Next Article in Journal
Quality of Life Scores Remained Different among the Genotypic Groups of Patients with Suspected Hemochromatosis, Even after Treatment Period
Next Article in Special Issue
Pharmacogenetics of Drugs Used in the Treatment of Cancers
Previous Article in Journal
Molecular Investigation of miRNA Biomarkers as Chemoresistance Regulators in Melanoma: A Protocol for Systematic Review and Meta-Analysis
Previous Article in Special Issue
Pharmacogenomics: An Update on Biologics and Small-Molecule Drugs in the Treatment of Psoriasis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of Actionable Genes in Pursuing a True Approach of Precision Medicine in Monogenic Diabetes

1
Research Unit of Diabetes and Endocrine Diseases, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy
2
Pediatric Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy
3
Department of Experimental Medicine, Sapienza University, 00161 Rome, Italy
*
Authors to whom correspondence should be addressed.
Genes 2022, 13(1), 117; https://doi.org/10.3390/genes13010117
Submission received: 30 November 2021 / Revised: 4 January 2022 / Accepted: 5 January 2022 / Published: 9 January 2022
(This article belongs to the Special Issue Pharmacogenomics: Precision Medicine and Drug Response)

Abstract

:
Monogenic diabetes is a genetic disorder caused by one or more variations in a single gene. It encompasses a broad spectrum of heterogeneous conditions, including neonatal diabetes, maturity onset diabetes of the young (MODY) and syndromic diabetes, affecting 1–5% of patients with diabetes. Some of these variants are harbored by genes whose altered function can be tackled by specific actions (“actionable genes”). In suspected patients, molecular diagnosis allows the implementation of effective approaches of precision medicine so as to allow individual interventions aimed to prevent, mitigate or delay clinical outcomes. This review will almost exclusively concentrate on the clinical strategy that can be specifically pursued in carriers of mutations in “actionable genes”, including ABCC8, KCNJ11, GCK, HNF1A, HNF4A, HNF1B, PPARG, GATA4 and GATA6. For each of them we will provide a short background on what is known about gene function and dysfunction. Then, we will discuss how the identification of their mutations in individuals with this form of diabetes, can be used in daily clinical practice to implement specific monitoring and treatments. We hope this article will help clinical diabetologists carefully consider who of their patients deserves timely genetic testing for monogenic diabetes.

1. Introduction

Precision medicine is meant as the attempt to provide the most specific management (i.e., prediction, prevention, diagnosis, follow-up and treatment) for subgroups of individuals who share similar features such as those of epidemiological, phenotypic, clinical and molecular origin. In the last few years, genome and exome sequencing have helped identify genetic variants which shape the risk of various diseases. Some of these variants are harbored by genes whose altered function can be tackled by specific approaches, so as to allow individual interventions aimed to prevent, mitigate or delay clinical outcomes (“actionable genes”) [1].
Diabetes mellitus has a current prevalence of 463 million (equivalent to 9.3% of the world population) that is expected to raise to 578 million by the year 2030 [2]. Diabetes imposes heavy burdens on health care systems, patients, and their families and is associated with serious economic implications, making any effort to tackle it of paramount importance [3]. As much as 95% of patients with diabetes are affected by either type 1 (approximately 10%) or type 2 diabetes, both typical examples of complex diseases based on the interaction between environmental factors and a predisposing genetic background [4]. Conversely, a small proportion of patients with diabetes (up to 5%), which, however, given the huge number of affected individuals is not trivial in absolute terms, is affected by monogenic forms of the disease [4,5]. Of interest several actionable genes that cause monogenic diabetes have been described and this allows the implementation of effective approaches of precision medicine in carrier patients [6,7].
This review will offer a brief overview of our present knowledge of the actionable genes that cause monogenic diabetes. For each of them we will provide a short background on what is known about gene function and dysfunction. Then we will discuss how the identification of their mutations in individuals with this form of diabetes can be used in daily clinical practice to implement specific monitoring and treatments.

2. Monogenic Diabetes

Monogenic diabetes is a genetic disorder caused by one or more variations in a single gene. It encompasses a broad spectrum of heterogeneous conditions, including neonatal diabetes, maturity onset diabetes of the young (MODY) and syndromic diabetes [8], affecting 1–5% of patients with diabetes [5,9]. These forms of diabetes have either dominant or recessive or non-Mendelian inheritance. To date, 40 different subtypes of monogenic diabetes have been identified [5]. By far, the most frequent form of monogenic diabetes is MODY, which according to its historical definition [10,11] is inherited in an autosomal dominant manner and occurs in lean individuals before the age of 25 years, with no need of insulin therapy. This strict definition is now outdated, since MODY also occurs in middle age and/or overweight/obese patients and may need insulin as the most appropriate treatment [12]. Mutations in fourteen different genes have been so far reported to cause several MODY subtypes (OMIM # 606391), the most common of which are due to mutations in GCK, HNF1A, HNF4A and HNF1B [7].
A well-known additional form of monogenic diabetes is neonatal diabetes, a rare disorder characterized by marked insulin-requiring hyperglycemia within the first 6 months of life [13]. Between 50 to 60% of cases resolve within 18 months and are, therefore, termed transient neonatal diabetes. The remaining patients require insulin treatment for life and are termed permanent neonatal diabetes [14]. Approximately 20–30% of patients with neonatal diabetes also present neuro-motor developmental delay [15,16,17]. Additional neurological implications including autism, attention deficit hyperactivity disorder, anxiety, sleep disorders and learning difficulties with impaired attention and memory [18,19,20,21,22,23] have been also described.
Finally, very rare forms of monogenic diabetes are those presenting in combination with several additional extra-pancreatic abnormalities (i.e., syndromic diabetes) which are autosomal, X-linked, recessively and dominantly inherited or due to mitochondrial mutations. Syndromic forms of diabetes can derive from severe defect of either insulin secretion (e.g., Wolfram syndrome, Wolcott–Rallison syndrome, thiamine-responsive megaloblastic anemia, mitochondrial mutations) or insulin action (i.e., insulin receptor mutations, Bardet–Biedl syndrome, Alstrom syndrome, Berardinelli–Seip congenital lipodystrophies) [5,24].
In general, a monogenic form of diabetes should be suspected either in children or in young individuals with diabetes when hyperglycemia ensues with no typical features of type 1 or type 2 diabetes [4,25]. These includes some degree of ketoacidosis and especially the ineludible need of insulin treatment for type 1 diabetes. At variance, the typical characteristics of type 2 diabetes are the presence of obesity and other comorbidities clustering in the metabolic syndrome, including which hypertension and atherogenic dyslipidemia are the most common. Overall, the great heterogeneity of all forms of hyperglycemia, makes sometimes difficult to differentiate monogenic diabetes from the type 1 and type 2 diabetes from a simple clinical point of view. It is therefore very useful that, in suspected patients, the simultaneous analysis of multiple genes by NGS, currently available in many contexts, gives the possibility of a rapid and precise molecular diagnosis.
Since several excellent comprehensive reviews have been recently published on both molecular and clinical aspects of monogenic diabetes [5,8,9], this article will almost exclusively concentrate on the clinical strategy that can be specifically pursued in carriers of mutations in actionable genes, including ABCC8, KCNJ11, GCK, HNF1A, HNF4A, HNF1B, PPARG, GATA4 and GATA6 [6] see Table 1.

3. ABCC8 and KCNJ11

ABCC8 and KCNJ11 encode respectively four sulfonylurea receptor 1 (SUR1) and four inward rectifier potassium channel Kir6 (Kir6.2) subunits both belonging to the ATP-sensitive potassium (KATP) channel in the pancreatic β-cells [26,27,28]. KATP channel links cellular glucose metabolism to electrical activity of the plasma membrane thereby regulating insulin secretion [27,29,30,31]. In details, at sub-stimulatory glucose concentration, the KATP channel is open, thus permitting the efflux of K+ from the β-cell that eventually causes membrane hyperpolarization. Channel opening is the result of a fine regulation and activation exerted by the binding of intracellular MgADP [27,32]. Conversely, when blood glucose level rises, β-cells metabolize glucose, converting ADP to ATP, which then binds to and closes the KATP channel causing membrane depolarization. This in turn activates voltage-dependent calcium channels, allows Ca+ influx into the cell and finally triggers insulin granule release [27].
More than 700 ABCC8 and 200 KCNJ11 pathogenic or likely pathogenic mutations have been reported [33]. As described below, mutations in these two genes result in a variety of phenotypes depending on the hyper- or hypo-activity of the KATP channel they induce.
ABCC8/KCNJ11 gain-of-function (GOF) mutations cause the permanent opening of the KATP channel [34], and thus reduce insulin secretion and eventually cause hyperglycemia with a wide spectrum of diabetes phenotypes, ranging from neonatal diabetes to MODY to adult-onset diabetes [35,36,37,38]. In ABCC8 about 100 dominant and 100 recessive activating mutations have been described [39,40] to cause neonatal diabetes. Conversely, nearly 100 dominant and only one recessive activating KCNJ11 mutations have been linked to neonatal diabetes [15,41]. About 60% of dominant mutations causing neonatal diabetes occur “de novo”; a germline mosaicism has been also observed in some families [42,43].
In contrast, far fewer MODY families with heterozygous ABCC8 [44,45,46] and KCNJ11 [47,48] mutations have been described worldwide.
ABCC8/KCNJ11 loss-of-function (LOF) mutations affect KATP channel by both preventing its posttranscriptional moving to the plasma membrane or by reducing its responsiveness to MgADP activation. These mutations, mostly observed in ABCC8 (nearly 600 vs. approximately 100 in KCNJ11) [49,50,51,52], are predominantly recessive inherited [53] and account for 36–70% cases of congenital hyperinsulinemic hypoglycemia (HH) [54,55], which is not the focus of this article and which has been discussed in details elsewhere [56,57].

ABCC8/KCNJ11 Actionability

When a diagnosis of neonatal diabetes due to a KATP channel activating mutation is made, sulphonylureas is the treatment of choice, being effective as monotherapy in most of these patients. Sulphonylureas bind specifically to the SUR1 subunit and close the KATP channel via an ATP-independent mechanism, thus bypassing the genetic defect on ABCC8/KCNJ11h [58]. In fact, the great majority of mutation carrying individuals treated with insulin can be shifted to sulphonylureas, with a significant improvement of quality of life and disease management [59,60]. Though generally very good, the efficacy of this treatment seems to be partly determined by both the type of mutation and the duration of diabetes [61,62].
Despite the relatively high doses of sulfonylureas used in these patients, no episodes of hypoglycaemia have been reported during a long-term follow-up while persistent efficacy and safety of the treatment was described [63].
Moreover, in some patients with intermediated neuro-motor developmental delay, high dose of sulphonylureas initially improves the neurological impairment [18,21,63,64,65].
During pregnancy, the benefit of sulphonylureas treatment is dependent of the combination of ABCC8 or KCNJ11 genotype in the mother and the fetus:
(a)
If the mother carries an ABCC8 or KCNJ11 activating mutation, and the fetus has a normal genotype, sulfonylureas therapy can result in excessive insulin secretion that induce macrosomia and neonatal HH in the baby [66,67,68]; when ultrasound monitoring results suggest this possibility, transfer from sulphonylureas to insulin is recommended [69].
(b)
If both the mother and the fetus carry an ABCC8 or KCNJ11 activating mutation, babies benefit from early exposure to sulphonylureas treatment which prevents the low birth weight caused by reduced insulin secretion in the uterus [40,59]; sulphonylureas treatment should be, therefore, continued at the lowest dose required to obtain the optimal glycemic control [69].
Coincidentally, because of reduced insulin secretion in the uterus birth weight will be low also when only the fetus has an activating ABCC8 or KCNJ11 mutation (e.g., either inherited from the father or because of a “de novo” mutation) [70].
When one of the above conditions is known or suspected (e.g., father known to carry an activating ABCC8 or KCNJ11 mutation), ultrasound should be performed every two weeks from the 26th week of gestation to strictly follow fetal growth [71].

4. GCK

Glucokinase (GCK) is one of four members of hexokinase enzymes family [72,73]. GCK acts in the first reaction of glycolytic pathway converting glucose in glucose-6-phosphate. This will be completely metabolized through the next glycolytic steps and eventually increase intracellular ATP concentration, thus inducing pancreatic β-cells insulin secretion. In fact, GCK is considered the glucose sensor in β-cells, with the rate of glucose phosphorylation being directly related to plasma glucose concentration in the physiological range [73]. In addition to pancreatic islets, GCK is expressed in several tissues such as brain, liver and endocrine cells of the gut [74]. To date, over 900 GCK variants have been identified spanning all over the gene (e.g., missense/nonsense mutations, splicing mutations, small and gross deletions, insertions). GCK mutation should be suspected in the presence of blood glucose levels in the range of 5.5–8.0 mmol/l and a relative low increment at the oral glucose tolerance test (OGTT) [75]. According to the type of mutation (e.g., heterozygous or homozygous and LOF or GOF), different phenotypes can ensue.
GCK heterozygous LOF mutations have been associated with a reduced GCK activity that alters the glucose threshold for stimulating insulin secretion and eventually a mild form of fasting hyperglycemia present from birth and named as GCK-MODY (traditionally known as MODY2). Patients with GCK-MODY are also characterized by reduced hyperglycemia-induced inhibition of hepatic glucose output and decreased post-prandial liver glycogen synthesis. Despite lifelong hyperglycemia, GCK-MODY phenotype is characterized by a very low prevalence of microvascular and macrovascular complications [76].
Conversely as described below, pregnancy management is particularly important in patients with GCK-MODY [77,78].
GCK homozygous or compounds heterozygous LOF mutations usually cause neonatal diabetes, although few carriers of such mutations have been reported to be indistinguishable from those with typical GCK-MODY [79,80].
GCK heterozygous GOF mutations cause HH due to increased glucose-GCK affinity that causes excessive insulin secretion not commensurate with blood glucose levels [74,81]. Details on HH, which is not the focus of this review, have been already extensively discussed [56,57].

