Sie sind auf Seite 1von 32

Accepted Manuscript

Parathyroid hormone resistance syndromes – Inactivating PTH/PTHrP Signaling


Disorders (iPPSDs)

Francesca Marta Elli, MSc, PhD, Arrate Pereda, MSc, PhD, Agnès Linglart, MD, PhD,
Guiomar Perez de Nanclares, MSc, PhD, Giovanna Mantovani, MD, PhD

PII: S1521-690X(18)30108-8
DOI: 10.1016/j.beem.2018.09.008
Reference: YBEEM 1242

To appear in: Best Practice & Research Clinical Endocrinology & Metabolism

Please cite this article as: Elli FM, Pereda A, Linglart A, de Nanclares GP, Mantovani G, Parathyroid
hormone resistance syndromes – Inactivating PTH/PTHrP Signaling Disorders (iPPSDs), Best
Practice & Research Clinical Endocrinology & Metabolism (2018), doi: https://doi.org/10.1016/
j.beem.2018.09.008.

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to
our customers we are providing this early version of the manuscript. The manuscript will undergo
copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please
note that during the production process errors may be discovered which could affect the content, and all
legal disclaimers that apply to the journal pertain.
ACCEPTED MANUSCRIPT
Best Practice and Research Clinical Endocrinology and Metabolism

Disorders of the Parathyroid Gland

Parathyroid hormone resistance syndromes – Inactivating PTH/PTHrP Signaling Disorders (iPPSDs)

PT
Francesca Marta Elli, MSc, PhD

RI
Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Endocrinology Unit; Department of Clinical

Sciences and Community Health, University of Milan, Milan, Italy

SC
email francesca.elli@unimi.it ; phone +39 02 55033512 ; fax +39 02 55033361

U
Arrate Pereda, MSc, PhD (co-first author) AN
Molecular (Epi)Genetics Laboratory, BioAraba National Health Institute, OSI Araba University Hospital,

Vitoria-Gasteiz, Spain.
M

email arrate.peredaaguirre@osakidetza.eus; phone +34945007097 ; fax +34945007118


D

Agnès Linglart, MD, PhD


TE

APHP, Department of paediatric endocrinology and diabetes for children, Bicêtre Paris-Sud Hospital, Le

Kremlin-Bicêtre, France,
EP

APHP, Reference center for rare disorders of calcium and phosphate metabolism, filière OSCAR and
C

Plateforme d'Expertise Maladies Rares Paris-Sud, Bicêtre Paris-Sud Hospital, Le Kremlin Bicêtre, France,
AC

email agnes.linglart@aphp.fr

Tel : +33145217853 ; fax : +33145217850

Guiomar Perez de Nanclares, MSc, PhD (co-corresponding author)

Molecular (Epi)Genetics Laboratory, BioAraba National Health Institute, OSI Araba University Hospital,

Vitoria-Gasteiz, Spain.

email gnanclares@osakidetza.eus; phone +34945007097 ; fax +34945007118


ACCEPTED MANUSCRIPT
Giovanna Mantovani, MD, PhD (corresponding author)

Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Endocrinology Unit; Department of Clinical

Sciences and Community Health, University of Milan, Milan, Italy

email giovanna.mantovani@unimi.it ; phone +39 02 50320607 ; fax +39 02 50320605

PT
RI
U SC
AN
M
D
TE
C EP
AC
ACCEPTED MANUSCRIPT
Abstract (max 150 words)

Metabolic disorders caused by impairments of the Gsα/cAMP/PKA pathway affecting the signaling of

PTH/PTHrP lead to features caused by non-responsiveness of target organs, in turn leading to manifestations

similar to the deficiency of the hormone itself.

Pseudohypoparathyroidism (PHP) and related disorders derive from a defect of the α subunit of the

PT
stimulatory G protein (Gsα) or of downstream effectors of the same pathway, such as the PKA regulatory

RI
subunit 1A and the phosphodyestarase type 4D.

The increasing knowledge on these diseases made the actual classification of PHP outdated as it does not

SC
include related conditions such as acrodysostosis (ACRDYS) or progressive osseous heteroplasia (POH), so

that a new nomenclature and classification has been recently proposed grouping these disorders under the

U
term “inactivating PTH/PTHrP signaling disorder” (iPPSD).
AN
This review will focus on the pathophysiology, clinical and molecular aspects of these rare, heterogeneous
M

but closely related diseases.

Key words:
D
TE

Hormone resistance

Pseudohypoparathyroidism (PHP)
EP

Inactivating PTH/PTHrP signaling disorders (iPPSD)

GNAS
C

PRKAR1A
AC

PDE4D

PDE3A
ACCEPTED MANUSCRIPT
1. Introduction

The non-responsiveness of target organs to a specific hormone usually derives from molecular alterations

affecting hormone receptors, while pathologic features caused by multiple hormone resistance arise from the

lack of the activation of effectors downstream the receptor itself.

Parathyroid hormone (PTH) is produced and secreted by the parathyroid glands and is a key modulator of

PT
mineral homeostasis. Its deficiency leads to hypocalcemia as key clinical and biochemical characteristic.

RI
In clinical practice, the diagnosis of PTH resistance, usually referred to as Pseudohypoparathyroidism (PHP),

is defined by the co-existence of elevated serum levels of parathyroid hormone (PTH), low or normal

SC
calcemia, elevated phosphatemia, normal renal function, normal magnesium levels and in absence of vitamin

D deficiency. Besides cramps, seizures and neuromuscular consequences of hypocalcemia, the clinical

U
manifestations of PTH resistance are the intracranial calcifications of the basal ganglia and cataracts due to
AN
the combination of long-standing of chronic hypocalcemia and hyperphosphatemia. PTH resistance can be

definitively ascertained using recombinant PTH (1–34), the Ellsworth-Howard test or a subcutaneous
M

challenge (1).
D

Over the last few decades our understanding on both the molecular basis and the clinical manifestations of
TE

disorders whose hallmark is PTH resistance has deeply expanded. This review will summarize the current

knowledge on this group of rare metabolic disorders.


C EP

2. Pathophysiology of hormone resistance


AC

The serum calcium level regulates PTH secretion via a negative feedback through the parathyroid calcium

sensing receptor (CASR). Decreased calcium levels stimulate PTH release. Secreted PTH interacts with its

specific G-protein-coupled receptor (GPCR), the type 1 receptor (PTHR1), which is highly expressed in

bone and kidney and mediates the rapid increases in renal tubular reabsorption of calcium and decreased

phosphorus reabsorption. It also participates in long-term calciostatic functions by enhancing mobilization of

calcium and phosphorus from bone and increasing renal synthesis of 1,25-dihydroxy vitamin D, which, in
ACCEPTED MANUSCRIPT
turn, increases intestinal calcium absorption (2-4). The PTHR1 also mediates the paracrine actions of PTH-

related protein (PTHrP), which plays a particularly vital role in the process of endochondral bone formation.

GPCRs activate a superfamily of heterotrimeric guanine nucleotide binding proteins called G proteins, that

trigger the activation of downstream effectors (enzymes and ion channels) inducing both short-term effects

on hormone secretion, neurotransmission and muscle contraction and long term effects on gene transcription

PT
(5-9). The specificity of G proteins depends on the alpha subunit, which vary in range of expression and

selectivity of receptor-effector coupling. The alpha subunit of the stimulatory G protein (Gsα) is a key

RI
element of the cAMP-mediated signaling, which stimulates the cAMP formation and the subsequent protein

SC
kinase A (PKA) activation (Figure 1).

In human disease, the impairment of the Gsα/cAMP/PKA pathway mostly affects the signaling of

U
PTH/PTHrP, and, with different severity, the signaling of additional hormones, including TSH, GHRH,
AN
calcitonin and gonadotropins, so their resistance can be detected at different stages of life in the patients,

with different severity and time course (10-20).


