Next Article in Journal
Role of Dendritic Cell in Diabetic Nephropathy
Previous Article in Journal
Hydrophilic and Hydrophobic Effects on the Structure and Themodynamic Properties of Confined Water: Water in Solutions
Previous Article in Special Issue
The Multifactorial Progression from the Islet Autoimmunity to Type 1 Diabetes in Children
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Maturity Onset Diabetes of the Young—New Approaches for Disease Modelling

by
Dawid Skoczek
1,2,
Józef Dulak
2 and
Neli Kachamakova-Trojanowska
1,*
1
Malopolska Centre of Biotechnology, Jagiellonian University, 30-387 Krakow, Poland
2
Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(14), 7553; https://doi.org/10.3390/ijms22147553
Submission received: 1 June 2021 / Revised: 4 July 2021 / Accepted: 9 July 2021 / Published: 14 July 2021
(This article belongs to the Special Issue Molecular Metabolic Regulation in Diabetes 2.0)

Abstract

:
Maturity-onset diabetes of the young (MODY) is a genetically heterogeneous group of monogenic endocrine disorders that is characterised by autosomal dominant inheritance and pancreatic β-cell dysfunction. These patients are commonly misdiagnosed with type 1 or type 2 diabetes, as the clinical symptoms largely overlap. Even though several biomarkers have been tested none of which could be used as single clinical discriminator. The correct diagnosis for individuals with MODY is of utmost importance, as the applied treatment depends on the gene mutation or is subtype-specific. Moreover, in patients with HNF1A-MODY, additional clinical monitoring can be included due to the high incidence of vascular complications observed in these patients. Finally, stratification of MODY patients will enable better and newer treatment options for MODY patients, once the disease pathology for each patient group is better understood. In the current review the clinical characteristics and the known disease-related abnormalities of the most common MODY subtypes are discussed, together with the up-to-date applied diagnostic criteria and treatment options. Additionally, the usage of pluripotent stem cells together with CRISPR/Cas9 gene editing for disease modelling with the possibility to reveal new pathophysiological mechanisms in MODY is discussed.

1. Diabetes—Current Classification and Place for Maturity-Onset Diabetes of the Young (MODY)

Diabetes is a chronic, metabolic disease characterised by elevated levels of blood glucose, which uncontrolled over time leads to serious damage of central organs such as the heart, kidneys, eyes and blood vessels. According to World Health Organization (WHO) about 422 million people worldwide have diabetes with 1.6 million deaths directly attributed to diabetes each year. Both the number of cases and the prevalence of diabetes have been steadily increasing over the past few decades. The first WHO classification system for diabetes was published in 1965 and was simple mainly based on the age of the patient: infantile, young, adult and elderly [1].
The next WHO system, published in 1980 and updated five years later, was globally accepted and widely adopted. In these classifications, the two major classes of diabetes were included as insulin dependent diabetes mellitus (IDDM) or type 1 diabetes mellitus (T1DM), and non-insulin dependent diabetes mellitus (NIDDM) or type 2 diabetes mellitus (T2DM). In addition to these, two other classes were added the so-called “other types” plus gestational diabetes mellitus (GDM). In 1999, WHO recommended that the classification of diabetes should encompass not only different aetiological types of diabetes, but also the clinical stages of the disease and reintroduced the terms T1DM and T2DM, as they were omitted in the report from 1985 [2].
Since the last classification, advances in the knowledge of pathophysiological pathways related to the disease have been made. It became clear that diabetes is a much more heterogeneous disease than the simple classification of T1DM and T2DM. Moreover, many patients have genetic predispositions to both forms of diabetes, which in combination to rapid changes in the environment leads to an increased incidence of the disease worldwide [3]. It was shown that molecular genetics can be used to help in the identification of specific subtypes of monogenic diabetes. Importantly, genetic diagnosis can not only reveal clinical subgroups, but can result in improved treatment outcomes for these patients [4]. All this led to updating the classification system in 2019 [5], that is centred on the β-cell. This enables that appropriate clinical care is delivered according to the international standards set out in the classification system. The β-cell-centric model recognises dysfunctional β-cells as the common denominator of diabetes, which may be caused by either monogenic or polygenic defects in combination to lifestyle factors and environmental changes [6]. In the new classification system, the two major groups (T1DM and T2DM) are included along with so-called “hybrid forms”, “other specific types”, hyperglycaemia during pregnancy and unclassified diabetes. In “other specific types” several subclasses were defined, among them “monogenic diabetes” (Figure 1).
The monogenic diabetes subclass was additionally divided into monogenic defects of β-cell function, monogenic defects in insulin action and other genetic syndromes associated with diabetes (Figure 2). Clinical manifestation of monogenic defects in β-cell function include maturity-onset diabetes of the young (MODY) and other genetic syndromes where insulin-deficient diabetes is associated with additional clinical features [7,8].

2. MODY Types

MODY is a rare condition, accounting for 1–5% of all diabetes cases [9,10] and 1–6% of paediatric cases of diabetes [11], which means that 1:10,000 adults can be affected [12]. Usually, the definitions of the monogenic subtype come from the gene symbol of the mutated gene followed by the clinical syndrome. The most common forms of MODY are caused by mutations in the glucokinase gene (GCK-MODY) and hepatocyte nuclear factor genes (HNF1A-, HNF1B- and HNF4A-MODY) which together are responsible for around 99% of all MODY cases [13]. HNF1A-MODY is the most frequently found (Figure 3) [14] but the incidence rates vary among different populations. For example, in Japan, around 51.9% cases are caused by unidentified mutations, and among the identified ones GCK mutations (22.8%) are the most prevalent [15]. In European countries (UK, Germany, The Netherlands, Norway, Poland), the most common subtype is HNF1A-MODY, followed by GCK-, HNF4A- and HNF1B-MODY [16,17,18,19,20]. Recently, the first MODY cases were reported in patients from Africa, where the predominant subtype was HNF1A-MODY representing 5.9% of the study population [21]. Additionally, mutations in MODY-associated genes were shown to be a significant risk factor for T2DM [22,23,24,25]. However, these big population studies also highlight the high rate of misdiagnosed individuals, which subsequently leads to inappropriate treatment strategies [23,25].
MODY is a monogenic, autosomal dominant form of diabetes with onset before the age of 25, absence of β-cell autoimmunity and impaired β-cell function [10,13]. Currently, 14 MODY subtypes caused by mutations in 14 different genes (Table 1) were described [10,26,27], although mutations in other genes were also described in relation to MODY [28].

3. Molecular Pathophysiology of the Most Common MODY Subtypes

All types of MODY are caused by a heterozygous mutation in a specific gene and many of them are considered as haploinsufficiency disorders [29], meaning that one defective allele results in insufficient dose of the gene product leading to failure of the normal cell phenotype. Haploinsufficiency genes are usually expressed during early development and are involved in developmental processes, transcription, cell cycle and nucleic acid metabolism [30,31]. Recently, a homozygous mutation causing MODY has been described [32].

3.1. HNF1A MODY

The most common form of MODY is caused by mutations in hepatocyte nuclear factor 1A gene (HNF1A). HNF1A is a transcription factor comprised of short N-terminal dimerisation domain, a DNA-binding domain and a C-terminal transactivation domain [33]. It is expressed in liver, kidney, intestine and pancreatic islets [34]. Three functional isoforms of HNF1A are described, which are regulated by the same promoter, however they present a different pattern of polyadenylation and are formed by alternative splicing [35]. The longest isoform—HNF1A(A)—consists of 10 exons; the shorter is HNF1A(B) and the shortest is HNF1A(C). These have lengths of 7 and 6 exons, respectively. The expression of these isoforms varies between tissues. In the pancreas, isoform B is the predominant form, while in the liver isoform A is the most abundantly expressed. Isoform C is detected at very low levels in diabetes-associated organs [36].
In the liver, HNF1A is responsible for gluconeogenesis and apolipoprotein synthesis, in the pancreas for the synthesis of the insulin receptor (INSR) and glucose transporter 1 and 2 (GLUT1, GLUT2), whereas in the gut it is supposed to play an important role in controlling terminal functions of the epithelium [37,38,39]. In humans, biallelic deletions of HNF1A are not observed because of its impact on development and homeostasis [40,41]. Recently, however, a biallelic mutation of this gene was found to be related to MODY [32] and to cause primary hepatocellular adenoma co-occurring with HNF1A-MODY [41]. Studies with murine HNF1A-/- model show that knock-out of this gene causes Fanconi syndrome, phenylketonuria, hepatic dysfunction, muscular atrophy, and eventually leads to death in the first weeks after the birth [42,43]. Despite its crucial role in homeostasis, there are studies suggesting a relationship of some HNF1A derivatives (especially long noncoding RNA—lncRNA) in progression of cancers such as pancreatic or gastric cancer [44,45].
The mutations in HNF1A, linked to MODY, are heterozygous, scattered throughout the protein coding region, promoter and 5′-UTR and consist of missense, nonsense and frameshift mutations [46,47]. Even though about 375 mutations in the HNF1A gene have been reported to date, 198 of these are associated with HNF1A-MODY or diabetes in general (Supplementary Table S1, based on ClinVar data [48]). It was confirmed that the type of mutation may affect the functionality and stability of the HNF1A protein, and eventually the age of HNF1A-MODY diagnosis [35,49,50,51]. The most common mutations found in the HNF1A gene are missense mutations, which are concentrated in the dimerisation and DNA-binding domains (Figure 4). These truncations are caused by nonsense mutations or, more frequently, by indels resulting in frameshifts and consequently leading to premature stop codons. Depending on the mutations found within HNF1A, their effects can be benign, cause MODY, or act as a risk factor for T2DM [52]. Recently, an evaluation of different HNF1A mutations and their effects on clinical parameters were made. The results showed that the best and superior readout that correlated to clinical phenotype was for the transcriptional activity of the gene, therefore it was suggested that new high-throughput functional screens should be developed [53]. Interestingly, a homozygous missense HNF1A (pA251T) variant related to MODY was described recently [32]. The new variant was found to be located in the highly conserved DNA binding domain and the in vitro functional assays demonstrated a modest reduction in transactivation activity and DNA binding of the mutated variant. The homozygous patients presented a similar clinical phenotype to the heterozygous HNF1A-MODY individuals, such as low levels of high sensitivity C-reactive protein (hsCRP) and good responses to sulfonylureas [32].
Recently it was shown that HNF1A may upregulate the transactivation of the anti-apoptotic gene, BCL2 Like 1 (BCL2L1). This upregulation was reduced in HNF1A-mutated variants, through inhibiting the transition of G1 to S phase of the cell cycle, thus affecting the cell growth [54]. Additionally, it was suggested that HNF1A-MODY patients can have alterations in their steroid metabolism pathways, which may also explain some of the phenotypic differences, such as normal body weight, in comparison to T2DM individuals [55].
Importantly, HNF1A-MODY patients often develop micro- and macrovascular complications, with retinopathy being one of the most prevalent [56,57]. The incidence of vascular complications is similar to patients with T1DM and T2DM [58]. Moreover, Steele et al. showed that 66% of deaths in HNF1A-MODY carriers may be caused by cardiovascular disease or cerebral vascular events, which is a severe threat to this group of patients. Hyperglycaemia has a negative impact on the cardiovascular system, although the glycaemic index of HNF1A-MODY patients may not be the main reason for this high mortality [57]. The mechanism of cardiovascular impairment in HNF1A-MODY is unclear, but there are suggestions that reduced levels of apolipoprotein M (apoM) in patients with HNF1A mutation may be key since the protective role of apoM/S1P axis on the endothelium has been thoroughly described [59,60,61]. Even though the high prevalence of vascular complications in HNF1A-MODY patients was shown in many studies, there are limited data on the underlying mechanisms for such an increased risk for vascular complications. An indirect assessment of the endothelial function in HNF1A-MODY patients showed that these individuals have an increase in intima-media thickness, suggesting abnormalities in endothelial function and presence of early atherosclerotic phenotype [62]. In a more direct approach, an increase in endothelial permeability in response to a proinflammatory cytokine was found in endothelial cells with heterozygous mutation in the HNF1A gene, which were cultured under normal glycaemic conditions, suggesting that endothelial dysfunction was not a due to hyperglycaemia [63]. Other endothelial functions of these cells, such as an increase of intracellular adhesion molecule 1 (ICAM-1) after TNFα stimulation and angiogenic response, were not affected by the HNF1A mutation [63].