GCK Actionability

With the exception of pregnancy, GCK-MODY patients do not need treatment because its mild hyperglycemia is not progressive. Only diet and lifestyle interventions should, therefore, be recommended [76,82].
In order to avoid abnormal fetal growth, hyperglycemia in women with GCK-MODY should be treated or not, according to the GCK genotype of both the mother and the fetus as it follows:
(a)
If the mother carries a GCK mutation a non-mutated fetus senses maternal hyperglycemia and consequently increases insulin secretion that, in turn, causes macrosomia (suggested by accelerated fetal growth at ultrasound and resulting in a final birth weight increased by 550–700 g) [83,84]. In these cases, insulin treatment of the pregnant mother is necessary.
(b)
If both the mother and the fetus carry a GCK mutation, insulin treatment is not recommended because fetal growth will be normal [72].
(c)
If only the fetus has a LOF GCK mutation no treatment is needed, but usually the babies have reduced birth weight (by approximately 400 g) due to reduced insulin secretion in the uterus [83,84].
When one of the above conditions is known or suspected (e.g., father known to carry a LOF GCK mutation), ultrasound should be performed every two weeks from the 26th week of gestation to strictly follow fetal growth [71].

5. Hepatocyte Nuclear Factors (HNF) Family

Hepatocyte nuclear factors (HNFs), which are classified into 4 families, were first identified as liver-enriched transcription factors and are known to be important regulator of pancreas, kidney and liver development and/or function [85,86,87]. LOF mutations in HNF1A, HNF1B and HNF4A, all leading to defective insulin secretion and reduced β-cells mass, cause some forms of MODY possibly associated with kidney and/or liver abnormalities [88].
For all these genes clinical phenotype of the mutation carriers is highly variable from one family to another and also within the same family [89].

5.1. HNF1A

Until now more than 400 different variants [90] spanning from the HNF1A promoter to the 3′UTR region (e.g., missense mutations, frame shift, nonsense, splicing mutations, in-frame amino acid deletions, insertions, duplications or partial and whole-gene deletions) have been described, many of which occurring in exon 2 and 4 [90,91]. HNF1A heterozygous and homozygous [92] LOF mutations cause HNF1A-MODY (MODY3) [93,94,95], by impairing the transcriptional activity of the gene which, in turn, affects, several target proteins involved in glucose metabolism, insulin secretion [96,97,98] and cell proliferation [99,100,101]. Some of these genes are implicated in glycolysis and ATP production which is blunted by LOF HNF1A mutation [94,102,103,104].
HNF1A-MODY manifests, usually, in the first 2–3 decades of life with mild symptoms (polyuria, polydipsia) or as asymptomatic postprandial hyperglycemia, without ketosis or ketoacidosis. Fasting glucose may be normal at the disease’s onset and increases gradually with age, while 2-h glucose at the oral glucose tolerance test (OGTT) is frankly increased (4.4–5 mmol/l above the normal range) [105]. Insulin deficiency is progressive, C-peptide values are lower than in healthy individuals, but generally higher than in type 1 diabetes [105].
In kidneys, HNF1A LOF mutations reduce the threshold of glucose reuptake from the glomerular filtrate [106]. As a consequence, carriers of HNF1A mutations show glycosuria [105] that begins several years before hyperglycemia and is likely due to down regulation of SLC5A2 [106] which encodes SGLT2, a sodium-dependent glucose transporter known to play a major role in renal glucose reabsorption. Liver adenomatosis may also be present [107,108,109,110]. Finally, increased fatty acid synthesis and altered transportation and eventually lipid accumulation into liver cells have been described [111].
In individuals with poor glycemic control, chronic microvascular complications (retinopathy, nephropathy and neuropathy) are common and ketoacidosis can develop [112,113]. The risk of hypertension and ischemic heart disease are similar to what observed in type 1 diabetes, much rarer than in type 2 diabetes, but certainly increased than in healthy controls [114]. Children exposed to high glucose levels in the uterus who inherit HNF1A mutation from their mother have an earlier onset of diabetes (5–10 years before) than those who inherit the mutation from their father [115].

HNF1A Actionability

Patients with HNF1A-MODY are sensitive to low-dose sulfonylureas [116,117]. In fact, SUR1 is downstream the HNF1A effects on glycolysis and mitochondrial ATP production. Sulphonylureas, therefore, bypass the β-cell defects induced by HNF1A mutations [107], so as to properly activate the downstream preserved machinery which is perfectly able to promote insulin secretion. Sulphonylureas are, therefore, the first-choice treatment in patients with HNF1A-MODY. Since in children and in adolescents sulfonylureas are not yet licensed, the use of meglitinides can be considered [118,119,120].
Sulfonylureas are usually effective for several decades, but in patients with severe insulin deficiency or after a long duration of diabetes (>11years), insulin treatment may eventually become necessary, either alone or on top of sulfonylureas [121].
GLP-1 Receptor Activation (GLP1-RA) stimulates insulin secretion bypassing the genetic HNF1A defect through the elevation of cyclic adenosine monophosphate (cAMP) and activation of protein kinase A [122,123,124]. GLP1-RA has been, therefore, recently suggested as an add-on treatment in patients who do not achieve optimal glycemic control with sulfonylurea or who experience frequent hypoglycemic events [125,126,127,128].
Although rare cases of macrosomic HNF1A mutation carriers presenting with neonatal transient HH that resolve with age have been described [129,130], HNF1A mutations do not generally influence birth weight because insulin secretion in the uterus remains normal [69,131]. Therefore, if only the fetus is suspected to carry an HNF1A mutation (e.g., father known to be mutated) no treatment is needed [69]. As far as the optimal treatment of gestational hyperglycemia is concerned, insulin is necessary if only the mother carries an HNF1A mutation [69,132].

5.2. HNF4A

HNF4A heterozygous LOF mutations are 10 times less frequent than those harbored by HNF1A. Until now more than 100 different variants [90] spanning the whole gene (e.g., missense mutations, frame shift, nonsense, splicing mutations, in-frame amino acid deletions, insertions, duplications or partial and whole-gene deletions) have been described, particularly in exon 7 and 8 [90,91].
HNF4A heterozygous mutations usually cause haploinsufficiency that impairs insulin secretion and causes HNF4A-MODY (MODY1) [102,103,104] with the same mechanisms described above for HNF1A mutations [133]. Of note, HNF4A regulates the expression of HNF1A [134] and is therefore expected that, with the exception of normoglycemic glycosuria [135], the HNF4A-MODY phenotype is similar to that of HNF1A-MODY, with glucose intolerance becoming evident during adolescence or early adulthood and deteriorating with age [105]. Chronic diabetes complications are frequent and their development is related to the degree of metabolic control [107,136,137]. In addition to their effects on β-cell function, HNF4A deficiencies affect liver function, triglyceride serum levels (50% reduction) and apolipoprotein biosynthesis (25% reduction of apolipoproteins AII and CIII and Lp(a) lipoprotein serum concentrations). Reduced HDL and increased LDL cholesterol levels have been also described [138,139] in patients with HNF4A-MODY.

HNF4A Actionability

As described for carriers of HNF1A mutations, patients with HNF4A-MODY are also sensitive to low-dose sulfonylureas. In fact, HNF4A-MODY patients can initially be treated with diet while sulfonylureas are recommended if glycemic control deteriorates [107]. Especially in young adults, this treatment results in better glycemic control than that achieved with insulin [102]. If hypoglycemia is not a problem, this treatment can be maintained for decades [116,140]. A short-acting agent such as a meglitinide can be considered in children [118,119,120].
An alternative treatment for HNF4A-MODY patients are GLP-1 agonists that should be taken into consideration in case of either non-optimal glycemic control or recurrent hypoglycemia due to sulfonylureas [122,126].
Given the very high risk of massive macrosomia (birth weight > 5000 g) and related clinical outcome of babies delivered by women with HNF4A-MODY, tight maternal glycemic control is instrumental [69,131,141].
Of note, 15% of newborns with HNF4A mutations have diazoxide-responsive neonatal HH which remits during infancy [131,141,142].

5.3. HNF1B

HNF1B encodes the hepatocyte nuclear factor 1β (HNF1B), a transcription factor with 80% homology with HNF1A, which binds DNA either as a homodimer or a heterodimer with HNF1A. Heterozygous mutations (e.g., missense mutations, complete gene deletions, little fragment deletions or insertions), half of which are de novo [143], show autosomal dominant inheritance and cause a clinical spectrum with renal cysts and hyperglycemia, (HNF1B-MODY, MODY5). HNF1B is a rare cause of MODY, accounting for <2% of cases [144]. In HNF1B-MODY hyperglycemia is usually diagnosed in the young adulthood (mean age of 26 with a range of 10–61) [145]. The range of glucose homeostasis goes from normoglycemia to insulin-treated diabetes with ketoacidosis [146]. Only few cases of neonatal diabetes have been described in carriers of HNF1B mutations [147,148].
The combination of diabetes with congenital anomalies in the kidney (cysts) and urinary tract is the most consistent clinical presentation in individuals with HNF1B mutations [149,150,151,152,153]. Alterations in pancreas, brain, parathyroid gland, and female genital tract (e.g., rudimental uterus, vaginal aplasia, bicornuate uterus and double vagina) are frequently described and are compatible with the involvement of HNF1B in their development [146,154,155,156,157,158].
Liver impairment can also manifest as neonatal jaundice, lack of intrahepatic bile ducts, rise of alanine aminotransferase and γ-glutamyl transferase serum levels [159]. Hypercholesterolemia and hyperuricemia have been also described [160,161].

HNF1B Actionability

HNF1B mutation carriers have insulin deficiency due to pancreatic hypoplasia [154]. Therefore, not unexpectedly, these patients require early insulin therapy [162,163] and do not/or rarely respond adequately to sulfonylureas [164].
Moreover, patients with HNF1B mutation have reduced exocrine pancreatic function that should, therefore, be monitored by measuring fecal elastase levels [165]. Uric acid levels should also be monitored in order to prevent gout. Systematic screening for all potential morphological anomalies, in particular for renal cysts and pancreatic and genital (especially in females) abnormalities [88], should be made.
During pregnancy, women with HNF1B-MODY typically require insulin for glycemic control. Generally, because of maternal hyperglycemia, birth weight of their babies is increased [166]. Conversely, when the fetus has a HNF1B mutation (e.g., inherited from the father) birth weight can be diminished reflecting reduced insulin secretion in the uterus [166].

6. PPARG

The Peroxisome proliferator-activated receptor γ (PPARG) gene encodes a ligand-activated transcription factor (PPARγ) belonging to the family of nuclear PPARs. PPARγ is predominantly expressed in white and brown adipose tissue [167,168,169] and exists as two isoforms, PPARγ1 and PPARγ2, with the latter containing an additional 30 amino acids at its N-terminus [169,170]. PPARγ is a key regulator of adipocyte differentiation as well as a potent modulator of whole-body energy balance, lipid biosynthesis, and insulin sensitivity [171,172]. Several SNPs and/or rare variants and mutations have been associated with a spectrum of metabolic diseases including obesity, syndromic form of monogenic diabetes and type 2 diabetes [173,174,175,176].
Heterozygous LOF mutations of PPARG are associated with a familial partial lipodystrophy type 3 (FPLD3) considered as a monogenic model of the common “metabolic syndrome” [177,178]. FPLD3 is a dominantly inherited syndrome, characterized by specific loss of subcutaneous fat from the limbs and gluteal region. It is also associated with early diabetes onset, hypertension, severe insulin resistance, extreme dyslipidemia, and hepatic steatosis [177,179]. The severity of lipodystrophy is related to the deleterious effect of each specific mutation on PPARγ function [180].
To date approximately 40 FPLD3 LOF PPARG mutations have been reported, most of which involving the ligand-binding domain or the DNA-binding domain [173,180]. Mechanisms of negative dominance and haploinsufficiency have both been suggested to explain the pathogenicity of PPARG mutations [181]. Notably, approximately 0.2% of the general population carries a rare missense variant in PPARG gene, 20% of which are functionally relevant and are associated with metabolic diseases, not necessarily leading to overt FPLD3 [182]. These evidences support the hypothesis that additional mechanisms such as gene-gene and gene-environment interactions might contribute to the variable phenotype [173].

PPARG Actionability

In 1995, thiazolidinediones (TZDs) have been identified as a class of potent activators of PPARγ, able to promote adipogenesis and improve insulin sensitivity [183,184,185,186]. Due to their ability to preserve pancreatic β-cell function and reduce insulin resistance, TZDs have become an established medication for type 2 diabetes [187]. Unfortunately, data on the use of TZDs in individuals with monogenic syndromic form of diabetes caused by LOF mutations in PPARG are still inconclusive. This is partly due to the rarity of this form, but also to the heterogeneous pharmacological response, probably secondary to the specific molecular defect [188,189,190]. At this regard, there is some evidence suggesting that knowing the specific molecular defect in a given patient may be helpful in choosing the most appropriate TZD [188,191]. For example, patients carrying the R308P mutation in PPARγ show reduced binding affinity to rosiglitazone, but not to pioglitazone [192].
Overall, when TZD is able to bind a mutated PPARγ some metabolic improvement including reductions of glycated hemoglobin, triglycerides, free fatty acids and increased body fat has been described [189,190,192,193].
Therefore, with due care and waiting for further and larger studies possibly testing also new TZD-derived compounds, PPARG can be envisioned as a likely “actionable gene” for patients with FPLD3 [194].

