- and hypoglycemia (Table 1). Although
Albert et al. Orphanet Journal of Rare Diseases (2015) 10:27 Page 7 of 13
we hypothesized that rare mutations in other genes contrib-
uted to these features, only a maternally inherited X-linked
variant in FRMPD4 (c.583A > G, p.K195E) segregated with
the disease and had a predicted pathogenicity of at least
probably damaging in Polyphen [27]; SIFT and CDPred pre-
dict the variant as tolerated. FRMPD4, which has been asso-
ciated with autism and schizophrenia [41], encodes a
product that regulates dendritic spine morphogenesis [42].
No pathogenic mutations were observed in genes associated
with retinal pigmentary changes.
Bone formation is decreased in patient II-1
SMS is widely expressed (Additional file 1: Figure S1);
consequently, we questioned why specific tissues such as
bone are particularly affected in SRS patients. To define
better the osteoporosis of SRS, we performed a bicortical
transiliac crest core biopsy of Patient II-1. The specimen
was soft and fragmented when removed from the tre-
phine (Figure 3A). Histomorphometry revealed an ab-
sence of a trabecular meshwork, a low bone volume and
a thin cortex. Cancellous bone volume was markedly de-
creased at 4.7%, compared with 23% ± 4.4 in healthy
controls. The cortical mean width measured only 238
microns compared to a mean of 1202 microns in con-
trols. Osteoblastic activity was markedly reduced with
osteoblasts occupying only 1% of the osteoid surface
(normal range 12.1% ± 4.6). There was no observable
osteoclastic activity in the patientsspecimen,dem-
onstrated by an eroded surface of 0% (normal range
4.1% ± 2.3) and an osteoclast surface of 0% (normal
range 0.7% ± 0.6). Surface bone formation rate was
11.6 μm
3
/μm
2
/y (normal range: 35.8 μm
3
/μm
2
/y ± 8.9).
CSF neurotransmitter levels do not suggest sympathetic
nervous system dysfunction in Patient II-1
Because bone density and turnover are controlled in part
by the sympathetic nervous system [43-49], we measured
CSF neurotransmitters in Patient II-1 for evidence of in-
creased sympathetic activity. The 5-methyltetrahydrofolate
(methyl donor), 5-hydroxyindoleacetic acid (serotonin me-
tabolite), 3-O-methyldopa (metabolite of L-Dopa), and tet-
rahydrobiopterin (cofactor for the synthesis of serotonin,
dopamine, norepinephrine) were all within normal limits.
Although homovanillic acid (catecholamine metabolite)
(459 nmol/L) and neopterin (catabolic product of GTP)
(30 nmol/L) were slightly above normal limits (324 and 28
nmol/L, respectively), these slight elevations were consid-
ered clinically insignificant.
Endocrine evaluation in patient II-1
Because bone density is also modulated by multiple
endocrine regulators [19], we assessed these in Patient
II-1. Intact PTH, Vitamin D, magnesium, cortisol, insulin-
like growth factor 1, thyroxine, thyroid stimulating hor-
mone, prolactin and ACTH were unremarkable (Table 2).
Also, manual inspection of the exome results did not
identify any predicted deleterious mutations of IGF1 or
IGF1R despite >20 fold depth of short read coverage
across the length of each exon. Prior to admission to the
NIH, patients II-1 and II-3 had fluctuations in blood cal-
cium and phosphate levels (Table 3). These fluctuations
had no apparent pattern or mediating factor based on diet,
fluid intake, or clinical status, and at the time of admission
to NIH, the blood levels of calcium and phosphorus were
normal. Tubular reabsorption of phosphate was normal,
but, the calculated 24-hr urine calcium excretion (derived
from an 18-hr collection) was elevated. Osteocalcin levels
were low for pubertal stage, consistent with decreased
bone formation. At age 15 years, patient II-1 had undes-
cended testes, remarkably delayed puberty, low LH and
FSH levels and undetectable testosterone. By age 18 years,
he had entered early puberty as evidenced by an increase
in LH and FSH, and testosterone levels in the Tanner II
range. Testicular ultrasound at that time identified both tes-
tes in the inguinal canal, with volumes of 5.7 mL and 1.7
mL. At a chronological age of 18 years and 4 months, his
hone age was 12 years and 6 months.
Table 1 Comparison of clinical features of all reported
SRS patients
Features Reported
patients
Patient II-1 Patient II-3
Cognitive impairment 15/15 + +
Seizures 8/15 + +
Myopia 2/13 + * + *
High, narrow palate 3/13 + +
Prominent lower lip 12/15 + -
Speech abnormalities 15/15 + ** + **
Diminished body bulk 15/15 + +
Kyphoscoliosis 13/15 + +
Osteoporosis 11/11 + +
Long finger/toes 12/15 + *** #
Unsteady gait 10/15 + **** + ****
Renal abnormalities 3/13 + +
Nephrocalcinosis
a
0/13 + +
Frequent infections 0/13 + +
Retinal pigment changes 0/13 + +
Hypo-/Hyper-glycemia 1/13 + +
Muscle fiber
abnormalities
1/13 + +
#Not assessed.
*Partial blindness.
**No vocalizations.
***Contractures of fingers.
****Non-ambulatory.
a
Note: Nephrocalcinosis in these patients previously recorded in
GeneReviews [20].
The phenotype data used to generate this table was collected from [11-17].
Albert et al. Orphanet Journal of Rare Diseases (2015) 10:27 Page 8 of 13
Figure 3 (See legend on next page.)
Albert et al. Orphanet Journal of Rare Diseases (2015) 10:27 Page 9 of 13
Compared to cultured SRS fibroblasts, SRS hBMSC have
comparable SMS mRNA and protein levels but pronounced
disturbances of polyamine levels
While absolute polyamine levels are not substantially
abnormal in cultured fibroblasts and lymphoblastoid cells
of SRS individuals, spermidine/spermine ratios reflected
the SMS enzyme deficiency [12-17]. Neither lymphoblas-
toid cells nor skin fibroblasts, however, are affected tissues
in SRS. We hypothesized therefore that cells from affected
tissues such as bone had differences in SMS expression or
polyamine metabolism accounting for their clinical mani-
festations. To test this, we compared SMS expression and
polyamine levels among cultured skin fibroblasts and
hBMSC. Cultured skin fibroblasts and hBMSC had com-
parable steady state SMS mRNA levels (Figure 3B) and
SMS protein levels (data not shown), suggesting that differ-
ential SMS expression is an unlikely basis for expression of
SRS features. Additionally, excluding differential degrad-
ation of the mutant SMS protein as the basis for expression
of SRS features, the cultured hBMSC derived from Patient
II-1 had SMS protein levels only 1.7-fold lower than for
control hBMSC (Figure 3C-K), whereas his fibroblasts had
2.6-fold less SMS protein compared to control fibroblasts.