3.2. HNF4A-MODY

Hepatocyte nuclear factor (HNF) 4A is a transcription factor that belongs to the steroid/thyroid hormone receptor superfamily and controls the expression of many genes associated with critical metabolic pathways [64,65,66]. It contains two zing finger domains, one DNA binding region and requires homodimerisation to bind its recognition DNA site in the nucleus. HNF4A is expressed mainly in the liver, but it can be also found in kidneys, pancreatic islets, and intestine [67,68]. It is engaged in many processes such as glucose entry, lipid and drug metabolism pathways and it may be involved in amino acid metabolism [64,67,69].
HNF4A-MODY was identified back in 1978 in a family known as the RW family and at that time was the first well-identified MODY type [70,71]. Heterozygous mutations in HNF4A are relatively rare and constitute 5–10% of all MODY cases. Till 2013, 103 different mutations were reported in 173 families [72], whereas currently there are around 180 mutations in HNF4A associated with MODY (ClinVar [48]). Mutations in HNF4A were shown to cause increased insulin production in the human foetus, causing faster growth, higher body weight and even macrosomia. The hyperinsulinaemic hypoglycaemia can be detected in the early life of HNF4A-MODY carriers, leading to MODY manifestation later on [73,74,75]. This is thought to reflect a switch later in life to defective insulin secretion, although a prolonged hyperinsulinaemic phase in adulthood was described as well [74]. Therefore, HNF4A-MODY induced transient neonatal hyperglycaemia may precede the later diabetes onset [76]. However, the precise mechanism and timing of this transition remains unclear.
Several types of mutations (missense and nonsense) in HNF4A have been linked to altered insulin secretion [77], where the KATP channel in murine β-cells may play a role in the dysfunction [78]. In β-cells, HNF4A mutations cause dysfunction or lipid profile disruption, which is probably mediated through genes involved in glucose metabolism and biosynthesis [64]. These include GLUT2, aldolase B, liver pyruvate kinase, insulin, mitochondrial uncoupling protein-2 and glyceraldehyde-3-phosphate dehydrogenase [79,80]. However, most of the HNF4A mutations do not demonstrate a dominant-negative effect on the gene expression profiles, and it is still not clear how moderate decreases in HNF4A activity cause disease in MODY patients [81].
In addition, some studies suggest that single nucleotide polymorphism in the HNF4A gene may predispose to T2DM, at least in some populations [82,83,84,85].

3.3. GCK-MODY

The glucokinase (GCK) gene has a crucial role as a glucose sensor and integrates glucose metabolism with insulin secretion in pancreatic β-cells [86]. It codes for an enzyme responsible for catalysis of the first glycolysis reaction. GCK is highly expressed in hepatocytes, pancreatic β-cells and in the brain. Mutations in this gene are responsible for various glucose regulation disorders, with GCK-MODY cases among them. The most frequent mutations are heterozygous inactivating mutations leading to alterations in the kinetic parameters of this enzyme [87]. Patients with GCK-MODY show a defect in glucose sensing, therefore the glucose homeostasis is maintained at a higher set point. This results in mild and asymptomatic fasting hyperglycaemia, which is present from birth [88]. The median age of hyperglycaemia diagnosis is around 24 years of age [87]. GCK-MODY patients are characterised by increased fasting glucose levels, but the majority do not require any pharmacotherapy. The exception are pregnant women, who should be treated with insulin to reduce the risk for diabetic foetopathy in the child, which is not a carrier of the mutation [89]. The foetal genotype is not usually known, but an assumption can be made based on serial ultrasound measurements. In case there is no evidence for accelerated growth, the foetus is assumed to have inherited the GCK mutation, and in such circumstance the maternal hyperglycaemia is not treated [90].
Even though patients with GCK-MODY have long-lasting hyperglycaemia, these individuals show low prevalence of micro- and macrovascular complications [91].

3.4. HNF1B-MODY

Hepatocyte nuclear factor 1B (HNF1B) is a transcription factor expressed in many organs, predominantly in the liver, intestine, pancreas and kidney. Importantly, the expression of HNF1B also occurs in the pre-pancreatic foregut endoderm and in pancreatic multipotent progenitor cells (MPCs), therefore it has an important role in pancreatic differentiation. Lineage tracing studies have revealed that embryonic cells expressing HNF1B are precursors of acinar, duct and endocrine lineages in the pancreas [92]. Using murine models, the important role of HNF1B in the proliferation and survival of the MPCs was confirmed [93]. Heterozygous mutations in the human HNF1B are associated with MODY, which is characterised by early onset diabetes, pancreas hypoplasia and multicystic kidney dysplasia, but also with kidney diseases and multi-organ disorders [94,95,96]. Based on murine studies, it was suggested that MODY might occur not only as a consequence of β-cell dysfunction, but also as a consequence of developmental defects, leading to diabetes later in life [93].
Recently, several novel mutations in HNF1B and their relation to MODY were reported [97,98,99]. In a human-related study, it was shown that a heterozygous mutation in HNF1B caused decreased transcriptional activity, reduced DNA binding and decreased expression of the GLUT2 gene. Based on these results, the authors conclude that the impaired insulin secretion in this family is related to the reduced GLUT-2 expression in β-cells rather than decreased insulin expression [100]. In patients with HNF1B-MODY presence of cystic kidneys, pancreatic abnormalities and elevated liver enzymes are common and were used as predictors of HNF1B mutations [101]. Similarly, the presence of renal/pancreatic abnormalities in young patients with diabetes are suggestive for genetic testing for HNF1B-MODY [102,103].
Diabetes complications and cardiovascular risk factors are highly prevalent in individuals with HNF1B-MODY. In these patients, both diabetic retinopathy and neuropathy were found; however, the major complications were related to kidneys, as chronic kidney disease was reported for about 44% of the studied HNF1B-MODY patients [104].

3.5. Other MODY Types

The MODY subtypes described above account for more than 99% of all MODY cases. With the increase in next-generation sequencing (NGS) capabilities, other rarer MODY cases have been reported [105,106]. The current knowledge for possible molecular pathophysiology in rarer forms of MODY were extensively summarised elsewhere [107,108,109].

4. Diagnosis and Current Treatment Options

4.1. Diagnosis of MODY Patients

The diagnosis of MODY is relatively difficult, as many of the symptoms are highly similar to T1DM and T2DM, which usually leads to misdiagnosis. On the other hand, there is a big clinical variability between the different MODY subtypes, which makes proper diagnosis extremely hard without genetic testing. It should be noted that mutations in some MODY-associated genes (HNF1A, ABCC8, HNF4A, GCK, KCNJ11) can cause congenital hyperinsulinism and hypoglycaemia in infants and children preceding the later diabetes onset [110,111,112]. Moreover, even though a family history of diabetes is highly suggestive for MODY, some mutations in MODY-associated genes can occur in high frequencies also de novo, showing the importance of genetic testing in individuals without a family history of diabetes [113].
Even though patients with MODY were found to differ from T1DM patients in several clinical predictors, such as C-peptide concentration [114], hsCRP [115,116,117], lipid levels, polyuria or age at diagnosis, still around 38% of the MODY patients are misdiagnosed with T1DM or T2DM [27,87,118,119,120]. In 2013 Steele et al. reported that age-related glycated haemoglobin (HbA1c) reference ranges can be used as diagnostic criteria for GCK-MODY discriminator [121]. Later on, a combination of markers were tested for a discrimination of individuals with common types of diabetes and MODY [58]. It was shown that hsCRP and 1,5-anhydroglucitol (1,5-AG) could only be used to distinguish HNF1A-MODY from other MODY subtypes, but not from T1DM or T2DM [58]. In two recent population studies, it was suggested that screening for monogenic biomarkers (endogenous insulin secretion, the ratio of urinary C-peptide and creatinine (UCPCR), islet autoantibodies), is an effective, cheap and easily implemented approach for systematic screening of young patients [122,123]. The diagnostic process included three stages, where positive UCPCR patients were further tested for islet autoantibodies, and if negative were then selected for genetic testing [123]. However, the clinical reliability of UCPCR for distinguishing MODY patients from T2DM, was not confirmed [124]. Therefore, the improved diagnosis will require the application of several biomarkers together, but ultimately the genetic test is the best form of diagnosis of MODY patients.
To complicate matters further, some MODY types have similar pathology, which can also lead to misdiagnosis. Some symptoms of HNF4A-MODY, such as transient neonatal hyperinsulinemic hypoglycaemia, progressive insulin secretory defect or microvascular complications, are very similar to symptoms observed in patients with HNF1A-MODY [119,125,126]. Due to these very similar phenotypes, and because HNF1A-MODY is more prevalent, some HNF4A-MODY cases may be incorrectly recognised as HNF1A-MODY. One study suggests that up to 29% of HNF4A-MODY cases may be incorrectly credited to HNF1A-MODY and sequencing of HNF4A is proposed when there is no mutation in HNF1A [127]. Recently, it was shown that single nucleotide polymorphism located in the HNF1A gene promoter can affect the binding of HNF4A and subsequently regulate HNF1A gene expression [128].
Additionally, HNF4A-MODY can have mild and atypical clinical presentation, again hampering correct diagnosis [129]. There are also individual features of HNF4A-MODY. One of the most described phenotypes is a lower concentration of HDL-cholesterol corresponding to reduced levels of apoA2, B and C3 [64,127,130]. However, the reduced level of triglycerides and apoC3 is questionable [127]. On the other hand, in a study which screened diabetic patients under the age of 20, it was showed that 10% of all HNF4A-MODY cases present dyslipidaemia [118]. The clinical presentation of HNF1A- and HNF4A-MODY could be similar, because these two genes can regulate each other [131].
Currently it is suggested that genetic tests for MODY should be performed when paediatric diabetes is diagnosed, together with modest hyperglycaemia and absence of all four islet autoantibodies (antibodies against GAD, insulinoma antigen-2, zinc transporter 8 and insulin), but there is no standardised diagnostic algorithm [12,132,133]. At present, genetic tests for MODY are conducted with NGS methodology due to lower costs and increased diagnostic accuracy [134].