7. GATA4 and GATA6

GATA family includes a group of six transcription factors (GATA1–6), all of which contain two tandem zinc-finger domains that bind a consensus site (A/T) GATA (A/G) of DNA target region and play a crucial role in the development and differentiation of all eukaryotic organisms [195].
LOF in GATA4 and GATA6 are associated with pancreatic agenesis/hypoplasia and diabetes, along with congenital heart abnormalities and several cancers [196].

7.1. GATA4

GATA4 heterozygous LOF mutations and complete deletion of the gene (deletion from 1 to 17 Mb that includes 8 additional genes), have been associated with congenital heart malformations [197,198,199], and more rarely with pancreatic agenesis and neonatal diabetes [200,201].

7.2. GATA6

GATA6 heterozygous mutations (e.g., missense, non-sense, frameshift mutations and small deletions mainly located in, or near to, tandem zinc-finger domains or in the splicing site), most of which occur de novo, can cause a wide spectrum of diabetes manifestations, ranging from pancreatic agenesis and neonatal diabetes to adult-onset diabetes. Usually, age at diabetes diagnosis ranges from 12–46 years [202]. In addition, exocrine pancreatic insufficiency and, more frequently, congenital heart malformations, together with congenital biliary tract anomalies, gut developmental disorders, neurocognitive abnormalities and additional endocrine abnormalities have been described [203]. Sometimes, the same mutation has been associated with alternative phenotypes showing a dissimilar penetrance in different patients [204]. No data in pregnant women are available, while low birth weight has been reported for babies carrying GATA4 or GATA6 mutations [201,202].

7.3. GATA4 and GATA6 Actionability

For these two genes, actionability consists in the early identification of the possible severe abnormalities mutations carriers can show. In fact, in GATA4/6 mutations carriers, particular in newborns, the evaluation and monitoring over time of congenital heart malformations and heart failure is mandatory. As a consequence of pancreatic agenesis/hypoplasia, these patients not only require insulin as the treatment of choice for hyperglycemia but may also need pancreatic enzyme supplementation. Since the same GATA4/6 mutations can generate variable phenotypes in different subjects, a case-by-case evaluation is necessary.

8. Conclusions

There is no doubt that a correct, precise and timely diagnosis is essential for the rapid management of most diseases and for the prevention of related complications. This general assumption also often applies when dealing with genetic diseases [205], thus stressing that timely recognition of patients who deserve genetic testing is extremely important.
Hopefully this article makes it clear that some forms of monogenic diabetes are optimal examples of precision medicine and of translating research findings into clinical practice. Rare forms of monogenic diabetes due to mutations of “actionable genes” have, in fact, paved the way for new therapeutic strategies and specific management of disease outcomes. Clinical diabetologists should, therefore, carefully consider who of their patients deserves timely genetic testing for monogenic diabetes, possibly using NGS approaches. Of note, the advent of NGS not only has allowed a prompt diagnosis but also has increased the chance of discovering new “diabetes genes” [206,207], a prerequisite to understand in depth glucose homeostasis and to possibly invent new drugs. It is conceivable that some of the genes we will discover in the future will be “actionable”, thus further increasing the number of patients who can be specifically managed according to their specific genetic defects.

