ArticlePDF Available

Haploinsufficiency of the NF1 gene is associated with protection against diabetes

Authors:

Abstract and Figures

Background The hereditary predisposition to diabetes is only partially explained by genes identified so far. Neurofibromatosis type 1 (NF1) is a rare monogenic dominant syndrome caused by aberrations of the NF1 gene. Here, we used a cohort of 1410 patients with NF1 to study the association of the NF1 gene with type 1 (T1D) and type 2 diabetes (T2D). Methods A total of 1410 patients were confirmed to fulfil the National Institutes of Health diagnostic criteria for NF1 by individually reviewing their medical records. The patients with NF1 were compared with 14 017 controls matched for age, sex and area of residence as well as 1881 non-NF1 siblings of the patients with NF1. Register-based information on purchases of antidiabetic medication and hospital encounters related to diabetes were retrieved. The Cox proportional hazards model was used to calculate the relative risk for diabetes in NF1. Results Patients with NF1 showed a lower rate of T2D when compared with a 10-fold control cohort (HR 0.27, 95% CI 0.17 to 0.43) or with their siblings without NF1 (HR 0.28, 95% CI 0.16 to 0.47). The estimates remained practically unchanged after adjusting the analyses for history of obesity and dyslipidaemias. The rate of T1D in NF1 was decreased although statistically non-significantly (HR 0.58, 95% CI 0.27 to 1.25). Conclusion Haploinsufficiency of the NF1 gene may protect against T2D and probably T1D. Since NF1 negatively regulates the Ras signalling pathway, the results suggest that the Ras pathway may be involved in the pathogenesis of diabetes.
Content may be subject to copyright.
1
Kallionpää RA, etal. J Med Genet 2020;0:1–7. doi:10.1136/jmedgenet-2020-107062
ORIGINAL RESEARCH
Haploinsufficiency of the NF1 gene is associated with
protection againstdiabetes
Roope A Kallionpää,1 Sirkku Peltonen,2,3 Jussi Leppävirta,4,5 Minna Pöyhönen,4,5
Kari Auranen,6 Hannu Järveläinen,1,7 Juha Peltonen 1
Genotype- phenotype correlations
To cite: Kallionpää RA,
Peltonen S, Leppävirta J, etal.
J Med Genet Epub ahead of
print: [please include Day
Month Year]. doi:10.1136/
jmedgenet-2020-107062
Additional material is
published online only. To view
please visit the journal online
(http:// dx. doi. org/ 10. 1136/
jmedgenet- 2020- 107062).
1Institute of Biomedicine,
University of Turku, Turku,
Finland
2Department of Dermatology
and Venereology, University of
Turku, Turku, Finland
3Department of Dermatology,
Turku University Hospital, Turku,
Finland
4Department of Clinical
Genetics, HUSLAB, Helsinki
University Hospital (HUS)
Diagnostic Center, Helsinki,
Finland
5Department of Medical and
Clinical Genetics, University of
Helsinki, Helsinki, Finland
6Department of Mathematics
and Statistics and Department
of Clinical Medicine, University
of Turku, Turku, Finland
7Department of Internal
Medicine, Satakunta Central
Hospital, Pori, Finland
Correspondence to
Professor Juha Peltonen,
Institute of Biomedicine,
University of Turku, Turku FI
20520, Finland; juhpel@ utu. fi
Received 1 April 2020
Revised 8 May 2020
Accepted 11 May 2020
© Author(s) (or their
employer(s)) 2020. Re- use
permitted under CC BY- NC. No
commercial re- use. See rights
and permissions. Published
by BMJ.
ABSTRACT
Background The hereditary predisposition to diabetes
is only partially explained by genes identified so far.
Neurofibromatosis type 1 (NF1) is a rare monogenic
dominant syndrome caused by aberrations of the NF1
gene. Here, we used a cohort of 1410 patients with NF1
to study the association of the NF1 gene with type 1
(T1D) and type 2 diabetes (T2D).
Methods A total of 1410 patients were confirmed to
fulfil the National Institutes of Health diagnostic criteria
for NF1 by individually reviewing their medical records.
The patients with NF1 were compared with 14 017
controls matched for age, sex and area of residence as
well as 1881 non- NF1 siblings of the patients with NF1.
Register- based information on purchases of antidiabetic
medication and hospital encounters related to diabetes
were retrieved. The Cox proportional hazards model was
used to calculate the relative risk for diabetes in NF1.
Results Patients with NF1 showed a lower rate of T2D
when compared with a 10- fold control cohort (HR 0.27,
95% CI 0.17 to 0.43) or with their siblings without NF1
(HR 0.28, 95% CI 0.16 to 0.47). The estimates remained
practically unchanged after adjusting the analyses for
history of obesity and dyslipidaemias. The rate of T1D
in NF1 was decreased although statistically non-
significantly (HR 0.58, 95% CI 0.27 to 1.25).
Conclusion Haploinsufficiency of the NF1 gene may
protect against T2D and probably T1D. Since NF1
negatively regulates the Ras signalling pathway, the
results suggest that the Ras pathway may be involved in
the pathogenesis of diabetes.
INTRODUCTION
Diabetes is characterised by impaired insulin produc-
tion or action, leading to chronically increased
blood glucose concentrations. Type 2 diabetes
(T2D) is the most common form of diabetes.1 It
typically manifests in adulthood and is strongly
linked to lifestyle factors such as obesity and lack
of exercise.1 2 The development of T2D involves
decreased insulin sensitivity and commonly leads
to impaired insulin production.1 3 Type 1 diabetes
(T1D) is an autoimmune disorder characterised
by destruction of the insulin- producing β-cells in
the pancreas.4 T1D is usually diagnosed already in
childhood. While T2D can be treated with a variety
of medications and lifestyle changes, patients with
T1D are fully dependent on insulin treatment.
Both types of diabetes are associated with increased
mortality.1 4
Familial aggregation and twin studies clearly
indicate a genetic component in diabetes risk.2 4–6
However, the genetic risk for especially T2D is highly
complex with relatively limited contributions from
individual genes. Genome- wide association studies
(GWAS) and sequencing have identified numerous
variants associated with T2D.7–11 These are mostly
common variants with small or modest effects or
low frequency variants with larger effects.9 12 Vari-
ants protective against T2D have been described
in, for example, CCND2,8 9 SLC30A8,11 13 TCF2,14
MC4R15 and PPARG,16 some of which can also
harbour risk- increasing variants.8 15 Regarding
T1D, up to 50% of its heredity can be explained
by human leucocyte antigen (HLA) genes, espe-
cially with HLA- DR3- DQ2 and HLA- DR4- DQ8
conferring increased risk as well as certain HLA
haplotypes providing protection against T1D.5 17 18
Other major T1D- associated genes include INS,
PTPN22 and IL2RA while most of the known non-
HLA risk genes only confer minor increases in T1D
susceptibility.18 The genes contributing to the initial
development of autoimmunity may differ from
those involved in the progression from islet autoan-
tibodies to clinical T1D, which further complicates
the genetics of T1D.19 Since family history of T1D
associates with higher risk for T2D and vice versa,
these diseases are thought to have also shared risk
genes but only a few (eg, MTNR1B and HNF1A)
have been identified.20–22 The identification of the
genetic underpinnings of diabetes elucidates the
disease pathogenesis, and knowledge of the protec-
tive variants may also suggest novel therapeutic
approaches.12
Neurofibromatosis type 1 (NF1; MIM 162200)
is a dominantly inherited monogenic multiorgan
syndrome caused by aberrations of the NF1 gene
in chromosome 17q11.2. NF1 has a prevalence
of 1/3000–1/2000 (0.03%–0.05%).23 The diag-
nosis is based on the National Institutes of Health
(NIH) diagnostic criteria,24 which include café-
au- lait macules of the skin, cutaneous and plexi-
form neurofibromas, skinfold freckling, Lisch
nodules of the eye, optic glioma, distinct osseous
lesions and NF1 in a family member. The NF1
gene encodes a tumour suppressor protein neuro-
fibromin that downregulates Ras activity.25 Thus,
NF1 is a Rasopathy. The disease- causing variants
are spread throughout the gene, and only few
mutational hotspots have been identified.26 NF1
is associated with increased mortality and a variety
of comorbidities including predisposition to many
on June 22, 2020 by guest. Protected by copyright.http://jmg.bmj.com/J Med Genet: first published as 10.1136/jmedgenet-2020-107062 on 22 June 2020. Downloaded from
2Kallionpää RA, etal. J Med Genet 2020;0:1–7. doi:10.1136/jmedgenet-2020-107062
Genotype- phenotype correlations
types of cancers and cardiovascular complications.27–32 Lower
fasting blood glucose concentrations have been reported in NF1
compared with control persons.33–35 A reduced rate of gestational
diabetes was observed in one cohort of patients with NF136 but
not in another.37 An analysis of insurance claims reported OR of
0.4 for diabetes among patients with NF1 in the USA.38 In addi-
tion, two previous studies based on death certificates have noted
a lower than expected number of diabetes- related deaths among
patients with NF1.39 40 However, these studies have not excluded
premature mortality associated with NF1 as the cause for the
lower rate of diabetes among patients with NF1. Interestingly, a
recent Danish cohort study reported a significantly reduced rate
of T1D- related hospitalisations and non- significantly reduced
rate of T2D- related hospitalisations among patients with NF1.32
Here, we aimed at further dissecting the risk for diabetes in NF1.
METHODS
Research permissions
Research permissions were obtained from the Finnish Insti-
tute for Health and Welfare, The Social Insurance Institution
of Finland, Finnish Population Register Centre and all partici-
pating hospitals. The study was register- based and exempt from
obtaining informed consent from participants.
Patients
The collection of the NF1 cohort has previously been
described.29 In brief, the cohort was collected by searching for
neurofibromatosis- related hospital visits in the 5 University
Hospitals and 15 Central Hospitals of mainland Finland in
1987–2011. The medical records of the patients identified in this
initial search were individually reviewed to confirm NF1 by the
NIH diagnostic criteria.24 Thus, the cohort is fully ascertained
and based on the complete population of mainland Finland.
The procedure yielded a total of 1410 patients with confirmed
NF1 and availability of valid minimum necessary information.
All Finnish residents have a unique personal identity code that
can be used as a key for retrieving information from national
registers.
The patients with NF1 were compared with two cohorts that
were formed using data from the Finnish Population Register
Centre: (1) For each patient with NF1, 10 controls matched for
date of birth, sex and place of residence were retrieved. First-
degree relatives of patients with NF1 were excluded. Due to the
small size of some municipalities, a total of 14 017 controls were
obtained. (2) Siblings of patients with NF1, defined by at least
one shared parent, were retrieved and persons with known or
suspected NF1 diagnosis were excluded. A total of 1881 non-
NF1 siblings of patients with NF1 were available.