To test if differences in polyamine metabolism might
contribute to the differential expression of SRS features,
we compared polyamine levels in cultured skin fibro-
blasts and hBMSCs. Skin fibroblast lysates derived from
Patients II-1 and II-3 had mean spermidine levels 1.78-
fold higher than control fibroblasts (p = 0.28) and mean
spermine levels 7.40-fold lower than control fibroblasts
(p = 0.21). In contrast, lysates of cultured hBMSCs from
Patient II-1 contained mean spermidine levels 5.07-fold
higher (SEM = 22.8, p = 0.001) than control hBMSCs
(Figure 3L) and had no detectable spermine.
SRS hBMSCs have decreased osteogenic activity
To determine the differences in osteogenic potential,
hBMSCs isolated from an unaffected control and Patient
II-1 were treated with osteogenic differentiation media
and stained with Alizarin Red S, an anthraquinone dye
that stains the calcium deposits indicative of mature os-
teocytes [50,51]. Based on the intensity of Alizarin Red S
Table 2 Markers of bone and endocrine function in
Patient II-1 at 18 years of age
Metabolite Patient II-1
values
Reference
range
Intact parathyroid hormone (pg/mL) 28.9 (15-65)
1,25-dihydroxycholecalciferol (pg/mL) 70 (18-64)
25-dihydroxycholecalciferol (ng/mL) 39 (33-100)
Osteocalcin (ng/mL) 35.7 (7.3-38.5)
1
,
(49-167)
2
Free thyroxine (ng/dL, direct dialysis) 1.6 (1-2.4)
Thyroid stimulating hormone (μIU/mL) 1.8 (0.4-4)
Testosterone (ng/mL) 43.6 (100-740)
1
,
(8-418)
2
Free testosterone (ng/dL) 0.5 (7.4-22.6)
1
Sex hormone binding
globulin (nmol/L)
63 (10-60)
1
,
(44-160)
2
Follicle stimulating hormone (U/L) 8.7 (1-11)
1
Luteinizing hormone (U/L) 2.5 (1-8)
1
Prolactin (mcg/dL) 9.4 (2-25)
Cortisol (mcg/dL, morning) 16.5 (5-25)
Adrenocorticotropic hormone (pg/mL) 12.7 (0.0-46.0)
Insulin-like growth factor (ng/mL) 347 (75-420)
2
pH Urine 8.5 (5-8)
18-hr urine volume (mL/24 h) 2885 (600-1800)
Calculated urine calcium excretion
(mmol/kg/24 h)
0.134 (<0.1)
Tubular reabsorption of
phosphorus (%TRP)
93.5 (85-95)
1
Reference ranges for age,
2
Reference range for pubertal stage.
(See figure on previous page.)
Figure 3 Characterization of the bone and osteoblast pathology. A. Photograph of the bone biopsy. B. Steady state SMS mRNA levels
relative to GAPDH expression in cultured fibroblasts and osteoblasts. The patients cells did not differ significantly from controls. Data were
derived by qRT-PCR analysis of 3 independent extractions of total RNA. C. Immunoblot showing steady state SMS protein expression in patient
and control osteoblasts. ß-tubulin is shown as a loading control. D. Graph showing steady state SMS protein levels in the patient and control
hBMSCs relative to ß-tubulin levels; there was no significant difference. The data are based on 3 independent experiments for each cell line.
E-J. Immunofluorescent detection of SMS protein subcellular distribution in unaffected (E-G) and Patient II-1 (H-J) hBMSCs. SMS protein is shown
in red and the nucleus is shown in blue. K. Graph quantifying immunoblot detected steady state SMS protein levels in the cytoplasm and nuclei
of patient and control hBMSCs. The cytoplasmic expression was normalized to β-tubulin expression and the nuclear expression to p84 expression.
L. Polyamine quantification in fibroblasts and osteoblasts. Note that the patient hBMSCs have a more striking imbalance of spermidine and spermine
levels than do the patient fibroblasts, * p < 0.05, *** p < 0.005. M. Osteogenic potential of bone marrow stromal cells (hBMSCs) isolated from Patient II-1
sample is markedly lower than that of an unaffected control (cnt). The hBMSCs were seeded in triplicates (6x10
4
/12-well) and either kept untreated (-)
or treated (+) with osteogenic differentiation media (see Methods) for 18 days. After the treatment, cells were fixed and were stained with Alizarin Red
S to check for calcium deposition, a marker of osteogenic differentiation.
Table 3 Calcium phosphate levels in patients II-1 and II-3
Serum Metabolite Patient II-1 Patient II-3 Reference range
Calcium (mg/dL) 7.8-10.6 6.6-10.1 8.78-10.5
Phosphorus (mg/dL) 2.4-5.1 3.5-5.3 3.1-5.1
Albert et al. Orphanet Journal of Rare Diseases (2015) 10:27 Page 10 of 13
staining, the differentiated hBMSCs from Patient II-1
produced markedly fewer calcium deposits than did
those from the control (Figure 3M). Addition of 1μM
spermine did not alter the Alizarin Red S staining (data
not shown).
Discussion
We report two brothers with SRS in whom we identified
aSMS mutation (NM_004595.4:c.443A > G, p.Gln148Arg)
resulting in near absence of enzyme activity and decreased
steady state SMS protein levels. To better delineate the
tissue-specificity of SRS features, we investigated the
low bone density of SMS and observed functional osteo-
blast and osteoclast deficiencies, a marked spermidine
and spermine imbalance in hBMSCs and poor calcium
phosphate mineralization by differentiated hBMSCs.
We therefore speculate that polyamines play a critical
role in osteoblasts that is not required by other cells,
such as lymphoblasts and fibroblasts.
The p.Gln148Arg mutation represents the first SMS
mutation to alter the MTA binding site [52]. Disturbance
of MTA homeostasis could contribute to the more severe
phenotype of the propositi relative to that of other SRS
patients. MTA is needed for the transfer of the aminopro-
pyl group from decarboxylated S-adenosylmethionine
(dcAdoMet) to spermidine; it is also an inhibitor of SRM
and SMS [53], and consequently decreases cellular sperm-
ine concentrations [54]. Indeed, the spermine:spermidine
ratio of our patientscells was reduced 3-fold more than
the ratio for cells tested from any other SRS patient (C.S.,
unpublished data).
The severity of the phenotype in the propositi relative to
other individuals with SRS might alternatively be attribut-
able to differing insults or genetic backgrounds. For ex-
ample, the perinatal intraventricular hemorrhage of patient
II-1 might contribute to the severity of his neurological fea-
tures, although the presence of similar neurological features
in his brother, who did not have an ischemic or
hemorrhagic brain insult, suggests this is not a substantial
contributor. On the other hand, the variant in FRMPD4
might modify the genetic background and thereby contrib-
ute to the intellectual disability of the propositi. We did not
identify other environmental insults or appropriately segre-
gating, pathogenic variants to explain the additional fea-
tures of the propositi. Consequently, if other genetic
contributors have a significant role in modifying the pheno-
type of SRS, they were either not detected by our exome se-
quencing or were common polymorphisms excluded by
our analyses. It remains possible that epigenetic and sto-
chastic factors also modulate the expressivity of SRS.