Author Contributions

Conceptualization, A.M., I.R., G.F., S.P., C.M., R.D.P. and V.T.; data curation, A.M., I.R. and S.P.; writing—original draft preparation, A.M., I.R., G.F., S.P., C.M., R.D.P. and V.T.; writing—review and editing, A.M., V.T.; supervision, A.M., V.T.; funding acquisition, A.M., C.M., R.D.P. and V.T. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by Italian Ministry of Health: Ricerca Corrente RC2020; RC2021 (A.M., C.M., R.D.P. and V.T.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We would like to thank Emiliano Giardina (UILDM Lazio Onlus Foundation, Department of Biomedicine and Prevention, Tor Vergata University, Rome), for inviting us to write this review.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Rego, S.; Dagan-Rosenfeld, O.; Zhou, W.; Sailani, M.R.; Limcaoco, P.; Colbert, E.; Avina, M.; Wheeler, J.; Craig, C.; Salins, D.; et al. High-frequency actionable pathogenic exome variants in an average-risk cohort. Cold Spring Harb. Mol. Case Stud. 2018, 4. [Google Scholar] [CrossRef]
  2. Saeedi, P.; Petersohn, I.; Salpea, P.; Malanda, B.; Karuranga, S.; Unwin, N.; Colagiuri, S.; Guariguata, L.; Motala, A.A.; Ogurtsova, K.; et al. Global and regional diabetes prevalence estimates for 2019 and projections for 2030 and 2045: Results from the international diabetes federation diabetes atlas. Diabetes Res. Clin. Pract. 2019, 157, 107843. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Nathanson, D.; Sabale, U.; Eriksson, J.W.; Nyström, T.; Norhammar, A.; Olsson, U.; Bodegård, J. Healthcare cost development in a type 2 diabetes patient population on glucose-lowering drug treatment: A nationwide observational study 2006–2014. PharmacoEconomics-Open 2018, 2, 393–402. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. American Diabetes Association. 2. Classification and Diagnosis of Diabetes: Standards of Medical Care in Diabetes—2020. Diabetes Care 2020, 43, S14–S31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Hattersley, A.T.; Greeley, S.A.W.; Polak, M.; Rubio-Cabezas, O.; Njølstad, P.R.; Mlynarski, W.; Castano, L.; Carlsson, A.; Raile, K.; Chi, D.V.; et al. Ispad clinical practice consensus guidelines 2018: The diagnosis and management of monogenic diabetes in children and adolescents. Pediatr. Diabetes 2018, 19, 47–63. [Google Scholar] [CrossRef] [PubMed]
  6. Bonnefond, A.; Boissel, M.; Bolze, A.; Durand, E.; Toussaint, B.; Vaillant, E.; Gaget, S.; de Graeve, F.; Dechaume, A.; Allegaert, F.; et al. Pathogenic variants in actionable mody genes are associated with type 2 diabetes. Nat. Metab. 2020, 2, 1126–1134. [Google Scholar] [CrossRef] [PubMed]
  7. Vaxillaire, M.; Froguel, P.; Bonnefond, A. How recent advances in genomics improve precision diagnosis and personalized care of maturity-onset diabetes of the young. Curr. Diab. Rep. 2019, 19, 79. [Google Scholar] [CrossRef] [PubMed]
  8. Vaxillaire, M.; Froguel, P. Monogenic diabetes: Implementation of translational genomic research towards precision medicine. J. Diabetes 2016, 8, 782–795. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Zhang, H.; Colclough, K.; Gloyn, A.L.; Pollin, T.I. Monogenic diabetes: A gateway to precision medicine in diabetes. J. Clin. Investig. 2021, 131, e142244. [Google Scholar] [CrossRef]
  10. Tattersall, R.B.; Fajans, S.S.; Arbor, A. A difference between the inheritance of classical juvenile-onset and maturity-onset type diabetes of young people. Diabetes 1975, 24, 44–53. [Google Scholar] [CrossRef]
  11. Thanabalasingham, G.; Pal, A.; Selwood, M.P.; Dudley, C.; Fisher, K.; Bingley, P.J.; Ellard, S.; Farmer, A.J.; McCarthy, M.I.; Owen, K.R. Systematic assessment of etiology in adults with a clinical diagnosis of young-onset type 2 diabetes is a successful strategy for identifying maturity-onset diabetes of the young. Diabetes Care 2012, 35, 1206–1212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Kleinberger, J.W.; Copeland, K.C.; Gandica, R.G.; Haymond, M.W.; Levitsky, L.L.; Linder, B.; Shuldiner, A.R.; Tollefsen, S.; White, N.H.; Pollin, T.I. Monogenic diabetes in overweight and obese youth diagnosed with type 2 diabetes: The today clinical trial. Genet. Med. 2018, 20, 583–590. [Google Scholar] [CrossRef] [Green Version]
  13. Naylor, R.N.; Greeley, S.A.W.; Bell, G.I.; Philipson, L.H. Genetics and pathophysiology of neonatal diabetes mellitus. J. Diabetes Investig. 2011, 2, 158–169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Polak, M.; Cavé, H. Neonatal diabetes mellitus: A disease linked to multiple mechanisms. Orphanet J. Rare Dis. 2007, 2, 12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Gloyn, A.L.; Pearson, E.R.; Antcliff, J.F.; Proks, P.; Bruining, G.J.; Slingerland, A.S.; Howard, N.; Srinivasan, S.; Silva, J.M.; Molnes, J.; et al. Activating mutations in the gene encoding the atp-sensitive potassium-channel subunit kir6.2 and permanent neonatal diabetes. N. Engl. J. Med. 2004, 350, 1838–1849. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Koster, J.C.; Cadario, F.; Peruzzi, C.; Colombo, C.; Nichols, C.G.; Barbetti, F. The g53d mutation in kir6.2 (kcnj11) is associated with neonatal diabetes and motor dysfunction in adulthood that is improved with sulfonylurea therapy. J. Clin. Endocrinol. Metab. 2008, 93, 1054–1061. [Google Scholar] [CrossRef] [PubMed]
  17. Masia, R.; Koster, J.C.; Tumini, S.; Chiarelli, F.; Colombo, C.; Nichols, C.G.; Barbetti, F. An atp-binding mutation (g334d) in kcnj11 is associated with a sulfonylurea-insensitive form of developmental delay, epilepsy, and neonatal diabetes. Diabetes 2007, 56, 328–336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Beltrand, J.; Elie, C.; Busiah, K.; Fournier, E.; Boddaert, N.; Bahi-Buisson, N.; Vera, M.; Bui-Quoc, E.; Ingster-Moati, I.; Berdugo, M.; et al. Sulfonylurea therapy benefits neurological and psychomotor functions in patients with neonatal diabetes owing to potassium channel mutations. Diabetes Care 2015, 38, 2033–2041. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Bowman, P.; Broadbridge, E.; Knight, B.A.; Pettit, L.; Flanagan, S.E.; Reville, M.; Tonks, J.; Shepherd, M.H.; Ford, T.J.; Hattersley, A.T. Psychiatric morbidity in children with kcnj11 neonatal diabetes. Diabet. Med. 2016, 33, 1387–1391. [Google Scholar] [CrossRef] [PubMed]
  20. Bowman, P.; Hattersley, A.T.; Knight, B.A.; Broadbridge, E.; Pettit, L.; Reville, M.; Flanagan, S.E.; Shepherd, M.H.; Ford, T.J.; Tonks, J. Neuropsychological impairments in children with kcnj11 neonatal diabetes. Diabet. Med. 2017, 34, 1171–1173. [Google Scholar] [CrossRef] [PubMed]
  21. Bowman, P.; Day, J.; Torrens, L.; Shepherd, M.H.; Knight, B.A.; Ford, T.J.; Flanagan, S.E.; Chakera, A.; Hattersley, A.T.; Zeman, A. Cognitive, neurological, and behavioral features in adults with. Diabetes Care 2019, 42, 215–224. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Busiah, K.; Drunat, S.; Vaivre-Douret, L.; Bonnefond, A.; Simon, A.; Flechtner, I.; Gérard, B.; Pouvreau, N.; Elie, C.; Nimri, R.; et al. Neuropsychological dysfunction and developmental defects associated with genetic changes in infants with neonatal diabetes mellitus: A prospective cohort study [corrected]. Lancet Diabetes Endocrinol. 2013, 1, 199–207. [Google Scholar] [CrossRef]
  23. Landmeier, K.A.; Lanning, M.; Carmody, D.; Greeley, S.A.W.; Msall, M.E. Adhd, learning difficulties and sleep disturbances associated with kcnj11-related neonatal diabetes. Pediatr. Diabetes 2017, 18, 518–523. [Google Scholar] [CrossRef]
  24. Barrett, T.G. Differential diagnosis of type 1 diabetes: Which genetic syndromes need to be considered? Pediatr. Diabetes 2007, 8, 15–23. [Google Scholar] [CrossRef]
  25. Trischitta, V.; Prudente, S.; Doria, A. Disentangling the heterogeneity of adulthood-onset non-autoimmune diabetes: A little closer but lot more to do. Curr. Opin. Pharm. 2020, 55, 157–164. [Google Scholar] [CrossRef]
  26. Inagaki, N.; Gonoi, T.; Clement, J.P.; Namba, N.; Inazawa, J.; Gonzalez, G.; Aguilar-Bryan, L.; Seino, S.; Bryan, J. Reconstitution of ikatp: An inward rectifier subunit plus the sulfonylurea receptor. Science 1995, 270, 1166–1170. [Google Scholar] [CrossRef]
  27. Pipatpolkai, T.; Usher, S.; Stansfeld, P.J.; Ashcroft, F.M. New insights into K ATP channel gene mutations and neonatal diabetes mellitus. Nat. Rev. Endocrinol. 2020, 16, 378–393. [Google Scholar] [CrossRef]
  28. Haghvirdizadeh, P.; Sadat Haerian, M.; Sadat Haerian, B. Abcc8 genetic variants and risk of diabetes mellitus. Gene 2014, 545, 198–204. [Google Scholar] [CrossRef] [PubMed]
  29. Ashcroft, F.M.; Harrison, D.E.; Ashcroft, S.J. Glucose induces closure of single potassium channels in isolated rat pancreatic β-cells. Nature 1984, 312, 446–448. [Google Scholar] [CrossRef]
  30. Miki, T.; Seino, S. Roles of katp channels as metabolic sensors in acute metabolic changes. J. Mol. Cell Cardiol. 2005, 38, 917–925. [Google Scholar] [CrossRef] [PubMed]
  31. Tarasov, A.; Dusonchet, J.; Ashcroft, F. Metabolic regulation of the pancreatic β-cell atp-sensitive k+ channel: A pas de deux. Diabetes 2004, 53, S113–S122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Zhou, Q.; Garin, I.; Castaño, L.; Argente, J.; Muñoz-Calvo, M.T.; Perez de Nanclares, G.; Shyng, S.L. Neonatal diabetes caused by mutations in sulfonylurea receptor 1: Interplay between expression and mg-nucleotide gating defects of atp-sensitive potassium channels. J. Clin. Endocrinol. Metab. 2010, 95, E473–E478. [Google Scholar] [CrossRef] [Green Version]
  33. De Franco, E.; Saint-Martin, C.; Brusgaard, K.; Knight Johnson, A.E.; Aguilar-Bryan, L.; Bowman, P.; Arnoux, J.B.; Larsen, A.R.; Sanyoura, M.; Greeley, S.A.W.; et al. Update of variants identified in the pancreatic β-cell k. Hum. Mutat. 2020, 41, 884–905. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Massa, O.; Iafusco, D.; d’Amato, E.; Gloyn, A.L.; Hattersley, A.T.; Pasquino, B.; Tonini, G.; Dammacco, F.; Zanette, G.; Meschi, F.; et al. Kcnj11 activating mutations in italian patients with permanent neonatal diabetes. Hum. Mutat. 2005, 25, 22–27. [Google Scholar] [CrossRef] [PubMed]
  35. Stanik, J.; Gasperikova, D.; Paskova, M.; Barak, L.; Javorkova, J.; Jancova, E.; Ciljakova, M.; Hlava, P.; Michalek, J.; Flanagan, S.E.; et al. Prevalence of permanent neonatal diabetes in slovakia and successful replacement of insulin with sulfonylurea therapy in kcnj11 and abcc8 mutation carriers. J. Clin. Endocrinol. Metab. 2007, 92, 1276–1282. [Google Scholar] [CrossRef] [PubMed]
  36. Slingerland, A.S.; Shields, B.M.; Flanagan, S.E.; Bruining, G.J.; Noordam, K.; Gach, A.; Mlynarski, W.; Malecki, M.T.; Hattersley, A.T.; Ellard, S. Referral rates for diagnostic testing support an incidence of permanent neonatal diabetes in three european countries of at least 1 in 260,000 live births. Diabetologia 2009, 52, 1683–1685. [Google Scholar] [CrossRef] [Green Version]
  37. Gloyn, A.L.; Reimann, F.; Girard, C.; Edghill, E.L.; Proks, P.; Pearson, E.R.; Temple, I.K.; Mackay, D.J.; Shield, J.P.; Freedenberg, D.; et al. Relapsing diabetes can result from moderately activating mutations in kcnj11. Hum. Mol. Genet. 2005, 14, 925–934. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Liu, L.; Nagashima, K.; Yasuda, T.; Liu, Y.; Hu, H.R.; He, G.; Feng, B.; Zhao, M.; Zhuang, L.; Zheng, T.; et al. Mutations in kcnj11 are associated with the development of autosomal dominant, early-onset type 2 diabetes. Diabetologia 2013, 56, 2609–2618. [Google Scholar] [CrossRef] [Green Version]
  39. Edghill, E.L.; Flanagan, S.E.; Ellard, S. Permanent neonatal diabetes due to activating mutations in abcc8 and kcnj11. Rev. Endocr. Metab. Disord. 2010, 11, 193–198. [Google Scholar] [CrossRef] [PubMed]
  40. Ellard, S.; Flanagan, S.E.; Girard, C.A.; Patch, A.M.; Harries, L.W.; Parrish, A.; Edghill, E.L.; Mackay, D.J.; Proks, P.; Shimomura, K.; et al. Permanent neonatal diabetes caused by dominant, recessive, or compound heterozygous sur1 mutations with opposite functional effects. Am. J. Hum. Genet. 2007, 81, 375–382. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Vedovato, N.; Cliff, E.; Proks, P.; Poovazhagi, V.; Flanagan, S.E.; Ellard, S.; Hattersley, A.T.; Ashcroft, F.M. Neonatal diabetes caused by a homozygous kcnj11 mutation demonstrates that tiny changes in atp sensitivity markedly affect diabetes risk. Diabetologia 2016, 59, 1430–1436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Gloyn, A.L.; Cummings, E.A.; Edghill, E.L.; Harries, L.W.; Scott, R.; Costa, T.; Temple, I.K.; Hattersley, A.T.; Ellard, S. Permanent neonatal diabetes due to paternal germline mosaicism for an activating mutation of the kcnj11 gene encoding the kir6.2 subunit of the β-cell potassium adenosine triphosphate channel. J. Clin. Endocrinol. Metab. 2004, 89, 3932–3935. [Google Scholar] [CrossRef] [Green Version]