M

The loss of Gsα signaling in the kidney, in particular in proximal tubular cells, is the crucial event leading to
D

PTH resistance. In most patients, PTH resistance develops during infancy or childhood. The most accepted
TE

hypothesis to explain this latency in PTH resistance development is the temporary physiological biallelic

expression of maternal and paternal Gsα alleles renal proximal tubules during early postnatal development
EP

and/or the rescue of the cAMP/PKA signaling XLαs, the extra-large splice variant of Gsα that is expresed in

early life (21-24).


C

It is proposed that the conserved anticalciuric action exerted at the level of the distal tubule allows most
AC

patients to preserve normo-hypocalciuria and life-long normal renal function without forming kidney stones

(25).

The defective generation of 1,25-dihydroxyvitamin D caused by the defective signaling of PTH, further

contributing to the decrease of serum calcium, is enhanced by untreated hyperphosphatemia (21-23).

In clinical practice, the diagnosis of TSH resistance is defined by the detection of elevated TSH levels, with

normal/slightly low free thyroxine levels, usually in the absence of goiter and antithyroid antibodies. It may
ACCEPTED MANUSCRIPT
develops over childhood or adolescence, although it is seldom detected during the neonatal screening for

congenital hypothyroidism (14-16).

Hypogonadism, delayed or incomplete sexual maturation, amenorrhea/oligomenorrhea and/or infertility in

women are manifestations of gonadotropins resistance (17). The GH deficiency derived from GHRH

resistance contributes to short stature (17-20).

PT
RI
After an extensive review of the literature and discussion about a novel classification/nomenclature to

describe disorders related to abnormalities of the PTH signaling pathway, a proposal to group these

SC
disorders, previously referred mainly to as Pseudohypoparathyroidism, under the term “inactivating

PTH/PTHrP signaling disorder” (iPPSD) has recently been published (Table 1) (26). We will use this

nomenclature through the text.


U
AN
M

3. Defects at the PTH/PTHrP receptor

3.1. PTHR1 and iPPSD1


D

The classical PTH/PTHrP receptor, otherwise known as PTHR1, binds intact PTH and biologically active
TE

amino-terminal fragments of PTH, such as PTH 1-34 (27). It recognizes both PTH and PTHrP due to the

substantial degree of homology in the amino-terminal parts of these two peptides. This receptor is heavily
EP

expressed in bone and kidney and is also present in additional tissues such as breast, skin, heart, blood
C

vessels, pancreas, and others that are not regarded as classical PTH target tissues (28).
AC

Mutations of the PTHR1 gene (MIM*168468), lying on the short arm of the chromosome 3 (3p21.31), are

associated with the iPPSD1 cluster (i.e., Blomstrand chondrodysplasia (BOCD, MIM#215045) and the Eiken

syndrome (MIM#600002)), with a recessive transmission pattern of inheritance.

BOCD is a lethal form of dwarfism (short limbs, polyhydramnios, hydrops fetalis, facial anomalies,

increased bone density and advanced skeletal maturation) and the Eiken syndrome is a skeletal dysplasia

(retarded bone maturation, principally of the epiphyses, pelvis, hands and feet) with slightly elevated levels

of PTH in some cases (29-31).


ACCEPTED MANUSCRIPT
BOCD type 1 is caused by the complete inactivation of PTHR1, leading to a most severe and often lethal

form, while the type 2, which derives from an incomplete inactivation of PTHR1, is associated with a

slightly less severe phenotype. So far only 5 pathogenetic genetic variants have been reported in exon 5

(c.310C>T), exon 6 (c.395C>T), intron 11 (c.1049+27G>A), exon 12 (c.1093del) and exon 13 (c.1148G>A).

Nowadays, only 1 variant in exon 16 (c.1453C>T, p.Arg485*) associated with the Eiken syndrome has been

PT
described (29-31).

All reported sequence are described following the Human Genome variation Society (HGVS)

RI
recommendations according with the reference transcript NM_000316.2 and are recorded in the publicly

SC
available Leiden Open Variation Database (LOVD) database at

https://databases.lovd.nl/shared/genes/PTH1R.

U
Heterozygous PTHR1 mutations have identified in patients with primary failure of tooth eruption (33-34).
AN
3.2. PTHR2
M

The PTH receptor 2 (PTHR2) selectively binds PTH but not PTHrP (28). It is heavily expressed in the

central nervous system, cardiovascular, and gastrointestinal systems, as well as in lung and testes, and may
D

be involved in the perception of pain (28).


TE

An intronic break of the PTHR2 gene was detected by whole genome sequencing in a 15-month-old boy with

sagital and metopic synostosis. The disruption of the PTHR2 gene may cause uncontrolled proliferation and
EP

differentiation of chondrocytes, which in turn results in premature closure of sutures (32). Experiments with

transgenic mice suggest a role for PTHR2 signaling in postnatal growth plate development and subsequent
C

bone mass acquisition as overexpression of human PTHR2 in chondrocytes of these mice has key
AC

consequences on postnatal development of endochondral skeleton (35).

4. Defects at the Gsα/cAMP/PKA pathway

Molecular defects affecting different members the PTH/PTHrP pathway are associated with a group of

heterogeneous but related disorders that represent a challenge for differential diagnosis and genetic

counseling. As the defects are affecting the pathway, not only PTH resistance, but also resistance to other
ACCEPTED MANUSCRIPT
hormones (TSH, gonadotropins, GHRH and probably others) that mediate their action through GPCR

coupled to Gsα/cAMP/PKA is observed.

In particular, genetic defects at the gene coding the Gsα determine a clinical condition classified as iPPSD2

(encompassing the previously named PHP type 1A and 1C, pseudopseudohypoparathyroidism, Progressive

Osseous Heteroplasia and Osteoma Cutis) whereas epigenetic defects at the same locus are causative of

PT
iPPSD3 (previously termed PHP type 1B). Alterations of other downstream molecular effectors, including

the Protein Kinase cAMP-Dependent Type I Regulatory Subunit Alpha (PRKAR1A) and the

RI
Phosphodiesterase 4D (PDE4D), cause iPPSD4 and iPPSD5 (previously defined as ACRDYS1 and

SC
ACRDYS2, being ACRDYS the acronym for acrodysostosis) whereas genetic variants affecting the

Phosphodiesterase 3A (PDE3A) have been associated to iPPSD6 (Table 1) (21-22, 36-40).

U
4.1 Genetic defects at the Gsα coding gene cause iPPSD2
AN
The cluster named iPPSD2 comprises Pseudohypoparathyroidism type 1A (PHP1A, MIM103580),

Pseudohypoparathyroidism type 1C (PHP1C, MIM612462), Pseudopseudohypoparathyroidism (PPHP,


M

MIM612463), Progressive Osseous Heteroplasia and Osteoma Cutis (POH, MIM166350).


D

PHP1A patients are characterized by features of Albright Hereditary Osteodystrophy (AHO) and resistance
TE

to hormones acting through GPCRs (PTH, TSH, gonadotropins and GHRH), with important interindividual

variability in severity and time course (10-13, 37, 41-44). Patients develop hyperphosphatemia,
EP

hypocalcemia and elevated PTH over the first years of life (10). Hypothyroidism may be detected at neonatal

screening and, in most cases, patients develop mild resistance to TSH with normal/slightly low thyroid
C

hormone levels early in infancy, sometimes over childhood or adolescence, often being already present at the
AC

time of PTH resistance detection (14-16). Female patients usually present delayed or incomplete sexual

maturation, amenorrhea/oligomenorrhea and/or infertility, but no causal relation between gonadotropins

levels and hypogonadism has been confirmed (17). In about two third of investigated patients resistance to

GHRH causing GH deficiency is reported (17-19).