Data sources
Dates of death and emigration were retrieved from the Finnish
Population Register Centre, yielding complete follow- up.
The Finnish Institute for Health and Welfare maintains the
Care Register for Health Care. The register was founded in 1969
as Hospital Discharge Register covering inpatient care. Since
1998, specialised public outpatient care has also been included.
The hospital visits and stays have been recorded using Interna-
tional Classification of Diseases, 9th revision (ICD-9) diagnosis
codes in 1987–1995 and ICD-10 diagnosis codes since 1996.
Each hospital visit and stay can be associated with up to six
diagnoses, all of which were taken into account in the present
study. These diagnoses do not necessarily reflect the primary
reason for the hospital encounter but may also be used to record
pre- existing chronic diseases. Hospital visits and stays associ-
ated with diabetes in overall were identified using ICD-10 codes
E10–E14. In order to identify T1D, ICD-10 code E10 was used.
For T2D, ICD-10 code E11 was used in 1996–2014 and ICD-9
code 250xA, where x can be any digit, was used pre-1996. The
ICD-10 code E66 was used to retrieve diagnoses of obesity, E78
for disorders of lipoprotein metabolism and other lipidaemias,
and I10 for primary hypertension.
The Social Insurance Institution of Finland is a govern-
ment agency that reimburses part of medication costs for most
prescription drugs bought by Finnish residents. The reimburse-
ments are paid directly to pharmacies when the drug is dispensed,
and the patient only pays the remaining part of the price. Since
the reimbursement does not require application by the patient,
the system encompasses the vast majority of all prescription
drug purchases in Finland. Reimbursed medication purchases
have been recorded since 1995 using Anatomical Therapeutic
Chemical (ATC) coding. Insulins and analogues are included in
class A10A and blood glucose lowering drugs other than insulins
in class A10B. Patients may also receive a higher special reim-
bursement of drugs prescribed for certain conditions, including
insulin- dependent diabetes, where an application by the physi-
cian is required.
Statistical methods
The primary study period was from 1 January 1998 to 31
December 2014 since all data types were available starting from
1998. The follow- up of patients with NF1 started at the age of
their first neurofibromatosis- related hospital visit or at their age
at the beginning of the study period, whichever was later. The
follow- up of controls started at the latter of the age at the first
neurofibromatosis- related hospital visit of the respective patient
with NF1 or the age at the beginning of the study period. The
follow- up of the siblings without NF1 started at birth, age at
the first neurofibromatosis- related hospital visit of their NF1
sibling(s) or the age at the beginning of the study period, which-
ever occurred last. For all study subjects, the follow- up ended at
the age of the first occurrence of a diagnosis of interest, death,
emigration or end of study period, whichever came first.
When all types of diabetes or T2D only were analysed, the
first occurrence of diabetes was defined by the first hospital
visit or stay with a relevant diagnosis code or the first purchase
of drugs classified as ATC A10A or A10B. In the T2D analysis
adjusted by the features of metabolic syndrome, the history
of having had a diagnosis of obesity or dyslipidaemias at any
time during the follow- up was included as a covariate. In the
T1D analysis, only the hospital visits and stays were considered
when defining the follow- up, but since T1D always progresses
to insulin- dependency, having purchased insulin or its analogue
(ATC A10A) at any time was also required to exclude any coding
errors. Patients with T1D were excluded from the analyses of
T2D. As a sensitivity analysis, the rate of T2D was also assessed
in the years 1987–2014 but medication purchases were not
included due to the availability of this information only since
1995. Another sensitivity analysis restricted the age range of
follow- up to 0–50 years.
Rates were estimated using the Poisson distribution. The esti-
mation of HRs was based on the Cox proportional hazard models
with delayed entry. In these analyses, the proportional hazards
assumption was fulfilled as assessed using scaled Schoenfeld
residuals. To allow heterogeneity between subgroups formed by
each patient with NF1 and the respective controls, a subgroup-
specific frailty term was included in the models. Similarly, in the
on June 22, 2020 by guest. Protected by copyright.http://jmg.bmj.com/J Med Genet: first published as 10.1136/jmedgenet-2020-107062 on 22 June 2020. Downloaded from
3
Kallionpää RA, etal. J Med Genet 2020;0:1–7. doi:10.1136/jmedgenet-2020-107062
Genotype- phenotype correlations
Table 1 Characteristics of the study cohorts in the primary study period 1998–2014 and in the sensitivity analysis study period 1987–2014
Follow- up 1998–2014 Follow- up 1987–2014
Patients with NF1 Controls Siblings without NF1 Patients with NF1 Controls Siblings without NF1
n 1349 13 870 1871 1410 14 017 1881
Sex
Females, n (%) 694 (51.4) 7189 (51.8) 893 (47.7) 732 (51.9) 7256 (51.8) 895 (47.6)
Males, n (%) 655 (48.6) 6681 (48.2) 978 (52.3) 678 (48.1) 6761 (48.2) 986 (52.4)
Year of birth, mean (SD) 1975.2 (21.6) 1974.4 (22.0) 1976.5 (18.5) 1974.0 (22.4) 1974.0 (22.4) 1976.4 (18.5)
Start of follow- up (cohort entry)
Age, mean (SD) 25.7 (20.7) 26.3 (21.0) 24.4 (17.4) 23.6 (20.7) 23.6 (20.7) 21.5 (16.7)
Year, mean (SD) 2001.1 (4.2) 2001.0 (4.1) 2001.1 (4.1) 1997.6 (7.3) 1997.6 (7.3) 1997.9 (7.2)
End of follow- up
Age, mean (SD) 38.3 (20.8) 39.7 (21.5) 37.8 (18.4) 38.6 (21.0) 40.0 (21.6) 37.7 (18.4)
Year, mean (SD) 2013.1 (2.8) 2013.7 (1.8) 2013.7 (1.5) 2012.2 (4.9) 2013.5 (2.7) 2013.6 (2.1)
Follow- up time, mean
(SD)
12.7 (4.9) 13.4 (4.5) 13.4 (4.4) 15.0 (7.5) 16.3 (7.4) 16.2 (7.2)
NF1, neurofibromatosis type 1.
analyses comparing patients with NF1 and their siblings without
NF1, each family was included as a frailty term. Results are
reported as point estimates and 95% CIs. Statistical significance
is defined at the two- sided 5% level. The R software (V.3.6.1)
and package survival (V.3.1–8) were used in the analyses.
RESULTS
We explored the incidence of diabetes in NF1 using a cohort
of 1410 Finnish patients with NF1 (table 1). Patients with
NF1 were compared with 14 017 control subjects individually
matched for age, sex and place of residence. Another comparison
cohort consisted of 1881 non- NF1 siblings of the patients with
NF1. We used register- based information on hospital visits and
stays and purchases of antidiabetic medication in 1998–2014 to
identify those with diabetes. The overall rate of diabetes among
patients with NF1 was 1.7 cases per 1000 person- years (95%
CI 1.1 to 2.4) while the rate was 5.1 (95% CI 4.8 to 5.4) in the
control cohort and 4.3 (95% CI 3.5 to 5.2) among the non- NF1
siblings. Consequently, the relative rate of diabetes was markedly
lower among patients with NF1 when compared with controls
(HR 0.34, 95% CI 0.23 to 0.49) or the siblings without NF1
(HR 0.35, 95% CI 0.23 to 0.55).
Type 1 diabetes
We next analysed T1D and T2D separately. Seven persons with
T1D were observed in the NF1 cohort during the follow- up
1998–2014, while there were 129 persons with T1D in the
control cohort and 19 siblings with T1D (tables 2 and 3). The
HR for T1D in NF1 was 0.58 (95% CI 0.27 to 1.25) when
compared with controls and 0.55 (95% CI 0.23 to 1.33) when
compared with the siblings without NF1 but the associations
were not statistically significant (table 2). One patient with NF1
and eight control persons were observed to have both T1D
and coeliac disease, yet no cases of autoimmune thyroiditis or
Addison's disease were seen among the patients with T1D. No
complication of T1D was particularly common among patients
with NF1 (online supplementary table S1).
Type 2 diabetes
During the follow- up 1998–2014, T2D was significantly less
common among patients with NF1 than among controls (HR
0.27, 95% CI 0.17 to 0.43) or siblings without NF1 (HR 0.28,
95% CI 0.16 to 0.47) and the HRs for T2D in NF1 were even
lower than for T1D (table 2). Although statistically significant
in both sexes, the reduction in the rate was more pronounced
among males (HR 0.14, 95% CI 0.06 to 0.35 in comparison NF1
versus controls) than among females (HR 0.40, 95% CI 0.23 to
0.68). Approximately 90%–98% of the persons had purchased
antidiabetic medication while only 33%–40% had a T2D- related
hospital encounter, indicating that the treatment was primarily
carried out in primary care setting (table 4). Moreover, 5/7
patients with NF1 and T2D- related hospital encounters had at
least one hospital encounter due to T2D without complications
(online supplementary table S2).
To assess whether the lower rate of T2D among patients
with NF1 was affected by premature mortality, the analysis was
restricted to ages<50 years. The decreased rate of T2D in NF1
persisted at ages<50 years with HR 0.35 (95% CI 0.16 to 0.79)
in comparison with controls and HR 0.30 (95% CI 0.13 to 0.71)
in comparison with the siblings without NF1. In this analysis,
T2D was observed in six patients with NF1, 184 control subjects
and 36 siblings without NF1. We had to restrict the time range of
the primary analysis to 1998–2014 because outpatient hospital
visits were only available since 1998 and medication purchases
since 1995. However, the relative risk estimates remained stable
when the study period was extended to 1987–2014 and only
hospital visits and stays were considered (NF1 versus controls:
7 and 338 persons with T2D, respectively; HR 0.26, 95% CI
0.12 to 0.54; NF1 versus siblings: 7 and 28 persons with T2D,
respectively; HR 0.20, 95% CI 0.07 to 0.53).