4.2. Current Treatment Options

As mentioned above, the majority of MODY patients are initially misdiagnosed with T1DM or T2DM and because of that are inappropriately treated [16,135,136]. The diagnosis of MODY has significant implications for diabetes management. For example, in patients with GCK-MODY, due to a higher basal glucose level the glucose-lowering therapies are ineffective, therefore the treatment of these patients is not recommended [88]. On the other hand, patients with HNF1A- or HNF4A-MODY are responsive to low-dose sulfonylureas, due to the increased pancreatic insulin secretion [126,137,138,139].
The importance of the correct diagnosis was highlighted in a UK population study [16]. Following genetic diagnosis for MODY, patients with GCK-MODY, who were inappropriately treated at the time of diagnosis, were able to stop this treatment without any effect on HbA1c levels. However, only 58% of HNF1A/HNF4A-MODY patients, who were also treated inappropriately in the past, could change the treatment to sulfonylurea/diet alone. In those individuals with longer duration of diabetes, it was recommended that sulfonylurea should be used together with the previous treatment started at the time of misdiagnosis [16]. The lack of response to sulfonylurea treatment in HNF1A-MODY, when diabetes duration is long-standing and mistreated, was recently confirmed in a pair of siblings with a novel HNF1A variant [140].
Even though HNF1A- and HNF4A-MODY patients are responsive to sulfonylurea, this therapy has a significant risk of hypoglycaemia. Therefore, a combination with other glucose-lowering agents such as dipeptidyl peptidase-4 inhibitor [141,142] or monotherapy with glucagon-like peptide (GLP-1) receptor agonists were also tested [143,144]. Recently, the usage of incretin hormone glucose-dependent insulinotropic peptide (GIP) and GLP-1 were tested in patients with HNF1A-MODY. These therapeutics were used together with sulfonylurea, and the results suggest that such combinations could be beneficial for HNF1A-MODY patients due to the increase of the glucose-stimulated insulin secretion [139].
To summarise, the proper recognition and MODY diagnosis is of utmost importance for the proper treatment of diabetes. The patients should be followed to determine the efficacy of the treatment and to monitor the possible vascular complications in HNF1A-and HNF4A-MODY subtypes. The most recent therapeutic approaches for MODY patients were summarised recently by Broome et al. [145].

5. Pluripotent Stem Cells for MODY Disease Modelling and Drug Research

In most MODY subtypes, the exact pathological mechanisms of disease progression are still unknown. This is due to the inaccessibility of human pancreatic tissue and the fact that rodent models do not recapitulate the MODY disease phenotype [146,147,148], with the exception of HNF1B-MODY [149]. Therefore, currently human-induced pluripotent stem cells (hiPSCs)-based disease modelling tools are being developed aiming to resolve some of the pathological mechanisms of MODY, and possibly reveal new therapeutic strategies for the patients. However, for the development of disease models, which may be used as drug-target platforms, hiPSCs must effectively differentiate and fully recapitulate the hallmarks of diseased cells and tissues. Moreover, the differentiated cells should represent homogeneous and if possible mature cell population, thus reducing the inter- and intra-experimental variation [150,151]. Additionally, to ensure valid disease modelling, the proper control lines should be selected. For monogenic diseases, as MODY, the most stringent controls are the so-called isogenic cell lines (Figure 5). These can be obtained through CRISPR/Cas9 gene editing in two ways: (1) using hiPSCs from healthy individual and introducing the mutation in the gene of interest [63], or (2) through repair of the gene-specific mutation in iPSCs derived from the diseased individual [152]. These two approaches have advantages and drawbacks. The advantages of the first approach are the relatively fast delivery of such mutated hiPSCs lines and that the observed effects will be mutation-specific, excluding the contribution of the genetic background. In the second approach, more patient-specific effects could be identified; however, such repaired hiPSCs line are hard to obtain and are dependent on the patient-specific genetic background. However, both approaches give the opportunity to identify disease-relevant phenotypes through mechanistic studies.
Up to date several hiPSC lines from different MODY types were developed [153,154,155,156]. Even though MODY is a monogenic disease, meaning that this disease is easy to model, there are limited studies using MODY patient-derived cells as disease modelling tools. One such study used patient-derived hiPSCs with a mutation in the HNF1B gene and, as a control, the authors used a non-family control hiPSCs line, and also hiPSCs from a non-diseased family member [157]. The authors were able to show that the mutant HNF1B gene expression was responsible for the compensatory increase in PDX1 gene expression in differentiated pancreatic progenitors [157]. In a different study, patient-derived iPSCs with HNF4A mutation upon differentiation toward insulin producing β-cells showed that mutation in this gene is not affecting the expression of insulin genes, nor the development of insulin-producing cells in vitro [158]. The results were obtained with patient-derived hiPSCs from several family members, and one of them, without mutation in HNF4A gene, was used as a non-diseased control [158]. A similar approach was used in a more recent study; however, the iPSCs were differentiated toward hepatopancreatic progenitors (HPPs), and alterations in hepatic and pancreatic β-cell gene signatures were found [156]. Moreover, immunofluorescent analysis showed that HNF4A protein is predominantly localised in the cytoplasm, and this mislocalisation could further account for the loss of function of HNF4A as a transcription factor [156].
There are only a few studies that used isogenic pluripotent stem cell lines to model MODY [63,152,159,160]. In all studies, CRISPR/Cas9 was used to introduce or repair the mutation in the gene of interest; however, the pluripotent stem cell source was different. In two of the studies, embryonic stem cells (ESCs) were used to introduce the mutation in the gene of interest and subsequently they were differentiated toward pancreatic β-like cells [159,160]. Cardenas-Diaz et al. found that loss of HNF1A led to an increase in alpha cell gene expression markers such as glucagon, and decreased PAX4 expression, which is crucial in regulating the development of β-cells. Moreover, these cells had impaired insulin secretion together with defects in glycolysis and mitochondrial respiration [159]. In the other study with ESCs, Zeng et al. introduced biallelic mutation in KCNJ11 gene and found that there was impaired insulin secretion together with defective glucose homeostasis. However, these cells did not show increased sensitivity to gluco- or lipotoxicity, checked with 35 mM D-glucose or 1 mM palmitate treatment [160].
In our study, hiPSCs derived from healthy donors were used to introduce a mutation in the HNF1A gene [63]. As HNF1A-MODY patients frequently develop vascular complications, the study aimed to check whether a mutation in the HNF1A gene could affect endothelial cell function. The differentiation of hiPSCs toward endothelial cells (hiPSC-ECs) was not affected by introducing HNF1A mutations. As shown in Figure 6, hiPSC-ECs derived from healthy or HNF1A-MODY individuals have similar expression of endothelial markers that are crucial for their functioning. Moreover, the majority of the endothelial functional parameters, such aspro-angiogenic responses, were not changed; however, an increase in the vascular permeability after stimulation with a pro-inflammatory cytokine was observed in hiPSC-ECs with the HNF1A mutation. These results could suggest that patients with HNF1A-MODY have increased susceptibility to the development of vascular complications [63]. The only study in which repaired patient-specific lines were used was performed by Balboa et al. [152]. They used hiPSCs from Finnish people with a mutation in the INS gene and differentiated them together with the respective isogenic control lines to β-like cells. The single-cell RNA sequencing showed increased expression of endoplasmic reticulum (ER) stress-associated genes, together with reduced proliferation. In vivo, the mutated cells had lower insulin secretion and increased levels of ER-stress markers [152].
All studies using pluripotent stem cells as disease modelling tools are summarised in Table 2.
While in vitro analysis of single cell subtypes is an excellent resource to study linage-specific disease mechanisms, currently more attention is given to developing more complex disease modelling tools. Organoid cultures require 3D growth, and during this process stem cells aggregate and differentiate in response to biophysical cues, resulting in complex structures that mimic the mature organ [161,162,163]. The advances in this field were recently reviewed by Sharma et al. [164]. In diabetes research, the current trend is moving toward developing islet organoids, which can provide large amounts of functional islets and therefore be used in underpinning disease mechanisms in vitro [165,166].

6. Final Remarks

Pathophysiological mechanisms of MODY are still not well understood. However, the usage of genomic-based approaches gives an excellent opportunity for gaining knowledge in the biology that results from the specific gene mutations in MODY, and can likely help to refine the treatment options for these patients.

Supplementary Materials

The following are available online at https://www.mdpi.com/article/10.3390/ijms22147553/s1.

Author Contributions

Conceptualisation N.K.-T.; Writing—original draft preparation D.S. and N.K.-T.; Writing—review and editing D.S., J.D. and N.K.-T.; Funding acquisition N.K.-T.; All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Science Centre Opus (2016/23/B/NZ1/01804) and Sonata-Bis (2020/38/E/NZ3/00516) research grants.