Central neuroendocrine signaling does not appear to be
impaired in SRS; however, peripheral neuroendocrine sig-
naling including hypogonadism and altered calcium and
phosphate homeostasis might contribute as might other
issues such as immobilization, renal tubular acidosis, low
muscle mass and medications. It is thought that male
hypogonadism decreases bone mineral density because
androgens promote osteoblast differentiation and prolifer-
ation and decrease the activity of osteoclasts [55]. Add-
itionally, impaired calcium and phosphate homeostasis
impede osteoclast function [56], and hypercalciuria is as-
sociated with decreased bone density, as well as the
nephrocalcinosis seen in this patient. Several anticonvul-
sants, including clonazepam and topiramate, are also asso-
ciated with decreases in bone mineral density [57]. These
factors alone are unlikely to fully account for the de-
creased bone mineral density observed in our patients,
since other SRS individuals have had low bone density in
the absence of these issues [11]. Rather, the predominant
mechanism of osteoporosis in SRS is likely related to im-
paired polyamine metabolism.
The disease mechanisms and phenotypic expansion re-
ported herein provide some insight for the management of
SRS. Optimal control of the metabolic abnormalities, lim-
ited use of medications known to affect bone, and appro-
priate physical therapy should be part of the management
plan in any chronically ill, immobilized individual. Since
analysis of the bone did not detect increased osteoclastic
activity, bisphosphonate therapy would likely be of minimal
effectiveness unless further studies refute our observations.
In addition, if study of additional patients establishes the
association of renal disease, retinal pigmentary changes,
and perturbations of glucose homeostasis with SRS, then
screening for and symptomatic management of these prob-
lems has the potential to improve patient care.
Conclusions
This report identifies a novel SRS-associated SMS muta-
tion, p. Gln148Arg, and expands the SRS phenotype. It
also provides the first evidence that SRS patients have a
loss of osteoblast and osteoclast activity and that the low
bone density of SRS likely arises by a cell intrinsic process.
Consent
The patientsparents gave written, informed consent for
publication of this case report and any accompanying
photographs.
Additional file
Additional file 1: Table S1. CNVs detected in the propositi. Table S2.
Exome variants meeting rarity and predicted deleteriousness requirements
and segregating with disease. Figure S1. Expression profile of SMS mRNA
and protein. (A) Graph showing qRT-PCR detection of SMS mRNA levels in
total RNA extracted from the respective tissues. SMS mRNA levels
were normalized to the mRNA levels of GAPDH. (B) Immunoblot
showing SMS protein levels in lysates from the respective tissues. (C)
SMS protein expression in each tissue plotted relative to GAPDH protein
expression.
Albert et al. Orphanet Journal of Rare Diseases (2015) 10:27 Page 11 of 13
Competing interests
The authors declare that they have no competing interests.
Authorscontributions
JSA designed, performed and interpreted all of thepolyamine assays, the
protein and RNA expression and imaging assays and drafted the manuscript.
Coordinated the re-admission of the patient so, bone biopsy and further
endocrinology studies could be obtained. NB cultured human bone marrow
stromal cells and performed the differentiation experiments and the calcification
assays. LW was the primary nurse practitioner involved with thepatients. She
coordinated the clinical sample testing and performed history and physical
exam. WB aided in the culturing human bone marrow stromal cells and
performed the differentiation experiments and the calcification assay and
aided in the analysis of the NGS and SNP chip data. VM cultured human
bone marrow stromal cells and performed the differentiation experiments
and the calcification assays. JA did the sanger confirmation. DA was the
attending physician when the patient was admitted, and coordinated
clinical care while at the NIH. CS helped with coordination of the enzyme
assay and provided samples from other individuals with SRS and expertise
about the disorder. JN performed the spermine synthase enzymatic assay.
TW helped with interpretation of the enzymatic assays. RG Was the
endocrinologist involved with the case that helped partition the bone
biopsy sample in the operating room and coordinated bone marrow stem
cell culture. MC provided consultation for endocrinology findings. LT performed
the bone biopsy. TM performed the NGS analysis that identified the variant in
SMS. WG coordinated the admission of these individuals into the UDP, curated
the chart, provided insights into interpretation of data and obtained funding for
the studies. All authors read and approved the final manuscript.
Acknowledgements
We thank Drs. Paul Lee, May Christine Malicdan and Grace Zhai for critical
review of the manuscript. We would like to thank Dr. Edward McCarthy at
The Johns Hopkins School of Medicine for performing the bone
histomorphometry, Dr. Camilo Toro for imaging interpretations and Shira
Ziegler for technical guidance for the alkaline phosphatase assay. This work
was supported by the Intramural Research Programs of the National Human
Genome Research Institute and National Institute of Dental and Craniofacial
Research and the Common Fund of the NIH Office of the Director, National
Institutes of Health. This work as also supported by extramural funding from
NINDS (NS073854) and South Carolina Department of Disabilities and Special
Needs (SCDDSN).
Author details
1
Undiagnosed Diseases Program, Common Fund, Office of the Director,
National Institutes of Health, Bethesda, MD 20814, USA.
2
Medical Genetics
Branch, National Human Genome Research Institute, Bethesda, MD, USA.
3
Skeletal Clinical Studies Unit, Craniofacial and Skeletal Disease Branch,
National Institute of Dental and Craniofacial Research, National Institutes of
Health, Bethesda, MD 20892, USA.
4
J.C. Self Research Institute, Greenwood
Genetics Centre, Greenwood, SC 29646, USA.
5
George Washington University
School of Medicine, Washington, DC, USA.
6
Childrens National Medical
Center, Washington, DC, USA.
Received: 9 July 2014 Accepted: 28 January 2015
References
1. D'Agostino L, Di Luccia A. Polyamines interact with DNA as molecular
aggregates. Eur J Biochem. 2002;269(17):431725.
2. Watanabe S, Ksama-Equchi K, Kobayashi H, Igarashi K. Estimation of polyamine
binding to macromolecules and ATP in bovine lymphocytes and rat liver. J Biol
Chem. 1991;266(31):208039.
3. Minois N, Carmona-Gutierrez D, Madeo F. Polyamines in aging and disease.
Aging. 2011;3(8):71632.
4. Raina A, Janne J. Physiology of the natural polyamines putrescine,
spermidine and spermine. Med Biol. 1975;53(3):12147.
5. Pegg AE, Michael AJ. Spermine synthase. Cell Mol Life Sci. 2010;67(1):11321.
6. Williams K. Interactions of polyamines with ion channels. Biochem J.
1997;325(Pt 2):28997.