We next explored whether the reduced rate of T2D in NF1
was related to metabolic syndrome and overweight. The HR for
a diagnosis of obesity in NF1 was 0.94 (95% CI 0.59 to 1.48)
compared with the matched controls and 1.44 (95% CI 0.76 to
2.72) compared with the siblings without NF1. When the rate of
any of the features of metabolic syndrome other than hypergly-
caemia were considered, that is, obesity, primary hypertension
and disorders of lipoprotein metabolism and other lipidaemias,
their HR in NF1 was 1.37 (95% CI 1.14 to 1.66) compared with
controls and 1.66 (95% CI 1.25 to 2.21) compared with the
siblings without NF1. After exclusion of hypertension, which is
known to be more prevalent among patients with NF1, the HRs
were 1.04 (95% CI 0.76 to 1.43) and 1.20 (95% CI 0.77 to 1.87),
respectively. Thus, the other components of metabolic syndrome
do not seem to be less frequent among patients with NF1 despite
the decreased rate of T2D. Consequently, after adjusting the
on June 22, 2020 by guest. Protected by copyright.http://jmg.bmj.com/J Med Genet: first published as 10.1136/jmedgenet-2020-107062 on 22 June 2020. Downloaded from
4Kallionpää RA, etal. J Med Genet 2020;0:1–7. doi:10.1136/jmedgenet-2020-107062
Genotype- phenotype correlations
Table 2 Observed follow- up times, diabetes cases during the follow- up and the resulting HRs in NF1 as compared with controls without NF1 or siblings without NF1 in 1998–2014
Patients with NF1 vs controls Patients with NF1 vs siblings without NF1
All Females Males All Females Males
Type 1 diabetes HR (95% CI) 0.58 (0.27 to 1.25) 0.95 (0.38 to 2.36) 0.29 (0.07 to 1.2) 0.55 (0.23 to 1.33) 1.11 (0.35 to 3.52) 0.26 (0.06 to 1.16)
NF1 Number of diabetes cases 7 5 2 7 5 2
Follow- up (person- years) 17 005.9 8814.0 8191.9 17 005.9 8814.0 8191.9
Rate (n/1000 person- years) (95% CI) 0.41 (0.17 to 0.85) 0.57 (0.18 to 1.32) 0.24 (0.03 to 0.88) 0.41 (0.17 to 0.85) 0.57 (0.18 to 1.32) 0.24 (0.03 to 0.88)
Reference Number of diabetes cases 129 57 72 19 7 12
Follow- up (person- years) 184 594.4 97 064.6 87 529.8 24 928.7 12 138.2 12 790.5
Rate (n/1000 person- years) (95% CI) 0.7 (0.58 to 0.83) 0.59 (0.44 to 0.76) 0.82 (0.64 to 1.04) 0.76 (0.46 to 1.19) 0.58 (0.23 to 1.19) 0.94 (0.48 to 1.64)
Type 2 diabetes HR (95% CI) 0.27 (0.17 to 0.43) 0.40 (0.23 to 0.68) 0.14 (0.06 to 0.35) 0.28 (0.16 to 0.47) 0.37 (0.19 to 0.71) 0.15 (0.06 to 0.41)
NF1 Number of diabetes cases 19 14 5 19 14 5
Follow- up (person- years) 16 877.2 8714.6 8162.6 16 877.2 8714.6 8162.6
Rate (n/1000 person- years) (95% CI) 1.13 (0.68 to 1.76) 1.61 (0.88 to 2.70) 0.61 (0.20 to 1.43) 1.13 (0.68 to 1.76) 1.61 (0.88 to 2.70) 0.61 (0.20 to 1.43)
Reference Number of diabetes cases 779 402 377 83 42 41
Follow- up (person- years) 179 213.0 94 286.5 84 926.6 24 337.8 11 890.2 12 447.6
Rate (n/1000 person- years) (95% CI) 4.35 (4.05 to 4.66) 4.26 (3.86 to 4.70) 4.44 (4.00 to 4.91) 3.41 (2.72 to 4.23) 3.53 (2.55 to 4.77) 3.29 (2.36 to 4.47)
NF1, neurofibromatosis type 1.
T2D analyses for both history of obesity and dyslipidaemias, the
relative rate for T2D among patients with NF1 remained low
with HRs of 0.28 (95% CI 0.18 to 0.44) in comparison to the
control cohort and 0.27 (95% CI 0.16 to 0.46) in comparison to
the siblings without NF1.
DISCUSSION
Here, we have shown that NF1 is associated with a reduced rate
of T2D and a statistically non- significant reduction in the rate of
T1D. Since NF1 is caused by pathogenic variants of the NF1 gene
with full penetrance, the results suggest that germline aberrations
of the NF1 gene and the resulting deficiency of neurofibromin
protein confer protection against diabetes. NF1 syndrome is
rare with the prevalence <0.05%,23 and any individual patho-
genic variant of the NF1 gene is even rarer. Rare variants are
generally hard to detect in GWAS,12 and the case of NF1 is even
more difficult as the variants are spread throughout the gene.
Variants of NF1 are also associated with reduced fitness, further
decreasing their prevalence in older age groups.23 It is thus not
surprising that the association between the NF1 gene and T1D
or T2D has not been recognised in the previous studies looking
for genes associated with diabetes. Our approach of studying
a curated clinical cohort of patients who share aberrations of
the NF1 gene overcomes some of these limitations. The clinical
features of NF1 allow combining a variety of different variants
with similar functional consequences. Assuming a prevalence of
1/2000–3000 for NF1, observing as many persons with NF1
haploinsufficiency and diabetes as in our cohort would require
an unselected population of 2.8–4.2 million persons, which is
much more than in the largest diabetes- GWAS published so far.9
Only few genes affecting the risk of both T1D and T2D have
been identified despite their overlapping heritability patterns.20 21
Although the ages of onset and risk factors of T1D and T2D are
largely different, the two types of diabetes share pathological
processes. Both types may involve inflammatory loss of pancre-
atic β-cell mass, which is due to autoimmunity in T1D and toxic
metabolites in T2D.3 4 20 21 Moreover, T1D may present with
insulin resistance.20 21 The association between NF1 and T1D
reported here was not statistically significant, yet a reduced rate
of T1D- related hospitalisations among persons with NF1 was
also observed in a recent cohort study.32 Together, these results
suggest that NF1 aberrations may be a mechanism affecting both
types of diabetes. Unfortunately, our register- based data do not
allow dissecting whether the observed low HRs for T1D and
T2D in NF1 are related to, for example, inflammation or meta-
bolic activity. Previous data show NF1- associated alterations in
the immune system,41 and gene expression data from Schwann
cells suggest association of NF1 gene dosage with HLA class II
expression (NCBI GEO database, accession GSE32029), which
may speak in favour of an inflammation- mediated mechanism.
NF1 is known to affect especially the cells of the neural crest
during development. Interestingly, neural crest cells have been
observed to regulate the size of the β-cell population in a murine
model during embryonic development.42
Previous publications have suggested lower fasting blood
glucose levels, increased insulin sensitivity and higher resting
energy expenditure in NF1.34 35 43 Hypothetically, the volumi-
nous benign neurofibroma tumour mass could be one mecha-
nism increasing the energy consumption of patients with NF1.
The increased energy expenditure in NF1 is in concordance with
results from other Rasopathies, as high resting energy expendi-
ture has also been reported in patients with Costello syndrome,44
and a murine model of Noonan syndrome with multiple
on June 22, 2020 by guest. Protected by copyright.http://jmg.bmj.com/J Med Genet: first published as 10.1136/jmedgenet-2020-107062 on 22 June 2020. Downloaded from
5
Kallionpää RA, etal. J Med Genet 2020;0:1–7. doi:10.1136/jmedgenet-2020-107062
Genotype- phenotype correlations
Table 3 Descriptive characteristics of patients with T1D during follow- up 1998–2014
Patients with NF1 Controls Siblings without NF1
n 7 129 19
Sex
Female, n (%) 5 (71.4) 57 (44.2) 7 (36.8)
Male, n (%) 2 (28.6) 72 (55.8) 12 (63.2)
Age at first T1D- related encounter during follow- up, mean (SD) 31.05 (19.15) 30.17 (20.42) 33.16 (20.99)
Number of T1D- related encounters/patient, median (range) 25 (2 to 72) 21 (1 to 1195) 22 (1 to 62)
Number of insulin purchases (ATC A10A)/patient, median (range) 50 (2 to 115) 66 (1 to 152) 62 (15 to 118)
Special drug reimbursement for insulin, n (%) 7 (100) 129 (100) 19 (100)
ATC, Anatomical Therapeutic Chemical classification; NF1, neurofibromatosis type 1; T1D, type 1 diabetes.
Table 4 Descriptive characteristics of patients with T2D during
follow- up 1998–2014
Patients with
NF1 Controls
Siblings
without NF1
n 19 779 83
Sex
Female, n (%) 14 (73.7) 402 (51.6) 42 (50.6)
Male, n (%) 5 (26.3) 377 (48.4) 41 (49.4)
Age at first T2D- related
encounter or drug purchase
during follow- up, mean (SD)
56.49 (17.45) 58.64 (14.18) 49.48 (12.63)
T2D- related encounters
Patients, n (%) 7 (36.8) 312 (40.1) 27 (32.5)
Number/patient among
those with at least
one encounter, median
(range)
2 (1 to 8) 2 (1 to 190) 2 (1 to 504)
Purchases of insulins and
analogues (ATC A10A)
Patients, n (%) 6 (31.6) 201 (25.8) 17 (20.5)
Number/patient among
those with at least one
purchase, median (range)
15 (2 to 22) 17 (1 to 99) 10 (1 to 85)
Purchases of other
antidiabetic medication
(ATC A10B)
Patients, n (%) 17 (89.5) 723 (92.8) 76 (91.6)
Number/patient among
those with at least one
purchase, median (range)
10 (1 to 81) 18 (1 to 128) 15 (1 to 104)
Special drug reimbursement
for insulin, n (%)
10 (52.6) 554 (71.1) 56 (67.5)
ATC, Anatomical Therapeutic Chemical classification; NF1, neurofibromatosis type 1;
T2D, type 2 diabetes.
lentigines showed increased energy expenditure.45 Costello
and Noonan syndromes and Noonan syndrome with multiple
lentigines have been reported to associate with lower weight
and a decreased rate of obesity.44–46 These findings suggest that
the present observation of reduced risk of T2D in NF1 may be
related to Ras signalling pathway activity. Interestingly, NF1 of
the child has been found to increase birth weight,37 while low
birth weight is known to be associated with increased risk for
T2D.47 Therefore, the increased birth weight of children with
NF1 may be an underlying factor of the lower risk for T2D in
NF1, or both these observations may reflect the same biological
process. Our present analysis of obesity and other components
of metabolic syndrome do not suggest lower incidence of these
risk factors among patients with NF1, nor did we previously find
altered rates of gestational diabetes in NF1.37
In light of the previous studies,48 the association between the
NF1 gene and T2D highlights the role of the Ras pathway in the
pathogenesis of T2D. The increased phosphorylation of Erk1/2
has previously been linked to protection against T2D by gain-
of- function variants of MC4R,15 and the protection conferred
by AKNRD55 is linked to MAP3K1 (MEK kinase).7 The lean
phenotype observed in a mouse model of the Noonan syndrome
with multiple lentigines was observed to revert on MEK inhi-
bition.45 Moreover, a study using mouse embryonic fibroblasts
reported altered mitochondrial function attributed to ERK1/2
activity induced by NF1 loss.49 While the pharmacological inhi-
bition of the tumour suppressor protein neurofibromin is not a
feasible therapeutic approach, downstream components of the
Ras pathway may provide targets for interfering with the patho-
genesis of T2D.