Acknowledgments

We would like to thank Paulina Podkalicka for taking the confocal images of hiPSC-ECs presented in Figure 6. Figure 4 and Figure 5 were prepared using BioRender tool (biorender.com), access granted for the period 25 May–25 June 2021 and 2 July–2 August 2021. We would like to thank Danuta E. Mossakowska for the English style corrections.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. WHO Expert Committee on Diabetes Mellitus; World Health Organization. Diabetes Mellitus: Report of a WHO Expert Committee [Meeting Held in Geneva from 24 to 30 November 1964]; World Health Organization: Geneva, Switzerland, 1965. [Google Scholar]
  2. World Health Organization. Definition, Diagnosis and Classification of Diabetes Mellitus and Its Complications: Report of a WHO Consultation. Part. 1, Diagnosis and Classification of Diabetes Mellitus; World Health Organization: Geneva, Switzerland, 1999. [Google Scholar]
  3. Tuomi, T.; Santoro, N.; Caprio, S.; Cai, M.; Weng, J.; Groop, L. The many faces of diabetes: A disease with increasing heterogeneity. Lancet 2014, 383, 1084–1094. [Google Scholar] [CrossRef]
  4. Malecki, M.T.; Mlynarski, W. Monogenic diabetes: Implications for therapy of rare types of disease. Diabetes Obes. Metab. 2008, 10, 607–616. [Google Scholar] [CrossRef] [PubMed]
  5. World Health Organization. Classification of Diabetes Mellitus; World Health Organization: Geneva, Switzerland, 2019; p. 40. [Google Scholar]
  6. Schwartz, S.S.; Epstein, S.; Corkey, B.E.; Grant, S.F.A.; Gavin, J.R.; Aguilar, R.B. The Time is Right for a New Classification System for Diabetes: Rationale and Implications of the β-Cell–Centric Classification Schema. Diabetes Care 2016, 39, 179–186. [Google Scholar] [CrossRef] [Green Version]
  7. Hattersley, A.; Bruining, J.; Shield, J.; Njolstad, P.; Donaghue, K.; International Society for Pediatric and Adolescent Diabetes. ISPAD Clinical Practice Consensus Guidelines 2006–2007. The diagnosis and management of monogenic diabetes in children. Pediatr. Diabetes 2006, 7, 352–360. [Google Scholar] [CrossRef]
  8. Hattersley, A.; Bruining, J.; Shield, J.; Njolstad, P.; Donaghue, K.C. The diagnosis and management of monogenic diabetes in children and adolescents. Pediatr. Diabetes 2009, 10 (Suppl. 12), 33–42. [Google Scholar] [CrossRef] [PubMed]
  9. Kim, S.-H. Maturity-Onset Diabetes of the Young: What Do Clinicians Need to Know? Diabetes Metab. J. 2015, 39, 468–477. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Urakami, T. Maturity-onset diabetes of the young (MODY): Current perspectives on diagnosis and treatment. Diabetes Metab. Syndr. Obes. 2019, 12, 1047–1056. [Google Scholar] [CrossRef] [Green Version]
  11. 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 (Suppl. 27), 47–63. [Google Scholar] [CrossRef] [PubMed]
  12. Nkonge, K.M.; Nkonge, D.K.; Nkonge, T.N. The epidemiology, molecular pathogenesis, diagnosis, and treatment of maturity-onset diabetes of the young (MODY). Clin. Diabetes Endocrinol. 2020, 6. [Google Scholar] [CrossRef]
  13. Heuvel-Borsboom, H.; de Valk, H.W.; Losekoot, M.; Westerink, J. Maturity onset diabetes of the young: Seek and you will find. Neth J. Med. 2016, 74, 193–200. [Google Scholar]
  14. Kavvoura, F.K.; Owen, K.R. Maturity onset diabetes of the young: Clinical characteristics, diagnosis and management. Pediatr. Endocrinol. Rev. 2012, 10, 234–242. [Google Scholar]
  15. Yorifuji, T.; Fujimaru, R.; Hosokawa, Y.; Tamagawa, N.; Shiozaki, M.; Aizu, K.; Jinno, K.; Maruo, Y.; Nagasaka, H.; Tajima, T.; et al. Comprehensive molecular analysis of Japanese patients with pediatric-onset MODY-type diabetes mellitus. Pediatr. Diabetes 2012, 13, 26–32. [Google Scholar] [CrossRef]
  16. Shepherd, M.H.; Shields, B.M.; Hudson, M.; Pearson, E.R.; Hyde, C.; Ellard, S.; Hattersley, A.T.; Patel, K.A. UNITED study 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]
  17. Irgens, H.U.; Molnes, J.; Johansson, B.B.; Ringdal, M.; Skrivarhaug, T.; Undlien, D.E.; Søvik, O.; Joner, G.; Molven, A.; Njølstad, P.R. Prevalence of monogenic diabetes in the population-based Norwegian Childhood Diabetes Registry. Diabetologia 2013, 56, 1512–1519. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Johansson, B.B.; Irgens, H.U.; Molnes, J.; Sztromwasser, P.; Aukrust, I.; Juliusson, P.B.; Søvik, O.; Levy, S.; Skrivarhaug, T.; Joner, G.; et al. Targeted next-generation sequencing reveals MODY in up to 6.5% of antibody-negative diabetes cases listed in the Norwegian Childhood Diabetes Registry. Diabetologia 2017, 60, 625–635. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Weinreich, S.S.; Bosma, A.; Henneman, L.; Rigter, T.; Spruijt, C.M.; Grimbergen, A.J.; Breuning, M.H.; de Koning, E.J.; Losekoot, M.; Cornel, M.C. A decade of molecular genetic testing for MODY: A retrospective study of utilization in The Netherlands. Eur. J. Hum. Genet. 2015, 23, 29–33. [Google Scholar] [CrossRef] [Green Version]
  20. Fendler, W.; Borowiec, M.; Baranowska-Jazwiecka, A.; Szadkowska, A.; Skala-Zamorowska, E.; Deja, G.; Jarosz-Chobot, P.; Techmanska, I.; Bautembach-Minkowska, J.; Mysliwiec, M.; et al. Prevalence of monogenic diabetes amongst Polish children after a nationwide genetic screening campaign. Diabetologia 2012, 55, 2631–2635. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Matsha, T.E.; Raghubeer, S.; Tshivhase, A.M.; Davids, S.F.G.; Hon, G.M.; Bjørkhaug, L.; Erasmus, R.T. Incidence of HNF1A and GCK MODY Variants in a South African Population. Appl. Clin. Genet. 2020, 13, 209–219. [Google Scholar] [CrossRef]
  22. Fuchsberger, C.; Flannick, J.; Teslovich, T.M.; Mahajan, A.; Agarwala, V.; Gaulton, K.J.; Ma, C.; Fontanillas, P.; Moutsianas, L.; McCarthy, D.J.; et al. The genetic architecture of type 2 diabetes. Nature 2016, 536, 41–47. [Google Scholar] [CrossRef] [Green Version]
  23. Bansal, V.; Gassenhuber, J.; Phillips, T.; Oliveira, G.; Harbaugh, R.; Villarasa, N.; Topol, E.J.; Seufferlein, T.; Boehm, B.O. Spectrum of mutations in monogenic diabetes genes identified from high-throughput DNA sequencing of 6888 individuals. BMC Med. 2017, 15, 213. [Google Scholar] [CrossRef] [Green Version]
  24. Flannick, J.; Mercader, J.M.; Fuchsberger, C.; Udler, M.S.; Mahajan, A.; Wessel, J.; Teslovich, T.M.; Caulkins, L.; Koesterer, R.; Barajas-Olmos, F.; et al. Exome sequencing of 20,791 cases of type 2 diabetes and 24,440 controls. Nature 2019, 570, 71–76. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Bonnefond, A.; Boissel, M.; Bolze, A.; Durand, E.; Toussaint, B.; Vaillant, E.; Gaget, S.; Graeve, F.D.; 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]
  26. Oliveira, S.C.; Neves, J.S.; Pérez, A.; Carvalho, D. Maturity-onset diabetes of the young: From a molecular basis perspective toward the clinical phenotype and proper management. Endocrinol. Diabetes Nutr. 2020, 67, 137–147. [Google Scholar] [CrossRef]
  27. Peixoto-Barbosa, R.; Reis, A.F.; Giuffrida, F.M.A. Update on clinical screening of maturity-onset diabetes of the young (MODY). Diabetol. Metab. Syndr. 2020, 12, 50. [Google Scholar] [CrossRef]
  28. Mohan, V.; Radha, V.; Nguyen, T.T.; Stawiski, E.W.; Pahuja, K.B.; Goldstein, L.D.; Tom, J.; Anjana, R.M.; Kong-Beltran, M.; Bhangale, T.; et al. Comprehensive genomic analysis identifies pathogenic variants in maturity-onset diabetes of the young (MODY) patients in South India. BMC Med. Genet. 2018, 19, 22. [Google Scholar] [CrossRef] [PubMed]
  29. Ferrer, J. A genetic switch in pancreatic beta-cells: Implications for differentiation and haploinsufficiency. Diabetes 2002, 51, 2355–2362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Huang, N.; Lee, I.; Marcotte, E.M.; Hurles, M.E. Characterising and Predicting Haploinsufficiency in the Human Genome. PLoS Genet. 2010, 6, e1001154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Dang, V.T.; Kassahn, K.S.; Marcos, A.E.; Ragan, M.A. Identification of human haploinsufficient genes and their genomic proximity to segmental duplications. Eur. J. Hum. Genet. 2008, 16, 1350–1357. [Google Scholar] [CrossRef] [Green Version]
  32. 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 HNF1A Alleles Are a Novel Cause of Young-Onset Diabetes and Result in Sulfonylurea-Sensitive Diabetes. Diabetes Care 2020, 43, 909–912. [Google Scholar] [CrossRef] [Green Version]
  33. Mendel, D.B.; Crabtree, G.R. HNF-1, a member of a novel class of dimerizing homeodomain proteins. J. Biol. Chem. 1991, 266, 677–680. [Google Scholar] [CrossRef]
  34. Blumenfeld, M.; Maury, M.; Chouard, T.; Yaniv, M.; Condamine, H. Hepatic nuclear factor 1 (HNF1) shows a wider distribution than products of its known target genes in developing mouse. Development 1991, 113, 589–599. [Google Scholar] [CrossRef]
  35. Harries, L.W.; Ellard, S.; Stride, A.; Morgan, N.G.; Hattersley, A.T. Isomers of the TCF1 gene encoding hepatocyte nuclear factor-1 alpha 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]
  36. Harries, L.W.; Brown, J.E.; Gloyn, A.L. Species-Specific Differences in the Expression of the HNF1A, HNF1B and HNF4A Genes. PLoS ONE 2009, 4, e7855. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Odom, D.T.; Zizlsperger, N.; Gordon, D.B.; Bell, G.W.; Rinaldi, N.J.; Murray, H.L.; Volkert, T.L.; Schreiber, J.; Rolfe, P.A.; Gifford, D.K.; et al. Control of pancreas and liver gene expression by HNF transcription factors. Science 2004, 303, 1378–1381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Luni, C.; Marth, J.D.; Doyle, F.J. Computational modeling of glucose transport in pancreatic β-cells identifies metabolic thresholds and therapeutic targets in diabetes. PLoS ONE 2012, 7, e53130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. D’Angelo, A.; Bluteau, O.; Garcia-Gonzalez, M.A.; Gresh, L.; Doyen, A.; Garbay, S.; Robine, S.; Pontoglio, M. Hepatocyte nuclear factor 1alpha and beta control terminal differentiation and cell fate commitment in the gut epithelium. Development 2010, 137, 1573–1582. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Valkovicova, T.; Skopkova, M.; Stanik, J.; Gasperikova, D. Novel insights into genetics and clinics of the HNF1A-MODY. Endocr. Regul. 2019, 53, 110–134. [Google Scholar] [CrossRef] [Green Version]
  41. Fu, J.; Wang, T.; Zhai, X.; Xiao, X. Primary hepatocellular adenoma due to biallelic HNF1A mutations and its co-occurrence with MODY 3: Case-report and review of the literature. Endocrine 2020, 67, 544–551. [Google Scholar] [CrossRef] [Green Version]
  42. Pontoglio, M.; Barra, J.; Hadchouel, M.; Doyen, A.; Kress, C.; Bach, J.P.; Babinet, C.; Yaniv, M. Hepatocyte nuclear factor 1 inactivation results in hepatic dysfunction, phenylketonuria, and renal Fanconi syndrome. Cell 1996, 84, 575–585. [Google Scholar] [CrossRef] [Green Version]
  43. 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 1alpha-deficient mice. J. Clin. Investig. 1998, 101, 2215–2222. [Google Scholar] [CrossRef] [PubMed]
  44. Abel, E.V.; Goto, M.; Magnuson, B.; Abraham, S.; Ramanathan, N.; Hotaling, E.; Alaniz, A.A.; Kumar-Sinha, C.; Dziubinski, M.L.; Urs, S.; et al. HNF1A is a novel oncogene that regulates human pancreatic cancer stem cell properties. eLife 2018, 7, e33947. [Google Scholar] [CrossRef] [PubMed]
  45. Liu, H.-T.; Liu, S.; Liu, L.; Ma, R.-R.; Gao, P. EGR1-Mediated Transcription of lncRNA-HNF1A-AS1 Promotes Cell-Cycle Progression in Gastric Cancer. Cancer Res. 2018, 78, 5877–5890. [Google Scholar] [CrossRef] [Green Version]
  46. 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] [PubMed] [Green Version]
  47. Çubuk, H.; Yalçın Çapan, Ö. A Review of Functional Characterization of Single Amino Acid Change Mutations in HNF Transcription Factors in MODY Pathogenesis. Protein J. 2021, 40, 348–360. [Google Scholar] [CrossRef]
  48. Landrum, M.J.; Lee, J.M.; Benson, M.; Brown, G.R.; Chao, C.; Chitipiralla, S.; Gu, B.; Hart, J.; Hoffman, D.; Jang, W.; et al. ClinVar: Improving access to variant interpretations and supporting evidence. Nucleic Acids Res. 2018, 46, D1062–D1067. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Vaxillaire, M.; Abderrahmani, A.; Boutin, P.; Bailleul, B.; Froguel, P.; Yaniv, M.; Pontoglio, M. Anatomy of a homeoprotein revealed by the analysis of human MODY3 mutations. J. Biol. Chem. 1999, 274, 35639–35646. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Malikova, J.; Kaci, A.; Dusatkova, P.; Aukrust, I.; Torsvik, J.; Vesela, K.; Kankova, P.D.; Njølstad, P.R.; Pruhova, S.; Bjørkhaug, L. Functional Analyses of HNF1A-MODY Variants Refine the Interpretation of Identified Sequence Variants. J. Clin. Endocrinol. Metab. 2020, 105, e1377–e1386. [Google Scholar] [CrossRef]
  51. Locke, J.M.; Saint-Martin, C.; Laver, T.W.; Patel, K.A.; Wood, A.R.; Sharp, S.A.; Ellard, S.; Bellanné-Chantelot, C.; Hattersley, A.T.; Harries, L.W.; et al. The Common HNF1A Variant I27L Is a Modifier of Age at Diabetes Diagnosis in Individuals with HNF1A-MODY. Diabetes 2018, 67, 1903–1907. [Google Scholar] [CrossRef] [Green Version]
  52. Najmi, L.A.; Aukrust, I.; Flannick, J.; Molnes, J.; Burtt, N.; Molven, A.; Groop, L.; Altshuler, D.; Johansson, S.; Bjørkhaug, L.; et al. Functional Investigations of HNF1A Identify Rare Variants as Risk Factors for Type 2 Diabetes in the General Population. Diabetes 2017, 66, 335–346. [Google Scholar] [CrossRef] [Green Version]
  53. Althari, S.; Najmi, L.A.; Bennett, A.J.; Aukrust, I.; Rundle, J.K.; Colclough, K.; Molnes, J.; Kaci, A.; Nawaz, S.; van der Lugt, T.; et al. Unsupervised Clustering of Missense Variants in HNF1A Using Multidimensional Functional Data Aids Clinical Interpretation. Am. J. Hum. Genet. 2020, 107, 670–682. [Google Scholar] [CrossRef]
  54. Sujjitjoon, J.; Charoensuk, C.; Thanyaphon, T.; Kooptiwut, S.; Thamtarana, P.J.; Tangjittipokin, W.; Chongjaroen, N.; Chanprasert, C.; Abubakar, Z.; Lapbenjakul, S.; et al. Defective functions of HNF1A variants on BCL2L1 transactivation and beta-cell growth. Biochem. Biophys. Res. Commun. 2020, 529, 826–833. [Google Scholar] [CrossRef] [PubMed]
  55. Juszczak, A.; Gilligan, L.C.; Hughes, B.A.; Hassan-Smith, Z.K.; McCarthy, M.I.; Arlt, W.; Tomlinson, J.W.; Owen, K.R. Altered cortisol metabolism in individuals with HNF1A-MODY. Clin. Endocrinol. 2020, 93, 269–279. [Google Scholar] [CrossRef] [PubMed]
  56. Szopa, M.; Wolkow, J.; Matejko, B.; Skupien, J.; Klupa, T.; Wybrańska, I.; Trznadel-Morawska, I.; Kiec-Wilk, B.; Borowiec, M.; Malecki, M.T. Prevalence of Retinopathy in Adult Patients with GCK-MODY and HNF1A-MODY. Exp. Clin. Endocrinol. Diabetes 2015, 123, 524–528. [Google Scholar] [CrossRef]
  57. 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]
  58. Szopa, M.; Klupa, T.; Kapusta, M.; Matejko, B.; Ucieklak, D.; Glodzik, W.; Zapala, B.; Sani, C.M.; Hohendorff, J.; Malecki, M.T.; et al. A decision algorithm to identify patients with high probability of monogenic diabetes due to HNF1A mutations. Endocrine 2019, 64, 75–81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Wolfrum, C.; Poy, M.N.; Stoffel, M. Apolipoprotein M is required for preβ-HDL formation and cholesterol efflux to HDL and protects against atherosclerosis. Nat. Med. 2005, 11, 418–422. [Google Scholar] [CrossRef]
  60. Kurano, M.; Yatomi, Y. Sphingosine 1-Phosphate and Atherosclerosis. J. Atheroscler. Thromb. 2018, 25, 16–26. [Google Scholar] [CrossRef] [Green Version]
  61. Richter, S.; Shih, D.Q.; Pearson, E.R.; Wolfrum, C.; Fajans, S.S.; Hattersley, A.T.; Stoffel, M. Regulation of apolipoprotein M gene expression by MODY3 gene hepatocyte nuclear factor-1alpha: Haploinsufficiency is associated with reduced serum apolipoprotein M levels. Diabetes 2003, 52, 2989–2995. [Google Scholar] [CrossRef] [Green Version]
  62. Szopa, M.; Osmenda, G.; Wilk, G.; Matejko, B.; Skupien, J.; Zapala, B.; Młynarski, W.; Guzik, T.; Malecki, M.T. Intima-media thickness and endothelial dysfunction in GCK and HNF1A-MODY patients. Eur. J. Endocrinol. 2015, 172, 277–283. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Kachamakova-Trojanowska, N.; Stepniewski, J.; Dulak, J. Human iPSCs-Derived Endothelial Cells with Mutation in HNF1A as a Model of Maturity-Onset Diabetes of the Young. Cells 2019, 8, 1440. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Lehto, M.; Bitzén, P.O.; Isomaa, B.; Wipemo, C.; Wessman, Y.; Forsblom, C.; Tuomi, T.; Taskinen, M.R.; Groop, L. Mutation in the HNF-4alpha gene affects insulin secretion and triglyceride metabolism. Diabetes 1999, 48, 423–425. [Google Scholar] [CrossRef]
  65. Gonzalez, F.J. Regulation of hepatocyte nuclear factor 4 alpha-mediated transcription. Drug Metab. Pharm. 2008, 23, 2–7. [Google Scholar] [CrossRef]
  66. Dubois, V.; Staels, B.; Lefebvre, P.; Verzi, M.P.; Eeckhoute, J. Control of Cell Identity by the Nuclear Receptor HNF4 in Organ Pathophysiology. Cells 2020, 9, 2185. [Google Scholar] [CrossRef] [PubMed]
  67. Marchesin, V.; Pérez-Martí, A.; Le Meur, G.; Pichler, R.; Grand, K.; Klootwijk, E.D.; Kesselheim, A.; Kleta, R.; Lienkamp, S.; Simons, M. Molecular Basis for Autosomal-Dominant Renal Fanconi Syndrome Caused by HNF4A. Cell Rep. 2019, 29, 4407.e5–4421.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Cattin, A.-L.; Le Beyec, J.; Barreau, F.; Saint-Just, S.; Houllier, A.; Gonzalez, F.J.; Robine, S.; Pinçon-Raymond, M.; Cardot, P.; Lacasa, M.; et al. Hepatocyte nuclear factor 4alpha, a key factor for homeostasis, cell architecture, and barrier function of the adult intestinal epithelium. Mol. Cell. Biol. 2009, 29, 6294–6308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Fang, B.; Mane-Padros, D.; Bolotin, E.; Jiang, T.; Sladek, F.M. Identification of a binding motif specific to HNF4 by comparative analysis of multiple nuclear receptors. Nucleic Acids Res. 2012, 40, 5343–5356. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Fajans, S.S.; Cloutier, M.C.; Crowther, R.L. Clinical and Etiologic Heterogeneity of Idiopathic Diabetes Mellitus. Diabetes 1978, 27, 1112–1125. [Google Scholar] [CrossRef]
  71. Fajans, S.S.; Bell, G.I. MODY. Diabetes Care 2011, 34, 1878–1884. [Google Scholar] [CrossRef] [Green Version]
  72. 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 Alpha and 4 Alpha in Maturity-Onset Diabetes of the Young and Hyperinsulinemic Hypoglycemia. Hum. Mutat. 2013, 34, 669–685. [Google Scholar] [CrossRef] [PubMed]
  73. Arya, V.B.; Rahman, S.; Senniappan, S.; Flanagan, S.E.; Ellard, S.; Hussain, K. HNF4A mutation: Switch from hyperinsulinaemic hypoglycaemia to maturity-onset diabetes of the young, and incretin response. Diabet. Med. 2014, 31, e11–e15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Bacon, S.; Kyithar, M.P.; Condron, E.M.; Vizzard, N.; Burke, M.; Byrne, M.M. Prolonged episodes of hypoglycaemia in HNF4A-MODY mutation carriers with IGT. Evidence of persistent hyperinsulinism into early adulthood. Acta Diabetol. 2016, 53, 965–972. [Google Scholar] [CrossRef] [PubMed]
  75. Clemente, M.; Vargas, A.; Ariceta, G.; Martínez, R.; Campos, A.; Yeste, D. Hyperinsulinaemic hypoglycaemia, renal Fanconi syndrome and liver disease due to a mutation in the HNF4A gene. Endocrinol. Diabetes Metab. Case Rep. 2017, 2017. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. 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] [Green Version]
  77. Barroso, I.; Luan, J.; Middelberg, R.P.S.; Harding, A.-H.; Franks, P.W.; Jakes, R.W.; Clayton, D.; Schafer, A.J.; O’Rahilly, S.; Wareham, N.J. Candidate gene association study in type 2 diabetes indicates a role for genes involved in beta-cell function as well as insulin action. PLoS Biol. 2003, 1, E20. [Google Scholar]
  78. Miura, A.; Yamagata, K.; Kakei, M.; Hatakeyama, H.; Takahashi, N.; Fukui, K.; Nammo, T.; Yoneda, K.; Inoue, Y.; Sladek, F.M.; et al. Hepatocyte nuclear factor-4alpha is essential for glucose-stimulated insulin secretion by pancreatic beta-cells. J. Biol. Chem. 2006, 281, 5246–5257. [Google Scholar] [CrossRef] [Green Version]
  79. Stoffel, M.; Duncan, S.A. The maturity-onset diabetes of the young (MODY1) transcription factor HNF4alpha regulates expression of genes required for glucose transport and metabolism. Proc. Natl. Acad. Sci. USA 1997, 94, 13209–13214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Wang, H.; Maechler, P.; Antinozzi, P.A.; Hagenfeldt, K.A.; Wollheim, C.B. Hepatocyte nuclear factor 4alpha regulates the expression of pancreatic beta -cell genes implicated in glucose metabolism and nutrient-induced insulin secretion. J. Biol. Chem. 2000, 275, 35953–35959. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Guo, S.; Lu, H. Novel Mechanisms of Regulation of the Expression and Transcriptional Activity of HNF4α. J. Cell. Biochem. 2019, 120, 519–532. [Google Scholar] [CrossRef] [Green Version]
  82. Jafar-Mohammadi, B.; Groves, C.J.; Gjesing, A.P.; Herrera, B.M.; Winckler, W.; Stringham, H.M.; Morris, A.P.; Lauritzen, T.; Doney, A.S.F.; Morris, A.D.; et al. A role for coding functional variants in HNF4A in type 2 diabetes susceptibility. Diabetologia 2011, 54, 111–119. [Google Scholar] [CrossRef] [Green Version]
  83. Andrulionytė, L.; Laukkanen, O.; Chiasson, J.-L.; Laakso, M. STOP-NIDDM Study Group Single nucleotide polymorphisms of the HNF4α gene are associated with the conversion to type 2 diabetes mellitus: The STOP-NIDDM trial. J. Mol. Med. 2006, 84, 701–708. [Google Scholar] [CrossRef] [PubMed]
  84. Granados-Silvestre, M.A.; Ortiz-López, M.G.; Granados, J.; Canizales-Quinteros, S.; Peñaloza-Espinosa, R.I.; Lechuga, C.; Acuña-Alonzo, V.; Sánchez-Pozos, K.; Menjivar, M. Susceptibility background for type 2 diabetes in eleven Mexican Indigenous populations: HNF4A gene analysis. Mol. Genet. Genom. 2017, 292, 1209–1219. [Google Scholar] [CrossRef] [PubMed]
  85. Azizi, S.M.; Sarhangi, N.; Afshari, M.; Abbasi, D.; Aghaei Meybodi, H.R.; Hasanzad, M. Association Analysis of the HNF4A Common Genetic Variants with Type 2 Diabetes Mellitus Risk. Int. J. Mol. Cell. Med. 2019, 8, 56–62. [Google Scholar]
  86. Matschinsky, F.M. Regulation of pancreatic beta-cell glucokinase: From basics to therapeutics. Diabetes 2002, 51 (Suppl. 3), S394–S404. [Google Scholar] [CrossRef] [Green Version]
  87. Wang, Z.; Diao, C.; Liu, Y.; Li, M.; Zheng, J.; Zhang, Q.; Yu, M.; Zhang, H.; Ping, F.; Li, M.; et al. Identification and functional analysis of GCK gene mutations in 12 Chinese families with hyperglycemia. J. Diabetes Investig. 2019, 10, 963–971. [Google Scholar] [CrossRef] [PubMed]
  88. 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] [PubMed] [Green Version]
  89. Hulín, J.; Škopková, M.; Valkovičová, T.; Mikulajová, S.; Rosoľanková, M.; Papcun, P.; Gašperíková, D.; Staník, J. Clinical implications of the glucokinase impaired function-GCK MODY today. Physiol. Res. 2020, 69, 995–1011. [Google Scholar] [CrossRef]
  90. Rudland, V.L. Diagnosis and management of glucokinase monogenic diabetes in pregnancy: Current perspectives. Diabetes Metab. Syndr. Obes. 2019, 12, 1081–1089. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. 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]
  92. Solar, M.; Cardalda, C.; Houbracken, I.; Martín, M.; Maestro, M.A.; De Medts, N.; Xu, X.; Grau, V.; Heimberg, H.; Bouwens, L.; et al. Pancreatic exocrine duct cells give rise to insulin-producing beta cells during embryogenesis but not after birth. Dev. Cell 2009, 17, 849–860. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. De Vas, M.G.; Kopp, J.L.; Heliot, C.; Sander, M.; Cereghini, S.; Haumaitre, C. Hnf1b controls pancreas morphogenesis and the generation of Ngn3+ endocrine progenitors. Development 2015, 142, 871–882. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. 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-1beta mutations. Ann. Intern. Med. 2004, 140, 510–517. [Google Scholar] [CrossRef] [PubMed]
  95. Chen, Y.-Z.; Gao, Q.; Zhao, X.-Z.; Chen, Y.-Z.; Bennett, C.L.; Xiong, X.-S.; Mei, C.-L.; Shi, Y.-Q.; Chen, X.-M. Systematic review of TCF2 anomalies in renal cysts and diabetes syndrome/maturity onset diabetes of the young type 5. Chin. Med. J. 2010, 123, 3326–3333. [Google Scholar] [PubMed]
  96. Bockenhauer, D.; Jaureguiberry, G. HNF1B-associated clinical phenotypes: The kidney and beyond. Pediatr. Nephrol. 2016, 31, 707–714. [Google Scholar] [CrossRef]
  97. Kato, T.; Tanaka, D.; Muro, S.; Jambaljav, B.; Mori, E.; Yonemitsu, S.; Oki, S.; Inagaki, N. A Novel p.L145Q Mutation in the HNF1B Gene in a Case of Maturity-onset Diabetes of the Young Type 5 (MODY5). Intern. Med. 2018, 57, 2035–2039. [Google Scholar] [CrossRef] [Green Version]
  98. Fujita, Y.; Tanaka, D.; Tatsuoka, H.; Matsubara, M.; Hyo, T.; Hamamoto, Y.; Komiya, T.; Inagaki, N.; Seino, Y.; Yamazaki, Y. A novel splice-site mutation of the HNF1B gene in a family with maturity onset diabetes of the young type 5 (MODY5). Endocrinol. Diabetes Metab. Case Rep. 2020, 2020. [Google Scholar] [CrossRef] [PubMed]
  99. Tao, T.; Yang, Y.; Hu, Z. A novel HNF1B mutation p.R177Q in autosomal dominant tubulointerstitial kidney disease and maturity-onset diabetes of the young type 5: A pedigree-based case report. Medicine 2020, 99, e21438. [Google Scholar] [CrossRef]
  100. Kim, E.K.; Lee, J.S.; Cheong, H.I.; Chung, S.S.; Kwak, S.H.; Park, K.S. Identification and Functional Characterization of P159L Mutation in HNF1B in a Family with Maturity-Onset Diabetes of the Young 5 (MODY5). Genom. Inform. 2014, 12, 240–246. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Sztromwasser, P.; Michalak, A.; Małachowska, B.; Młudzik, P.; Antosik, K.; Hogendorf, A.; Zmysłowska, A.; Borowiec, M.; Młynarski, W.; Fendler, W. A cross-sectional study of patients referred for HNF1B-MODY genetic testing due to cystic kidneys and diabetes. Pediatr. Diabetes 2020, 21, 422–430. [Google Scholar] [CrossRef] [Green Version]
  102. Bustamante, C.; Sanchez, J.; Seeherunvong, T.; Ukarapong, S. Early Onset of Mody5 Due to Haploinsufficiency of HNF1B. AACE Clin. Case Rep. 2020, 6, e243–e246. [Google Scholar] [CrossRef]
  103. Motyka, R.; Kołbuc, M.; Wierzchołowski, W.; Beck, B.B.; Towpik, I.E.; Zaniew, M. Four Cases of Maturity Onset Diabetes of the Young (MODY) Type 5 Associated with Mutations in the Hepatocyte Nuclear Factor 1 Beta (HNF1B) Gene Presenting in a 13-Year-Old Boy and in Adult Men Aged 33, 34, and 35 Years in Poland. Am. J. Case Rep. 2021, 22, e928994. [Google Scholar] [PubMed]
  104. Dubois-Laforgue, D.; Cornu, E.; Saint-Martin, C.; Coste, J.; Bellanné-Chantelot, C.; Timsit, J. 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 (HNF1B) Molecular Defects. Diabetes Care 2017, 40, 1436–1443. [Google Scholar] [CrossRef] [Green Version]
  105. Ivanoshchuk, D.E.; Shakhtshneider, E.V.; Rymar, O.D.; Ovsyannikova, A.K.; Mikhailova, S.V.; Orlov, P.S.; Ragino, Y.I.; Voevoda, M.I. Analysis of APPL1 Gene Polymorphisms in Patients with a Phenotype of Maturity Onset Diabetes of the Young. J. Pers. Med. 2020, 10, 100. [Google Scholar] [CrossRef]
  106. De Santana, L.S.; Caetano, L.A.; Costa-Riquetto, A.D.; Franco, P.C.; Dotto, R.P.; Reis, A.F.; Weinert, L.S.; Silveiro, S.P.; Vendramini, M.F.; do Prado, F.A.; et al. Targeted sequencing identifies novel variants in common and rare MODY genes. Mol. Genet. Genom. Med. 2019, 7, e962. [Google Scholar] [CrossRef] [Green Version]
  107. Pipatpolkai, T.; Usher, S.; Stansfeld, P.J.; Ashcroft, F.M. New insights into KATP channel gene mutations and neonatal diabetes mellitus. Nat. Rev. Endocrinol. 2020, 16, 378–393. [Google Scholar] [CrossRef] [PubMed]
  108. Beltrand, J.; Busiah, K.; Vaivre-Douret, L.; Fauret, A.L.; Berdugo, M.; Cavé, H.; Polak, M. Neonatal Diabetes Mellitus. Front. Pediatr. 2020, 8, 540718. [Google Scholar] [CrossRef] [PubMed]
  109. Aarthy, R.; Aston-Mourney, K.; Mikocka-Walus, A.; Radha, V.; Amutha, A.; Anjana, R.M.; Unnikrishnan, R.; Mohan, V. Clinical features, complications and treatment of rarer forms of maturity-onset diabetes of the young (MODY)—A review. J. Diabetes Its Complicat. 2020, 35, 107640. [Google Scholar] [CrossRef]
  110. 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]
  111. 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]
  112. Rozenkova, K.; Malikova, J.; Nessa, A.; Dusatkova, L.; Bjørkhaug, L.; Obermannova, B.; Dusatkova, P.; Kytnarova, J.; Aukrust, I.; Najmi, L.A.; et al. High Incidence of Heterozygous ABCC8 and HNF1A Mutations in Czech Patients with Congenital Hyperinsulinism. J. Clin. Endocrinol. Metab. 2015, 100, E1540–E1549. [Google Scholar] [CrossRef] [Green Version]
  113. Stanik, J.; Dusatkova, P.; Cinek, O.; Valentinova, L.; Huckova, M.; Skopkova, M.; Dusatkova, L.; Stanikova, D.; Pura, M.; Klimes, I.; et al. De novo mutations of GCK, HNF1A and HNF4A may be more frequent in MODY than previously assumed. Diabetologia 2014, 57, 480–484. [Google Scholar] [CrossRef] [PubMed]
  114. Besser, R.E.J.; Shepherd, M.H.; McDonald, T.J.; Shields, B.M.; Knight, B.A.; Ellard, S.; Hattersley, A.T. Urinary C-peptide creatinine ratio is a practical outpatient tool for identifying hepatocyte nuclear factor 1-α/hepatocyte nuclear factor 4-α maturity-onset diabetes of the young from long-duration type 1 diabetes. Diabetes Care 2011, 34, 286–291. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Owen, K.R.; Thanabalasingham, G.; James, T.J.; Karpe, F.; Farmer, A.J.; McCarthy, M.I.; Gloyn, A.L. Assessment of high-sensitivity C-reactive protein levels as diagnostic discriminator of maturity-onset diabetes of the young due to HNF1A mutations. Diabetes Care 2010, 33, 1919–1924. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. McDonald, T.J.; Shields, B.M.; Lawry, J.; Owen, K.R.; Gloyn, A.L.; Ellard, S.; Hattersley, A.T. High-sensitivity CRP discriminates HNF1A-MODY from other subtypes of diabetes. Diabetes Care 2011, 34, 1860–1862. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Thanabalasingham, G.; Shah, N.; Vaxillaire, M.; Hansen, T.; Tuomi, T.; Gašperíková, D.; Szopa, M.; Tjora, E.; James, T.J.; Kokko, P.; et al. A large multi-centre European study validates high-sensitivity C-reactive protein (hsCRP) as a clinical biomarker for the diagnosis of diabetes subtypes. Diabetologia 2011, 54, 2801–2810. [Google Scholar] [CrossRef]
  118. Warncke, K.; Kummer, S.; Raile, K.; Grulich-Henn, J.; Woelfle, J.; Steichen, E.; Prinz, N.; Holl, R.W. Frequency and Characteristics of MODY 1 (HNF4A Mutation) and MODY 5 (HNF1B Mutation): Analysis From the DPV Database. J. Clin. Endocrinol. Metab. 2019, 104, 845–855. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Chandran, S.; Rajadurai, V.S.; Hoi, W.H.; Flanagan, S.E.; Hussain, K.; Yap, F. A Novel HNF4A Mutation Causing Three Phenotypic Forms of Glucose Dysregulation in a Family. Front. Pediatr 2020, 8, 320. [Google Scholar] [CrossRef]
  120. Fu, J.; Wang, T.; Liu, J.; Wang, X.; Zhang, Q.; Li, M.; Xiao, X. Using Clinical Indices to Distinguish MODY2 (GCK Mutation) and MODY3 (HNF1A Mutation) from Type 1 Diabetes in a Young Chinese Population. Diabetes Ther. 2019, 10, 1381–1390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Steele, A.M.; Wensley, K.J.; Ellard, S.; Murphy, R.; Shepherd, M.; Colclough, K.; Hattersley, A.T.; Shields, B.M. Use of HbA1c in the identification of patients with hyperglycaemia caused by a glucokinase mutation: Observational case control studies. PLoS ONE 2013, 8, e65326. [Google Scholar] [CrossRef]
  122. Shields, B.M.; Shepherd, M.; Hudson, M.; McDonald, T.J.; Colclough, K.; Peters, J.; Knight, B.; Hyde, C.; Ellard, S.; Pearson, E.R.; et al. Population-Based Assessment of a Biomarker-Based Screening Pathway to Aid Diagnosis of Monogenic Diabetes in Young-Onset Patients. Diabetes Care 2017, 40, 1017–1025. [Google Scholar] [CrossRef] [Green Version]
  123. Shepherd, M.; Shields, B.; Hammersley, S.; Hudson, M.; McDonald, T.J.; Colclough, K.; Oram, R.A.; Knight, B.; Hyde, C.; Cox, J.; et al. Systematic Population Screening, Using Biomarkers and Genetic Testing, Identifies 2.5% of the U.K. Pediatric Diabetes Population With Monogenic Diabetes. Diabetes Care 2016, 39, 1879–1888. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Wang, Y.; Gao, Y.; Cai, X.; Chen, L.; Zhou, L.; Ma, Y.; Gong, S.; Han, X.; Ji, L. Clinical Implications of Urinary C-Peptide Creatinine Ratio in Patients with Different Types of Diabetes. J. Diabetes Res. 2019, 2019, 1747684. [Google Scholar] [CrossRef] [Green Version]
  125. Naylor, R.; Knight Johnson, A.; del Gaudio, D. Maturity-Onset Diabetes of the Young Overview. In GeneReviews®; Adam, M.P., Ardinger, H.H., Pagon, R.A., Wallace, S.E., Bean, L.J., Stephens, K., Amemiya, A., Eds.; University of Washington: Seattle, WA, USA, 1993. [Google Scholar]
  126. Gardner, D.S.; Tai, E.S. Clinical features and treatment of maturity onset diabetes of the young (MODY). Diabetes Metab. Syndr. Obes. 2012, 5, 101–108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Pearson, E.R.; Pruhova, S.; Tack, C.J.; Johansen, A.; Castleden, H.A.J.; 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]
  128. Španinger, E.; Potočnik, U.; Bren, U. Molecular Dynamics Simulations Predict that rSNP Located in the HNF-1α Gene Promotor Region Linked with MODY3 and Hepatocellular Carcinoma Promotes Stronger Binding of the HNF-4α Transcription Factor. Biomolecules 2020, 10, 1700. [Google Scholar] [CrossRef]
  129. Spiro, A.J.; Vu, K.N.; Warnock, A.L. An Atypical HNF4A Mutation Which Does Not Conform to the Classic Presentation of HNF4A-MODY. Case Rep. Endocrinol. 2018, 2018, 1560472. [Google Scholar] [CrossRef] [Green Version]
  130. Shih, D.Q.; Dansky, H.M.; Fleisher, M.; Assmann, G.; Fajans, S.S.; Stoffel, M. Genotype/phenotype relationships in HNF-4alpha/MODY1: Haploinsufficiency is associated with reduced apolipoprotein (AII), apolipoprotein (CIII), lipoprotein(a), and triglyceride levels. Diabetes 2000, 49, 832–837. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Yamagata, K. Roles of HNF1α and HNF4α in pancreatic β-cells: Lessons from a monogenic form of diabetes (MODY). Vitam. Horm. 2014, 95, 407–423. [Google Scholar]
  132. Carlsson, A.; Shepherd, M.; Ellard, S.; Weedon, M.; Lernmark, Å.; Forsander, G.; Colclough, K.; Brahimi, Q.; Valtonen-Andre, C.; Ivarsson, S.A.; et al. Absence of Islet Autoantibodies and Modestly Raised Glucose Values at Diabetes Diagnosis Should Lead to Testing for MODY: Lessons From a 5-Year Pediatric Swedish National Cohort Study. Diabetes Care 2020, 43, 82–89. [Google Scholar] [CrossRef] [Green Version]
  133. Marchand, L.; Li, M.; Leblicq, C.; Rafique, I.; Alarcon-Martinez, T.; Lange, C.; Rendon, L.; Tam, E.; Courville-Le Bouyonnec, A.; Polychronakos, C. Monogenic Causes in the Type 1 Diabetes Genetics Consortium Cohort: Low Genetic Risk for Autoimmunity in Case Selection. J. Clin. Endocrinol. Metab. 2021, 106, 1804–1810. [Google Scholar] [CrossRef]
  134. Tatsi, E.B.; Kanaka-Gantenbein, C.; Scorilas, A.; Chrousos, G.P.; Sertedaki, A. Next generation sequencing targeted gene panel in Greek MODY patients increases diagnostic accuracy. Pediatr. Diabetes 2020, 21, 28–39. [Google Scholar] [CrossRef]
  135. Pihoker, C.; Gilliam, L.K.; Ellard, S.; Dabelea, D.; Davis, C.; Dolan, L.M.; Greenbaum, C.J.; Imperatore, G.; Lawrence, J.M.; Marcovina, S.M.; et al. Prevalence, Characteristics and Clinical Diagnosis of Maturity Onset Diabetes of the Young Due to Mutations in HNF1A, HNF4A, and Glucokinase: Results From the SEARCH for Diabetes in Youth. J. Clin. Endocrinol. Metab. 2013, 98, 4055–4062. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Shields, B.M.; Hicks, S.; Shepherd, M.H.; Colclough, K.; Hattersley, A.T.; Ellard, S. Maturity-onset diabetes of the young (MODY): How many cases are we missing? Diabetologia 2010, 53, 2504–2508. [Google Scholar] [CrossRef] [PubMed]
  137. 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]
  138. Bacon, S.; Kyithar, M.P.; Rizvi, S.R.; Donnelly, E.; McCarthy, A.; Burke, M.; Colclough, K.; Ellard, S.; Byrne, M.M. Successful maintenance on sulphonylurea therapy and low diabetes complication rates in a HNF1A-MODY cohort. Diabet. Med. 2016, 33, 976–984. [Google Scholar] [CrossRef]
  139. 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]
  140. Tan, C.S.H.; Ang, S.F.; Lim, S.C. Response to multiple glucose-lowering agents in a sib-pair with a novel HNF1α (MODY3) variant. Eur. J. Hum. Genet. 2020, 28, 518–520. [Google Scholar] [CrossRef] [PubMed]
  141. Sidelmann Christensen, A.; Storgaard, H.; Hædersdal, S.; Hansen, T.; Knop, F.K.; Vilsbøll, T. Glimepiride monotherapy versus combination of glimepiride and linagliptin therapy in patients with HNF1A-diabetes: A protocol for a randomised, double-blinded, placebo-controlled trial. BMJ Open 2018, 8, e022517. [Google Scholar] [CrossRef] [Green Version]
  142. Christensen, A.S.; Hædersdal, S.; Støy, J.; Storgaard, H.; Kampmann, U.; Forman, J.L.; Seghieri, M.; Holst, J.J.; Hansen, T.; Knop, F.K.; et al. Efficacy and Safety of Glimepiride with or without Linagliptin Treatment in Patients With HNF1A Diabetes (Maturity-Onset Diabetes of the Young Type 3): A Randomized, Double-Blinded, Placebo-Controlled, Crossover Trial (GLIMLINA). Diabetes Care 2020, 43, 2025–2033. [Google Scholar] [CrossRef] [PubMed]
  143. 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] [Green Version]
  144. 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]
  145. 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]
  146. Lee, Y.H.; Sauer, B.; Gonzalez, F.J. Laron dwarfism and non-insulin-dependent diabetes mellitus in the Hnf-1alpha knockout mouse. Mol. Cell. Biol. 1998, 18, 3059–3068. [Google Scholar] [CrossRef] [Green Version]
  147. Hagenfeldt-Johansson, K.A.; Herrera, P.L.; Wang, H.; Gjinovci, A.; Ishihara, H.; Wollheim, C.B. Beta-cell-targeted expression of a dominant-negative hepatocyte nuclear factor-1 alpha induces a maturity-onset diabetes of the young (MODY)3-like phenotype in transgenic mice. Endocrinology 2001, 142, 5311–5320. [Google Scholar] [CrossRef] [PubMed]
  148. Shih, D.Q.; Heimesaat, M.; Kuwajima, S.; Stein, R.; Wright, C.V.E.; Stoffel, M. Profound defects in pancreatic β-cell function in mice with combined heterozygous mutations in Pdx-1, Hnf-1α, and Hnf-3β. Proc. Natl. Acad. Sci. USA 2002, 99, 3818–3823. [Google Scholar] [CrossRef] [Green Version]
  149. Quilichini, E.; Fabre, M.; Nord, C.; Dirami, T.; Le Marec, A.; Cereghini, S.; Pasek, R.C.; Gannon, M.; Ahlgren, U.; Haumaitre, C. Insights into the etiology and physiopathology of MODY5/HNF1B pancreatic phenotype with a mouse model of the human disease. J. Pathol. 2021, 254, 31–45. [Google Scholar]
  150. Saha, K.; Jaenisch, R. Technical challenges in using human induced pluripotent stem cells to model disease. Cell Stem Cell 2009, 5, 584–595. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Volpato, V.; Webber, C. Addressing variability in iPSC-derived models of human disease: Guidelines to promote reproducibility. Dis. Models Mech. 2020, 13, dmm042317. [Google Scholar] [CrossRef] [Green Version]
  152. Balboa, D.; Saarimäki-Vire, J.; Borshagovski, D.; Survila, M.; Lindholm, P.; Galli, E.; Eurola, S.; Ustinov, J.; Grym, H.; Huopio, H.; et al. Insulin mutations impair beta-cell development in a patient-derived iPSC model of neonatal diabetes. eLife 2018, 7, e38519. [Google Scholar] [CrossRef] [PubMed]
  153. Stepniewski, J.; Kachamakova-Trojanowska, N.; Ogrocki, D.; Szopa, M.; Matlok, M.; Beilharz, M.; Dyduch, G.; Malecki, M.T.; Jozkowicz, A.; Dulak, J. Induced pluripotent stem cells as a model for diabetes investigation. Sci. Rep. 2015, 5, 8597. [Google Scholar] [CrossRef] [Green Version]
  154. Teo, A.K.K.; Gupta, M.K.; Doria, A.; Kulkarni, R.N. Dissecting diabetes/metabolic disease mechanisms using pluripotent stem cells and genome editing tools. Mol. Metab. 2015, 4, 593–604. [Google Scholar] [CrossRef] [PubMed]
  155. Braverman-Gross, C.; Nudel, N.; Ronen, D.; Beer, N.L.; McCarthy, M.I.; Benvenisty, N. Derivation and molecular characterization of pancreatic differentiated MODY1-iPSCs. Stem Cell Res. 2018, 31, 16–26. [Google Scholar] [CrossRef] [PubMed]
  156. Ng, N.H.J.; Jasmen, J.B.; Lim, C.S.; Lau, H.H.; Krishnan, V.G.; Kadiwala, J.; Kulkarni, R.N.; Ræder, H.; Vallier, L.; Hoon, S.; et al. HNF4A Haploinsufficiency in MODY1 Abrogates Liver and Pancreas Differentiation from Patient-Derived Induced Pluripotent Stem Cells. iScience 2019, 16, 192–205. [Google Scholar] [CrossRef] [Green Version]
  157. Teo, A.K.K.; Lau, H.H.; Valdez, I.A.; Dirice, E.; Tjora, E.; Raeder, H.; Kulkarni, R.N. Early Developmental Perturbations in a Human Stem Cell Model of MODY5/HNF1B Pancreatic Hypoplasia. Stem Cell Rep. 2016, 6, 357–367. [Google Scholar] [CrossRef] [Green Version]
  158. Vethe, H.; Bjørlykke, Y.; Ghila, L.M.; Paulo, J.A.; Scholz, H.; Gygi, S.P.; Chera, S.; Ræder, H. Probing the missing mature β-cell proteomic landscape in differentiating patient iPSC-derived cells. Sci. Rep. 2017, 7, 4780. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Cardenas-Diaz, F.L.; Osorio-Quintero, C.; Diaz-Miranda, M.A.; Kishore, S.; Leavens, K.; Jobaliya, C.; Stanescu, D.; Ortiz-Gonzalez, X.; Yoon, C.; Chen, C.S.; et al. Modeling Monogenic Diabetes using Human ESCs Reveals Developmental and Metabolic Deficiencies Caused by Mutations in HNF1A. Cell Stem Cell 2019, 25, 273.e5–289.e5. [Google Scholar] [CrossRef] [PubMed]
  160. Zeng, H.; Guo, M.; Zhou, T.; Tan, L.; Chong, C.N.; Zhang, T.; Dong, X.; Xiang, J.Z.; Yu, A.S.; Yue, L.; et al. An Isogenic Human ESC Platform for Functional Evaluation of Genome-wide-Association-Study-Identified Diabetes Genes and Drug Discovery. Cell Stem Cell 2016, 19, 326–340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  161. Dutta, D.; Heo, I.; Clevers, H. Disease Modeling in Stem Cell-Derived 3D Organoid Systems. Trends Mol. Med. 2017, 23, 393–410. [Google Scholar] [CrossRef]
  162. Liu, C.; Oikonomopoulos, A.; Sayed, N.; Wu, J.C. Modeling human diseases with induced pluripotent stem cells: From 2D to 3D and beyond. Development 2018, 145, dev156166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Rowe, R.G.; Daley, G.Q. Induced pluripotent stem cells in disease modelling and drug discovery. Nat. Rev. Genet. 2019, 20, 377–388. [Google Scholar] [CrossRef] [PubMed]
  164. Sharma, A.; Sances, S.; Workman, M.J.; Svendsen, C.N. Multi-lineage Human iPSC-Derived Platforms for Disease Modeling and Drug Discovery. Cell Stem Cell 2020, 26, 309–329. [Google Scholar] [CrossRef]
  165. Zhang, X.; Ma, Z.; Song, E.; Xu, T. Islet organoid as a promising model for diabetes. Protein Cell 2021, 1–19. [Google Scholar]
  166. Balboa, D.; Iworima, D.G.; Kieffer, T.J. Human Pluripotent Stem Cells to Model Islet Defects in Diabetes. Front. Endocrinol. 2021, 12, 149. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Classification system of diabetes cases (based on WHO, 2019 [5]).
Figure 1. Classification system of diabetes cases (based on WHO, 2019 [5]).
Ijms 22 07553 g001
Figure 2. Subclassification of the monogenic diabetes (based on WHO, 2019 [5]).
Figure 2. Subclassification of the monogenic diabetes (based on WHO, 2019 [5]).
Ijms 22 07553 g002
Figure 3. Prevalence of MODY types in some European countries.
Figure 3. Prevalence of MODY types in some European countries.
Ijms 22 07553 g003
Figure 4. Schematic representation of the HNF1A (isoform A) structure with highlighted functional elements: dimerisation (blue), DNA-binding (green and yellow) and transactivation (red) domains together with the respective amino acids region shown in brackets. The mutations depicted in the figure show 196 of the 198 HNF1A-MODY-related mutations found in ClinVar database and located in the coding sequence of the gene. Two mutations from the database were not included, as they are located in the HNF1A promoter region (Supplementary Table S1). The mutations are additionally stratified by clinical significance and the total number of variations in the particular functional domain is presented. Grey boxes—disordered regions; NLS—nuclear localisation signal (range 197–205 amino acids); POUS—POU specific domain; POUH—POU homeodomain. Figure created with BioRender.com.
Figure 4. Schematic representation of the HNF1A (isoform A) structure with highlighted functional elements: dimerisation (blue), DNA-binding (green and yellow) and transactivation (red) domains together with the respective amino acids region shown in brackets. The mutations depicted in the figure show 196 of the 198 HNF1A-MODY-related mutations found in ClinVar database and located in the coding sequence of the gene. Two mutations from the database were not included, as they are located in the HNF1A promoter region (Supplementary Table S1). The mutations are additionally stratified by clinical significance and the total number of variations in the particular functional domain is presented. Grey boxes—disordered regions; NLS—nuclear localisation signal (range 197–205 amino acids); POUS—POU specific domain; POUH—POU homeodomain. Figure created with BioRender.com.
Ijms 22 07553 g004
Figure 5. Approaches for generation of human disease-relevant isogenic lines: (1) starting from healthy or (2) diseased individual. Figure created with BioRender.com.
Figure 5. Approaches for generation of human disease-relevant isogenic lines: (1) starting from healthy or (2) diseased individual. Figure created with BioRender.com.
Ijms 22 07553 g005
Figure 6. hiPSC-derived endothelial cells differentiated as described in [63]. Cells were derived from healthy (control) and two HNF1A-MODY individuals (HNF1A-MODYa and HNF1A-MODYb). No difference in the expression of VE-cadherin (red) or phosphorylated form of endothelial nitric oxide synthase (phospho-eNOS, green) could be observed.
Figure 6. hiPSC-derived endothelial cells differentiated as described in [63]. Cells were derived from healthy (control) and two HNF1A-MODY individuals (HNF1A-MODYa and HNF1A-MODYb). No difference in the expression of VE-cadherin (red) or phosphorylated form of endothelial nitric oxide synthase (phospho-eNOS, green) could be observed.
Ijms 22 07553 g006
Table 1. Current MODY subtypes—gene function, clinical manifestation and complications.
Table 1. Current MODY subtypes—gene function, clinical manifestation and complications.
Gene SymbolFull NameGene FunctionClinical ManifestationDiabetic Complications
HNF1AHepatocyte nuclear factor 1 alphatranscription factorprogressive insulin secretory defect; diminished renal threshold for glycosuria;common
GCKGlucokinaseenzyme in the first step of glucose metabolismstable, mild fasting hyperglycemia;rare
HNF4AHepatocyte nuclear factor 4 alphatranscription factortransient neonatal diabetes; progressive insulin secretory defectcommon
HNF1BHepatocyte nuclear factor 1 betatranscription factorrenal abnormalities and insufficiency at young age; liver test abnormalities; exocrine pancreatic dysfunction; hyperuricemiacommon
PDX1Pancreatic and duodenal homebox-1transcription factorpermanent neonatal diabetes in homozygote; pancreas agenesisunknown
NEUROD1Neurogenic differentiation 1transcription factorneonatal diabetes; pancreatic abnormalities; child or adult-onset diabetes neurological abnormalitiesunknown
KLF11Krupell-like factor 11transcription factorpancreatic malignancy; similar to T2DMunknown
CELCarboxyl ester lipasecontrols exocrine and endocrine functions of pancreasexocrine pancreatic dysfunction; lipomatosis and fibrosis with posterior diabetes developmentunknown
PAX4Paired box 4transcription factorpossible ketoacidosisunknown
INSInsulinencode the insulin precursorpermanent neonatal diabetesunknown
BLKB-lymphoid tyrosine kinasetyrosine kinase functions in signal transductionoverweightunknown
ABCC8ATP-binding cassette C8regulating insulin releasepermanent or transient neonatal diabetesunknown
KCNJ11Inwardly rectifying potassium channel subfamily J member 11regulating insulin releaseneonatal diabetes in homozygoteunknown
APPL1Adaptor protein, phosphotyrosine interacting with PH domain and leucine zipper 1insulin signaling pathwayinsulin secretion defect; child or adult-onset diabetesunknown
Table 2. Summary of studies using pluripotent stem cells as disease modelling tool, hiPSCs—human induced pluripotent stem cells, ESCs—embryonic stem cells, ER—endoplasmic reticulum.
Table 2. Summary of studies using pluripotent stem cells as disease modelling tool, hiPSCs—human induced pluripotent stem cells, ESCs—embryonic stem cells, ER—endoplasmic reticulum.
MODY SubtypePluripotent CellsDifferentiated Cell TypeControl LinesMechanism RevealedReference
HNF1B-MODYPatient-derived hiPSCsPancreatic progenitorsFamily non-diseased and non-family control individualsCompensatory increase in PDX1 in mutant pancreatic progenitors.[157]
HNF4A-MODYPatient-derived hiPSCsInsulin-producing beta-cellsFamily non-diseased and non-family control individualsNo effect on expression of insulin genes, nor in the development of insulin-producing beta cells[158]
HNF4A-MODYPatient-derived hiPSCsHepatopancreatic progenitors (HPPs)Family non-diseased and non-family control individualsAlterations in hepatic and pancreatic beta-cell signatures and abnormal cytoplasmic localisation of HNF4A.[156]
HNF1A-MODYESCsPancreatic beta-like cellsIsogenic controlIncrease in alpha-cell gene expression markers, impaired insulin secretion, defect in glycolysis and mitochondrial respiration.[159]
KCNJ11-MODYESCs (biallelic mutation introduced)Pancreatic beta-like cellsIsogenic controlImpaired insulin secretion, defective glucose homeostasis[160]
INS-MODYPatient-derived hiPSCsPancreatic beta-like cellsIsogenic controlIncreased expression of ER-stress associated genes, reduced proliferation in vitro, lower insulin secretion in vivo together with increased ER-stress markers.[152]
HNF1A-MODYhiPSCsEndothelial cellsIsogenic controlIncreased vascular permeability in response to pro-inflammatory cytokine, no difference in pro-angiogenic response.[63]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Skoczek, D.; Dulak, J.; Kachamakova-Trojanowska, N. Maturity Onset Diabetes of the Young—New Approaches for Disease Modelling. Int. J. Mol. Sci. 2021, 22, 7553. https://doi.org/10.3390/ijms22147553

AMA Style

Skoczek D, Dulak J, Kachamakova-Trojanowska N. Maturity Onset Diabetes of the Young—New Approaches for Disease Modelling. International Journal of Molecular Sciences. 2021; 22(14):7553. https://doi.org/10.3390/ijms22147553

Chicago/Turabian Style

Skoczek, Dawid, Józef Dulak, and Neli Kachamakova-Trojanowska. 2021. "Maturity Onset Diabetes of the Young—New Approaches for Disease Modelling" International Journal of Molecular Sciences 22, no. 14: 7553. https://doi.org/10.3390/ijms22147553

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