7. Janne O, Bardin CW, Jacob ST. DNA-dependent RNA polymerases I and II
from kidney. Effect of polyamines on the in vitro transcription of DNA and
chromatin. Biochemistry. 1975;14(16):358997.
8. Friedman ME, Bachrach U. Inhibition of protein synthesis by spermine in
growing cells of Staphylococcus aureus. J Bacteriol. 1966;92(1):4955.
9. Nichols CG, Lopatin AN. Inward rectifier potassium channels. Annu Rev
Physiol. 1997;59:17191.
10. Snyder RD, Robinson A. Recessive sex-linked mental retardation in the
absence of other recognizable abnormalities. Report of a family. Clin Pediatr.
1969;8(11):66974.
11. Arena JF, Schwartz C, Ouzts L, Stevenson R, Miller M, Garza J, et al. X-linked
mental retardation with thin habitus, osteoporosis, and kyphoscoliosis:
linkage to Xp21.3-p22.12. Am J Med Genet. 1996;64(1):508.
12. Cason AL, Ikequchi Y, Skinner C, Wood TC, Holder KR, Lubs JA, et al. X-linked
spermine synthase gene (SMS) defect: the first polyamine deficiency
syndrome. Eur J Hum Genet. 2003;11(12):93744.
13. de Alencastro G, McCloskey DE, Kliemann SE, Maranduba CM, Pegg AE,
Wang X, et al. New SMS mutation leads to a striking reduction in spermine
synthase protein function and a severe form of Snyder-Robinson X-linked
recessive mental retardation syndrome. J Med Genet. 2008;45(8):53943.
14. Becerra-Solano LE, Butler J, Castaneda-Cisneros G, McCloskey DE, Wang X,
Pegg AE, et al. A missense mutation, p.V132G, in the X-linked spermine
synthase gene (SMS) causes Snyder-Robinson syndrome. Am J Med Genet.
2009;149A(3):32835.
15. Schwartz CE, Wang X, Stevenson RE, Pegg AE. Spermine Synthase
Deficiency Resulting in X-Linked Intellectual Disability (SnyderRobinson
Syndrome). In: Pegg AE, Castero Jr RA, editors. Polyamines: Methods and
Protocols. New York: Humana Press; 2011. p. 43745.
16. Peron A, Spaccini L, Norris J, Bova SM, Selicorni A, Weber G, et al.
Snyder-Robinson syndrome: a novel nonsense mutation in spermine
synthase and expansion of the phenotype. Am J Med Genet A.
2013;161(9):231620.
17. Zhang Z, Norris J, Kalscheuer V, Wood T, Wang L, Schwartz C, et al. A Y328C
missense mutation in spermine synthase causes a mild form of Snyder-Robinson
syndrome. Hum Mol Genet. 2013;22(18):378997.
18. Raisz LG. Pathogenesis of osteoporosis: concepts, conflicts, and prospects.
J Clin Invest. 2005;115(12):331825.
19. Guntur AR, Rosen CJ. Bone as an endocrine organ. Endocr Pract.
2012;18(5):75862.
20. Albert J, Schwartz CE, Boerkoel CF, Stevenson RE. Snyder-Robinson Syndrome.
In: Pagon RA, Adam MP, Ardinger HH, Bird TD, Dolan CR, Fong CT, et al.,
editors. Seattle (WA): GeneReviews(R); 1993.
21. Zemel BS, Leonard MB, Kelly A, Lappe JM, Gilsanz V, Oberfield S, et al.
Height adjustment in assessing dual energy x-ray absorptiometry
measurements of bone mass and density in children. J Clin Endocrinol
Metab. 2010;95(3):126573.
22. Wang K, Li M, Hadley D, Liu R, Glessner J, Grant SF, et al. PennCNV: an
integrated hidden Markov model designed for high-resolution copy
number variation detection in whole-genome SNP genotyping data.
Genome Res. 2007;17(11):166574.
23. Manoli I, Golas G, Westbroek W, Vilboux T, Markello TC, Introne W, et al.
Chediak-Higashi syndrome with early developmental delay resulting from
paternal heterodisomy of chromosome 1. Am J Med Genet A.
2010;152A(6):147483.
24. Markello TC, Adams DR, Genome-scale sequencing to identify genes
involved in Mendelian disorders. Current protocols in human genetics /
editorial board, Jonathan L. Haines[et al], 2013. 79: p. Unit 6 13.
25. Adams DR, Sincan M, Fuentes Fajardo K, Mullikin JC, Pierson TM, et al.
Analysis of DNA sequence variants detected by highthroughput
sequencing. Hum Mutat. 2012;33(4):599608.
26. Markello TC, Han T, Carlson-Donohoe H, Ahaghotu C, Harper U, Jones M,
et al. Recombination mapping using Boolean logic and highdensity SNP
genotyping for exome sequence filtering. Mol Genet Metab.
2012;105(3):3829.
27. Adzhubei IA, Schmidt S, Peshkin L, Ramensky VE, Gerasimova A, Bork P,
et al. A method and server for predicting damaging missense mutations.
Nat Methods. 2010;7(4):2489.
28. Johnston JJ, Teer JK, Cherukuri PF, Hansen NF, Loftus SK, NIH Intramural
Sequencing Center (NISC), et al. Massively parallel sequencing of exons on
the X chromosome identifies RBM10 as the gene that causes a syndromic
form of cleft palate. Am J Hum Genet. 2010;86(5):7438.
Albert et al. Orphanet Journal of Rare Diseases (2015) 10:27 Page 12 of 13
29. Kumar P, Henikoff S, Ng PC. Predicting the effects of coding
nonsynonymous variants on protein function using the SIFT algorithm. Nat
Protoc. 2009;4(7):107381.
30. Ng PC, Henikoff S. Predicting the effects of amino acid substitutions on
protein function. Annu Rev Genomics Hum Genet. 2006;7:6180.
31. Ng PC, Henikoff S. SIFT: Predicting amino acid changes that affect protein
function. Nucleic Acids Res. 2003;31(13):38124.
32. Ng PC, Henikoff S. Accounting for human polymorphisms predicted to
affect protein function. Genome Res. 2002;12(3):43646.
33. Ng PC, Henikoff S. Predicting deleterious amino acid substitutions. Genome
Res. 2001;11(5):86374.
34. Henikoff S, Henikoff JG. Amino acid substitution matrices from protein
blocks. Proc Natl Acad Sci U S A. 1992;89(22):109159.
35. McCarthy EF, Earnest K, Rossiter K, Shapiro J. Bone histomorphometry in
adults with type IA osteogenesis imperfecta. Clin Orthop Relat Res.
1997;336:25462.
36. Sowell J, Norris J, Jones K, Schwartz C, Wood T. Diagnostic screening for
spermine synthase deficiency by liquid chromatography tandem mass
spectrometry. Clin Chim Acta. 2011;412(7-8):65560.
37. Bhattacharyya N, Wiench M, Dumitrescu C, Connolly BM, Bugge TH, Patel
HV, et al. Mechanism of FGF23 processing in fibrous dysplasia. J Bone Miner
Res Off J Am Soc Bone Miner Res. 2012;27(5):113241.
38. Kodiha M, Umar R, Stochaj U. Optimized immunofluorescence staining
protocol to detect the nucleoporin Nup98 in different subcellular
compartments. Protocol Exchange, 2009.
39. Wu H, Min J, Zeng H, McCloskey DE, Ikeguchi Y, Loppnau P, et al. Crystal
structure of human spermine synthase: implications of substrate binding
and catalytic mechanism. J Biol Chem. 2008;283(23):1613546.
40. Schrodinger, LLC. The PyMOL Molecular Graphics System, Version 1.5.0.5, 2010.
41. Piton A, Gauthier J, Hamdan FF, Lafreniere RG, Yang Y, Henrion E, et al.
Systematic resequencing of X-chromosome synaptic genes in autism
spectrum disorder and schizophrenia. Mol Psychiatry. 2011;16(8):86780.
42. Lee HW, Choi J, Shin H, Kim K, Yang J, Na M, et al. Preso, a novel
PSD-95-interacting FERM and PDZ domain protein that regulates
dendritic spine morphogenesis. J Neurosci Off J Soc Neurosci.
2008;28(53):1454656.
43. Nagao M, Feinstein TN, Ezura Y, Hayata T, Notomi T, Saita Y, et al.
Sympathetic control of bone mass regulated by osteopontin. Proc Natl
Acad Sci U S A. 2011;108(43):1776772.
44. Kajimura D, Hinoi E, Ferron M, Kode A, Riley KJ, Zhou B, et al. Genetic
determination of the cellular basis of the sympathetic regulation of bone
mass accrual. J Exp Med. 2011;208(4):84151.
45. Takeda S, Karsenty G. Molecular bases of the sympathetic regulation of
bone mass. Bone. 2008;42(5):83740.
46. Takeda S, Elefteriou F, Levasseur R, Liu X, Zhao L, Parker KL, et al. Leptin
regulates bone formation via the sympathetic nervous system. Cell.
2002;111(3):30517.
47. Elefteriou F, Ahn JD, Takeda S, Starbuck M, Yang X, Liu X, et al. Leptin
regulation of bone resorption by the sympathetic nervous system and
CART. Nature. 2005;434(7032):51420.
48. Kondo H, Nifuji A, Takeda S, Ezura Y, Rittling SR, Denhardt DT, et al.
Unloading induces osteoblastic cell suppression and osteoclastic cell
activation to lead to bone loss via sympathetic nervous system. J Biol
Chem. 2005;280(34):30192200.
49. Fu L, Patel MS, Bradley A, Wagner EF, Karsenty G. The molecular clock
mediates leptin-regulated bone formation. Cell. 2005;122(5):80315.
50. Jackson L, Jones DR, Scotting P, Sottile V. Adult mesenchymal stem cells:
differentiation potential and therapeutic applications. J Postgrad Med.
2007;53(2):1217.
51. Sottile V, Halleux C, Bassilana F, Keller H, Seuwen K. Stem cell characteristics
of human trabecular bone-derived cells. Bone. 2002;30(5):699704.
52. Ikeguchi Y, Bewley MC, Pegg AE. Aminopropyltransferases: function,
structure and genetics. J Biochem. 2006;139(1):19.
53. Williams-Ashman HG, Seidenfeld J, Galletti P. Trends in the biochemical
pharmacology of 5'-deoxy-5'-methylthioadenosine. Biochem Pharmacol.
1982;31(3):27788.
54. Raina A, Tuomi K, Pajula RL. Inhibition of the synthesis of polyamines and
macromolecules by 5'-methylthioadenosine and 5'-alkylthiotubercidins in
BHK21 cells. Biochem J. 1982;204(3):697703.
55. Dupree K, Dobs A. Osteopenia and male hypogonadism. Rev Urol.
2004;6 Suppl 6:S304.
56. Ross AC. The 2011 report on dietary reference intakes for calcium and
vitamin D. Public Health Nutr. 2011;14(5):9389.
57. Lee RH, Lyles KW, Colon-Emeric C. A review of the effect of anticonvulsant
medications on bone mineral density and fracture risk. Am J Geriatr
Pharmacother. 2010;8(1):3446.
Submit your next manuscript to BioMed Central
and take full advantage of:
Convenient online submission
Thorough peer review
No space constraints or color figure charges
Immediate publication on acceptance
Inclusion in PubMed, CAS, Scopus and Google Scholar
Research which is freely available for redistribution
Submit your manuscript at
www.biomedcentral.com/submit
Albert et al. Orphanet Journal of Rare Diseases (2015) 10:27 Page 13 of 13
... Various groups have reported on the effect of polyamines on osteogenic differentiation of MSCs in culture, with widely discordant results [6][7][8][9][10] that may be attributed to the low number of biological replicates, tissue of origin of MSCs (adipose vs. bone marrow), species, outcome measurements, and concentrations of both polyamines and inhibitors tested. Work by Albert et al. has suggested that mesenchymal stem cells/multipotent stromal cells (MSCs) derived from SRS patients show impaired osteogenic differentiation in vitro [11]. To model the disease in a dish, we have previously silenced Sms expression using shRNA in MSCs derived from healthy donors and confirmed that SMS deficiency impairs osteogenesis, both in vitro and after implantation into immune-deficient mice [12]. ...
... To modify intracellular polyamine levels, we chemically inhibited SMS and ODC1 using CDAP and DFMO, respectively ( Figure 1). CDAP is expected to lead to increased spermidine and reduced spermine, as previously described in SRS patients [11] and when using a shRNA targeting SMS [12]. Inhibition of ODC1 is expected to cause a reduction of putrescine, spermidine, and spermine [9,15]. ...