The present findings are corroborated by previous studies,
which suggested a lower rate of diabetes in NF1.32 38–40 Our
total population- based approach reduces biases inherent in
hospital- based patient ascertainment and the individual curation
of NF1 diagnoses ensures reliability. Since we assessed the rates
of diabetes using patient- specific person times conditionally on
survival, the premature mortality associated with NF1 cannot
explain the findings. This conclusion is further supported by
the robustness of the decreased rate of T2D in NF1 also among
persons aged <50 years. Moreover, we were able to analyse T1D
and T2D separately to show that especially the rate of T2D is
reduced among patients with NF1. A limitation of the present
study is the lack of biospecimens that would allow elucidating
the reasons of the decreased rate of diabetes, and T2D in partic-
ular, seen in NF1. The use of register- based data also leaves some
uncertainty in the diabetes diagnoses as a large proportion of
T2D is known to be undiagnosed in the population. However,
patients with NF1 are not expected to have a higher rate of
missing diagnoses than persons without NF1, since NF1 and
its comorbidities are likely to increase the number of healthcare
contacts. Furthermore, the comprehensive data on medication
purchases should allow identification of all patients undergoing
treatment.
All the patients with NF1 included in the present study fulfilled
the diagnostic criteria of NF1 and thus carried a pathogenic
variant of the NF1 gene. Comparing patients with NF1 with both
the matched controls and siblings allows ruling out confounding
related to, for example, calendar time, age, parental education
or hereditary factors other than NF1. Thus, we conclude that the
lower rate of at least T2D and probably T1D in NF1 is indeed
associated with the NF1 gene.
Acknowledgements Turku University Hospital is a member of ERN GENTURIS.
Contributors Data acquisition: RAK, SP, JL, MP, JP. Data analysis: RAK, JL, KA.
Drafting the manuscript: RAK. All authors have made contributions to the conception
on June 22, 2020 by guest. Protected by copyright.http://jmg.bmj.com/J Med Genet: first published as 10.1136/jmedgenet-2020-107062 on 22 June 2020. Downloaded from
6Kallionpää RA, etal. J Med Genet 2020;0:1–7. doi:10.1136/jmedgenet-2020-107062
Genotype- phenotype correlations
and design of the study, taken part in data interpretation, revised the manuscript
and approved the submitted version.
Funding The study was funded with grants from the Finnish Cancer Foundation,
Turku University Hospital, University of Turku, Turku University Foundation, Orion
Research Foundation and Ida Montin Foundation.
Competing interests JP, consultant to AstraZeneca.
Patient consent for publication Not required.
Ethics approval The study was approved by the Ethics Committee of the Hospital
District of Southwest Finland and adhered to the principles of the Declaration of
Helsinki.
Provenance and peer review Not commissioned; externally peer reviewed.
Data availability statement Data may be obtained from a third party and are
not publicly available. Data are available for researchers with appropriate research
permissions from Finnish Institute for Health and Welfare, The Social Insurance
Institution of Finland and Finnish Population Register Centre.
Open access This is an open access article distributed in accordance with the
Creative Commons Attribution Non Commercial (CC BY- NC 4.0) license, which
permits others to distribute, remix, adapt, build upon this work non- commercially,
and license their derivative works on different terms, provided the original work is
properly cited, appropriate credit is given, any changes made indicated, and the use
is non- commercial. See:http:// creativecommons. org/ licenses/ by- nc/ 4. 0/.
ORCID iD
JuhaPeltonen http:// orcid. org/ 0000- 0002- 5732- 4167
REFERENCES
1 Zheng Y, Ley SH, Hu FB. Global aetiology and epidemiology of type 2 diabetes mellitus
and its complications. Nat Rev Endocrinol 2018;14:88–98.
2 Fletcher B, Gulanick M, Lamendola C. Risk factors for type 2 diabetes mellitus. J
Cardiovasc Nurs 2002;16:17–23.
3 Donath MY, Shoelson SE. Type 2 diabetes as an inflammatory disease. Nat Rev
Immunol 2011;11:98–107.
4 DiMeglio LA, Evans- Molina C, Oram RA. Type 1 diabetes. The Lancet
2018;391:2449–62.
5 Noble JA, Valdes AM. Genetics of the HLA region in the prediction of type 1 diabetes.
Curr Diab Rep 2011;11:533–42.
6 Jerram S, Leslie RD. The genetic architecture of type 1 diabetes. Genes 2017;8:209.
7 Morris AP, Voight BF, Teslovich TM, Ferreira T, Segrè AV, Steinthorsdottir V, Strawbridge
RJ, Khan H, Grallert H, Mahajan A, Prokopenko I, Kang HM, Dina C, Esko T, Fraser
RM, Kanoni S, Kumar A, Lagou V, Langenberg C, Luan Jian’an, Lindgren CM, Müller-
Nurasyid M, Pechlivanis S, Rayner NW, Scott LJ, Wiltshire S, Yengo L, Kinnunen L,
Rossin EJ, Raychaudhuri S, Johnson AD, Dimas AS, Loos RJF, Vedantam S, Chen H,
Florez JC, Fox C, Liu C- T, Rybin D, Couper DJ, Kao WHL, Li M, Cornelis MC, Kraft P,
Sun Q, van Dam RM, Stringham HM, Chines PS, Fischer K, Fontanillas P, Holmen OL,
Hunt SE, Jackson AU, Kong A, Lawrence R, Meyer J, Perry JRB, Platou CGP, Potter S,
Rehnberg E, Robertson N, Sivapalaratnam S, Stančáková A, Stirrups K, Thorleifsson
G, Tikkanen E, Wood AR, Almgren P, Atalay M, Benediktsson R, Bonnycastle LL, Burtt
N, Carey J, Charpentier G, Crenshaw AT, Doney ASF, Dorkhan M, Edkins S, Emilsson
V, Eury E, Forsen T, Gertow K, Gigante B, Grant GB, Groves CJ, Guiducci C, Herder C,
Hreidarsson AB, Hui J, James A, Jonsson A, Rathmann W, Klopp N, Kravic J, Krjutškov
K, Langford C, Leander K, Lindholm E, Lobbens S, Männistö S, Mirza G, Mühleisen TW,
Musk B, Parkin M, Rallidis L, Saramies J, Sennblad B, Shah S, Sigurðsson G, Silveira
A, Steinbach G, Thorand B, Trakalo J, Veglia F, Wennauer R, Winckler W, Zabaneh D,
Campbell H, van Duijn C, Uitterlinden AG, Hofman A, Sijbrands E, Abecasis GR, Owen
KR, Zeggini E, Trip MD, Forouhi NG, Syvänen A- C, Eriksson JG, Peltonen L, Nöthen
MM, Balkau B, Palmer CNA, Lyssenko V, Tuomi T, Isomaa B, Hunter DJ, Qi L, Shuldiner
AR, Roden M, Barroso I, Wilsgaard T, Beilby J, Hovingh K, Price JF, Wilson JF, Rauramaa
R, Lakka TA, Lind L, Dedoussis G, Njølstad I, Pedersen NL, Khaw K- T, Wareham NJ,
Keinanen- Kiukaanniemi SM, Saaristo TE, Korpi- Hyövälti E, Saltevo J, Laakso M,
Kuusisto J, Metspalu A, Collins FS, Mohlke KL, Bergman RN, Tuomilehto J, Boehm
BO, Gieger C, Hveem K, Cauchi S, Froguel P, Baldassarre D, Tremoli E, Humphries SE,
Saleheen D, Danesh J, Ingelsson E, Ripatti S, Salomaa V, Erbel R, Jöckel K- H, Moebus S,
Peters A, Illig T, de Faire U, Hamsten A, Morris AD, Donnelly PJ, Frayling TM, Hattersley
AT, Boerwinkle E, Melander O, Kathiresan S, Nilsson PM, Deloukas P, Thorsteinsdottir
U, Groop LC, Stefansson K, Hu F, Pankow JS, Dupuis J, Meigs JB, Altshuler D, Boehnke
M, McCarthy MI, Wellcome Trust Case Control Consortium, Meta- Analyses of Glucose
and Insulin- related traits Consortium (MAGIC) Investigators, Genetic Investigation
of ANthropometric Traits (GIANT) Consortium, Asian Genetic Epidemiology
Network–Type 2 Diabetes (AGEN- T2D) Consortium, South Asian Type 2 Diabetes
(SAT2D) Consortium, DIAbetes Genetics Replication And Meta- analysis (DIAGRAM)
Consortium. Large- Scale association analysis provides insights into the genetic
architecture and pathophysiology of type 2 diabetes. Nat Genet 2012;44:981–90.
8 Steinthorsdottir V, Thorleifsson G, Sulem P, Helgason H, Grarup N, Sigurdsson A,
Helgadottir HT, Johannsdottir H, Magnusson OT, Gudjonsson SA, Justesen JM, Harder
MN, Jørgensen ME, Christensen C, Brandslund I, Sandbæk A, Lauritzen T, Vestergaard
H, Linneberg A, Jørgensen T, Hansen T, Daneshpour MS, Fallah M- S, Hreidarsson AB,
Sigurdsson G, Azizi F, Benediktsson R, Masson G, Helgason A, Kong A, Gudbjartsson
DF, Pedersen O, Thorsteinsdottir U, Stefansson K. Identification of low- frequency and
rare sequence variants associated with elevated or reduced risk of type 2 diabetes.
Nat Genet 2014;46:294–8.