  43. Edghill, E.L.; Gloyn, A.L.; Goriely, A.; Harries, L.W.; Flanagan, S.E.; Rankin, J.; Hattersley, A.T.; Ellard, S. Origin of de novo kcnj11 mutations and risk of neonatal diabetes for subsequent siblings. J. Clin. Endocrinol. Metab. 2007, 92, 1773–1777. [Google Scholar] [CrossRef] [Green Version]
  44. Bowman, P.; Flanagan, S.E.; Edghill, E.L.; Damhuis, A.; Shepherd, M.H.; Paisey, R.; Hattersley, A.T.; Ellard, S. Heterozygous abcc8 mutations are a cause of mody. Diabetologia 2012, 55, 123–127. [Google Scholar] [CrossRef]
  45. Cattoni, A.; Jackson, C.; Bain, M.; Houghton, J.; Wei, C. Phenotypic variability in two siblings with monogenic diabetes due to the same abcc8 gene mutation. Pediatr. Diabetes 2019, 20, 482–485. [Google Scholar] [CrossRef]
  46. Gonsorcikova, L.; Vaxillaire, M.; Pruhova, S.; Dechaume, A.; Dusatkova, P.; Cinek, O.; Pedersen, O.; Froguel, P.; Hansen, T.; Lebl, J. Familial mild hyperglycemia associated with a novel abcc8-v84i mutation within three generations. Pediatr. Diabetes 2011, 12, 266–269. [Google Scholar] [CrossRef]
  47. Bonnefond, A.; Philippe, J.; Durand, E.; Dechaume, A.; Huyvaert, M.; Montagne, L.; Marre, M.; Balkau, B.; Fajardy, I.; Vambergue, A.; et al. Whole-exome sequencing and high throughput genotyping identified kcnj11 as the thirteenth mody gene. PLoS ONE 2012, 7, e37423. [Google Scholar] [CrossRef]
  48. Ang, S.F.; Lim, S.C.; Tan, C.S.H.; Fong, J.C.; Kon, W.Y.; Lian, J.X.; Subramanium, T.; Sum, C.F. A preliminary study to evaluate the strategy of combining clinical criteria and next generation sequencing (ngs) for the identification of monogenic diabetes among multi-ethnic asians. Diabetes Res. Clin. Pract. 2016, 119, 13–22. [Google Scholar] [CrossRef]
  49. Ashcroft, F.M. Atp-sensitive potassium channelopathies: Focus on insulin secretion. J. Clin. Investig. 2005, 115, 2047–2058. [Google Scholar] [CrossRef] [Green Version]
  50. Nichols, C.G.; Shyng, S.L.; Nestorowicz, A.; Glaser, B.; Clement, J.P.; Gonzalez, G.; Aguilar-Bryan, L.; Permutt, M.A.; Bryan, J. Adenosine diphosphate as an intracellular regulator of insulin secretion. Science 1996, 272, 1785–1787. [Google Scholar] [CrossRef]
  51. Taschenberger, G.; Mougey, A.; Shen, S.; Lester, L.B.; LaFranchi, S.; Shyng, S.L. Identification of a familial hyperinsulinism-causing mutation in the sulfonylurea receptor 1 that prevents normal trafficking and function of katp channels. J. Biol. Chem. 2002, 277, 17139–17146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Thomas, P.; Ye, Y.; Lightner, E. Mutation of the pancreatic islet inward rectifier kir6.2 also leads to familial persistent hyperinsulinemic hypoglycemia of infancy. Hum. Mol. Genet. 1996, 5, 1809–1812. [Google Scholar] [CrossRef]
  53. Pinney, S.E.; Ganapathy, K.; Bradfield, J.; Stokes, D.; Sasson, A.; Mackiewicz, K.; Boodhansingh, K.; Hughes, N.; Becker, S.; Givler, S.; et al. Dominant form of congenital hyperinsulinism maps to hk1 region on 10q. Horm Res. Paediatr. 2013, 80, 18–27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Kapoor, R.R.; Flanagan, S.E.; Arya, V.B.; Shield, J.P.; Ellard, S.; Hussain, K. Clinical and molecular characterisation of 300 patients with congenital hyperinsulinism. Eur. J. Endocrinol. 2013, 168, 557–564. [Google Scholar] [CrossRef]
  55. Snider, K.E.; Becker, S.; Boyajian, L.; Shyng, S.L.; MacMullen, C.; Hughes, N.; Ganapathy, K.; Bhatti, T.; Stanley, C.A.; Ganguly, A. Genotype and phenotype correlations in 417 children with congenital hyperinsulinism. J. Clin. Endocrinol. Metab. 2013, 98, E355–E363. [Google Scholar] [CrossRef]
  56. Gϋemes, M.; Rahman, S.A.; Kapoor, R.R.; Flanagan, S.; Houghton, J.A.L.; Misra, S.; Oliver, N.; Dattani, M.T.; Shah, P. Hyperinsulinemic hypoglycemia in children and adolescents: Recent advances in understanding of pathophysiology and management. Rev. Endocr. Metab. Disord. 2020, 21, 577–597. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Giri, D.; Hawton, K.; Senniappan, S. Congenital hyperinsulinism: Recent updates on molecular mechanisms, diagnosis and management. J. Pediatr. Endocrinol. Metab. 2021, 10, 1515. [Google Scholar] [CrossRef]
  58. Carmody, D.; Bell, C.D.; Hwang, J.L.; Dickens, J.T.; Sima, D.I.; Felipe, D.L.; Zimmer, C.A.; Davis, A.O.; Kotlyarevska, K.; Naylor, R.N.; et al. Sulfonylurea treatment before genetic testing in neonatal diabetes: Pros and cons. J. Clin. Endocrinol. Metab. 2014, 99, E2709–E2714. [Google Scholar] [CrossRef] [PubMed]
  59. Pearson, E.R.; Flechtner, I.; Njølstad, P.R.; Malecki, M.T.; Flanagan, S.E.; Larkin, B.; Ashcroft, F.M.; Klimes, I.; Codner, E.; Iotova, V.; et al. Switching from insulin to oral sulfonylureas in patients with diabetes due to kir6.2 mutations. N. Engl. J. Med. 2006, 355, 467–477. [Google Scholar] [CrossRef] [Green Version]
  60. Rafiq, M.; Flanagan, S.E.; Patch, A.M.; Shields, B.M.; Ellard, S.; Hattersley, A.T.; Neonatal Diabetes International Collaborative Group. Effective treatment with oral sulfonylureas in patients with diabetes due to sulfonylurea receptor 1 (sur1) mutations. Diabetes Care 2008, 31, 204–209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Babiker, T.; Vedovato, N.; Patel, K.; Thomas, N.; Finn, R.; Männikkö, R.; Chakera, A.J.; Flanagan, S.E.; Shepherd, M.H.; Ellard, S.; et al. Successful transfer to sulfonylureas in kcnj11 neonatal diabetes is determined by the mutation and duration of diabetes. Diabetologia 2016, 59, 1162–1166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Thurber, B.W.; Carmody, D.; Tadie, E.C.; Pastore, A.N.; Dickens, J.T.; Wroblewski, K.E.; Naylor, R.N.; Philipson, L.H.; Greeley, S.A.; United States Neonatal Diabetes Working Group. Age at the time of sulfonylurea initiation influences treatment outcomes in kcnj11-related neonatal diabetes. Diabetologia 2015, 58, 1430–1435. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Bowman, P.; Sulen, Å.; Barbetti, F.; Beltrand, J.; Svalastoga, P.; Codner, E.; Tessmann, E.H.; Juliusson, P.B.; Skrivarhaug, T.; Pearson, E.R.; et al. Effectiveness and safety of long-term treatment with sulfonylureas in patients with neonatal diabetes due to kcnj11 mutations: An international cohort study. Lancet Diabetes Endocrinol. 2018, 6, 637–646. [Google Scholar] [CrossRef]
  64. Fendler, W.; Pietrzak, I.; Brereton, M.F.; Lahmann, C.; Gadzicki, M.; Bienkiewicz, M.; Drozdz, I.; Borowiec, M.; Malecki, M.T.; Ashcroft, F.M.; et al. Switching to sulphonylureas in children with idend syndrome caused by kcnj11 mutations results in improved cerebellar perfusion. Diabetes Care 2013, 36, 2311–2316. [Google Scholar] [CrossRef] [Green Version]
  65. Støy, J.; Greeley, S.A.; Paz, V.P.; Ye, H.; Pastore, A.N.; Skowron, K.B.; Lipton, R.B.; Cogen, F.R.; Bell, G.I.; Philipson, L.H.; et al. Diagnosis and treatment of neonatal diabetes: A united states experience. Pediatr. Diabetes 2008, 9, 450–459. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Myngheer, N.; Allegaert, K.; Hattersley, A.; McDonald, T.; Kramer, H.; Ashcroft, F.M.; Verhaeghe, J.; Mathieu, C.; Casteels, K. Fetal macrosomia and neonatal hyperinsulinemic hypoglycemia associated with transplacental transfer of sulfonylurea in a mother with kcnj11-related neonatal diabetes. Diabetes Care 2014, 37, 3333–3335. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Klupa, T.; Kozek, E.; Nowak, N.; Cyganek, K.; Gach, A.; Milewicz, T.; Czajkowski, K.; Tolloczko, J.; Mlynarski, W.; Malecki, M.T. The first case report of sulfonylurea use in a woman with permanent neonatal diabetes mellitus due to kcnj11 mutation during a high-risk pregnancy. Clin. Endocrinol. Metab. 2010, 95, 3599–3604. [Google Scholar] [CrossRef] [Green Version]
  68. Gaal, Z.; Klupa, T.; Kantor, I.; Mlynarski, W.; Albert, L.; Tolloczko, J.; Balogh, I.; Czajkowski, K.; Malecki, M.T. Sulfonylurea use during entire pregnancy in diabetes because of kcnj11 mutation: A report of two cases. Diabetes Care 2012, 35, e40. [Google Scholar] [CrossRef] [Green Version]
  69. Shepherd, M.; Brook, A.J.; Chakera, A.J.; Hattersley, A.T. Management of sulfonylurea-treated monogenic diabetes in pregnancy: Implications of placental glibenclamide transfer. Diabet. Med. 2017, 34, 1332–1339. [Google Scholar] [CrossRef] [PubMed]
  70. Slingerland, A.S.; Hattersley, A.T. Activating mutations in the gene encoding kir6.2 alter fetal and postnatal growth and also cause neonatal diabetes. J. Clin. Endocrinol. Metab. 2006, 91, 2782–2788. [Google Scholar] [CrossRef] [Green Version]
  71. Dickens, L.T.; Naylor, R.N. Clinical management of women with monogenic diabetes during pregnancy. Curr. Diab. Rep. 2018, 18, 12. [Google Scholar] [CrossRef]
  72. Chakera, A.J.; Steele, A.M.; Gloyn, A.L.; Shepherd, M.H.; Shields, B.; Ellard, S.; Hattersley, A.T. Recognition and management of individuals with hyperglycemia because of a heterozygous glucokinase mutation. Diabetes Care 2015, 38, 1383–1392. [Google Scholar] [CrossRef] [Green Version]
  73. Nolan, C.J.; Prentki, M. The islet β-cell: Fuel responsive and vulnerable. Trends. Endocrinol. Metab. 2008, 19, 285–291. [Google Scholar] [CrossRef] [PubMed]
  74. Osbak, K.K.; Colclough, K.; Saint-Martin, C.; Beer, N.L.; Bellanné-Chantelot, C.; Ellard, S.; Gloyn, A.L. Update on mutations in glucokinase (gck), which cause maturity-onset diabetes of the young, permanent neonatal diabetes, and hyperinsulinemic hypoglycemia. Hum. Mutat. 2009, 30, 1512–1526. [Google Scholar] [CrossRef]
  75. Ellard, S.; Beards, F.; Allen, L.I.; Shepherd, M.; Ballantyne, E.; Harvey, R.; Hattersley, A.T. A high prevalence of glucokinase mutations in gestational diabetic subjects selected by clinical criteria. Diabetologia 2000, 43, 250–253. [Google Scholar] [CrossRef] [Green Version]
  76. Steele, A.M.; Shields, B.M.; Wensley, K.J.; Colclough, K.; Ellard, S.; Hattersley, A.T. Prevalence of vascular complications among patients with glucokinase mutations and prolonged, mild hyperglycemia. JAMA 2014, 311, 279–286. [Google Scholar] [CrossRef] [Green Version]
  77. Chakera, A.J.; Spyer, G.; Vincent, N.; Ellard, S.; Hattersley, A.T.; Dunne, F.P. The 0.1% of the population with glucokinase monogenic diabetes can be recognized by clinical characteristics in pregnancy: The atlantic diabetes in pregnancy cohort. Diabetes Care 2014, 37, 1230–1236. [Google Scholar] [CrossRef] [Green Version]
  78. Dickens, L.T.; Letourneau, L.R.; Sanyoura, M.; Greeley, S.A.W.; Philipson, L.H.; Naylor, R.N. Management and pregnancy outcomes of women with gck-mody enrolled in the us monogenic diabetes registry. Acta Diabetol. 2019, 56, 405–411. [Google Scholar] [CrossRef]
  79. Raimondo, A.; Chakera, A.J.; Thomsen, S.K.; Colclough, K.; Barrett, A.; de Franco, E.; Chatelas, A.; Demirbilek, H.; Akcay, T.; Alawneh, H.; et al. Phenotypic severity of homozygous gck mutations causing neonatal or childhood-onset diabetes is primarily mediated through effects on protein stability. Hum. Mol. Genet. 2014, 23, 6432–6440. [Google Scholar] [CrossRef] [PubMed]
  80. Marucci, A.; Biagini, T.; Di Paola, R.; Menzaghi, C.; Fini, G.; Castellana, S.; Cardinale, G.M.; Mazza, T.; Trischitta, V. Association of a homozygous gck missense mutation with mild diabetes. Mol. Genet. Genomic Med. 2019, 7, e00728. [Google Scholar] [CrossRef] [Green Version]
  81. Gloyn, A.L. Glucokinase (gck) mutations in hyper- and hypoglycemia: Maturity-onset diabetes of the young, permanent neonatal diabetes, and hyperinsulinemia of infancy. Hum. Mutat. 2003, 22, 353–362. [Google Scholar] [CrossRef]
  82. Bishay, R.H.; Greenfield, J.R. A review of maturity onset diabetes of the young (mody) and challenges in the management of glucokinase-mody. Med. J. Aust. 2016, 205, 480–485. [Google Scholar] [CrossRef]
  83. Spyer, G.; Hattersley, A.T.; Sykes, J.E.; Sturley, R.H.; MacLeod, K.M. Influence of maternal and fetal glucokinase mutations in gestational diabetes. Am. J. Obstet. Gynecol. 2001, 185, 240–241. [Google Scholar] [CrossRef]
  84. Spyer, G.; Macleod, K.M.; Shepherd, M.; Ellard, S.; Hattersley, A.T. Pregnancy outcome in patients with raised blood glucose due to a heterozygous glucokinase gene mutation. Diabet. Med. 2009, 26, 14–18. [Google Scholar] [CrossRef]
  85. Harries, L.W.; Ellard, S.; Stride, A.; Morgan, N.G.; Hattersley, A.T. Isomers of the tcf1 gene encoding hepatocyte nuclear factor-1 α show differential expression in the pancreas and define the relationship between mutation position and clinical phenotype in monogenic diabetes. Hum. Mol. Genet. 2006, 15, 2216–2224. [Google Scholar] [CrossRef] [Green Version]
  86. Bellanné-Chantelot, C.; Carette, C.; Riveline, J.P.; Valéro, R.; Gautier, J.F.; Larger, E.; Reznik, Y.; Ducluzeau, P.H.; Sola, A.; Hartemann-Heurtier, A.; et al. The type and the position of hnf1a mutation modulate age at diagnosis of diabetes in patients with maturity-onset diabetes of the young (mody)-3. Diabetes 2008, 57, 503–508. [Google Scholar] [CrossRef] [Green Version]