Article
Full-text available
Despite the well-known relevance of polyamines to many forms of life, little is known about how polyamines regulate osteogenesis and skeletal homeostasis. Here, we report a series of in vitro studies conducted with human-bone-marrow-derived pluripotent stromal cells (MSCs). First, we show that during osteogenic differentiation, mRNA levels of most polyamine-associated enzymes are relatively constant, except for the catabolic enzyme spermidine/spermine N1-acetyltransferase 1 (SAT1), which is strongly increased at both mRNA and protein levels. As a result, the intracellular spermidine to spermine ratio is significantly reduced during the early stages of osteoblastogenesis. Supplementation of cells with exogenous spermidine or spermine decreases matrix mineralization in a dose-dependent manner. Employing N-cyclohexyl-1,3-propanediamine (CDAP) to chemically inhibit spermine synthase (SMS), the enzyme catalyzing conversion of spermidine into spermine, also suppresses mineralization. Intriguingly, this reduced mineralization is rescued with DFMO, an inhibitor of the upstream polyamine enzyme ornithine decarboxylase (ODC1). Similarly, high concentrations of CDAP cause cytoplasmic vacuolization and alter mitochondrial function, which are also reversible with the addition of DFMO. Altogether, these studies suggest that excess polyamines, especially spermidine, negatively affect hydroxyapatite synthesis of primary MSCs, whereas inhibition of polyamine synthesis with DFMO rescues most, but not all of these defects. These findings are relevant for patients with Snyder–Robinson syndrome (SRS), as the presenting skeletal defects—associated with SMS deficiency—could potentially be ameliorated by treatment with DFMO.
... SPM was also found to promote osteogenic differentiation in goat adipose tissue-derived mesenchymal stem cells (ADSCs) (Tjabringa et al. 2006(Tjabringa et al. , 2008. Patients with Snyder-Robinson Syndrome (SRS), an X-linked intellectual disability disorder caused by a mutation in spermine synthase, suffer from osteoporosis in which osteoblasts and osteoclasts are severely depleted (Albert et al. 2015). Recently, modulation of gut microbiota was reported to regulate bone remodeling, and treatment with probiotics has become a new approach to osteoporosis therapy (Lyu et al. 2023). ...
Article
Full-text available
Exogenous polyamines, including putrescine (PUT), spermidine (SPD), and spermine (SPM), and the irreversible inhibitor of the rate-limiting enzyme ornithine decarboxylase (ODC) of polyamine biosynthesis, α-difluoromethylornithine (DFMO), are implicated as stimulants for bone formation. We demonstrate in this study the osteogenic potential of exogenous polyamines and DFMO in human osteoblasts (hOBs), murine monocyte cell line RAW 264.7, and an ovariectomized rat model. The effect of polyamines and DFMO on hOBs and RAW 264.7 cells was studied by analyzing gene expression, alkaline phosphatase (ALP) activity, tartrate-resistant acid phosphatase (TRAP) activity, and matrix mineralization. Ovariectomized rats were treated with polyamines and DFMO and analyzed by micro computed tomography (micro CT). The mRNA level of the early onset genes of osteogenic differentiation, Runt-related transcription factor 2 (Runx2) and ALP, was significantly elevated in hOBs under osteogenic conditions, while both ALP activity and matrix mineralization were enhanced by exogenous polyamines and DFMO. Under osteoclastogenic conditions, the gene expression of both receptor activator of nuclear factor-κB (RANK) and nuclear factor of activated T-cells, cytoplasmic 1 (NFATc1) was reduced, and TRAP activity was suppressed by exogenous polyamines and DFMO in RAW 264.7 cells. In an osteoporotic animal model of ovariectomized rats, SPM and DFMO were found to improve bone volume in rat femurs, while trabecular thickness was increased in all treatment groups. Results from this study provide in vitro and in vivo evidence indicating that polyamines and DFMO act as stimulants for bone formation, and their osteogenic effect may be associated with the suppression of osteoclastogenesis.
... p.Q148R and p.I150T are located in the central domain. p.Q148R [34] alters the 5′-methylthioadenosine (MTA)binding site, whereas p.I150T [35] induces structural changes in the vicinity of the MTA-binding site and reduces the stability of the C-terminal domain. p.L277F, p.M303Kfs*3, and p.Y328C are located in the C-terminal domain. ...
Article
Full-text available
Polyamines are ubiquitous in almost all biological entities and involved in various crucial physiological processes. They are also closely associated with the onset and progression of many diseases. Polyaminopathies are a group of rare genetic disorders caused by alterations in the function of proteins within the polyamine metabolism network. Although the identified polyaminopathies are all rare diseases at present, they are genetically heritable, rendering high risks not only to the carriers but also to their descendants. Meanwhile, more polyaminopathic patients might be discovered with the increasing accessibility of gene sequencing. This review aims to provide a comprehensive overview of the structural variations of mutated proteins in current polyaminopathies, in addition to their causative genes, types of mutations, clinical symptoms, and therapeutic approaches. We focus on analyzing how alterations in protein structure lead to protein dysfunction, thereby facilitating the onset of diseases. We hope this review will offer valuable insights and references for the future clinical diagnosis and precision treatment of polyaminopathies.
... Deficiency of spermine synthase causes excessive spermidine accumulation and a lack of spermine. 141 Patients with Snyder-Robinson syndrome, a syndrome caused by loss-of-function mutations of the spermine synthase gene, exhibit severe OP and kyphoscoliosis and have BMSCs with impaired capacities for osteogenic differentiation and mineralization. 142 Hydrogen sulfide. ...
Article
Full-text available
The gut microbiota (GM) plays a crucial role in maintaining the overall health and well-being of the host. Recent studies have demonstrated that the GM may significantly influence bone metabolism and degenerative skeletal diseases, such as osteoporosis (OP). Interventions targeting GM modification, including probiotics or antibiotics, have been found to affect bone remodeling. This review provides a comprehensive summary of recent research on the role of GM in regulating bone remodeling and seeks to elucidate the regulatory mechanism from various perspectives, such as the interaction with the immune system, interplay with estrogen or parathyroid hormone (PTH), the impact of GM metabolites, and the effect of extracellular vesicles (EVs). Moreover, this review explores the potential of probiotics as a therapeutic approach for OP. The insights presented may contribute to the development of innovative GM-targeted therapies for OP.
... dietary SPM supplementation (5,6,(10)(11)(12)(13)(14)(15)(16). The degree to which this ratio is altered in individual patients depends on the specific SMS mutation and appears to correlate with SMS enzymatic activity as well as phenotype severity, though existing data are insufficient to conclude a genotype-phenotype correlation (14). ...
Preprint
Snyder-Robinson Syndrome (SRS) is caused by mutations in the spermine synthase ( SMS ) gene, the enzyme product of which converts the polyamine spermidine into spermine. Affecting primarily males, common manifestations of SRS include intellectual disability, osteoporosis, hypotonic musculature, and seizures, along with other more variable symptoms. Currently, medical management focuses on treating these symptoms without addressing the underlying molecular cause of the disease. Reduced SMS catalytic activity in cells of SRS patients causes the accumulation of spermidine, while spermine levels are reduced. The resulting exaggeration in spermidine-to-spermine ratio is a biochemical hallmark of SRS that tends to correlate with symptom severity in the patient. Our studies aim to pharmacologically manipulate polyamine metabolism to correct this polyamine imbalance and investigate the potential of this approach as a therapeutic strategy for affected individuals. Here we report the use of difluoromethylornithine (DFMO; eflornithine), an FDA-approved inhibitor of polyamine biosynthesis, in re-establishing normal spermidine-to-spermine ratios in SRS patient cells. Through mechanistic studies, we demonstrate that, while reducing spermidine biosynthesis, DFMO also stimulates the conversion of existing spermidine into spermine in cell lines with hypomorphic variants of SMS . Further, DFMO treatment induces a compensatory uptake of exogenous polyamines, including spermine and spermine mimetics, cooperatively reducing spermidine and increasing spermine levels. In a Drosophila SRS model characterized by reduced lifespan, adding DFMO to the feed extended lifespan. As nearly all known SRS patient mutations are hypomorphic, these studies form a foundation for future translational studies with significant therapeutic potential.
... Polyamine biosynthesis is upregulated during placentation 6 , and knockout or pharmacological inhibition of ODC in rodents results in embryo lethality 7 and abnormal placental development 8 . Moreover, human infants with loss-of-function mutations in spermine synthase (SMS), a condition known as Snyder Robinson Syndrome (SRS), are born small-for-gestational age (SGA) 9,10 . Despite these profound phenotypes, it is still unclear how impairments in polyamine metabolism cause these effects. ...
Article
Full-text available
Placental function and dysfunction differ by sex but the mechanisms are unknown. Here we show that sex differences in polyamine metabolism are associated with escape from X chromosome inactivation of the gene encoding spermine synthase (SMS). Female placental trophoblasts demonstrate biallelic SMS expression, associated with increased SMS mRNA and enzyme activity. Polyamine depletion in primary trophoblasts reduced glycolysis and oxidative phosphorylation resulting in decreased acetyl-coA availability and global histone hypoacetylation in a sex-dependent manner. Chromatin-immunoprecipitation sequencing and RNA-sequencing identifies progesterone biosynthesis as a target of polyamine regulated gene expression, and polyamine depletion reduced progesterone release in male trophoblasts. The effects of polyamine depletion can be attributed to spermine as SMS-silencing recapitulated the effects on energy metabolism, histone acetylation, and progesterone release. In summary, spermine metabolism alters trophoblast gene expression through acetyl-coA biosynthesis and histone acetylation, and SMS escape from X inactivation explains some features of human placental sex differences.
... Daily administration of spermine to rats ameliorated bone and cartilage destruction due to collagen-induced arthritis . Albert et al. (2015) reported that specific alterations in polyamines metabolism led to Snyder-Robinson Syndrome (SRS), a disease characterized by profound depletion of osteoclasts and osteoblasts. Recently, it has also been reported that warmth exposure prevents osteoporosis in mice by inducing the production of polyamines. ...
Article
Full-text available
A growing body of scientific evidence supports the notion that gut microbiota plays a key role in the regulation of various physiological and pathological processes related to human health. Recent findings have now established that gut microbiota also contributes to the regulation of bone homeostasis. Studies on animal models have unraveled various underlying mechanisms responsible for gut microbiota-mediated bone regulation. Normal gut microbiota is thus required for the maintenance of bone homeostasis. However, dysbiosis of gut microbiota communities is reported to be associated with several bone-related ailments such as osteoporosis, rheumatoid arthritis, osteoarthritis, and periodontitis. Dietary interventions in the form of probiotics, prebiotics, synbiotics, and postbiotics have been reported in restoring the dysbiotic gut microbiota composition and thus could provide various health benefits to the host including bone health. These dietary interventions prevent bone loss through several mechanisms and thus could act as potential therapies for the treatment of bone pathologies. In the present review, we summarize the current knowledge of how gut microbiota and its derived microbial compounds are associated with bone metabolism and their roles in ameliorating bone health. In addition to this, we also highlight the role of various dietary supplements like probiotics, prebiotics, synbiotics, and postbiotics as promising microbiota targeted interventions with the clinical application for leveraging treatment modalities in various inflammatory bone pathologies.
Article
Snyder–Robinson syndrome (SRS) results from mutations in spermine synthase (SMS), which converts the polyamine spermidine into spermine. Affecting primarily males, common manifestations of SRS include intellectual disability, osteoporosis, hypotonia, and seizures. Symptom management is the only treatment. Reduced SMS activity causes spermidine accumulation while spermine levels are reduced. The resulting exaggerated spermidine:spermine ratio is a biochemical hallmark of SRS that tends to correlate with symptom severity. Our studies aim to pharmacologically manipulate polyamine metabolism to correct this imbalance as a therapeutic strategy for SRS. Here we report the repurposing of 2‐difluoromethylornithine (DFMO), an FDA‐approved inhibitor of polyamine biosynthesis, in rebalancing spermidine:spermine ratios in SRS patient cells. Mechanistic in vitro studies demonstrate that, while reducing spermidine biosynthesis, DFMO also stimulates the conversion of spermidine into spermine in hypomorphic SMS cells and induces uptake of exogenous spermine, altogether reducing the aberrant ratios. In a Drosophila SRS model characterized by reduced lifespan, DFMO improves longevity. As nearly all SRS patient mutations are hypomorphic, these studies form a strong foundation for translational studies with significant therapeutic potential.
Technical Report
Full-text available
Snyder-Robinson syndrome is an X-linked condition that causes intellectual impairment. (sometimes referred to as spermine synthase deficiency) This condition is characterized by asthenic body habitus, facial dimorphisms, a wide-based gait, osteoporosis, and recurrent fractures. Here, we present the findings of a pediatric autopsy on a 4-year-old boy who had been previously identified as having Snyder-Robinson syndrome as a result of an SMS gene mutation and had a history of seizures, repeated fractures, and aberrant gait Hypoxic-ischemic encephalopathy brought on by persistent seizure activity was the cause of death. Present complaint and investigation: The patient, a 50-year-old male with modest mental retardation with adaptive skills, gait problems necessitating the use of an On December 9, 2021, a patient with numerous fractures, treatment-resistant myoclonic epilepsy, and a walker or holding hands was brought to the Hospital. He showed no evidence of being able to speak with numerous words and some signals despite behavioral concerns. To identify the diagnosis, a blood investigation is used. analysis of the enzyme synthase. reduced or nonexistent SMS enzyme activity in cultured lymphoblast or fresh white cells testing for molecular genetics discovery of a harmful homozygous loss-of-function mutant. x-ray Bilateral peripheral infiltrates on a chest x-ray were alarming for a viral infection. a cerebral spinal fluid and blood culture-based sepsis workup analgesia was initiated because of the changed mental status. As the developmental delay worsens, many boys with SRS experience some motor impairment. Main diagnosis and therapeutic interventions and outcome: The physician decide that the patient has Snyder Robinson syndrome after performing a physical examination and a general evaluation. An evaluation for sepsis using cerebral spinal fluid and blood cultures was prompted by the patient's changed mental status. Bilateral peripheral infiltrates on the chest x-ray were alarming for a viral infection. Conclusion: Our results are consistent with the theories that poor bone density results from a failure to mineralize, and that tissue-specific changes in the tissue specificity of SRS features are influenced by metabolism. Anew SMS mutation is the cause of Snyder-Robinson disease. A new SMS mutation results in Snyder-Robinson syndrome. Our patient's Additional indications that the digestive system is important to include severe failure to thrive, enteral feeding intolerance, hepatic fibrosis, pancreatic exocrine insufficiency, and jejunal stenosis.
Article
Full-text available
Snyder-Robinson Syndrome (SRS, OMIM: 309583) is an X-linked intellectual disability syndrome, characterized by a collection of clinical features including facial asymmetry, marfanoid habitus, hypertonia, osteoporosis and unsteady gait. It is caused by a significant decrease or loss of spermine synthase (SMS) activity. Here we report a new missense mutation, p.Y328C, (c.1084A>G), in SMS in a family with X-linked intellectual disability. The affected males available for evaluation had mild ID, speech and global delay, an asthenic build, short stature with long fingers and mild kyphosis. The spermine/spermidine ratio in lymphoblasts was 0.53, significantly reduced compared to normal (1.87 average). Activity analysis of SMS in the index patient failed to detect any activity above background. In silico modeling demonstrated that the Y328C mutation has a significant effect on SMS stability, resulting in decreased folding free energy and larger structural fluctuations compared with those of wild type SMS. The loss of activity was attributed to the increase of conformational dynamics in the mutant which affects the active site geometry, rather than preventing dimer formation. Taken together, the biochemical and in silico studies confirm the p.Y328C mutation in SMS is responsible for the patients having a mild form of SRS and reveal yet another molecular mechanism resulting in a non-functional SMS causing SRS.
Article
The analysis of genome-scale sequence data can be defined as the interrogation of a complete set of genetic instructions in a search for individual loci that produce or contribute to a pathological state. Bioinformatic analysis of sequence data requires sufficient discriminant power to find this needle in a haystack. Current approaches make choices about selectivity and specificity thresholds, and the quality, quantity, and completeness of the data in these analyses. There are many software tools available for individual, analytic component-tasks, including commercial and open-source options. Three major types of techniques have been included in most published exome projects to date: frequency/population genetic analysis, inheritance state consistency, and predictions of deleteriousness. The required infrastructure and use of each technique during analysis of genomic sequence data for clinical and research applications are discussed. Future developments will alter the strategies and sequence of using these tools and are also discussed. Curr. Protoc. Hum. Genet. 79:6.13.1-6.13.19. © 2013 by John Wiley & Sons, Inc.
Article
Snyder-Robinson syndrome is a rare form of X-linked intellectual disability caused by mutations in the spermine synthase (SMS) gene, and characterized by intellectual disability, thin habitus with diminished muscle mass, osteoporosis, kyphoscoliosis, facial dysmorphism (asymmetry, full lower lip), long great toes, and nasal or dysarthric speech. Physical signs seem to evolve from childhood to adulthood. We describe the first Italian patient with Snyder-Robinson syndrome and a novel nonsense mutation in SMS (c.200G>A; p.G67X). Apart from the typical features of the syndrome, the index patient presented with an ectopic right kidney and epilepsy from the first year of age that was characterized by focal motor seizures and negative myoclonus. The clinical and molecular evaluation of this family and the review of the literature expand the phenotype of Snyder-Robinson syndrome to include myoclonic or myoclonic-like seizures (starting even in the first years of life) and renal abnormalities in affected males. © 2013 Wiley Periodicals, Inc.
Chapter
Snyder-Robinson syndrome (SRS) is an X-linked intellectual disability syndrome characterized by asthenic build, facial dysmorphism with a prominent lower lip, kyphoscoliosis, osteoporosis, and speech abnormalities. Developmental delay usually presents as failure to meet early developmental milestones and then evolves to moderate to profound global intellectual disability (which appears to remain stable over time) and variable motor disability. Asthenic habitus and low muscle mass usually develop during the first year, even in males who are ambulatory. During the first decade, males with SRS develop osteoporosis, resulting in fractures in the absence of trauma. The diagnosis is suspected in males with characteristic clinical findings and is confirmed with molecular genetic testing when a hemizygous loss-of-function SMS mutation is identified. Treatment of manifestations: Speech, physical, and/or occupational therapy may be helpful. Seizures have shown varying responses to anti-seizure medications; carbamazepine, phenobarbital, and clobazan have been used successfully in some individuals. Calcium supplementation has slightly improved bone mineral density in a few patients. Surveillance: Because of the osteoporosis, individuals with SRS should be investigated for factures if medically indicated. While receiving calcium supplementation, patients should be monitored regularly for ectopic calcification. SRS is inherited in an X-linked manner. If the mother of the proband has a disease-causing mutation, the chance of transmitting it in each pregnancy is 50%: Males who inherit the mutation will be affected; females who inherit the mutation will be carriers. No known carriers have had features of SRS. No affected male has reproduced. Carrier testing for at-risk relatives and prenatal testing for pregnancies at increased risk are an option if the disease-causing SMS mutation has been identified in an affected family member.
Article
The past three years have seen remarkable progress in research on the molecular basis of inward rectification, with significant implications for basic understanding and pharmacological manipulation of cellular excitability. Expression cloning of the first inward rectifier K channel (Kir) genes provided the necessary break-through that has led to isolation of a family of related clones encoding channels with the essential functional properties of classical inward rectifiers, ATP-sensitive K channels, and muscarinic receptor-activated K channels. High-level expression of cloned channels led to the discovery that classical inward so-called anomalous rectification is caused by voltage-dependent block of the channel by polyamines and Mg2+ ions, and it is now clear that a similar mechanism results in inward rectification of alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionate (AMPA)-kainate receptor channels. Knowledge of the primary structures of Kir channels and the ability to mutate them also has led to the determination of many of the structural requirements of inward rectification.
Article
We reevaluated a family previously described as having nonspecific X-linked mental retardation (XLMR) by Snyder and Robinson [1969: Clin Pediatr 8:669–674] (MIM 309583). Clinical and DNA studies were conducted on 17 relatives, including 6 males with mild-to-moderate mental retardation, 3 carrier females, and 8 normal males. In contrast to the normal appearance and minimal clinical findings reported 22 years ago, affected males were found to have a characteristic set of clinical findings. These developed gradually over the first 2 decades, and included thin body build with diminished muscle mass, osteoporosis and kyphoscoliosis, slight facial asymmetry with a prominent lower lip, nasal speech, high narrow or cleft palate, and long great toes. Carrier females were clinically normal. Multipoint linkage analysis indicated linkage to markers distal to the 3′ end of DMD (DXS41 and DXS989), with a maximal lod score of 4.7. On the basis of these findings, this entity is redefined as XLMR syndrome. © 1996 Wiley-Liss, Inc.