9 Mahajan A, Taliun D, Thurner M, Robertson NR, Torres JM, Rayner NW, Payne AJ,
Steinthorsdottir V, Scott RA, Grarup N, Cook JP, Schmidt EM, Wuttke M, Sarnowski C,
Mägi R, Nano J, Gieger C, Trompet S, Lecoeur C, Preuss MH, Prins BP, Guo X, Bielak
LF, Below JE, Bowden DW, Chambers JC, Kim YJ, Ng MCY, Petty LE, Sim X, Zhang
W, Bennett AJ, Bork- Jensen J, Brummett CM, Canouil M, Ec kardt K- U, Fischer K,
Kardia SLR, Kronenberg F, Läll K, Liu C- T, Locke AE, Luan Jian’an, Ntalla I, Nylander
V, Schönherr S, Schurmann C, Yengo L, Bottinger EP, Brandslund I, Christensen C,
Dedoussis G, Florez JC, Ford I, Franco OH, Frayling TM, Giedraitis V, Hackinger S,
Hattersley AT, Herder C, Ikram MA, Ingelsson M, Jørgensen ME, Jørgensen T, Kriebel
J, Kuusisto J, Ligthart S, Lindgren CM, Linneberg A, Lyssenko V, Mamakou V, Meitinger
T, Mohlke KL, Morris AD, Nadkarni G, Pankow JS, Peters A, Sattar N, Stančáková A,
Strauch K, Taylor KD, Thorand B, Thorleifsson G, Thorsteinsdottir U, Tuomilehto J, Witte
DR, Dupuis J, Peyser PA, Zeggini E, Loos RJF, Froguel P, Ingelsson E, Lind L, Groop
L, Laakso M, Collins FS, Jukema JW, Palmer CNA, Grallert H, Metspalu A, Dehghan
A, Köttgen A, Abecasis GR, Meigs JB, Rotter JI, Marchini J, Pedersen O, Hansen T,
Langenberg C, Wareham NJ, Stefansson K, Gloyn AL, Morris AP, Boehnke M, McCarthy
MI. Fine- Mapping type 2 diabetes loci to single- variant resolution using high- density
imputation and islet- specific epigenome maps. Nat Genet 2018;50:1505–13.
10 Xue A, Wu Y, Zhu Z, Zhang F, Kemper KE, Zheng Z, Yengo L, Lloyd- Jones LR, Sidorenko
J, Wu Y, McRae AF, Visscher PM, Zeng J, Yang J. Genome- Wide association analyses
identify 143 risk variants and putative regulatory mechanisms for type 2 diabetes. Nat
Commun 2018;9:2941.
11 Flannick J, Mercader JM, Fuchsberger C, Udler MS, Mahajan A, Wessel J, Teslovich
TM, Caulkins L, Koesterer R, Barajas- Olmos F, Blackwell TW, Boerwinkle E, Brody JA,
Centeno- Cruz F, Chen L, Chen S, Contreras- Cubas C, Córdova E, Correa A, Cortes
M, DeFronzo RA, Dolan L, Drews KL, Elliott A, Floyd JS, Gabriel S, Garay- Sevilla ME,
García- Ortiz H, Gross M, Han S, Heard- Costa NL, Jackson AU, Jørgensen ME, Kang
HM, Kelsey M, Kim B- J, Koistinen HA, Kuusisto J, Leader JB, Linneberg A, Liu C- T, Liu J,
Lyssenko V, Manning AK, Marcketta A, Malacara- Hernandez JM, Martínez- Hernández
A, Matsuo K, Mayer- Davis E, Mendoza- Caamal E, Mohlke KL, Morrison AC, Ndungu
A, Ng MCY, O’Dushlaine C, Payne AJ, Pihoker C, Post WS, Preuss M, Psaty BM, Vasan
RS, Rayner NW, Reiner AP, Revilla- Monsalve C, Robertson NR, Santoro N, Schurmann
C, So WY, Soberón X, Stringham HM, Strom TM, Tam CHT, Thameem F, Tomlinson B,
Torres JM, Tracy RP, van Dam RM, Vujkovic M, Wang S, Welch RP, Witte DR, Wong
T- Y, Atzmon G, Barzilai N, Blangero J, Bonnycastle LL, Bowden DW, Chambers JC,
Chan E, Cheng C- Y, Cho YS, Collins FS, de Vries PS, Duggirala R, Glaser B, Gonzalez
C, Gonzalez ME, Groop L, Kooner JS, Kwak SH, Laakso M, Lehman DM, Nilsson P,
Spector TD, Tai ES, Tuomi T, Tuomilehto J, Wilson JG, Aguilar- Salinas CA, Bottinger E,
Burke B, Carey DJ, Chan JCN, Dupuis J, Frossard P, Heckbert SR, Hwang MY, Kim YJ,
Kirchner HL, Lee J- Y, Lee J, Loos RJF, Ma RCW, Morris AD, O’Donnell CJ, Palmer CNA,
Pankow J, Park KS, Rasheed A, Saleheen D, Sim X, Small KS, Teo YY, Haiman C, Hanis
CL, Henderson BE, Orozco L, Tusié-Luna T, Dewey FE, Baras A, Gieger C, Meitinger T,
Strauch K, Lange L, Grarup N, Hansen T, Pedersen O, Zeitler P, Dabelea D, Abecasis G,
Bell GI, Cox NJ, Seielstad M, Sladek R, Meigs JB, Rich SS, Rotter JI, Altshuler D, Burtt
NP, Scott LJ, Morris AP, Florez JC, McCarthy MI, Boehnke M, Broad Genomics Platform,
DiscovEHR Collaboration, CHARGE, LuCamp, ProDiGY, GoT2D, ESP, SIGMA- T2D, T2D-
GENES, AMP- T2D- GENES. Exome sequencing of 20,791 cases of type 2 diabetes and
24,440 controls. Nature 2019;570:71–6.
12 Manolio TA, Collins FS, Cox NJ, Goldstein DB, Hindorff LA, Hunter DJ, McCarthy MI,
Ramos EM, Cardon LR, Chakravarti A, Cho JH, Guttmacher AE, Kong A, Kruglyak L,
Mardis E, Rotimi CN, Slatkin M, Valle D, Whittemore AS, Boehnke M, Clark AG, Eichler
EE, Gibson G, Haines JL, Mackay TFC, McCarroll SA, Visscher PM. Finding the missing
heritability of complex diseases. Nature 2009;461:747–53.
13 Flannick J, Thorleifsson G, Beer NL, Jacobs SBR, Grarup N, Burtt NP, Mahajan A,
Fuchsberger C, Atzmon G, Benediktsson R, Blangero J, Bowden DW, Brandslund I,
Brosnan J, Burslem F, Chambers J, Cho YS, Christensen C, Douglas DA, Duggirala R,
Dymek Z, Farjoun Y, Fennell T, Fontanillas P, Forsén T, Gabriel S, Glaser B, Gudbjartsson
DF, Hanis C, Hansen T, Hreidarsson AB, Hveem K, Ingelsson E, Isomaa B, Johansson S,
Jørgensen T, Jørgensen ME, Kathiresan S, Kong A, Kooner J, Kravic J, Laakso M, Lee
J- Y, Lind L, Lindgren CM, Linneberg A, Masson G, Meitinger T, Mohlke KL, Molven A,
Morris AP, Potluri S, Rauramaa R, Ribel- Madsen R, Richard A- M, Rolph T, Salomaa
V, Segrè AV, Skärstrand H, Steinthorsdottir V, Stringham HM, Sulem P, Tai ES, Teo
YY, Teslovich T, Thorsteinsdottir U, Trimmer JK, Tuomi T, Tuomilehto J, Vaziri- Sani F,
Voight BF, Wilson JG, Boehnke M, McCarthy MI, Njølstad PR, Pedersen O, Groop L,
Cox DR, Stefansson K, Altshuler D, Go- T2D Consortium, T2D- GENES Consortium.
Loss- Of- Function mutations in Slc30a8 protect against type 2 diabetes. Nat Genet
2014;46:357–63.
14 Gudmundsson J, Sulem P, Steinthorsdottir V, Bergthorsson JT, Thorleifsson G,
Manolescu A, Rafnar T, Gudbjartsson D, Agnarsson BA, Baker A, Sigurdsson A,
Benediktsdottir KR, Jakobsdottir M, Blondal T, Stacey SN, Helgason A, Gunnarsdottir
S, Olafsdottir A, Kristinsson KT, Birgisdottir B, Ghosh S, Thorlacius S, Magnusdottir D,
Stefansdottir G, Kristjansson K, Bagger Y, Wilensky RL, Reilly MP, Morris AD, Kimber
on June 22, 2020 by guest. Protected by copyright.http://jmg.bmj.com/J Med Genet: first published as 10.1136/jmedgenet-2020-107062 on 22 June 2020. Downloaded from
7
Kallionpää RA, etal. J Med Genet 2020;0:1–7. doi:10.1136/jmedgenet-2020-107062
Genotype- phenotype correlations
CH, Adeyemo A, Chen Y, Zhou J, So W- Y, Tong PCY, Ng MCY, Hansen T, Andersen G,
Borch- Johnsen K, Jorgensen T, Tres A, Fuertes F, Ruiz- Echarri M, Asin L, Saez B, van
Boven E, Klaver S, Swinkels DW, Aben KK, Graif T, Cashy J, Suarez BK, van Vierssen
Trip O, Frigge ML, Ober C, Hofker MH, Wijmenga C, Christiansen C, Rader DJ, Palmer
CNA, Rotimi C, Chan JCN, Pedersen O, Sigurdsson G, Benediktsson R, Jonsson E,
Einarsson GV, Mayordomo JI, Catalona WJ, Kiemeney LA, Barkardottir RB, Gulcher
JR, Thorsteinsdottir U, Kong A, Stefansson K. Two variants on chromosome 17 confer
prostate cancer risk, and the one in Tcf2 protects against type 2 diabetes. Nat Genet
2007;39:977–83.
15 Lotta LA, Mokrosiński J, Mendes de Oliveira E, Li C, Sharp SJ, Luan Jian’an, Brouwers
B, Ayinampudi V, Bowker N, Kerrison N, Kaimakis V, Hoult D, Stewart ID, Wheeler
E, Day FR, Perry JRB, Langenberg C, Wareham NJ, Farooqi IS. Human gain- of-
function MC4R variants show signaling bias and protect against obesity. Cell
2019;177:597–607.
16 Deeb SS, Fajas L, Nemoto M, Pihlajamäki J, Mykkänen L, Kuusisto J, Laakso M,
Fujimoto W, Auwerx J. A Pro12Ala substitution in PPARγ2 associated with decreased
receptor activity, lower body mass index and improved insulin sensitivity. Nat Genet
1998;20:284–7.
17 Pociot F, Lernmark Åke. Genetic risk factors for type 1 diabetes. The Lancet
2016;387:2331–9.
18 Pociot F. Type 1 diabetes genome- wide association studies: not to be lost in
translation. Clin Trans Immunol 2017;6:e162.
19 Krischer JP, Liu X, Lernmark Åke, Hagopian WA, Rewers MJ, She J- X, Toppari J, Ziegler
A- G, Akolkar B. The influence of type 1 diabetes genetic susceptibility regions, age,
sex, and family history on the progression from multiple autoantibodies to type 1
diabetes: a TEDDY study report. Diabetes 2017;66:3122–9.
20 Tuomi T. Type 1 and type 2 diabetes: what do they have in common? Diabetes
2005;54:S40–5.
21 Basile KJ, Guy VC, Schwartz S, Grant SFA. Overlap of genetic susceptibility to type 1
diabetes, type 2 diabetes, and latent autoimmune diabetes in adults. Curr Diab Rep
2014;14:550.
22 Andersen MK, Sterner M, Forsén T, Käräjämäki A, Rolandsson O, Forsblom C, Groop
P- H, Lahti K, Nilsson PM, Groop L, Tuomi T. Type 2 diabetes susceptibility gene variants
predispose to adult- onset autoimmune diabetes. Diabetologia 2014;57:1859–68.
23 Kallionpää RA, Uusitalo E, Leppävirta J, Pöyhönen M, Peltonen S, Peltonen J.
Prevalence of neurofibromatosis type 1 in the Finnish population. Genet Med
2018;20:1082–6.
24 National Institutes of Health. National Institutes of health consensus development
conference statement: neurofibromatosis. Arch Neurol 1988;45:575–8.
25 Xu G, O’Connell P, Viskochil D, Cawthon R, Robertson M, Culver M, Dunn D, Stevens J,
Gesteland R, White R, Weiss R. The neurofibromatosis type 1 gene encodes a protein
related to gap. Cell 1990;62:599–608.
26 Koczkowska M, Callens T, Chen Y, Gomes A, Hicks AD, Sharp A, Johns E, Uhas KA,
Armstrong L, Bosanko KA, Babovic- Vuksanovic D, Baker L, Basel DG, Bengala M,
Bennett JT, Chambers C, Clarkson LK, Clementi M, Cortés FM, Cunningham M,
D’Agostino MD, Delatycki MB, Digilio MC, Dosa L, Esposito S, Fox S, Freckmann M- L,
Fauth C, Giugliano T, Giustini S, Goetsch A, Goldberg Y, Greenwood RS, Griffis C, Gripp
KW, Gupta P, Haan E, Hachen RK, Haygarth TL, Hernández- Chico C, Hodge K, Hopkin
RJ, Hudgins L, Janssens S, Keller K, Kelly- Mancuso G, Kochhar A, Korf BR, Lewis AM,
Liebelt J, Lichty A, Listernick RH, Lyons MJ, Maystadt I, Martinez Ojeda M, McDougall
C, McGregor LK, Melis D, Mendelsohn N, Nowaczyk MJM, Ortenberg J, Panzer K,
Pappas JG, Pierpont ME, Piluso G, Pinna V, Pivnick EK, Pond DA, Powell CM, Rogers
C, Ruhrman Shahar N, Rutledge SL, Saletti V, Sandaradura SA, Santoro C, Schatz UA,
Schreiber A, Scott DA, Sellars EA, Sheffer R, Siqveland E, Slopis JM, Smith R, Spalice A,
Stockton DW, Streff H, Theos A, Tomlinson GE, Tran G, Trapane PL, Trevisson E, Ullrich
NJ, Van den Ende J, Schrier Vergano SA, Wallace SE, Wangler MF, Weaver DD, Yohay
KH, Zackai E, Zonana J, Zurcher V, Claes KBM, Eoli M, Martin Y, Wimmer K, De Luca A,
Legius E, Messiaen LM. Clinical spectrum of individuals with pathogenic NF1 missense
variants affecting p.Met1149, p.Arg1276, and p.Lys1423: genotype- phenotype study
in neurofibromatosis type 1. Hum Mutat 2020;41:299–315.
27 Zöller M, Rembeck B, Akesson HO, Angervall L, expectancy L. Mortality and prognostic
factors in neurofibromatosis type 1. A twelve- year follow- up of an epidemiological
study in Göteborg, Sweden. Acta Derm Venereol 1995;75:136–40.
28 Evans DGR, O’Hara C, Wilding A, Ingham SL, Howard E, Dawson J, Moran A, Scott-
Kitching V, Holt F, Huson SM. Mortality in neurofibromatosis 1: in North West England:
an assessment of actuarial survival in a region of the UK since 1989. Eur J Hum Genet
2011;19:1187–91.
29 Uusitalo E, Leppävirta J, Koffert A, Suominen S, Vahtera J, Vahlberg T, Pöyhönen
M, Peltonen J, Peltonen S. Incidence and mortality of neurofibromatosis: a total
population study in Finland. J Invest Dermatol 2015;135:904–6.
30 Uusitalo E, Rantanen M, Kallionpää RA, Pöyhönen M, Leppävirta J, Ylä-Outinen
H, Riccardi VM, Pukkala E, Pitkäniemi J, Peltonen S, Peltonen J. Distinctive cancer
associations in patients with neurofibromatosis type 1. JCO 2016;34:1978–86.
31 Gutmann DH, Ferner RE, Listernick RH, Korf BR, Wolters PL, Johnson KJ.
Neurofibromatosis type 1. Nat Rev Dis Primers 2017;3.
32 Kenborg L, Duun- Henriksen AK, Dalton SO, Bidstrup PE, Doser K, Rugbjerg K,
Pedersen C, Krøyer A, Johansen C, Andersen KK, Østergaard JR, Hove H, Sørensen SA,
Riccardi VM, Mulvihill JJ, Winther JF. Multisystem burden of neurofibromatosis 1 in
Denmark: registry- and population- based rates of hospitalizations over the life span.
Genet Med;26.
33 de Souza MLR, Jansen AK, Martins AS, Rodrigues LOC, de Rezende NA.
Nutrient intake in neurofibromatosis type 1: A cross- sectional study. Nutrition
2015;31:858–62.
34 Martins AS, Jansen AK, Rodrigues LOC, Matos CM, Souza MLR, de Souza JF, Diniz
MdeFHS, Barreto SM, Diniz LM, de Rezende NA, Riccardi VM. Lower fasting blood
glucose in neurofibromatosis type 1. Endocr Connect 2016;5:28–33.
35 Martins AS, Jansen AK, Rodrigues LOC, Matos CM, Souza MLR, Miranda DM, Rezende
NAde. Increased insulin sensitivity in individuals with neurofibromatosis type 1. Arch
Endocrinol Metab 2018;62:41–6.
36 Terry AR, Barker FG, Leffert L, Bateman BT, Souter I, Plotkin SR. Neurofibromatosis
type 1 and pregnancy complications: a population- based study. Am J Obstet Gynecol
2013;209:46.e1–46.e8.
37 Leppävirta J, Kallionpää RA, Uusitalo E, Vahlberg T, Pöyhönen M, Peltonen J, Peltonen
S. Neurofibromatosis type 1 of the child increases birth weight. Am J Med Genet A
2019;179:ajmg.a.61161.
38 Madubata CC, Olsen MA, Stwalley DL, Gutmann DH, Johnson KJ. Neurofibromatosis
type 1 and chronic neurological conditions in the United States: an administrative
claims analysis. Genet Med 2015;17:36–42.
39 Rasmussen SA, Yang Q, Friedman JM. Mortality in neurofibromatosis 1: an analysis
using U.S. death certificates. Am J Hum Genet 2001;68:1110–8.
40 Masocco M, Kodra Y, Vichi M, Conti S, Kanieff M, Pace M, Frova L, Taruscio D.
Mortality associated with neurofibromatosis type 1: a study based on Italian death
certificates (1995-2006). Orphanet J Rare Dis 2011;6:11.
41 Karmakar S, Reilly KM. The role of the immune system in neurofibromatosis type
1- associated nervous system tumors. CNS Oncol 2017;6:45–60.
42 Nekrep N, Wang J, Miyatsuka T, German MS. Signals from the neural crest regulate
beta- cell mass in the pancreas. Development 2008;135:2151–60.
43 de Souza MLR, Jansen AK, Rodrigues LOC. Vilela DL de S, Kakehasi am, Martins as, de
Souza JF, de Rezende Na. increased resting metabolism in neurofibromatosis type 1.
Clin Nutr ESPEN 2019;32:44–9.
44 Leoni C, Onesimo R, Giorgio V, Diamanti A, Giorgio D, Martini L, Rossodivita A,
Tartaglia M, Zampino G. Understanding growth failure in Costello syndrome:
increased resting energy expenditure. J Pediatr 2016;170:322–4.
45 Tajan M, Batut A, Cadoudal T, Deleruyelle S, Le Gonidec S, Saint Laurent
C, Vomscheid M, Wanecq E, Tréguer K, De Rocca Serra- Nédélec A, Vinel C,
Marques M- A, Pozzo J, Kunduzova O, Salles J- P, Tauber M, Raynal P, Cavé H,
Edouard T, Valet P, Yart A. Leopard syndrome- associated SHP2 mutation confers
leanness and protection from diet- induced obesity. Proc Natl Acad Sci U S A
2014;111:E4494–503.
46 Binder G, Grathwol S, von Loeper K, Blumenstock G, Kaulitz R, Freiberg C, Webel
M, Lissewski C, Zenker M, Paul T. Health and quality of life in adults with Noonan
syndrome. J Pediatr 2012;161:501–5.
47 Knop MR, Geng Ting-Ting, Gorny AW, Ding R, Li C, Ley SH, Huang T. Birth Weight and
Risk of Type 2 Diabetes Mellitus, Cardiovascular Disease, and Hypertension in Adults:
A Meta-Analysis of 7 646 267 Participants From 135 Studies. J Am Heart Assoc
2018;7:e008870.
48 Dard L, Bellance N, Lacombe D, Rossignol R. Ras signalling in energy metabolism and
rare human diseases. Biochim Biophys Acta Bioenerg 2018;1859:845–67.
49 Masgras I, Ciscato F, Brunati AM, Tibaldi E, Indraccolo S, Curtarello M, Chiara F,
Cannino G, Papaleo E, Lambrughi M, Guzzo G, Gambalunga A, Pizzi M, Guzzardo
V, Rugge M, Vuljan SE, Calabrese F, Bernardi P, Rasola A. Absence of neurofibromin
induces an oncogenic metabolic switch via mitochondrial ERK- mediated
phosphorylation of the chaperone TRAP1. Cell Rep 2017;18:659–72.
on June 22, 2020 by guest. Protected by copyright.http://jmg.bmj.com/J Med Genet: first published as 10.1136/jmedgenet-2020-107062 on 22 June 2020. Downloaded from
... One clear exception to this rule is the highly conserved anthropometric and metabolic phenotype featuring short stature, low body mass index (BMI), and protection from diabetes, which has been described in multiple NF1 cohorts and registry studies (3)(4)(5)(6). For example, a recently published registry study comparing 1,400 persons with NF1 to 14,000 matched controls revealed a 75% reduction in diabetes prevalence in persons with NF1 (7). Strikingly, the NF1 cohort was similarly protected from diabetes when compared with non-NF1 siblings suggesting that NF1 mutations, but not other genetic or environmental factors, convey this metabolic phenotype. ...
... When examining individual diagnoses within this category, the prevalence of diabetes mellitus type 1 was reduced 2.5-fold while diabetes mellitus type 2 prevenance was similar to the general population. A more recent Finnish registry study explored this potential relationship with a specific focus on diabetes mellitus (7). In comparison to non-NF1 siblings and non-related matched controls, the hazard ratio for diabetes mellitus type 1 in the NF1 cohort was 0.55 and 0.58, respectively. ...
Preprint
Full-text available
Persons with neurofibromatosis type 1 (NF1), a tumor predisposition syndrome, are largely protected from diabetes and exhibit evidence of enhanced glucose metabolism, which is replicated in mice harboring Nf1 mutations. A hallmark of NF1-associated neurofibromas and sarcomas is the high density of inflammatory macrophages and targeting macrophages appears efficacious in models of NF1. Inflammatory macrophages rely on glycolysis to rapidly generate ATP; thus, identifying whether neurofibromin, the protein encoded by the NF1 gene, controls glucose uptake and/or glycolysis in macrophages is therapeutically compelling. Using neurofibromin-deficient macrophages and macrophage-specific Nf1 knockout mice, we demonstrate that neurofibromin complexes with glucose transporter 1 (GLUT1) to restrain its activity and that loss of neurofibromin permits Akt2 to facilitate GLUT1 translocation to the membrane in macrophages. In turn, glucose internalization and glycolysis are highly up regulated and provoke putative reparative (M2) macrophages to undergo inflammatory phenotypic switch. Inflammatory M1 macrophages and inflammatory-like M2 macrophages invest the perivascular stroma of tumors and induce pathologic angiogenesis in mice harboring macrophage-specific Nf1 deletion. These studies identify a clear mechanism for the enhanced glycolysis and low risk for diabetes observed in persons with NF1 and provide a novel therapeutic target for manifestations of NF1.
... They also found a decreased rate of type 1 diabetes, which was not statistically significant (HR 0.58, 95% CI 0.27-1.25). 36 The authors suggested that the lower risk of ...
... Although the mechanism is not established, regulation of the Ras signaling pathway might play a role in the pathogenesis of type 2 diabetes. 36 We examined potential sex differences in NF1-associated endocrine morbidity but found only the well-known sex differences in thyroid disease 37 and osteoporosis. 38 The same differences were seen in the data on endocrine-associated prescription and partly in the data on endocrine surgery, which showed that women with NF1 had a higher risk of surgery on the thyroid but not for fractures. ...
Article
Objective: Previous studies have found that neurofibromatosis 1 (NF1) is associated with an increased risk for endocrine disorders, but no comprehensive overview of the risk for specific endocrine disorders has been published. We assessed endocrine morbidity in individuals with NF1 from information on hospital admissions, surgery for endocrine disorders, and relevant medication. Design: A nationwide population registry-based cohort study. Methods: We identified 2467 individuals with NF1 diagnosed between 1977-2013 from the Danish National Patient Register and the RAREDIS database and 20 132 randomly sampled age- and sex-matched population comparisons. Information on endocrine diseases was identified using registrations of discharge diagnoses, surgery, and medication prescriptions. The rates of endocrine disorders in individuals with NF1 were compared with those in the comparison cohort in Cox proportional hazard models. Results: Individuals with NF1 had a higher rate than the comparison group of any endocrine discharge diagnosis (hazard ratio (HR) 1.72, 95% confidence interval (CI): 1.58-1.87), endocrine-related surgery (2.03, 1.39-2.96), and prescribed medications (1.32, 1.23-1.42). Increased HRs were observed for diseases and surgical operations of several glands, including pheochromocytoma, and for osteoporosis, and osteoporotic fractures. Decreased rates were observed with drugs for type 2 diabetes. Women with NF1 had higher HRs for surgery of the ovaries, uterus, and sterilization, but lower rates of surgeries of cervix and prescriptions for birth control pills. Conclusions: NF1 is associated with a variety of endocrine disorders, surgery, and medication related to endocrine disease. Awareness of endocrine morbidity is important in the clinical follow-up of individuals with NF1.
... NF1 therapies blocking RAS signaling pathways, such as the MEKi Selumetinib, have shown partial shrinkage of NF1 plexiform tumors [176] . However, as neurofibromin's regulatory role is not limited to one signaling pathway, any therapy targeting a specific downstream effector will likely result in only partial NF1-phenotype resolution, as opposed to therapies that restore neurofibromin levels and thus correct all dysregulated pathways. ...
Article
Full-text available
Neurofibromatosis type 1 (NF1) is a genetic disorder with a wide range of manifestations and severity. Currently, the few available NF1 treatments target specific manifestations, with no available therapies targeted to correct the underlying driver of all NF1 manifestations. Evidence supports that haploinsufficiency in NF1 caused by a decreased amount of wild-type (WT) neurofibromin in all Nf1+/- cells directly causes or facilitates a range of NF1 manifestations. Consequently, NF1 haploinsufficiency correction therapy (NF1-HCT) represents a potentially effective approach to treat some NF1 manifestations. NF1-HCT would normalize the level of WT neurofibromin in all NF1-haploinsufficient cells, including those integral to the NF1 phenotype such as Schwann cells (SCs), melanocytes, neurons, bone cells, and cells of the tumor microenvironment. This would correct altered cellular signaling pathways and, in turn, restore normal function to cells with a retained WT allele. NF1-HCT will not restore WT neurofibromin in NF1-/- cells; however, by restoring function in the surrounding Nf1+/- microenvironment cells, NF1-HCT is predicted to have a beneficial effect on NF1-/- cells. NF1-HCT is expected to have a clinical effect in some NF1 manifestations, as follows: (i) prevention, or delay of onset, of potential manifestations; and (ii) reversal, or halting/slowing progression, of established manifestations. This review describes the rationale for NF1-HCT, including specific NF1 considerations (e.g., NF1 clinical phenotype, neurofibromin function/regulation, NF1 mutational spectrum, genotype-phenotype correlation, and the impact of haploinsufficiency in NF1), HCT in other haploinsufficient diseases, potential NF1-HCT drug treatment strategies, and the potential advantages/challenges of NF1-HCT.
... This could be caused by a general imbalance in the levels of several hormones, including lower levels of leptin and visfatin and higher adiponectin in NF1 patients with respect to control subjects. It remains to be explained the mechanistic connection between heterozygous loss of neurofibromin and these metabolic changes, confirmed in a large cohort of patients [11]. Moreover, NF1 individuals show reduced cerebral glucose metabolism, specifically in the thalamus [12]. ...
Chapter
Full-text available
Rewiring cellular metabolism is a key hallmark of cancer. Multiple evidences show that alterations in various metabolic circuits directly contribute to the tumorigenic process at different levels (e.g. cancer initiation, metastasis, resistance). However, the characterization of the metabolic profile of Neurofibromatosis type 1 (NF1)-related neoplastic cells has been only partially elucidated both in benign neurofibromas and in malignant peripheral nerve sheath tumors (MPNSTs). Here, we illustrate the state of the art on the knowledge of the metabolic features of tumors related to NF1 and discuss their potential implications for the development of novel therapeutic perspectives.
Article
Full-text available
Neurofibromatosis type 1 (OMIM 162200) affects ~ 1 in 3,000 individuals worldwide and is one of the most common monogenetic neurogenetic disorders that impacts brain function. The disorder affects various organ systems, including the central nervous system, resulting in a spectrum of clinical manifestations. Significant progress has been made in understanding the disorder’s pathophysiology, yet gaps persist in understanding how the complex signaling and systemic interactions affect the disorder. Two features of the disorder are alterations in neuronal function and metabolism, and emerging evidence suggests a potential relationship between them. This review summarizes neurofibromatosis type 1 features and recent research findings on disease mechanisms, with an emphasis on neuronal and metabolic features.
Article
Full-text available
Alternative splicing contributes to cancer development. Indeed, splicing analysis of cancer genome-wide association study (GWAS) risk variants has revealed likely causal variants. To systematically assess GWAS variants for splicing effects, we developed a prioritization workflow using a combination of splicing prediction tools, alternative transcript isoforms, and splicing quantitative trait locus (sQTL) annotations. Application of this workflow to candidate causal variants from 16 endometrial cancer GWAS risk loci highlighted single-nucleotide polymorphisms (SNPs) that were predicted to upregulate alternative transcripts. For two variants, sQTL data supported the predicted impact on splicing. At the 17q11.2 locus, the protective allele for rs7502834 was associated with increased splicing of an exon in a NF1 alternative transcript encoding a truncated protein in adipose tissue and is consistent with an endometrial cancer transcriptome-wide association study (TWAS) finding in adipose tissue. Notably, NF1 haploinsufficiency is protective for obesity, a well-established risk factor for endometrial cancer. At the 17q21.32 locus, the rs2278868 risk allele was predicted to upregulate a SKAP1 transcript that is subject to nonsense-mediated decay, concordant with a corresponding sQTL in lymphocytes. This is consistent with a TWAS finding that indicates decreased SKAP1 expression in blood increases endometrial cancer risk. As SKAP1 is involved in T cell immune responses, decreased SKAP1 expression may impact endometrial tumor immunosurveillance. In summary, our analysis has identified potentially causal endometrial cancer GWAS risk variants with plausible biological mechanisms and provides a splicing annotation workflow to aid interpretation of other GWAS datasets.
Article
Full-text available
We report 281 individuals carrying a pathogenic recurrent NF1 missense variants at p.Met1149, p.Arg1276 or p.Lys1423, representing three non‐truncating NF1 hotspots in the University of Alabama at Birmingham (UAB) cohort, together identified in 1.8% of unrelated NF1 individuals. About 25% (95% CI, 20.5%‐31.2%) of individuals heterozygous for a pathogenic NF1 p.Met1149, p.Arg1276 or p.Lys1423 missense variant had a Noonan‐like phenotype, which is significantly more compared to the “classic” NF1‐affected cohorts (all P<0.0001). Furthermore, p.Arg1276 and p.Lys1423 pathogenic missense variants were associated with a high prevalence of cardiovascular abnormalities, including pulmonic stenosis (all P<0.0001), while p.Arg1276 variants had a high prevalence of symptomatic spinal neurofibromas (P<0.0001) compared with “classic” NF1‐affected cohorts. However, p.Met1149‐positive individuals had a mild phenotype, characterized mainly by pigmentary manifestations without externally visible plexiform neurofibromas, symptomatic spinal neurofibromas or symptomatic optic pathway gliomas. As up to 0.4% of unrelated individuals in the UAB cohort carries a p.Met1149 missense variant, this finding will contribute to more accurate stratification of a significant number of NF1 individuals. Although clinically relevant genotype‐phenotype correlations are rare in NF1, each affecting only a small percentage of individuals, together they impact counseling and management of a significant number of the NF1 population. This article is protected by copyright. All rights reserved.