  87. Cereghini, S. Liver-enriched transcription factors and hepatocyte differentiation. FASEB J. 1996, 10, 267–282. [Google Scholar] [CrossRef]
  88. Fajans, S.S.; Bell, G.I.; Polonsky, K.S. Molecular mechanisms and clinical pathophysiology of maturity-onset diabetes of the young. N. Engl. J. Med. 2001, 345, 971–980. [Google Scholar] [CrossRef]
  89. Fajans, S.S.; Bell, G.I. Phenotypic heterogeneity between different mutations of mody subtypes and within mody pedigrees. Diabetologia 2006, 49, 1106–1108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Colclough, K.; Bellanne-Chantelot, C.; Saint-Martin, C.; Flanagan, S.E.; Ellard, S. Mutations in the genes encoding the transcription factors hepatocyte nuclear factor 1 α and 4 α in maturity-onset diabetes of the young and hyperinsulinemic hypoglycemia. Hum. Mutat. 2013, 34, 669–685. [Google Scholar] [CrossRef]
  91. Lek, M.; Karczewski, K.J.; Minikel, E.V.; Samocha, K.E.; Banks, E.; Fennell, T.; O’Donnell-Luria, A.H.; Ware, J.S.; Hill, A.J.; Cummings, B.B.; et al. Analysis of protein-coding genetic variation in 60,706 humans. Nature 2016, 536, 285–291. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Misra, S.; Hassanali, N.; Bennett, A.J.; Juszczak, A.; Caswell, R.; Colclough, K.; Valabhji, J.; Ellard, S.; Oliver, N.S.; Gloyn, A.L. Homozygous hypomorphic. Diabetes Care 2020, 43, 909–912. [Google Scholar] [CrossRef] [Green Version]
  93. Naqvi, A.A.T.; Hasan, G.M.; Hassan, M.I. Investigating the role of transcription factors of pancreas development in pancreatic cancer. Pancreatology 2018, 18, 184–190. [Google Scholar] [CrossRef]
  94. Vesterhus, M.; Haldorsen, I.S.; Raeder, H.; Molven, A.; Njølstad, P.R. Reduced pancreatic volume in hepatocyte nuclear factor 1a-maturity-onset diabetes of the young. J. Clin. Endocrinol. Metab. 2008, 93, 3505–3509. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Hansen, S.K.; Párrizas, M.; Jensen, M.L.; Pruhova, S.; Ek, J.; Boj, S.F.; Johansen, A.; Maestro, M.A.; Rivera, F.; Eiberg, H.; et al. Genetic evidence that hnf-1α-dependent transcriptional control of hnf-4α is essential for human pancreatic β cell function. J. Clin. Investig. 2002, 110, 827–833. [Google Scholar] [CrossRef] [PubMed]
  96. Wang, H.; Maechler, P.; Hagenfeldt, K.A.; Wollheim, C.B. Dominant-negative suppression of hnf-1α function results in defective insulin gene transcription and impaired metabolism-secretion coupling in a pancreatic β-cell line. EMBO J. 1998, 17, 6701–6713. [Google Scholar] [CrossRef] [Green Version]
  97. Wang, H.; Antinozzi, P.A.; Hagenfeldt, K.A.; Maechler, P.; Wollheim, C.B. Molecular targets of a human hnf1 α mutation responsible for pancreatic β-cell dysfunction. EMBO J. 2000, 19, 4257–4264. [Google Scholar] [CrossRef] [Green Version]
  98. Shih, D.Q.; Screenan, S.; Munoz, K.N.; Philipson, L.; Pontoglio, M.; Yaniv, M.; Polonsky, K.S.; Stoffel, M. Loss of hnf-1α function in mice leads to abnormal expression of genes involved in pancreatic islet development and metabolism. Diabetes 2001, 50, 2472–2480. [Google Scholar] [CrossRef] [Green Version]
  99. Yamagata, K.; Nammo, T.; Moriwaki, M.; Ihara, A.; Iizuka, K.; Yang, Q.; Satoh, T.; Li, M.; Uenaka, R.; Okita, K.; et al. Overexpression of dominant-negative mutant hepatocyte nuclear fctor-1 α in pancreatic β-cells causes abnormal islet architecture with decreased expression of e-cadherin, reduced β-cell proliferation, and diabetes. Diabetes 2002, 51, 114–123. [Google Scholar] [CrossRef] [Green Version]
  100. Yang, Q.; Yamagata, K.; Fukui, K.; Cao, Y.; Nammo, T.; Iwahashi, H.; Wang, H.; Matsumura, I.; Hanafusa, T.; Bucala, R.; et al. Hepatocyte nuclear factor-1α modulates pancreatic β-cell growth by regulating the expression of insulin-like growth factor-1 in ins-1 cells. Diabetes 2002, 51, 1785–1792. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Wobser, H.; Düssmann, H.; Kögel, D.; Wang, H.; Reimertz, C.; Wollheim, C.B.; Byrne, M.M.; Prehn, J.H. Dominant-negative suppression of hnf-1 α results in mitochondrial dysfunction, ins-1 cell apoptosis, and increased sensitivity to ceramide-, but not to high glucose-induced cell death. J. Biol. Chem. 2002, 277, 6413–6421. [Google Scholar] [CrossRef] [Green Version]
  102. Byrne, M.M.; Sturis, J.; Menzel, S.; Yamagata, K.; Fajans, S.S.; Dronsfield, M.J.; Bain, S.C.; Hattersley, A.T.; Velho, G.; Froguel, P.; et al. Altered insulin secretory responses to glucose in diabetic and nondiabetic subjects with mutations in the diabetes susceptibility gene mody3 on chromosome 12. Diabetes 1996, 45, 1503–1510. [Google Scholar] [CrossRef] [PubMed]
  103. Dukes, I.D.; Sreenan, S.; Roe, M.W.; Levisetti, M.; Zhou, Y.P.; Ostrega, D.; Bell, G.I.; Pontoglio, M.; Yaniv, M.; Philipson, L.; et al. Defective pancreatic β-cell glycolytic signaling in hepatocyte nuclear factor-1α-deficient mice. J. Biol. Chem. 1998, 273, 24457–24464. [Google Scholar] [CrossRef] [Green Version]
  104. Pontoglio, M.; Sreenan, S.; Roe, M.; Pugh, W.; Ostrega, D.; Doyen, A.; Pick, A.J.; Baldwin, A.; Velho, G.; Froguel, P.; et al. Defective insulin secretion in hepatocyte nuclear factor 1α-deficient mice. J. Clin. Investig. 1998, 101, 2215–2222. [Google Scholar] [CrossRef]
  105. Stride, A.; Vaxillaire, M.; Tuomi, T.; Barbetti, F.; Njølstad, P.R.; Hansen, T.; Costa, A.; Conget, I.; Pedersen, O.; Søvik, O.; et al. The genetic abnormality in the β cell determines the response to an oral glucose load. Diabetologia 2002, 45, 427–435. [Google Scholar] [CrossRef]
  106. Pontoglio, M.; Prié, D.; Cheret, C.; Doyen, A.; Leroy, C.; Froguel, P.; Velho, G.; Yaniv, M.; Friedlander, G. Hnf1α controls renal glucose reabsorption in mouse and man. EMBO Rep. 2000, 1, 359–365. [Google Scholar] [CrossRef] [Green Version]
  107. Pearson, E.R.; Starkey, B.J.; Powell, R.J.; Gribble, F.M.; Clark, P.M.; Hattersley, A.T. Genetic cause of hyperglycaemia and response to treatment in diabetes. Lancet 2003, 362, 1275–1281. [Google Scholar] [CrossRef]
  108. Bluteau, O.; Jeannot, E.; Bioulac-Sage, P.; Marqués, J.M.; Blanc, J.F.; Bui, H.; Beaudoin, J.C.; Franco, D.; Balabaud, C.; Laurent-Puig, P.; et al. Bi-allelic inactivation of tcf1 in hepatic adenomas. Nat. Genet. 2002, 32, 312–315. [Google Scholar] [CrossRef]
  109. Reznik, Y.; Dao, T.; Coutant, R.; Chiche, L.; Jeannot, E.; Clauin, S.; Rousselot, P.; Fabre, M.; Oberti, F.; Fatome, A.; et al. Hepatocyte nuclear factor-1 α gene inactivation: Cosegregation between liver adenomatosis and diabetes phenotypes in two maturity-onset diabetes of the young (mody)3 families. J. Clin. Endocrinol. Metab. 2004, 89, 1476–1480. [Google Scholar] [CrossRef]
  110. Menzel, R.; Kaisaki, P.J.; Rjasanowski, I.; Heinke, P.; Kerner, W.; Menzel, S. A low renal threshold for glucose in diabetic patients with a mutation in the hepatocyte nuclear factor-1α (hnf-1α) gene. Diabet Med. 1998, 15, 816–820. [Google Scholar] [CrossRef]
  111. Rebouissou, S.; Imbeaud, S.; Balabaud, C.; Boulanger, V.; Bertrand-Michel, J.; Tercé, F.; Auffray, C.; Bioulac-Sage, P.; Zucman-Rossi, J. Hnf1α inactivation promotes lipogenesis in human hepatocellular adenoma independently of srebp-1 and carbohydrate-response element-binding protein (chrebp) activation. J. Biol. Chem. 2007, 282, 14437–14446. [Google Scholar] [CrossRef] [Green Version]
  112. Nyunt, O.; Wu, J.Y.; McGown, I.N.; Harris, M.; Huynh, T.; Leong, G.M.; Cowley, D.M.; Cotterill, A.M. Investigating maturity onset diabetes of the young. Clin. Biochem. Rev. 2009, 30, 67–74. [Google Scholar]
  113. Pruhova, S.; Dusatkova, P.; Neumann, D.; Hollay, E.; Cinek, O.; Lebl, J.; Sumnik, Z. Two cases of diabetic ketoacidosis in hnf1a-mody linked to severe dehydration: Is it time to change the diagnostic criteria for mody? Diabetes Care 2013, 36, 2573–2574. [Google Scholar] [CrossRef] [Green Version]
  114. Steele, A.M.; Shields, B.M.; Shepherd, M.; Ellard, S.; Hattersley, A.T.; Pearson, E.R. Increased all-cause and cardiovascular mortality in monogenic diabetes as a result of mutations in the hnf1a gene. Diabet. Med. 2010, 27, 157–161. [Google Scholar] [CrossRef]
  115. Klupa, T.; Warram, J.H.; Antonellis, A.; Pezzolesi, M.; Nam, M.; Malecki, M.T.; Doria, A.; Rich, S.S.; Krolewski, A.S. Determinants of the development of diabetes (maturity-onset diabetes of the young-3) in carriers of hnf-1α mutations: Evidence for parent-of-origin effect. Diabetes Care 2002, 25, 2292–2301. [Google Scholar] [CrossRef] [Green Version]
  116. Shepherd, M.; Shields, B.; Ellard, S.; Rubio-Cabezas, O.; Hattersley, A.T. A genetic diagnosis of hnf1a diabetes alters treatment and improves glycaemic control in the majority of insulin-treated patients. Diabet. Med. 2009, 26, 437–441. [Google Scholar] [CrossRef]
  117. Pearson, E.R.; Liddell, W.G.; Shepherd, M.; Corrall, R.J.; Hattersley, A.T. Sensitivity to sulphonylureas in patients with hepatocyte nuclear factor-1α gene mutations: Evidence for pharmacogenetics in diabetes. Diabet. Med. 2000, 17, 543–545. [Google Scholar] [CrossRef]
  118. Becker, M.; Galler, A.; Raile, K. Meglitinide analogues in adolescent patients with hnf1a-mody (mody 3). Pediatrics 2014, 133, e775–e779. [Google Scholar] [CrossRef] [Green Version]
  119. Raile, K.; Schober, E.; Konrad, K.; Thon, A.; Grulich-Henn, J.; Meissner, T.; Wölfle, J.; Scheuing, N.; Holl, R.W.; Mellitus, D.I.T.G.B.C.N.D. Treatment of young patients with hnf1a mutations (hnf1a-mody). Diabet. Med. 2015, 32, 526–530. [Google Scholar] [CrossRef]
  120. Broome, D.T.; Pantalone, K.M.; Kashyap, S.R.; Philipson, L.H. Approach to the patient with mody-monogenic diabetes. J. Clin. Endocrinol. Metab. 2021, 106, 237–250. [Google Scholar] [CrossRef] [PubMed]
  121. Shepherd, M.H.; Shields, B.M.; Hudson, M.; Pearson, E.R.; Hyde, C.; Ellard, S.; Hattersley, A.T.; Patel, K.A. A UK nationwide prospective study of treatment change in mody: Genetic subtype and clinical characteristics predict optimal glycaemic control after discontinuing insulin and metformin. Diabetologia 2018, 61, 2520–2527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Østoft, S.H.; Bagger, J.I.; Hansen, T.; Pedersen, O.; Faber, J.; Holst, J.J.; Knop, F.K.; Vilsbøll, T. Glucose-lowering effects and low risk of hypoglycemia in patients with maturity-onset diabetes of the young when treated with a glp-1 receptor agonist: A double-blind, randomized, crossover trial. Diabetes Care 2014, 37, 1797–1805. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Thorens, B. Expression cloning of the pancreatic β cell receptor for the gluco-incretin hormone glucagon-like peptide 1. Proc. Natl. Acad. Sci. USA 1992, 89, 8641–8645. [Google Scholar] [CrossRef] [Green Version]
  124. Gromada, J.; Ding, W.G.; Barg, S.; Renström, E.; Rorsman, P. Multisite regulation of insulin secretion by camp-increasing agonists: Evidence that glucagon-like peptide 1 and glucagon act via distinct receptors. Pflugers. Arch. 1997, 434, 515–524. [Google Scholar] [CrossRef] [PubMed]
  125. Christensen, A.S.; Hædersdal, S.; Storgaard, H.; Rose, K.; Hansen, N.L.; Holst, J.J.; Hansen, T.; Knop, F.K.; Vilsbøll, T. Gip and glp-1 potentiate sulfonylurea-induced insulin secretion in hepatocyte nuclear factor 1α mutation carriers. Diabetes 2020, 69, 1989–2002. [Google Scholar] [CrossRef] [PubMed]
  126. Broome, D.T.; Tekin, Z.; Pantalone, K.M.; Mehta, A.E. Novel use of glp-1 receptor agonist therapy in hnf4a-mody. Diabetes Care 2020, 43, e65. [Google Scholar] [CrossRef] [Green Version]
  127. Docena, M.K.; Faiman, C.; Stanley, C.M.; Pantalone, K.M. Mody-3: Novel hnf1a mutation and the utility of glucagon-like peptide (glp)-1 receptor agonist therapy. Endocr. Pract. 2014, 20, 107–111. [Google Scholar] [CrossRef] [PubMed]
  128. Fantasia, K.L.; Steenkamp, D.W. Optimal glycemic control in a patient with hnf1a mody with glp-1 ra monotherapy: Implications for future therapy. J. Endocr. Soc. 2019, 3, 2286–2289. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Dusatkova, P.; Pruhova, S.; Sumnik, Z.; Kolouskova, S.; Obermannova, B.; Cinek, O.; Lebl, J. Hnf1a mutation presenting with fetal macrosomia and hypoglycemia in childhood prior to onset of overt diabetes. J. Pediatr. Endocrinol. Metab. 2011, 24, 187–189. [Google Scholar] [CrossRef] [PubMed]
  130. Stanescu, D.E.; Hughes, N.; Kaplan, B.; Stanley, C.A.; de León, D.D. Novel presentations of congenital hyperinsulinism due to mutations in the mody genes: Hnf1a and hnf4a. J. Clin. Endocrinol. Metab. 2012, 97, E2026–E2030. [Google Scholar] [CrossRef] [Green Version]
  131. Pearson, E.R.; Boj, S.F.; Steele, A.M.; Barrett, T.; Stals, K.; Shield, J.P.; Ellard, S.; Ferrer, J.; Hattersley, A.T. Macrosomia and hyperinsulinaemic hypoglycaemia in patients with heterozygous mutations in the hnf4a gene. PLoS Med. 2007, 4, e118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Dickens, L.T.; Thomas, C.C. Updates in gestational diabetes prevalence, treatment, and health policy. Curr. Diab. Rep. 2019, 19, 33. [Google Scholar] [CrossRef] [PubMed]