PHP1C is clinically indistinguishable from PHP1A, and, at present, the differential diagnosis depends on the

detection of a normal Gsα activity in the membranes of erythrocytes, fibroblasts and platelets of PHP1C
ACCEPTED MANUSCRIPT
patients, a technique available in very few laboratories, not validated for diagnostic use, and does not add

useful information for patients’ treatment and follow up (45).

In PPHP, AHO occurs in the absence of endocrine abnormalities and may present both sporadically or

coexist with PHP1A in the same family but never in the same sibship (46-49).

POH is a rare disorder of mesenchymal differentiation characterized by ectopic ossifications in dermis,

PT
skeletal muscle and deep connective tissues, whose location and extension are extremely variable. The

RI
ectopic bone formation begins as a primary osteoma cutis within the dermis at birth or during infancy, then it

extends into subcutaneous and deep connective tissues during childhood (50-52). Recently, POH patients

SC
showing also AHO features and growth disturbances during the prime infancy have been reported (51-56).

U
AN
iPPSD2 is caused by inactivating mutations affecting Gsα coding exons, encoded by the GNAS gene (MIM

*139320; 20q13.2-13.3), that lead to a partial deficiency of a reduction in the Gsα activity in the membranes
M

of various cell types (erythrocytes, fibroblasts, platelets). Mutations at the maternal allele are associated with

PHP1A and PHP1C, whereas the same mutations, when affecting the paternal allele lead to PPHP or POH
D

(44-45, 48-49, 52-54).


TE

Since the description of the first GNAS inactivating mutation in 1990, more than 200 variants (both point

mutations and small/large deletions or insertions) distributed through the gene have been discovered. Most
EP

defects are private mutations, and the few recurring ones derive from a common molecular mechanism rather

than from a founder effect. The only known mutational hot spot is the c.568_571del deletion in exon 7. All
C

GNAS exons are affected with different frequency, with the exception of exons 1 and 7 that host about 20%
AC

each (37, 43-44, 48-49, 57).

Due to the autosomal dominant inheritance of GNAS defects, offsprings of iPPSD2 patients have a 50%

recurrence risk and depending on the involved allele the clinical outcome will be different (PHP1A/PHP1C

versus PPHP/POH). The detection of pathogenetic variants provides the possibility of predictive genetic

testing in relatives as well as prenatal diagnosis (41-44, 48-49, 57).


ACCEPTED MANUSCRIPT
All known sequence variants, described following the HGVS recommendations according with the reference

transcript NM_001077488.2, are recorded in the in the publicly available LOVD database at

https://databases.lovd.nl/shared/genes/GNAS.

4.2. Epigenetic defects at GNAS locus cause iPPSD3 or Pseudohypoparathyroidism type 1B

PT
iPPSD3 is characterized by PTH renal resistance, frequently also TSH resistance, in the absence of other

RI
physical abnormalities. Recently, also the impairment of the GHRH-R signaling was proposed as GH

deficiency in affected monozygotic twins was reported (58-61).

SC
Common complications of chronic hypocalcemia together with hyperphosphatemia are basal ganglia

U
calcifications and cataracts.
AN
The iPPSD3 subgroup includes disorders associated with changes in the methylation pattern of GNAS

differentially methylated regions (DMRs), including primary epigenetic defects (both severe and partial
M

broad GNAS loss of imprinting) and conditions secondary to uniparental disomy of the paternal chromosome

20 (UPD(20)pat) and to deletions of the imprinting control region located within STX16 and/or NESP55
D

DMR (62-71).
TE

The Gsα expression is subject to tissue-specific imprinting and iPPDS3 patients display a paternal-specific
EP

patterns of cytosine methylation at GNAS DMRs of their maternally inherited alleles. Most cases are

sporadic, so that only one affected individual in a given family has the defect with no underlying known
C

genetic lesion, thus not permitting to determine the recurrence risk.


AC

The existence of patients with a partial loss of imprinting supports the hypothesis that this type of epigenetic

defect is of postzigotic derivation and leads to epigenetic mosaicism, which could justify its sporadicity and

phenotypic variability (72).

Only in a small subset of sporadic patients, the epigenetic defect is associated with UPD(20)pat, whose

recurrence, in the absence of a parental translocation, is expected to be less than 1% (62, 69-71).
ACCEPTED MANUSCRIPT
On the other hand, the familial form of the disease has a 50% recurrence risk because it is maternally

transmitted in an autosomal dominant manner. This autosomal dominant form is typically characterized by

an isolated loss of methylation affecting the exon GNAS A/B:TSS-DMR due to microdeletions disrupting the

imprinting control region located into the STX16 gene or, less frequently, by a methylation defect at all

maternal GNAS DMRs caused by deletions removing the NESP55 DMR (63-68).

PT
RI
4.3. Defects at PKA cause iPPSD4 or iPPSD5

iPPSD4 and iPPSD5 refer to Acrodysostosis type 1 (ACRDYS1, MIM101800) and type 2 (ACRDYS2,

SC
614613), respectively, that are rare congenital malformation syndromes characterized by skeletal dysplasia

frequently associated with AHO-like features (see section 4.5 for Albright’s hereditary osteodystrophy -

U
AHO), such as small broad hands and feet with brachydactyly type E, progressive growth failure with severe
AN
short stature with cone-shaped epiphyses and early epiphyseal fusion, hypoplasia of the skull and thickened

calvaria, advanced bone age, variable degrees of intellectual, developmental and/or behavioural disabilities
M

and facial dysostosis (broad face with widely spaced eyes, maxillonasal hypoplasia and flattening of the
D

nasal bridge) (9, 66-70). Additional recurring comorbidities are hearing loss, intracranial hypertension,
TE

shypo-deformity of knees and shoulders and atopy/rhinitis/eczema.

About 65% of ACRDYS1 and about 15% of ACRDYS2 patients present with PTH and TSH resistance, and
EP

up to 20% of ACRDYS2 patients show an altered response to FSH, cryptorchidism and/or lack of puberal

spurt (73-90).
C
AC

iPPSD4 and iPPSD5 derive from autosomal dominant genetic defects, with a 50% recurrence risk in the

offsprings of mutated patients, affecting PRKAR1A (MIM* 188830; 17q24.2) and PDE4D (MIM*600129;

5q12.1) genes, respectively.

All known variants are mainly private, exept a mutational hot spot in the PRKAR1A gene (c.1102C>T). As

for the PDE4D gene, mutations are confined to single kindreds, with the exception of 4 recurring variants

affecting the catalytic unit (c.803T>C, c.1586A>C, c.1835G>A and c.1850T>C) (78-79, 81, 83-88).
ACCEPTED MANUSCRIPT
All known sequence variants, described following the HGVS recommendations according with the reference

transcript NM_002734.4 for the PRKAR1A gene and NM_001165899.1 for the PDE4D gene, are recorded in

the in the publicly available LOVD database at https://databases.lovd.nl/shared/genes/PRKAR1A and

https://databases.lovd.nl/shared/genes/PDE4D.

PT
4.4. PDE3A mutations and iPPSD6

RI
The iPPSD6 subtype includes the previously named Hypertension with Brachydactyly Syndrome (HTNB,

MIM112410), characterized by brachydactyly type E, severe salt-independent but age-dependent

SC
hypertension, increased fibroblast growth rate, neurovascular contact at the rostral-ventrolateral medulla,

altered baroreflex blood pressure regulation and death from stroke before age 50 years when untreated (40).

U
Up to now few cases have been described, so there is a lack of a full clinical characterization of this disorder.
AN
M

iPPSD6 is associated with genetic variants affecting the PDE3A gene (MIM*123805; 3p21.31).
D

Genetic variants detected to date are located in exon 4 (c.1333A>G, c.1334C>A/G, c.1336T>C, c.1339G>A,
TE

c.1340C>T and c.1346G>A/T) and they increase the hydrolytic activity on cAMP (91).

All known sequence variants, described following the HGVS recommendations according with the reference
EP

transcript NM_000921.4, are recorded in the in the publicly available LOVD database at

https://databases.lovd.nl/shared/genes/PDE3A.
C
AC

4.5. Overlapping phenotypes in iPPSDs

Patients with iPPSD2 may present with heterogeneous and nonspecific clinical findings including a

dysmorphic trait such as rounded face, auxological parameters such as decreased growth velocity and/or

short stature and early-onset obesity, variable degrees of mental retardation and/or behavioural defects,

extraskeletal ossifications and brachydactyly, that are collectively described as a clinical entity termed

Albright hereditary osteodystrophy (AHO). Such phenotypic features are frequently observed also in patients
ACCEPTED MANUSCRIPT
affected with iPPSD3 (59-64) or iPPSD4 and iPPSD5, making the differential diagnosis even more

challenging (22, 79, 92-93).

Additional musculoskeletal abnormalities that seem to be associated with AHO include the spinal cord

compression and the carpal tunnel syndrome (94-95). In patients affected with iPPSD4 and iPPSD5, cone-

shaped epiphyses and facial dysmorphism are often observed (78).

PT
Recent data suggested that defects affecting the PTH/PTHrP pathway may also affect the intrauterine and

RI
postnatal growth (96). As a matter of fact, in subjects with the paternally-inherited GNAS disease a growth

retardation leading to low birth weight and length was described, while in subjects affected with the

SC
autosomal dominant form of the GNAS imprinting disorder an opposite phenotype with increased intrauterine

growth was documented (96-98). In about 15% of all the subjects with acrodysostosys bearing a PRKAR1A

U
genetic variant an intrauterine growth retardation was reported, but this percentage is likely to be under-
AN
estimated, this sign not being systematically collected in most series of the past (78).

As for neurocognitive abnormalities, including mental retardation, developmental delay and emotional
M

disorders, frequency and severity are not yet well established in iPPSD2 and iPPSD3 patients. Moreover, an
D

apparent discrepancy between the adult (27%) and the pediatric populations (64%) was reported (46, 92).
TE

About half of PRKAR1A mutated patients and up to 95% of the PDE4D mutated patients (iPPSD4&5)

presented neurocognitive impairment in a case series (78).


EP

Heterotopic intramembranous ossifications are one of the most specific features of GNAS mutations. Their

number and extension are highly heterogeneous and can be found on physical examination as palpable hard
C

nodules, often limited to the dermis and subcutaneous tissues in iPPSD2 patients but they may deepen and
AC

become highly invalidating in what we call Progressive Osseous Heteroplasia.

Premature closure of the growth plate, associated with coning of the epiphysis, determines the brachydactyly

(shortening of III, IV and V metacarpals, and I distal phalanx), that becomes apparent during

infancy/childhood. Due to its highly variable often asymmetric presentation, it is recommended to construct

the metacarpophalangeal pattern profile after posterioanterior left hand radiographs. Brachydactyly is present

in iPPSD2 and in more than 90% of iPPSD4 and iPPSD5 patients (78, 99).
ACCEPTED MANUSCRIPT
Further additional recurring comorbidities, such as hearing loss, recurrent otitis media, intracranial

hypertension, shypodeformity of knees and shoulders, and atophy/rhinitis/eczema, deserve the investigation

of larger cohorts in order to define their relationship with iPPSD4 and iPPSD5 (78).

5. Treatment and Follow-up

PT
PTH resistance should be treated in order to maintain normo-calcemia. Oral calcium supplementation and

RI
active vitamin D metabolites, preferentially calcitriol or alfa-calcidiol, represent the main therapeutic option.

Being preserved the calcium-reabsorbing action of PTH in the distal nephron, hypercalciuria rarely occurs

SC
during treatment so that calcium and vitamin D supplementation can be given at doses higher than those

usually used in primary hypoparathyroidism, although caution is still required as for potential side effects at

U
the kidney level. To avoid any potential skeletal demineralization effects of PTH excess, it is important to
AN
normalize calcium levels and lower circulating PTH as much as possible (100). Vitamin D active analogues

may be started during the development of PTH resistance, once PTH rises, to prevent severe hypocalcemia.
M

Mostly during infancy and childhood, phosphorus chelants may be necessary to ameliorate
D

hyperphosphoremia.
TE

TSH and gonadotropin resistance should be treated as any other form of hypothyroidism or hypogonadism

with levothyroxine and sex hormones, respectively.


EP

Short stature most probably results from a combination of the premature fusion of growth plates, causing the

loss of the puberal growth spurt, and a GH deficiency. A pilot study on rhGH replacement therapy showed
C

that height and growth velocity may be improved by the administration during the limited time window of
AC

the pre-pubertal linear growth, although, actually, no conclusive data are available on the treatment of GH

deficiency (101).

With regard to weight and BMI, patients should counteract the tendency to develop obesity by an appropriate

dietary and exercise intervention. No specific therapies for physical and neurocognitive manifestations are

available. As for heterotopic ossifications, the only opportunity is surgical excision, to be considered just in

case of discomfort, close relation with joints or disfigurement.


ACCEPTED MANUSCRIPT

6. Conclusions

iPPSDs are heterogeneous but closely related disorders caused by different and complex (epi)genetic defects.

Indeed, patients face a wide range of problems from early childhood to adulthood, including severe

alterations of mineral metabolism, endocrine disturbancies due to hormone resistance leading to

PT
hypothyroidism, hypogonadism and GH deficiency, growth impairment independently of hormonal status,

RI
skeletal issues, ectopic ossifications with potential severe limitation in mobility, and cognitive and/or

psychomotor impairment. For these reasons a multidisciplinary approach for getting a correct diagnosis,

SC
management and follow-up is mandatory, as a specialized expertise is required to manage each of the many

clinical aspects and potential complications (102).

U
AN
PRACTICE POINTS
M

• PTH resistance is defined by the co-existence of elevated serum levels of PTH, hypocalcemia, elevated

phosphatemia, normal renal function and normal magnesium levels, in the absence of vitamin D
D

deficiency.
TE

• The objectives of conventional management of PTH resistance include maintenance of serum levels of

calcium and phosphorus within the normal range, while avoiding hypercalciuria, and lowering PTH levels
EP

as permitted.

• The impairment of the Gsα/cAMP/PKA pathway mostly affects the signaling of PTH/PTHrP, and, with
C

different severity, the signaling of additional hormones, including TSH, GHRH, calcitonin and
AC

gonadotropins. These resistances can be detected at different stages of life in the patients, with different

severity and time course.

• The diagnosis of TSH resistance is defined by elevated TSH levels, with normal or low free thyroid

hormones levels, usually in the absence of goiter and antithyroid antibodies. It may develop over early

infancy, childhood or adolescence, although it is seldom detected at neonatal screening.

• iPPSD2 is caused by inactivating mutations at the GNAS gene. Mutations at the maternal allele are

associated with PHP1A and PHP1C, which curse with AHO and hormone resistance; when they affect the
ACCEPTED MANUSCRIPT
paternal allele, lead to PPHP or POH. PPHP is manifested with AHO without hormone resistance,

whereas POH is characterized by ectopic ossifications rarely associated with AHO features.

• iPPSD3 is characterized by PTH renal resistance, frequently also TSH resistance, in the absence of other

physical abnormalities. This subgroup includes disorders associated with changes in the methylation

pattern of GNAS DMRs, including primary epigenetic defects, UPD(20)pat and deletions of the

PT
imprinting control region located within STX16 and/or NESP55 DMR.

• iPPSD4 and iPPSD5 refer to ACRDYS1 and ACRDYS2, respectively, which are rare congenital skeletal

RI
displasias with AHO-like features. About 65% of ACRDYS1 and 15% of ACRDYS2 patients present

with PTH and TSH resistance. iPPSD4 and iPPSD5 derive from autosomal dominant genetic defects,

SC
affecting different functional domains of PRKAR1A and PDE4D, respectively.

• iPPSDs are heterogeneous but closely related disorders caused by different and complex (epi)genetic

U
defects. As patients face a wide range of problems from early childhood to adulthood, specialized and
AN
multidisciplinary expertise is required to manage each of the many clinical aspects and potential
M

complications.
D

RESEARCH AGENDA
TE

While considerable progress has been made over the past years in the genetic diagnosis as well as patients’

managment and follow up, challenges still exist as to the anticipation and prevention of long-term
EP

complications related to the natural history of these disorders. Thus, future research should focus on:
C

• The precise characterization of the natural history study of these disorders by a multicenter, international
AC

and systematic collection of clinical, biochemical and health-related quality of life data.

• The identification of new genes underlying unresolved cases with iPPSD.

• The prevention of long-term complications associated with PTH resistance through the optimization and

standardization of substitutive therapies.

• The improvement of final height through the optimization of indications, doses, efficacy and framework

for the treatment with rhGH in GH-deficient patients.


ACCEPTED MANUSCRIPT
• The identification of environmental factors and underlying molecular defects contributing to the

development and progression of ectopic ossifications, with the aim of preventing them and, when present,

of identifying novel targets for their treatment.

PT
RI
U SC
AN
M
D
TE
C EP
AC
ACCEPTED MANUSCRIPT
Summary

The non-responsiveness to the PTH/PTHrP pathway derives from molecular alterations affecting different

members of the GPCRs signaling in target organs, including both the PTHR1 receptor and downstream

effectors GNAS, PRKAR1A, PDE4D and PDE3A.

The complex (epi)genetic defects affecting signalling molecules of the Gsα/cAMP/PKA pathway cause

PT
different but related diseases, collectively named as inactivating PTH/PTHrP Signaling Disorder (iPPSD)

RI
(previously Pseudohypoparathyroidism, Pseudopseudohypoparathyroidism, Progressive Osseous

Heteroplasia, Acrodysostosis).

SC
Patients face a wide range of problems from early childhood to adulthood, including severe alterations of

mineral metabolism, endocrine disturbances due to other hormone resistances, growth impairment

U
independently of hormonal status, skeletal issues, ectopic ossifications, and cognitive and/or psychomotor
AN
impairment.
M

Genetic variants of the PTHR1 gene are associated with the iPPSD1 cluster that includes the Blomstrand

chondrodysplasia and the Eiken syndrome, with a recessive transmission pattern of inheritance.
D

The iPPSD2 cluster, associated with genetic GNAS defects, comprises Pseudohypoparathyroidism type 1A,
TE

Pseudohypoparathyroidism type 1C, Pseudopseudohypoparathyroidism and Progressive osseous

heteroplasia.
EP

The iPPSD3 subgroup includes disorders associated with changes in the methylation pattern of GNAS
C

DMRs, including primary epigenetic defects and conditions secondary to UPD(20)pat and to deletions of the
AC

imprinting control region located within STX16 and/or NESP55 DMR.

iPPSD4 and iPPSD5 derive from autosomal dominant genetic defects affecting PRKAR1A and PDE4D

genes, respectively. iPPSD6 is associated with genetic variants affecting the PDE3A gene.

iPPSDs show a molecular and clinical overlap that makes sometimes challenging and time-consuming to

perform a precise differential diagnosis. A multidisciplinary approach to reach a correct diagnosis together

with proper treatment and follow-up is mandatory.


ACCEPTED MANUSCRIPT

Acknowledgements: This work was supported by Ricerca Corrente Funds from Fondazione IRCCS Ca’

Granda Policlinico Ospedale Maggiore (to GM); a grant from the Instituto de Salud Carlos III (Institute of

Health Carlos III) of the Ministry of Economy and Competitiveness (Spain) (to GPdN and AP), co-financed

PT
by the European Regional Development Fund (PI16-00073), a grant from the Department of Health of the

RI
Basque Government (GV2017/111040 to GPdN). The authors are members of the Euro-

Pseudohypoparathyroidism network (EuroPHP). All members of the EuroPHP network have been members

SC
of the EUCID.net (COST action BM1208 on imprinting disorders; www.imprinting-disorders.eu).

U
AN
Conflict of interest statement: The authors declare no competing interests.
M
D
TE
C EP
AC
ACCEPTED MANUSCRIPT
LEGEND TO THE FIGURE

Figure 1. Schematic representacion of the Gsα/PKA/AMPc signaling pathway. (A) Several hormones

(PTH, PTHrP, TSH, gonadotropin, GHRH, calcitonin, epinephrin) act throught this pathway. (B) The

hormone binding to the G-protein-coupled receptor (GPCR) activates the Gsα subunit by exchanging GDP

with GTP, which is realeased from its heterotrimeric form and (C) travels by the cell membrane to interact

PT
with the adenylate ciclase (AC), promoting cAMP production. (D) cAMP binds to the regulatory subunits of

the cAMP-dependent PKA enzyme, causing them to break apart from the catalytic subunits and (E) allowing

RI
its activation and subsequent phosphorylation of transcription factors. The PDE4D convert cAMP into AMP

SC
to swith off the signal. (F) Phosphorylation of the transcription factors converts them into their active form,

so, after going into the nucleous, they regulate gene expression.

U
AN
M
D
TE
C EP
AC
ACCEPTED MANUSCRIPT
References (10 most important with*)

1. Todorova-Koteva K, Wood K, Imam S, Jaume JC. Screening for parathyroid hormone resistance in
patients with non-phenotypically evident pseudohypoparathyroidism. Endocr Pract 2012; 1–21.

2. Potts JT. Parathyroid hormone: past and present. J Endocrinol 2005; 187(3): 311-325.

3. Gensure RC, Gardella TJ, Juppner H. Parathyroid hormone and parathyroid hormone-related peptide and

PT
their receptors. Biochem Biophys Res Commun 2005; 328: 666-678.

4. Poole K, Reeve J. Parathyroid hormone – a bone anabolic and catabolic agent. Curr Opin Pharmacol 2005;

RI
5(6): 612–617.

5. Wettschureck N, Offermanns S. Mammalian G proteins and their cell type specific functions. Physiol Rev

SC
2005; 85(4): 1159-1204.

6. Spiegel AM, Shenker A, Weinstein LS. Receptor-effector coupling by G proteins: implications for normal

U
and abnormal signal transduction. Endocr Rev 1992; 13(3): 536-565.
AN
7. Spiegel AM. Mutations in G proteins and G protein-coupled receptors in endocrine disease. J Clin
Endocrinol Metab 1996; 81(7): 2434-2442.
M

8. Lania A, Mantovani G, Spada A. G protein mutations in endocrine diseases. Eur J Endocrinol 2001;
145(5): 543-559.
D

9. Lania AG, Mantovani G, Spada A. Mechanisms of disease: Mutations of G proteins and G-protein-
coupled receptors in endocrine diseases. Nat Clin Pract Endocrinol Metab 2006; 2(12): 681-693.
TE

10*. Weinstein LS, Yu S, Warner DR et al. Endocrine manifestations of stimulatory G protein α-subunit
mutations and the role of genomic imprinting. Endocr Rev 2001; 22: 675–705.
EP

11. Levine MA, Downs RW Jr, Moses AM et al. Resistance to multiple hormones in patients with
pseudohypoparathyroidism. Association with deficient activity of guanine nucleotide regulatory protein. Am
C

J Med 1983; 74(4): 545–556.


AC

12. Wemeau JL, Balavoine AS, Ladsous M et al. Multihormonal resistance to parathyroid hormone, thyroid
stimulating hormone, and other hormonal and neurosensory stimuli in patients with
pseudohypoparathyroidism. J Pediatr Endocrinol Metab 2006; 19(Suppl 2): 653–661.

13. Germain-Lee EL. Short stature, obesity, and growth hormone deficiency in pseudohypoparathyroidism
type Ia. Pediatr Endocrinol Rev 2006; 3: 318–327.

14. Levine MA, Jap TS, Hung W. Infantile hypothyroidism in two sibs: an unusual presentation of
pseudohypoparathyroidism type Ia. J Pediatr 1985; 107: 919–922.
ACCEPTED MANUSCRIPT
15. Pohlenz J, Ahrens W, Hiort O. A new heterozygous mutation (L338N) in the human Gsα (GNAS1) gene
as a cause for congenital hypothyroidism in Albright’s hereditary osteodystrophy. Eur J Endocrinol 2003;
148: 463–468.

16. Pinsker JE, Rogers W, McLean S et al. Pseudohypoparathyroidism type 1a with congenital
hypothyroidism. J Pediatr Endocrinol Metab 2006; 19: 1049–1052.

17. Mantovani G, Spada A. Resistance to growth hormone releasing hormone and gonadotropins in

PT
Albright’s hereditary osteodystrophy. J Pediatr Endocrinol Metab 2006; 19: 663–670.

18. Mantovani G, Maghnie M, Weber G et al. Growth hormone-releasing hormone resistance in

RI
pseudohypoparathyroidism type Ia: new evidence for imprinting of the Gsα gene. J Clin Endocrinol Metab
2003; 88: 4070–4074.

SC
19. Germain-Lee EL, Groman J, Crane JL et al. Growth hormone deficiency in pseudohypoparathyroidism
type 1a: another manifestation of multihormone resistance. J Clin Endocrinol Metab 2003; 88: 4059–4069.

U
20. de Sanctis L, Bellone J, Salerno M et al. GH secretion in a cohort of children with
AN
pseudohypoparathyroidism type Ia. J Endocrinol Invest 2007; 30: 97–103.

21. Linglart A, Silve C, Rothenbuhler A. Multiple hormonal resistances: Diagnosis, evaluation and therapy.
M

Ann Endocrinol (Paris) 2015;76(2): 98-100.

22*. Mantovani G, Spada A, Elli FM. Pseudohypoparathyroidism and Gsα-cAMP-linked disorders: current
D

view and open issues. Nat Rev Endocrinol 2016;12(6): 347-356.


TE

23. Turan S, Fernandez-Rebollo E, Aydin C et al. Postnatal establishment of allelic Gαs silencing as a
plausible explanation for delayed onset of parathyroid hormone resistance owing to heterozygous Gαs
disruption. J Bone Miner Res 2014; 29(3): 749-760.
EP

24. Usardi A, Mamoune A, Nattes E et al. Progressive Development of PTH Resistance in Patients with
Inactivating Mutations on the Maternal Allele of GNAS. J Clin Endocrinol Metab 2017; 102(6): 1844-1850.
C

25*. Yu S, Yu D, Lee E et al. Variable and tissue-specific hormone resistance in heterotrimeric Gs protein
AC

alpha-subunit (Gsalpha) knockout mice is due to tissue-specific imprinting of the gsalpha gene. Proc Natl
Acad Sci USA 1998; 95(15): 8715-8720.

26*. Thiele S, Mantovani G, Barlier A et al. From pseudohypoparathyroidism to inactivating PTH/PTHrP


signalling disorder (iPPSD), a novel classification proposed by the EuroPHP network. Eur J Endocrinol
2016; 175(6): P1-P17.

27*. Jüppner H, Abou-Samra AB, Freeman M, et al. A G protein-linked receptor for parathyroid hormone
and parathyroid hormone-related peptide. Science 1991; 254(5034): 1024-1026.
ACCEPTED MANUSCRIPT
28. Murray TM, Rao LG, Divieti P et al. Parathyroid hormone secretion and action: evidence for discrete
receptors for the carboxyl-terminal region and related biological actions of carboxyl- terminal ligands.
Endocr Rev 2005; 26(1): 78-113.

29. Blomstrand S, Claësson I, Säve-Söderbergh J. A case of lethal congenital dwarfism with accelerated
skeletal maturation. Pediatr Radiol. 1985; 15(2): 141-3.

30. Duchatelet S, Ostergaard E, Cortes D et al. Recessive mutations in PTHR1 cause contrasting skeletal

PT
dysplasias in Eiken and Blomstrand syndromes. Hum Mol Genet 2005; 14(1): 1-5.

31. Hoogendam J, Farih-Sips H, Wÿnaendts LC et al. Novel mutations in the parathyroid hormone

RI
(PTH)/PTH-related peptide receptor type 1 causing Blomstrand osteochondrodysplasia types I and II. J Clin
Endocrinol Metab 2007; 92(3): 1088-95.

SC
32. Kim J, Won HH, Kim Y et al. Breakpoint mapping by whole genome sequencing identifies PTH2R gene
disruption in a patient with midline craniosynostosis and a de novo balanced chromosomal rearrangement. J

U
Med Genet 2015; 52(10): 706-709.
AN
33. Decker E, Stellzig-Eisenhauer A, Fiebig BS et al. PTHR1 loss-of-function mutations in familial,
nonsyndromic primary failure of tooth eruption. Am J Hum Genet 2008; 83(6): 781-786.
M

34. Roth H, Fritsche LG, Meier C et al. Expanding the spectrum of PTH1R mutations in patients with
primary failure of tooth eruption. Clin Oral Investig 2014; 8(2): 377-384.
D

35. Panda DK, Goltzman D, Karaplis AC. Defective postnatal endochondral bone development by
chondrocyte-specific targeted expression of parathyroid hormone type 2 receptor. Am J Physiol Endocrinol
TE

Metab. 2012; 303(12): E1489-1501.

36. Levine MA, Downs RW, Singer M et al. Deficient activity of guanine nucleotide regulatory protein in
EP

erythrocytes from patients with pseudohypoparathyroidism. Biochem Biophys Res Commun 1980; 94:
1319–1324.
C

37. Patten JL, Johns DR, Valle D et al. Mutation in the gene encoding the stimulatory G protein of adenylate
AC

cyclase in Albright’s hereditary osteodystrophy. N Engl J Med 1990; 322(20): 1412–1419.

38. Mantovani G, Spada A. Mutations in the Gs alpha gene causing hormone resistance. Best Pract Res Clin
Endocrinol Metab 2006; 20(4): 501-513.

39. Silve C. Acrodysostosis: A new form of pseudohypoparathyroidism? Ann Endocrinol (Paris) 2015;
76(2): 110-112.

40. Maass PG, Aydin A, Luft FC et al. PDE3A mutations cause autosomal dominant hypertension with
brachydactyly. Nature Genet 2015; 47: 647-653.
ACCEPTED MANUSCRIPT
41. Mitsui T, Nagasaki K, Takagi M et al. A family of pseudohypoparathyroidism type Ia with an 850-kb
submicroscopic deletion encompassing the whole GNAS locus. Am J Med Genet A 2012; 158A(1): 261-264.

42. Garin I, Elli FM, Linglart A et al. Novel microdeletions affecting the GNAS locus in
pseudohypoparathyroidism: characterization of the underlying mechanisms. J Clin Endocrinol Metab 2015;
100(4): E681-687.

43. Thiele S, Werner R, Grötzinger J et al. A positive genotype-phenotype correlation in a large cohort of

PT
patients with Pseudohypoparathyroidism Type Ia and Pseudopseudohypoparathyroidism and 33 newly
identified mutations in the GNAS gene. Mol Genet Genomic Med 2015; 3(2): 111‑120.

RI
44. Lemos MC, Thakker RV. GNAS mutations in Pseudohypoparathyroidism type 1a and related disorders.
Hum Mutat 2015; 36(1): 11‑19.

SC
45. Aldred MA. Genetics of pseudohypoparathyroidism types Ia and Ic. J Pediatr Endocrinol Metab 2006;
19(2): 635–640.

U
46. Farfel Z, Friedman E. Mental deficiency in pseudohypoparathyroidism type I is associated with Ns-
AN
protein deficiency. Ann Intern Med 1986; 105(2): 197–199.

47. Eyre WG, Reed WB. Albright's hereditary osteodystrophy with cutaneous bone formation. ArchDermatol
M

1971;104: 634-642.

48. Wilson LC, Oude Luttikhuis ME, Clayton PT et al. Parental origin of Gs alpha gene mutations in
D

Albright's hereditary osteodystrophy. J Med Genet 1994; 31(11): 835-839.


TE

49. Nakamoto JM, Sandstrom AT, Brickman AS et al. Pseudohypoparathyroidism type Ia from maternal but
not paternal transmission of a Gsalpha gene mutation. Am J Med Genet 1998; 77(4): 261-267.
EP

50. Kaplan FS, Craver R, MacEwen GD et al. Progressive osseous heteroplasia: a distinct developmmental
disorder of heterotopic ossification two new case reports and follow-up of three previously reported cases, J
Bone Joint Surg 1994; 74A: 425-436.
C

51. Eddy MC, Jan De Beur SM, Yandow SM et al. Deficiency of the alpha-subunit of the stimulatory G
AC

protein and severe extraskeletal ossification, J Bone Miner Res 2000; 15(11): 2074-2083.

52. Adegbite NS, Xu M, Kaplan FS et al. Diagnostic and Mutational Spectrum of Progressive Osseous
Heteroplasia (POH) and Other Forms of GNAS-based Heterotopic Ossification. Am J Med Genet A 2008;
146A(14): 1788–1796.

53. Shore EM, Ahn J, Jan dB et al. Paternally inherited inactivating mutations of the GNAS1 gene in
progressive osseous heteroplasia. N Engl J Med 2002; 346: 99-106.

54. Elli FM, Barbieri AM, Bordogna P et al. Screening for GNAS genetic and epigenetic alterations in
progressive osseous heteroplasia: first Italian series. Bone 2013; 56: 276-280.
ACCEPTED MANUSCRIPT
55. L. Tresserra, F. Tresserra, P.J. Grases et al. Congenital plate-like osteoma cutis of the forehead: an
atypical presentation form, J Craniomaxillofac Surg 1998; 26(2): 102-106.

56. Lebrun M, Richard N, Abeguile´G et al. Progressive Osseous Heteroplasia: A Model for the Imprinting
Effects of GNAS Inactivating Mutations in Humans. J Clin Endocrinol Metab 2010; 95(6): 3028–3038.

57. Elli FM, deSanctis L, Ceoloni B et al. Pseudohypoparathyroidism type Ia and


pseudopseudohypoparathyroidism: the growing spectrum of GNAS inactivating mutations. Hum Mutat

PT
2013; 34(3): 411‑416.

58. Barret D, Breslau NA, Wax MB et al. New form of pseudohypoparathyroidism with abnormal catalytic

RI
adenylate cyclase. Am J Physiol 1989; 257: E277–283.

59. Winter JS, Hughes IA. Familial pseudohypoparathyroidism without somatic anomalies. Can Med Assoc J

SC
1986; 123: 26–31.

60. Nusynowitz ML, Frame B, Kolb FO. The spectrum of the hypoparathyroid states: a classification based

U
on physiologic principles. Medicine 1976; 55: 105–119.
AN
61. Liu J, Erlichman B, Weinstein LS. The stimulatory G protein α-subunit G s α is imprinted in human
thyroid glands: implications for thyroid function in pseudohypoparathyroidism types 1A and 1B. J Clin
M

Endocrinol Metab 2003; 88: 4336–4341.

62*. Liu J, Litman D, Rosenberg MJ et al. A GNAS1 imprinting defect in pseudohypoparathyroidism type
D

IB. J Clin Invest 2000; 106(9): 1167-1174.


TE

63. Bastepe M, Frohlich LF, Hendy GN et al. Autosomal dominant pseudohypoparathyroidism type Ib is
associated with a heterozygous microdeletion that likely disrupts a putative imprinting control element of
GNAS. J Clin Invest 2003; 112(8): 1255-1263.
EP

64*. Bastepe M, Frohlich LF, Linglart A et al. Deletion of the NESP55 differentially methylated region
causes loss of maternal GNAS imprints and pseudohypoparathyroidism type Ib. Nature genetics 2005; 37(1):
C

25-27.
AC

65. Linglart A, Gensure RC, Olney RC et al. A novel STX16 deletion in autosomal dominant
pseudohypoparathyroidism type Ib redefines the boundaries of a cis-acting imprinting control element of
GNAS. Am J Hum Genet 2005; 76: 804-814.

66. Chillambhi S, Turan S, Hwang D et al. Deletion of the noncoding GNAS antisense transcript causes
pseudohypoparathyroidism type Ib and biparental defects of GNAS methylation in cis. J Clin Endocrinol
Metab 2010; 95(8): 3993-4002.
ACCEPTED MANUSCRIPT
67. Richard N, Abeguilè G, Coudray N et al. A new deletion ablating NESP55 causes loss of maternal
imprint of A/B GNAS and autosomal dominant pseudohypoparathyroidism type Ib. J Clin Endocrinol Metab
2012; 97(5): E863-867.

68. Elli FM, de Sanctis L, Peverelli E et al. Autosomal dominant pseudohypoparathyroidism type Ib: a novel
inherited deletion ablating STX16 causes loss of imprinting at the A/B DMR. J Clin Endocrinol Metab 2014;
99(4): E724-728.

PT
69. Dixit A, Chandler KE, Lever M et al. Pseudohypoparathyroidism type 1b due to paternal uniparental
disomy of chromosome 20q. J Clin Endo38crinol Metab 2013; 98(1): E103-108.

RI
70. Takatani R, Minagawa M, Molinaro A et al. Similar frequency of paternal uniparental disomy involving
chromosome 20q (patUPD20q) in Japanese and Caucasian patients affected by sporadic

SC
pseudohypoparathyroidism type Ib (sporPHP1B). Bone 2015; 79: 15-20.

71. Bastepe M, Lane AH, Jüppner H. Paternal uniparental isodisomy of chromosome 20q--and the resulting

U
changes in GNAS1 methylation--as a plausible cause of pseudohypoparathyroidism. Am J Hum Genet 2001;
68(5): 1283-1289.
AN
72. Elli FM, Bordogna P, Arosio M et al. Mosaicism for GNAS methylation defects associated with
pseudohypoparathyroidism type 1B arose in early post-zygotic phases. Clin Epigenetics 2018; 10: 16.
M

73. Brailsford JF. The Radiology of Bones and Joints, 4th ed. 1948; London, Churchill
D

74. Maroteaux P, Malamut G. Acrodysostosis. Presse Med 1968; 76: 2189–2192.


TE

75. Robinow M, Pfeiffer RA, Gorlin RJ et al. Acrodysostosis. A syndrome of peripheral dysostosis, nasal
hypoplasia, and mental retardation. Am J Dis Child 1971; 121: 195–203.
EP

76. Reiter S. Acrodysostosis. A case of peripheral dysostosis, nasal hypoplasia, mental retardation and
impaired hearing. Pediatr Radiol 1978; 7: 53–55.

77. Davies SJ, Hughes HE. Familial acrodysostosis: can it be distinguished from Albright’s hereditary
C

osteodystrophy? Clin Dysmorphol 1992; 1: 207–215.


AC

78. Elli FM, Bordogna P, de Sanctis L et al. Screening of PRKAR1A and PDE4D in a Large Italian Series of
Patients Clinicaly Diagnosed with Albright Hereditary Osteodystrophy and/or Pseudohypoparathyroidism. J
Bone Miner Res 2016; 31(6): 1215-1224.

79. Linglart A, Menguy C, Couvineau A et al. Recurrent PRKAR1A mutation in acrodysostosis with
hormone resistance. N Engl J Med 2011; 364(23): 2218–2226.

80. Michot C, Le GC, Goldenberg A et al. Exome Sequencing Identifies PDE4D Mutations as Another
Cause of Acrodysostosis. Am J Hum Genet 2012; 90: 740-745
ACCEPTED MANUSCRIPT
81. Lee H, Graham JM Jr, Rimoin DL et al. Exome sequencing identifies PDE4D mutations in
acrodysostosis. Am J Hum Genet 2012; 90: 746-751.

82. Nagasaki K, Iida T, Sato H et al. PRKAR1A Mutation Affecting cAMP-Mediated G Protein-Coupled
Receptor Signaling in a Patient with Acrodysostosis and Hormone Resistance. J Clin Endocrinol Metab
2012; 97: E1808-E1813

83*. Linglart A, Fryssira H, Hiort O et al. PRKAR1A and PDE4D mutations cause acrodysostosis but two

PT
distinct syndromes with or without GPCR-signaling hormone resistance. J Clin Endocrinol Metab 2012;
97(12): E2328-2338.

RI
84. Lynch DC, Dyment DA, Huang L et al. Identification of novel mutations confirms PDE4D as a major
gene causing acrodysostosis. Hum Mutat 2013; 34(1): 97-102.

SC
85. Muhn F, Klopocki E, Graul-Neumann L et al. Novel mutations of the PRKAR1A gene in patients with
acrodysostosis. Clin Genet 2013; 84(6): 531-538.

U
86. Lindstrand A, Grigelioniene G, Nilsson D et al. Different mutations in PDE4D associated with
AN
developmental disorders with mirror phenotypes. J Med Genet 2014; 51(1) :45-54.

87. Kaname T, Ki CS, Niikawa N et al. Heterozygous mutations in cyclic AMP phosphodiesterase-4D
M

(PDE4D) and protein kinase A (PKA) provide new insights into the molecular pathology of acrodysostosis.
Cell Signal 2014; 26(11): 2446-2459.
D

88. Li N, Nie M, Li M et al. The first mutation identified in a Chinese acrodysostosis patient confirms a
p.G289E variation of PRKAR1A causes acrodysostosis. Int J Mol Sci 2014; 15(8): 13267-13274.
TE

89. Mitsui T, Kim OH, Hall CM et al. Acroscyphodysplasia as a phenotypic variation of


pseudohypoparathyroidism and acrodysostosis type 2. Am J Med Genet A 2014; 164A:2529-2534.
EP

90. Ablow RC, Hsia YE, Brandt IK. Acrodysostosis coinciding with pseudohypoparathyroidism and pseudo-
pseudohypoparathyroidism. AJR Am J Roentgenol 1977; 128: 95–99.
C

91. Maass PG, Aydin A, Luft FC et al. PDE3A mutations cause autosomal dominant hypertension with
AC

brachydactyly. Nat Genet 2015; 47(6): 647-53.

92. Mantovani G. Clinical review: Pseudohypoparathyroidism: diagnosis and treatment. J Clin Endocrinol
Metab 2011; 96: 3020-3030.

93. Silve C, Le-Stunff C, Motte E et al. Acrodysostosis syndromes. BoneKEy Reports 2012; 1, 225: 1-7.

94. Roberts TT, Khasnavis S, Papaliodis DN et al. Spinal cord compression in pseudohypoparathyroidism.
Spine J 2013; 13: e15–19.

95. Joseph AW, Shoemaker AH, Germain-Lee EL. Increased prevalence of carpal tunnel syndrome in
Albright hereditary osteodystrophy. J Clin Endocrinol Metab 2011; 96: 2065–2073.
ACCEPTED MANUSCRIPT
96*. Hanna P, Grybek V, de Nanclares GP et al. Genetic and epigenetic defects at the GNAS locus lead to
distinct patterns of skeletal growth but similar early-onset obesity. J Bone Miner Res 2018; Apr 25. doi:
10.1002/jbmr.3450. (Epub ahead of print)

97. Bréhin AC, Colson C, Maupetit-Méhouas S et al. Loss of methylation at GNAS exon A/B is associated
with increased intrauterine growth. J Clin Endocrinol Metab 2015; 100(4): E623-631.

98. Richard N, Molin A, Coudray N et al. Paternal GNAS mutations lead to severe intrauterine growth

PT
retardation (IUGR) and provide evidence for a role of XLas in fetal development. J Clin Endocrinol Metab
2013; 98(9): E1549-1556.

RI
99. de Sanctis L, Vai S, Andreo MR et al. Brachydactyly in 14 genetically characterized
pseudohypoparathyroidism type Ia patients. J Clin Endocrinol Metab 2004; 89(4): 1650-1655.

SC
100. Neary NM, El-Maouche D, Hopkins R et al. Development and treatment of tertiary
hyperparathyroidism in patients with pseudohypoparathyroidism type 1B. J Clin Endocrinol Metab 2012; 97:

U
3025–3030.
AN
101. Mantovani G, Ferrante E, Giavoli C et al. Recombinant human GH replacement therapy in children
with pseudohypoparathyroidism type Ia: first study on the effect on growth. J Clin Endocrinol Metab 2010;
95: 5011–5017.
M

102*. Mantovani G, Bastepe M, Monk D et al. Diagnosis and management of Pseudohypoparathyroidism


and related disorders: first international consensus statement. Nat Rev Endoc 2018; Accepted In press.
D
TE
C EP
AC
ACCEPTED MANUSCRIPT

A
B

PT
RI
C

SC
D

U
AN
M
D
E
TE F
EP
C
AC
ACCEPTED MANUSCRIPT

Table 1: Correlation between the novel and classical classifications for PTH/PTHrP signaling disorders

Novel
Classical classification OMIM Phenotype Molecular determinant

PT
classification
BLOMSTRAND
# 215045 skeletal dysplasia, advanced skeletal maturation, lethal
CHONDRODYSPLASIA PTHR1 mutations (AR, BOCD: LoF [PMID: 17164305]; Eiken: GoF [PMID:

RI
iPPSD1
7721741])
EIKEN SYNDROME # 600002 skeletal dysplasia, severely delayed skeletal maturation

SC
# 103580 Multihormonal resistance, AHO
PHP1A Maternal LoF point mutation/deletion GNAS [PMID: 2109828]

U
# 612462
PHP1C Maternal LoF point mutation GNAS exon 13; LoI at GNAS DMRs

AN
(few cases reported)

iPPSD2 AHO

M
PPHP Paternal LoF point mutation/deletion GNAS
# 612463

D
POH
# 166350 Heterotopic ossification
Paternal LoF point mutation/deletion GNAS

TE
EP
Sporadic: LoI at GNAS DMRs; UPD(20)pat
iPPSD3 PHP1B # 603233 Multihormonal resistance (rarely signs of AHO) Familial (AD): isolated LoM at GNAS A/B:TSS-DMR (maternal STX16
deletion); LoI at GNAS DMRs(maternal NESP and/or AS deletion)
C

iPPSD4 ACRDYS1 # 101800 skeletal dysplasia, AHO-like, multiple endocrine abnormalities PRKAR1A mutations (GoF of the subunit, LoF of PKA [PMID: 21651393])
AC

iPPSD5 ACRDYS2 # 614613 skeletal dysplasia, AHO-like, intellectual disability PDE4D mutations (GoF) [PMID: 14602083]

iPPSD6 HTNB # 112410 Brachydactyly, hypertension PDE3A mutations (GoF) [PMID:25961942]


ACCEPTED MANUSCRIPT

AR: autosomal recessive; LoF: loss of function; GoF: gain of function; LoI: loss of imprinting; LoM: loss of methylation; AD: autosomal dominant.

PT
RI
U SC
AN
M
D
TE
C EP
AC

Das könnte Ihnen auch gefallen