Article
Full-text available
Protein-coding genetic variants that strongly affect disease risk can yield relevant clues to disease pathogenesis. Here we report exome-sequencing analyses of 20,791 individuals with type 2 diabetes (T2D) and 24,440 non-diabetic control participants from 5 ancestries. We identify gene-level associations of rare variants (with minor allele frequencies of less than 0.5%) in 4 genes at exome-wide significance, including a series of more than 30 SLC30A8 alleles that conveys protection against T2D, and in 12 gene sets, including those corresponding to T2D drug targets (P = 6.1 × 10⁻³) and candidate genes from knockout mice (P = 5.2 × 10⁻³). Within our study, the strongest T2D gene-level signals for rare variants explain at most 25% of the heritability of the strongest common single-variant signals, and the gene-level effect sizes of the rare variants that we observed in established T2D drug targets will require 75,000–185,000 sequenced cases to achieve exome-wide significance. We propose a method to interpret these modest rare-variant associations and to incorporate these associations into future target or gene prioritization efforts.
Article
Full-text available
Neurofibromatosis type 1 (NF1) is associated with reduced adult height, but there are no cohort studies on birth size. This retrospective study includes a cohort of 1,410 persons with NF1 and a matched comparison cohort from the general population. Figures for birth size were retrieved from the administrative registers of Finland, and the data were converted to standard deviation scores (SDS), defined as standard deviation difference to the reference population. The birth weight among infants with NF1 was higher than among infants without the disorder (adjusted mean difference [95% confidence interval]: 0.53 SDS [0.19–0.87]), as was the head circumference at birth (0.58 SDS [0.26–0.90]). The birth length of the NF1 infants did not differ significantly from the comparison cohort. The birth weight in the group consisting of NF1 and non‐NF1 infants of NF1 mothers was lower than among infants of mothers in the comparison cohort (−0.28 SDS [−0.51 to −0.06]), as was the birth length (−0.22 SDS [−0.45 to 0.00]). In conclusion, the birth weight and head circumference of persons with NF1 are significantly higher than those of persons without the disorder. NF1 of the mother reduces birth weight and birth length of the infant.
Article
Full-text available
Background Low birth weight has been associated with increased risk of type 2 diabetes mellitus, cardiovascular disease, and hypertension, but the risk at high birth weight levels remains uncertain. This systematic review and meta‐analysis aimed to clarify the shape of associations between birth weight and aforementioned diseases in adults and assessed sex‐specific risks. Methods and Results We systematically searched PubMed, EMBASE, and Web of Science for studies published between 1980 and October 2016. Studies of birth weight and type 2 diabetes mellitus (T2DM), cardiovascular disease (CVD), and hypertension were included. Random‐effects models were used to derive the summary relative risks and corresponding 95% confidence intervals.We identified 49 studies with 4 053 367 participants assessing the association between birth weight and T2DM, 33 studies with 5 949 477 participants for CVD, and 53 studies with 4 335 149 participants for hypertension and high blood pressure. Sex‐specific binary analyses showed that only females had an increased risk of T2DM and CVD at the upper tail of the birth weight distribution. While categorical analyses of 6 birth weight groups and dose‐response analyses showed J‐shaped associations of birth weight with T2DM and CVD, the association was inverse with hypertension. The lowest risks for T2DM, CVD, and hypertension were observed at 3.5 to 4.0, 4.0 to 4.5, and 4.0 to 4.5 kg, respectively. Conclusions These findings indicate that birth weight is associated with risk of T2DM and CVD in a J‐shaped manner and that this is more pronounced among females.
Article
Full-text available
We expanded GWAS discovery for type 2 diabetes (T2D) by combining data from 898,130 European-descent individuals (9% cases), after imputation to high-density reference panels. With these data, we (i) extend the inventory of T2D-risk variants (243 loci, 135 newly implicated in T2D predisposition, comprising 403 distinct association signals); (ii) enrich discovery of lower-frequency risk alleles (80 index variants with minor allele frequency <5%, 14 with estimated allelic odds ratio >2); (iii) substantially improve fine-mapping of causal variants (at 51 signals, one variant accounted for >80% posterior probability of association (PPA)); (iv) extend fine-mapping through integration of tissue-specific epigenomic information (islet regulatory annotations extend the number of variants with PPA >80% to 73); (v) highlight validated therapeutic targets (18 genes with associations attributable to coding variants); and (vi) demonstrate enhanced potential for clinical translation (genome-wide chip heritability explains 18% of T2D risk; individuals in the extremes of a T2D polygenic risk score differ more than ninefold in prevalence). © 2018, The Author(s), under exclusive licence to Springer Nature America, Inc.
Article
Full-text available
Type 2 diabetes (T2D) is a very common disease in humans. Here we conduct a meta-analysis of genome-wide association studies (GWAS) with ~16 million genetic variants in 62,892 T2D cases and 596,424 controls of European ancestry. We identify 139 common and 4 rare variants associated with T2D, 42 of which (39 common and 3 rare variants) are independent of the known variants. Integration of the gene expression data from blood (n = 14,115 and 2765) with the GWAS results identifies 33 putative functional genes for T2D, 3 of which were targeted by approved drugs. A further integration of DNA methylation (n = 1980) and epigenomic annotation data highlight 3 genes (CAMK1D, TP53INP1, and ATP5G1) with plausible regulatory mechanisms, whereby a genetic variant exerts an effect on T2D through epigenetic regulation of gene expression. Our study uncovers additional loci, proposes putative genetic regulatory mechanisms for T2D, and provides evidence of purifying selection for T2D-associated variants.
Article
The aim was to assess lifetime risk for hospitalization in individuals with neurofibromatosis 1 (NF1). The 2467 individuals discharged with a diagnosis indicating NF1 or followed in a clinical center for NF1 were matched to 20,132 general population comparisons. Based on diagnoses in 12 main diagnostic groups and 146 subcategories, we calculated rate ratios (RRs), absolute excess risks (AERs), and hazard ratios for hospitalizations. The RR for any first hospitalization among individuals with NF1 was 2.3 (95% confidence interval 2.2–2.5). A high AER was seen for all 12 main diagnostic groups, dominated by disorders of the nervous system (14.5% of all AERs), benign (13.6%) and malignant neoplasms (13.4%), and disorders of the digestive (10.5%) and respiratory systems (10.3%). Neoplasms, nerve and peripheral ganglia disease, pneumonia, epilepsy, bone and joint disorders, and intestinal infections were major contributors to the excess disease burden caused by NF1. Individuals with NF1 had more hospitalizations and spent more days in hospital than the comparisons. The increased risk for any hospitalization was observed for both children and adults, with or without an associated cancer. NF1 causes an overall greater likelihood of hospitalization, with frequent and longer hospitalizations involving all organ systems throughout life.
Article
Background & aims: Neurofibromatosis type 1 (NF1) is an autosomal dominant genetic disease that is characterized by neurocutaneous changes with multisystem involvement. A previous study with adults with NF1 revealed that changes in total energy expenditure were related to food consumption and body composition. Resting energy expenditure (REE), a measure of energy that the body expends to maintain vital functions, has not been assessed in NF1 populations. This study aimed to assess REE in individuals with NF1 using indirect calorimetry (IC) and evaluate its correlation with body composition and muscle strength. Methods: Twenty-six adults with NF1 (14 men) aged 18-45 years underwent IC for assessing REE, respiratory quotient (RQ), and substrate utilization. Body composition was assessed by dual energy X-ray absorptiometry. Weight, height, and waist circumference (WC) were also measured. Maximum muscular strength (Smax) was measured by handgrip test using a dynamometer. Patients in the NF1 group were compared to 26 healthy controls in the control group, who were matched by sex, age, body mass index (BMI), and physical activity level. Results: There were no differences in weight, WC, fat mass, and body fat percentage (BFP). Appendicular lean mass (ALM) adjusted by BMI (ALMBMI) (0.828 ± 0.161 versus 0.743 ± 0.190; P = 0.048) and Smax (37.5 ± 10.6 versus 31.1 ± 12.2; P = 0.035) was lower in the NF1 group than in the control group. No differences in body composition, strength, and anthropometric parameters were observed in men, but women with NF1 presented lower body surface area (BSA), lean body mass (LBM), ALM, ALMBMI, and Smax. REE adjusted by weight, LBM, or ALM was higher in the NF1 group than in the control group (medians, 21.9 versus 26.3, P = 0.046; 36.5 versus 41.1, P = 0.012; and 82.3 versus 92.4, P = 0.006, respectively), and these differences were observed only among women. RQ was lower in the NF1 group than in the control group (0.9 ± 0.1 versus 0.8 ± 0.1; P = 0.008), revealing that individuals with NF1 oxidized more lipids and fewer carbohydrates than controls. REE correlated negatively with BFP and positively with weight, height, BMI, WC, BSA, LBM, ALM, ALMBMI, bone mineral content, and Smax. Conclusions: Individuals with NF1, particularly women, presented with increased REE (adjusted by weight, LBM, or ALM) and lower RQ compared to healthy controls. These findings were associated with lower ALMBMI and Smax, possibly indicating premature sarcopenia in this population. Further investigation concerning energy metabolism in NF1 and gender differences may be helpful in explaining underlying mechanisms of these changes.
Article
The melanocortin 4 receptor (MC4R) is a G protein-coupled receptor whose disruption causes obesity. We functionally characterized 61 MC4R variants identified in 0.5 million people from UK Biobank and examined their associations with body mass index (BMI) and obesity-related cardiometabolic diseases. We found that the maximal efficacy of β-arrestin recruitment to MC4R, rather than canonical Gαs-mediated cyclic adenosine-monophosphate production, explained 88% of the variance in the association of MC4R variants with BMI. While most MC4R variants caused loss of function, a subset caused gain of function; these variants were associated with significantly lower BMI and lower odds of obesity, type 2 diabetes, and coronary artery disease. Protective associations were driven by MC4R variants exhibiting signaling bias toward β-arrestin recruitment and increased mitogen-activated protein kinase pathway activation. Harnessing β-arrestin-biased MC4R signaling may represent an effective strategy for weight loss and the treatment of obesity-related cardiometabolic diseases.
Article
Type 1 diabetes is a chronic autoimmune disease characterised by insulin deficiency and resultant hyperglycaemia. Knowledge of type 1 diabetes has rapidly increased over the past 25 years, resulting in a broad understanding about many aspects of the disease, including its genetics, epidemiology, immune and β-cell phenotypes, and disease burden. Interventions to preserve β cells have been tested, and several methods to improve clinical disease management have been assessed. However, wide gaps still exist in our understanding of type 1 diabetes and our ability to standardise clinical care and decrease disease-associated complications and burden. This Seminar gives an overview of the current understanding of the disease and potential future directions for research and care.