  133. Wang, H.; Maechler, P.; Antinozzi, P.A.; Hagenfeldt, K.A.; Wollheim, C.B. Hepatocyte nuclear factor 4α regulates the expression of pancreatic β -cell genes implicated in glucose metabolism and nutrient-induced insulin secretion. J. Biol. Chem. 2000, 275, 35953–35959. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Yamagata, K.; Furuta, H.; Oda, N.; Kaisaki, P.J.; Menzel, S.; Cox, N.J.; Fajans, S.S.; Signorini, S.; Stoffel, M.; Bell, G.I. Mutations in the hepatocyte nuclear factor-4α gene in maturity-onset diabetes of the young (mody1). Nature 1996, 384, 458–460. [Google Scholar] [CrossRef] [PubMed]
  135. Stride, A.; Ellard, S.; Clark, P.; Shakespeare, L.; Salzmann, M.; Shepherd, M.; Hattersley, A.T. Β-cell dysfunction, insulin sensitivity, and glycosuria precede diabetes in hepatocyte nuclear factor-1α mutation carriers. Diabetes Care 2005, 28, 1751–1756. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Isomaa, B.; Henricsson, M.; Lehto, M.; Forsblom, C.; Karanko, S.; Sarelin, L.; Häggblom, M.; Groop, L. Chronic diabetic complications in patients with mody3 diabetes. Diabetologia 1998, 41, 467–473. [Google Scholar] [CrossRef]
  137. Pearson, E.R.; Pruhova, S.; Tack, C.J.; Johansen, A.; Castleden, H.A.; Lumb, P.J.; Wierzbicki, A.S.; Clark, P.M.; Lebl, J.; Pedersen, O.; et al. Molecular genetics and phenotypic characteristics of mody caused by hepatocyte nuclear factor 4α mutations in a large european collection. Diabetologia 2005, 48, 878–885. [Google Scholar] [CrossRef] [PubMed]
  138. Shih, D.Q.; Dansky, H.M.; Fleisher, M.; Assmann, G.; Fajans, S.S.; Stoffel, M. Genotype/phenotype relationships in hnf-4α/mody1: Haploinsufficiency is associated with reduced apolipoprotein (aii), apolipoprotein (ciii), lipoprotein(a), and triglyceride levels. Diabetes 2000, 49, 832–837. [Google Scholar] [CrossRef] [Green Version]
  139. Lehto, M.; Bitzén, P.O.; Isomaa, B.; Wipemo, C.; Wessman, Y.; Forsblom, C.; Tuomi, T.; Taskinen, M.R.; Groop, L. Mutation in the hnf-4α gene affects insulin secretion and triglyceride metabolism. Diabetes 1999, 48, 423–425. [Google Scholar] [CrossRef] [PubMed]
  140. Fajans, S.S.; Brown, M.B. Administration of sulfonylureas can increase glucose-induced insulin secretion for decades in patients with maturity-onset diabetes of the young. Diabetes Care 1993, 16, 1254–1261. [Google Scholar] [CrossRef] [PubMed]
  141. Fajans, S.S.; Bell, G.I. Macrosomia and neonatal hypoglycaemia in rw pedigree subjects with a mutation (q268x) in the gene encoding hepatocyte nuclear factor 4α (hnf4a). Diabetologia 2007, 50, 2600–2601. [Google Scholar] [CrossRef]
  142. Flanagan, S.E.; Kapoor, R.R.; Mali, G.; Cody, D.; Murphy, N.; Schwahn, B.; Siahanidou, T.; Banerjee, I.; Akcay, T.; Rubio-Cabezas, O.; et al. Diazoxide-responsive hyperinsulinemic hypoglycemia caused by hnf4a gene mutations. Eur. J. Endocrinol. 2010, 162, 987–992. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Faguer, S.; Chassaing, N.; Bandin, F.; Prouheze, C.; Garnier, A.; Casemayou, A.; Huart, A.; Schanstra, J.P.; Calvas, P.; Decramer, S.; et al. The hnf1b score is a simple tool to select patients for hnf1b gene analysis. Kidney Int. 2014, 86, 1007–1015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Ryffel, G.U. Mutations in the human genes encoding the transcription factors of the hepatocyte nuclear factor (hnf)1 and hnf4 families: Functional and pathological consequences. J. Mol. Endocrinol. 2001, 27, 11–29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Clissold, R.L.; Hamilton, A.J.; Hattersley, A.T.; Ellard, S.; Bingham, C. Hnf1b-associated renal and extra-renal disease-an expanding clinical spectrum. Nat. Rev. Nephrol. 2015, 11, 102–112. [Google Scholar] [CrossRef] [PubMed]
  146. Bingham, C.; Ellard, S.; Cole, T.R.; Jones, K.E.; Allen, L.I.; Goodship, J.A.; Goodship, T.H.; Bakalinova-Pugh, D.; Russell, G.I.; Woolf, A.S.; et al. Solitary functioning kidney and diverse genital tract malformations associated with hepatocyte nuclear factor-1β mutations. Kidney Int. 2002, 61, 1243–1251. [Google Scholar] [CrossRef] [Green Version]
  147. Edghill, E.L.; Bingham, C.; Ellard, S.; Hattersley, A.T. Mutations in hepatocyte nuclear factor-1β and their related phenotypes. J. Med. Genet. 2006, 43, 84–90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Adalat, S.; Bockenhauer, D.; Ledermann, S.E.; Hennekam, R.C.; Woolf, A.S. Renal malformations associated with mutations of developmental genes: Messages from the clinic. Pediatr. Nephrol. 2010, 25, 2247–2255. [Google Scholar] [CrossRef]
  149. Bingham, C.; Bulman, M.P.; Ellard, S.; Allen, L.I.; Lipkin, G.W.; Hoff, W.G.; Woolf, A.S.; Rizzoni, G.; Novelli, G.; Nicholls, A.J.; et al. Mutations in the hepatocyte nuclear factor-1β gene are associated with familial hypoplastic glomerulocystic kidney disease. Am. J. Hum. Genet. 2001, 68, 219–224. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Kolatsi-Joannou, M.; Bingham, C.; Ellard, S.; Bulman, M.P.; Allen, L.I.S.; Hattersley, A.T.; Woolf, A.S. Hepatocyte nuclear factor-1β: A new kindred with renal cysts and diabetes and gene expression in normal human development. J. Am. Soc. Nephrol. 2001, 12, 2175–2180. [Google Scholar] [CrossRef] [PubMed]
  151. Woolf, A.S.; Feather, S.A.; Bingham, C. Recent insights into kidney diseases associated with glomerular cysts. Pediatr. Nephrol. 2002, 17, 229–235. [Google Scholar] [CrossRef]
  152. Queisser-Luft, A.; Stolz, G.; Wiesel, A.; Schlaefer, K.; Spranger, J. Malformations in newborn: Results based on 30;940 infants and fetuses from the mainz congenital birth defect monitoring system (1990–1998). Arch. Gynecol Obstet. 2002, 266, 163–167. [Google Scholar] [CrossRef] [PubMed]
  153. Soliman, N.A.; Ali, R.I.; Ghobrial, E.E.; Habib, E.I.; Ziada, A.M. Pattern of clinical presentation of congenital anomalies of the kidney and urinary tract among infants and children. Nephrology 2015, 20, 413–418. [Google Scholar] [CrossRef] [PubMed]
  154. Bellanné-Chantelot, C.; Chauveau, D.; Gautier, J.F.; Dubois-Laforgue, D.; Clauin, S.; Beaufils, S.; Wilhelm, J.M.; Boitard, C.; Noël, L.H.; Velho, G.; et al. Clinical spectrum associated with hepatocyte nuclear factor-1β mutations. Ann. Intern. Med. 2004, 140, 510–517. [Google Scholar] [CrossRef]
  155. Haldorsen, I.S.; Vesterhus, M.; Raeder, H.; Jensen, D.K.; Søvik, O.; Molven, A.; Njølstad, P.R. Lack of pancreatic body and tail in hnf1b mutation carriers. Diabet. Med. 2008, 25, 782–787. [Google Scholar] [CrossRef]
  156. Adalat, S.; Woolf, A.S.; Johnstone, K.A.; Wirsing, A.; Harries, L.W.; Long, D.A.; Hennekam, R.C.; Ledermann, S.E.; Rees, L.; van’t Hoff, W.; et al. Hnf1b mutations associate with hypomagnesemia and renal magnesium wasting. J. Am. Soc. Nephrol. 2009, 20, 1123–1131. [Google Scholar] [CrossRef] [Green Version]
  157. Iwasaki, N.; Babazono, T.; Tomonaga, O.; Ogata, M.; Yokokawa, H.; Iwamoto, Y. Mutations in the hepatocyte nuclear factor-1β (mody5) gene are not a major factor contributing to end-stage renal disease in japanese people with diabetes mellitus. Diabetologia 2001, 44, 127–128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Lindner, T.H.; Njolstad, P.R.; Horikawa, Y.; Bostad, L.; Bell, G.I.; Sovik, O. A novel syndrome of diabetes mellitus; renal dysfunction and genital malformation associated with a partial deletion of the pseudo-pou domain of hepatocyte nuclear factor-1. Hum. Mol. Genet. 1999, 8, 2001–2008. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Kotalova, R.; Dusatkova, P.; Cinek, O.; Dusatkova, L.; Dedic, T.; Seeman, T.; Lebl, J.; Pruhova, S. Hepatic phenotypes of hnf1b gene mutations: A case of neonatal cholestasis requiring portoenterostomy and literature review. World J. Gastroenterol. 2015, 21, 2550–2557. [Google Scholar] [CrossRef]
  160. Beckers, D.; Bellanné-Chantelot, C.; Maes, M. Neonatal cholestatic jaundice as the first symptom of a mutation in the hepatocyte nuclear factor-1β gene (hnf-1β). J. Pediatr. 2007, 150, 313–314. [Google Scholar] [CrossRef]
  161. Raile, K.; Klopocki, E.; Holder, M.; Wessel, T.; Galler, A.; Deiss, D.; Müller, D.; Riebel, T.; Horn, D.; Maringa, M.; et al. Expanded clinical spectrum in hepatocyte nuclear factor 1b-maturity-onset diabetes of the young. J. Clin. Endocrinol. Metab. 2009, 94, 2658–2664. [Google Scholar] [CrossRef] [Green Version]
  162. Pearson, E.R.; Badman, M.K.; Lockwood, C.R.; Clark, P.M.; Ellard, S.; Bingham, C.; Hattersley, A.T. Contrasting diabetes phenotypes associated with hepatocyte nuclear factor-1α and -1β mutations. Diabetes Care 2004, 27, 1102–1117. [Google Scholar] [CrossRef] [Green Version]
  163. Dubois-Laforgue, D.; Cornu, E.; Saint-Martin, C.; Coste, J.; Bellanné-Chantelot, C.; Timsit, J.; for the Monogenic Diabetes Study Group of the Société Francophone du Diabète. Diabetes, associated clinical spectrum, long-term prognosis, and genotype/phenotype correlations in 201 adult patients with hepatocyte nuclear factor 1b. Diabetes Care 2017, 40, 1436–1443. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Murphy, R.; Ellard, S.; Hattersley, A.T. Clinical implications of a molecular genetic classification of monogenic β-cell diabetes. Nat. Clin. Pract. Endocrinol. Metab. 2008, 4, 200–213. [Google Scholar] [CrossRef]
  165. Tjora, E.; Wathle, G.; Erchinger, F.; Engjom, T.; Molven, A.; Aksnes, L.; Haldorsen, I.S.; Dimcevski, G.; Raeder, H.; Njølstad, P.R. Exocrine pancreatic function in hepatocyte nuclear factor 1β-maturity-onset diabetes of the young (hnf1b-mody) is only moderately reduced: Compensatory hypersecretion from a hypoplastic pancreas. Diabet. Med. 2013, 30, 946–955. [Google Scholar] [CrossRef] [PubMed]
  166. Edghill, E.L.; Bingham, C.; Slingerland, A.S.; Minton, J.A.; Noordam, C.; Ellard, S.; Hattersley, A.T. Hepatocyte nuclear factor-1 β mutations cause neonatal diabetes and intrauterine growth retardation: Support for a critical role of hnf-1β in human pancreatic development. Diabet. Med. 2006, 23, 1301–1306. [Google Scholar] [CrossRef] [PubMed]
  167. Boitier, E.; Gautier, J.C.; Roberts, R. Advances in understanding the regulation of apoptosis and mitosis by peroxisome-proliferator activated receptors in pre-clinical models: Relevance for human health and disease. Comp. Hepatol. 2003, 2, 3. [Google Scholar] [CrossRef]
  168. Berger, J.; Moller, D.E. The mechanisms of action of ppars. Annu. Rev. Med. 2002, 53, 409–435. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Tontonoz, P.; Spiegelman, B.M. Fat and beyond: The diverse biology of ppar. Annu. Rev. Biochem. 2008, 77, 289–312. [Google Scholar] [CrossRef] [PubMed]
  170. Medina-Gomez, G.; Gray, S.L.; Yetukuri, L.; Shimomura, K.; Virtue, S.; Campbell, M.; Curtis, R.K.; Jimenez-Linan, M.; Blount, M.; Yeo, G.S.; et al. Ppar γ 2 prevents lipotoxicity by controlling adipose tissue expandability and peripheral lipid metabolism. PLoS Genet. 2007, 3, e64. [Google Scholar] [CrossRef] [Green Version]
  171. Tontonoz, P.; Hu, E.; Spiegelman, B.M. Stimulation of adipogenesis in fibroblasts by ppar γ 2, a lipid-activated transcription factor. Cell 1994, 79, 1147–1156. [Google Scholar] [CrossRef]
  172. Ahmadian, M.; Suh, J.M.; Hah, N.; Liddle, C.; Atkins, A.R.; Downes, M.; Evans, R.M. Pparγ signaling and metabolism: The good, the bad and the future. Nat. Med. 2013, 19, 557–566. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Broekema, M.F.; Savage, D.B.; Monajemi, H.; Kalkhoven, E. Gene-gene and gene-environment interactions in lipodystrophy: Lessons learned from natural pparγ mutants. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2019, 1864, 715–732. [Google Scholar] [CrossRef]
  174. Floyd, Z.E.; Stephens, J.M. Controlling a master switch of adipocyte development and insulin sensitivity: Covalent modifications of ppar. Biochim. Biophys. Acta 2012, 1822, 1090–1095. [Google Scholar] [CrossRef] [Green Version]
  175. Choi, J.H.; Banks, A.S.; Estall, J.L.; Kajimura, S.; Boström, P.; Laznik, D.; Ruas, J.L.; Chalmers, M.J.; Kamenecka, T.M.; Blüher, M.; et al. Anti-diabetic drugs inhibit obesity-linked phosphorylation of pparγ by cdk5. Nature 2010, 466, 451–456. [Google Scholar] [CrossRef] [Green Version]
  176. Hegele, R.A. Monogenic forms of insulin resistance: Apertures that expose the common metabolic syndrome. Trends Endocrinol. Metab. 2003, 14, 371–377. [Google Scholar] [CrossRef]
  177. Barroso, I.; Gurnell, M.; Crowley, V.E.; Agostini, M.; Schwabe, J.W.; Soos, M.A.; Maslen, G.L.; Williams, T.D.; Lewis, H.; Schafer, A.J.; et al. Dominant negative mutations in human pparγ associated with severe insulin resistance, diabetes mellitus and hypertension. Nature 1999, 402, 880–883. [Google Scholar] [CrossRef]
  178. Jeninga, E.H.; van Beekum, O.; van Dijk, A.D.; Hamers, N.; Hendriks-Stegeman, B.I.; Bonvin, A.M.; Berger, R.; Kalkhoven, E. Impaired peroxisome proliferator-activated receptor γ function through mutation of a conserved salt bridge (r425c) in familial partial lipodystrophy. Mol. Endocrinol. 2007, 21, 1049–1065. [Google Scholar] [CrossRef] [Green Version]
  179. Semple, R.K.; Chatterjee, V.K.; O’Rahilly, S. Ppar γ and human metabolic disease. J. Clin. Investig. 2006, 116, 581–589. [Google Scholar] [CrossRef] [Green Version]
  180. Jeninga, E.H.; Gurnell, M.; Kalkhoven, E. Functional implications of genetic variation in human ppar. Trends Endocrinol. Metab. 2009, 20, 380–387. [Google Scholar] [CrossRef]
  181. Hegele, R.A.; Joy, T.R.; Al-Attar, S.A.; Rutt, B.K. Thematic review series: Adipocyte biology. Lipodystrophies: Windows on adipose biology and metabolism. J. Lipid Res. 2007, 48, 1433–1444. [Google Scholar] [CrossRef] [Green Version]
  182. Majithia, A.R.; Flannick, J.; Shahinian, P.; Guo, M.; Bray, M.A.; Fontanillas, P.; Gabriel, S.B.; Rosen, E.D.; Altshuler, D.; Consortium, G.D.; et al. Rare variants in pparg with decreased activity in adipocyte differentiation are associated with increased risk of type 2 diabetes. Proc. Natl. Acad. Sci. USA 2014, 111, 13127–13132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Lehmann, J.M.; Moore, L.B.; Smith-Oliver, T.A.; Wilkison, W.O.; Willson, T.M.; Kliewer, S.A. An antidiabetic thiazolidinedione is a high affinity ligand for peroxisome proliferator-activated receptor γ (ppar γ). J. Biol. Chem. 1995, 270, 12953–12956. [Google Scholar] [CrossRef] [Green Version]
  184. Tan, G.D.; Fielding, B.A.; Currie, J.M.; Humphreys, S.M.; Désage, M.; Frayn, K.N.; Laville, M.; Vidal, H.; Karpe, F. The effects of rosiglitazone on fatty acid and triglyceride metabolism in type 2 diabetes. Diabetologia 2005, 48, 83–95. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Day, C. Thiazolidinediones: A new class of antidiabetic drugs. Diabet. Med. 1999, 16, 179–192. [Google Scholar] [CrossRef] [PubMed]
  186. Pearson, S.L.; Cawthorne, M.A.; Clapham, J.C.; Dunmore, S.J.; Holmes, S.D.; Moore, G.B.; Smith, S.A.; Tadayyon, M. The thiazolidinedione insulin sensitiser, brl 49653, increases the expression of ppar-γ and ap2 in adipose tissue of high-fat-fed rats. Biochem. Biophys. Res. Commun. 1996, 229, 752–757. [Google Scholar] [CrossRef] [PubMed]
  187. Soccio, R.E.; Chen, E.R.; Lazar, M.A. Thiazolidinediones and the promise of insulin sensitization in type 2 diabetes. Cell Metab. 2014, 20, 573–591. [Google Scholar] [CrossRef] [Green Version]
  188. Savage, D.B.; Tan, G.D.; Acerini, C.L.; Jebb, S.A.; Agostini, M.; Gurnell, M.; Williams, R.L.; Umpleby, A.M.; Thomas, E.L.; Bell, J.D.; et al. Human metabolic syndrome resulting from dominant-negative mutations in the nuclear receptor peroxisome proliferator-activated receptor-. Diabetes 2003, 52, 910–917. [Google Scholar] [CrossRef] [Green Version]
  189. Francis, G.A.; Li, G.; Casey, R.; Wang, J.; Cao, H.; Leff, T.; Hegele, R.A. Peroxisomal proliferator activated receptor-γ deficiency in a canadian kindred with familial partial lipodystrophy type 3 (fpld3). BMC Med. Genet. 2006, 7, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  190. Demir, T.; Onay, H.; Savage, D.B.; Temeloglu, E.; Uzum, A.K.; Kadioglu, P.; Altay, C.; Ozen, S.; Demir, L.; Cavdar, U.; et al. Familial partial lipodystrophy linked to a novel peroxisome proliferator activator receptor -γ (pparg) mutation, h449l: A comparison of people with this mutation and those with classic codon 482 lamin a/c (lmna) mutations. Diabet. Med. 2016, 33, 1445–1450. [Google Scholar] [CrossRef] [Green Version]
  191. Agostini, M.; Gurnell, M.; Savage, D.B.; Wood, E.M.; Smith, A.G.; Rajanayagam, O.; Garnes, K.T.; Levinson, S.H.; Xu, H.E.; Schwabe, J.W.; et al. Tyrosine agonists reverse the molecular defects associated with dominant-negative mutations in human peroxisome proliferator-activated receptor. Endocrinology 2004, 145, 1527–1538. [Google Scholar] [CrossRef] [Green Version]
  192. Agostini, M.; Schoenmakers, E.; Beig, J.; Fairall, L.; Szatmari, I.; Rajanayagam, O.; Muskett, F.W.; Adams, C.; Marais, A.D.; O’Rahilly, S.; et al. A pharmacogenetic approach to the treatment of patients with. Diabetes 2018, 67, 1086–1092. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Arioglu, E.; Duncan-Morin, J.; Sebring, N.; Rother, K.I.; Gottlieb, N.; Lieberman, J.; Heroin, D.; Kleiner, D.E.; Reynolds, J.; Premkumar, A.; et al. Efficacy and safety of troglitazone in the treatment of lipodystrophy syndromes. Ann. Intern. Med. 2000, 133, 263–274. [Google Scholar] [CrossRef] [PubMed]
  194. Lu, P.; Zhao, Z. Advances on pparγ research in the emerging era of precision medicine. Curr. Drug. Targets 2018, 19, 663–673. [Google Scholar] [CrossRef] [Green Version]
  195. Molkentin, J.D. The zinc finger-containing transcription factors gata-4, -5, and -6. Ubiquitously expressed regulators of tissue-specific gene expression. J. Biol. Chem. 2000, 275, 38949–38952. [Google Scholar] [CrossRef] [Green Version]
  196. Lentjes, M.H.; Niessen, H.E.; Akiyama, Y.; de Bruïne, A.P.; Melotte, V.; van Engeland, M. The emerging role of gata transcription factors in development and disease. Expert Rev. Mol. Med. 2016, 18, e3. [Google Scholar] [CrossRef]
  197. Garg, V.; Kathiriya, I.S.; Barnes, R.; Schluterman, M.K.; King, I.N.; Butler, C.A.; Rothrock, C.R.; Eapen, R.S.; Hirayama-Yamada, K.; Joo, K.; et al. Gata4 mutations cause human congenital heart defects and reveal an interaction with tbx5. Nature 2003, 424, 443–447. [Google Scholar] [CrossRef]
  198. Nemer, G.; Fadlalah, F.; Usta, J.; Nemer, M.; Dbaibo, G.; Obeid, M.; Bitar, F. A novel mutation in the gata4 gene in patients with tetralogy of fallot. Hum. Mutat. 2006, 27, 293–294. [Google Scholar] [CrossRef]
  199. Wat, M.J.; Shchelochkov, O.A.; Holder, A.M.; Breman, A.M.; Dagli, A.; Bacino, C.; Scaglia, F.; Zori, R.T.; Cheung, S.W.; Scott, D.A.; et al. Chromosome 8p23.1 deletions as a cause of complex congenital heart defects and diaphragmatic hernia. Am. J. Med. Genet. A 2009, 149A, 1661–1677. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  200. D’Amato, E.; Giacopelli, F.; Giannattasio, A.; d’Annunzio, G.; Bocciardi, R.; Musso, M.; Lorini, R.; Ravazzolo, R. Genetic investigation in an italian child with an unusual association of atrial septal defect, attributable to a new familial gata4 gene mutation, and neonatal diabetes due to pancreatic agenesis. Diabet. Med. 2010, 27, 1195–1200. [Google Scholar] [CrossRef]
  201. Shaw-Smith, C.; de Franco, E.; Lango Allen, H.; Batlle, M.; Flanagan, S.E.; Borowiec, M.; Taplin, C.E.; van Alfen-van der Velden, J.; Cruz-Rojo, J.; Perez de Nanclares, G.; et al. Gata4 mutations are a cause of neonatal and childhood-onset diabetes. Diabetes 2014, 63, 2888–2894. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. De Franco, E.; Shaw-Smith, C.; Flanagan, S.E.; Shepherd, M.H.; Hattersley, A.T.; Ellard, S.; Consortium, I.N. Gata6 mutations cause a broad phenotypic spectrum of diabetes from pancreatic agenesis to adult-onset diabetes without exocrine insufficiency. Diabetes 2013, 62, 993–997. [Google Scholar] [CrossRef] [Green Version]
  203. Allen, H.L.; Flanagan, S.E.; Shaw-Smith, C.; de Franco, E.; Akerman, I.; Caswell, R.; International Pancreatic Agenesis Consortium; Ferrer, J.; Hattersley, A.T.; Ellard, S. Gata6 haploinsufficiency causes pancreatic agenesis in humans. Nat. Genet. 2011, 44, 20–22. [Google Scholar] [CrossRef]
  204. Raghuram, N.; Marwaha, A.; Greer, M.C.; Gauda, E.; Chitayat, D. Congenital hypothyroidism, cardiac defects, and pancreatic agenesis in an infant with gata6 mutation. Am. J. Med. Genet. A 2020, 182, 1496–1499. [Google Scholar] [CrossRef]
  205. Dzau, V.J.; Ginsburg, G.S. Realizing the full potential of precision medicine in health and health care. JAMA 2016, 316, 1659–1660. [Google Scholar] [CrossRef]
  206. Jungtrakoon Thamtarana, P.; Marucci, A.; Pannone, L.; Bonnefond, A.; Pezzilli, S.; Biagini, T.; Buranasupkajorn, P.; Hastings, T.; Mendonca, C.; Marselli, L.; et al. Gain of function of malate dehydrogenase 2 (mdh2) and familial hyperglycemia. J. Clin. Endocrinol. Metab. 2021. [Google Scholar] [CrossRef] [PubMed]
  207. Prudente, S.; Jungtrakoon, P.; Marucci, A.; Ludovico, O.; Buranasupkajorn, P.; Mazza, T.; Hastings, T.; Milano, T.; Morini, E.; Mercuri, L.; et al. Loss-of-function mutations in appl1 in familial diabetes mellitus. Am. J. Hum. Genet. 2015, 97, 177–185. [Google Scholar] [CrossRef] [Green Version]
Table 1. Summary of actionable genes with related diabetes phenotype and actionability.
Table 1. Summary of actionable genes with related diabetes phenotype and actionability.
GeneMutationPhenotypeDisease MechanismAdditional ComplicationBirth WeightActionability
ABCC8/
KCNJ11
Heterozygous and Homozygous GOFNeonatal diabetes
MODY
Adult-onset diabetes
KATP channel permanently open, K+ efflux/membrane hyperpolarization/defective insulin secretionNeurodevelopment dysfunctionNormal, as long as maternal hyperglycemia is properly treated
Low, when only the fetus is mutated
High dose of sulphonylureas (also in pregnancy, as long as the fetus is mutated; otherwise, insulin should be given)
GCKHeterozygous LOF
Homozygous/Compound heterozygous LOF
GCK-MODY (moderate fasting hyperglycemia from birth, low risk of chronic complication)
Neonatal diabetes
Increased glucose sensor threshold (glucose stimulated insulin secretion
begins at higher glucose level)
NoneNormal, as long as maternal hyperglycemia is properly treated
Low, when only the fetus is mutated
No treatment needed
(except during pregnancy when insulin is the treatment of choice)
HNF1AHeterozygous
and
Homozygous LOF
HNF1A-MODY (fasting glycemia increase with age, normoglycemic glycosuria, liver adenomatosis)Reduced HNF1A expression, reduced β-cell mass, blunted glycolysis and ATP production and eventually defective insulin secretionRetinopathy, nephropathy and neuropathy are common. Ketoacidosis can developNormal, as long as maternal hyperglycemia is properly treatedLow dose of sulphonylureas also in pregnancy for the first two trimesters (when both the mother and the fetus are mutated)
HNF4AHeterozygous LOF HNF4-MODY (fasting glycemia increase with age, liver dysfunction)Reduced HNF1A expression, reduced β-cell mass, blunted glycolysis and ATP production and eventually defective insulin secretionReduced triglycerides and lipoprotein serum concentrationNormal, as long as maternal hyperglycemia is properly treated
HNF1BHeterozygous LOF HNF1B-MODY (high fasting glycemia, ketoacidosis)Reduced HNF1B expression, pancreatic hypoplasia, blunted glycolysis and ATP production and eventually defective insulin secretionKidney cysts and urinary tract abnormalities, atrophic pancreas, genital abnormalities, hyperuricemia, goutNormal, as long as maternal hyperglycemia is properly treated
Low, when only the fetus is mutated
Systemic screening for renal cysts, exocrine pancreatic function and genital abnormalities (especially in females)
PPARGHeterozygous LOF
Severe insulin resistanceDefective adipocyte differentiation due to PPARG haploinsufficiency or dominant negative LOF mutationFamilial partial lipodystrophy type 3 (early-onset diabetes, hypertension, severe insulin resistance and dyslipidemia, hepatic steatosis)No clear data are availableThiazolidinediones
GATA4Heterozygous LOF or complete gene deletionNeonatal diabetesDysfunctional transcriptional activity, and altered embryonic organ developmentCongenital heart malformation, pancreatic agenesis or hypoplasiaLowEvaluation and follow up of congenital heart malformation and pancreatic agenesis/hypoplasia
GATA6Heterozygous LOFNeonatal diabetes
Adult-onset diabetes
Dysfunctional transcriptional activity, and altered embryonic organ developmentCongenital biliary tract anomalies, gut developmental disorders, neurocognitive abnormalities, additional endocrine abnormalitiesLow
GOF: gain of function. LOF: loss of function.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Marucci, A.; Rutigliano, I.; Fini, G.; Pezzilli, S.; Menzaghi, C.; Di Paola, R.; Trischitta, V. Role of Actionable Genes in Pursuing a True Approach of Precision Medicine in Monogenic Diabetes. Genes 2022, 13, 117. https://doi.org/10.3390/genes13010117

AMA Style

Marucci A, Rutigliano I, Fini G, Pezzilli S, Menzaghi C, Di Paola R, Trischitta V. Role of Actionable Genes in Pursuing a True Approach of Precision Medicine in Monogenic Diabetes. Genes. 2022; 13(1):117. https://doi.org/10.3390/genes13010117

Chicago/Turabian Style

Marucci, Antonella, Irene Rutigliano, Grazia Fini, Serena Pezzilli, Claudia Menzaghi, Rosa Di Paola, and Vincenzo Trischitta. 2022. "Role of Actionable Genes in Pursuing a True Approach of Precision Medicine in Monogenic Diabetes" Genes 13, no. 1: 117. https://doi.org/10.3390/genes13010117

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop