Entry - *142410 - HNF1 HOMEOBOX A; HNF1A - OMIM
* 142410

HNF1 HOMEOBOX A; HNF1A


Alternative titles; symbols

TRANSCRIPTION FACTOR 1; TCF1
HEPATOCYTE NUCLEAR FACTOR-1-ALPHA
HEPATOCYTE NUCLEAR FACTOR 1; HNF1
HEPATIC NUCLEAR FACTOR-1-ALPHA
ALBUMIN PROXIMAL FACTOR


HGNC Approved Gene Symbol: HNF1A

Cytogenetic location: 12q24.31     Genomic coordinates (GRCh38): 12:120,978,543-121,002,512 (from NCBI)


Gene-Phenotype Relationships
Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
12q24.31 {Diabetes mellitus, insulin-dependent} 222100 AR 3
{Diabetes mellitus, noninsulin-dependent, 2} 125853 AD 3
Diabetes mellitus, insulin-dependent, 20 612520 3
Hepatic adenoma, somatic 142330 3
MODY, type III 600496 AD 3
Renal cell carcinoma 144700 3

TEXT

Cloning and Expression

Using a rat Hnf1 cDNA-derived probe, Bach et al. (1990) isolated HNF1 clones from a human liver cDNA library. The deduced 631-amino acid human HNF1 protein contains a homeodomain in its N-terminal half and shares close similarity with the 628-amino acid rat protein.

The amino acid sequence of HNF1 displays distant sequence homology to the homeodomains of homeotic genes (see 142950) (Courtois et al., 1987).


Gene Function

The orderly and sequential activation of genes during development is thought to be related to the selective expression of groups of regulatory proteins acting primarily at the level of transcription. Courtois et al. (1987) found a nuclear protein in hepatocytes, but not in other cell types, that binds to a sequence required for hepatocyte-specific transcription of the genes for the alpha and beta chains of fibrinogen (134820, 134830) and alpha-1-antitrypsin (107400). This protein, called hepatocyte nuclear factor-1 (HNF1) by them, interacts with sequences required for optimal promoter function of the genes mentioned. The promoter or enhancer regions for several viral and cellular genes not expressed in the liver did not compete for binding to these sequences. HNF1 is predominantly expressed in liver and kidney. The restricted expression of HNF1 and its selective interaction with the control regions of several liver-specific genes suggested to Courtois et al. (1987) that it is involved in developmentally regulated gene expression in the liver. HNF1 binds to the promoters of a variety of genes that are expressed exclusively in the liver, e.g., fibrinogen-alpha and -beta, albumin (103600), alpha-fetoprotein (104150), alpha-1-antitrypsin, liver-type pyruvate kinase (609712), transthyretin (176300), aldolase B (612724), and hepatitis B virus large surface protein.

Dimerization among transcription factors is a frequent finding in the regulation of eukaryotic gene expression. HNF1-alpha functions as a dimer. Mendel et al. (1991) identified DCOH (126090), a dimerization cofactor of HNF1-alpha, which displayed a restricted tissue distribution and did not bind to DNA but, rather, selectively stabilized HNF1-alpha dimers. Hua et al. (2000) showed that the dimerization motif of HNF1-alpha forms an intermolecular 4-helix bundle. The bundle is destabilized by a subset of mutations associated with maturity-onset diabetes of the young (MODY; 606391). Impaired dimerization of the beta-cell transcription factor thus provides a molecular mechanism of metabolic deregulation in diabetes mellitus.

Van Wering et al. (2002) showed that mouse Gata5 (611496) and Hnf1-alpha interacted in vitro and in transfected COS-7 cells. The interaction required the C-terminal zinc finger and basic region of Gata5 and the homeodomain of Hnf1-alpha. Physical association of GATA5 and HNF1-alpha was required for synergistic activation of the human lactase-phlorizin hydrolase (LCT; 603202) promoter. Deletion of the HNF1-alpha activation domains or interruption of the HNF1-binding sites in the LCT promoter resulted in complete loss of transcriptional activity, whereas deletion of the GATA5 activation domains or interruption of the GATA-binding sites reduced, but did not eliminate, transcriptional activity.

To gain insight into the transcriptional regulatory networks that specify and maintain human tissue diversity, Odom et al. (2004) used chromatin immunoprecipitation combined with promoter microarrays to identify systematically the genes occupied by the transcriptional regulators HNF1-alpha, HNF4-alpha (600281), and HNF6 (604164), together with RNA polymerase II (see 180660), in human liver and pancreatic islets. Odom et al. (2004) identified tissue-specific regulatory circuits formed by HNF1-alpha, HNF4-alpha, and HNF6 with other transcription factors, revealing how these factors function as master regulators of hepatocyte and islet transcription. Odom et al. (2004) concluded that their results suggested how misregulation of HNF4-alpha can contribute to type 2 diabetes (125853). They found that HNF1-alpha is bound to at least 222 target genes in hepatocytes. HNF1-alpha occupied the promoter regions of 106 genes within pancreatic islets, 30% of which were also bound by HNF1-alpha in hepatocytes. In islets, fewer chaperones and enzymes were bound by HNF1-alpha than in hepatocytes, and the receptors and signal transduction machinery regulated by HNF1-alpha varied between the 2 tissues. Odom et al. (2004) found that HNF4-alpha bound to the promoters of about 12% of hepatocyte islet genes represented on the microarray. HNF4-alpha acted in a much larger number of hepatocyte and beta-cell genes than did HNF1-alpha, suggesting that HNF4-alpha has broad activities in these 2 tissues.

Odom et al. (2007) analyzed the binding of FOXA2 (600288), HNF1A, HNF4A, and HNF6 to 4,000 orthologous gene pairs in hepatocytes purified from human and mouse livers. Despite the conserved function of these factors, 41 to 89% of the binding events seemed to be species-specific. Importantly, the binding sites varied widely between species in ways that could not be predicted from human-mouse sequence alignments alone.


Mapping

Bach et al. (1990) assigned the human HNF1 gene to chromosome 12q24.3 by in situ hybridization, and the mouse gene to 5F by RFLP analysis of interspecific mouse backcrosses. One other gene, that for short chain acyl-CoA-dehydrogenase (606885), had also been assigned to chromosomes 12 and 5 in man and mouse, respectively. Kuo et al. (1990) also assigned the HNF1 gene to 12q22-qter in the human and to chromosome 5 in the mouse. By means of somatic cell hybrids segregating either human or rat chromosomes, Szpirer et al. (1992) independently assigned the TCF1 gene to human chromosome 12 and found that it was located also on rat chromosome 12, thereby defining a new segment of homology between the 2 species (and a segment of mouse chromosome 5).


Molecular Genetics

MODY, a single-gene disorder responsible for 2 to 5% of noninsulin-dependent (type II) diabetes mellitus (NIDDM; 125853), is characterized by autosomal dominant inheritance and an age of onset of 25 years or younger. Yamagata et al. (1996) narrowed the localization of a form of MODY (MODY3; 600496) to chromosome 12q24.2 by a combination of genetic linkage and fluorescence in situ hybridization. To identify the nature of the MODY3 gene, Yamagata et al. (1996) used a combination of approaches, including testing genes known to be on 12q to see if they mapped into the contig to which MODY3 mapped, exon trapping, and cDNA selection, for which human pancreatic islet cDNA was used (insulin secretion is abnormal in MODY3 patients, making islets a likely site of expression of MODY3 mRNA and protein). They identified 14 genes encoding known proteins, 12 known expressed sequence tags (ESTs), and 9 new ESTs. They found mutations in the gene encoding hepatocyte nuclear factor-1-alpha, a transcription factor that helps in the tissue-specific regulation of the expression of several liver genes and also functions as a weak transactivator of the rat insulin-I gene. In all, Yamagata et al. (1996) identified 6 different mutations which were associated with MODY3 (e.g., 142410.0001). In several pedigrees individuals were found who inherited the mutant allele and the at-risk chromosome 12 haplotype but were nondiabetic or only showed evidence of impaired glucose tolerance or diabetes during pregnancy. These individuals were expected eventually to develop diabetes mellitus. In one member of a family NIDDM was diagnosed at the age of 65 years, at which time he was mildly obese, suggesting that he had late-onset NIDDM rather than MODY.

Vaxillaire et al. (1997) examined 10 unrelated Caucasian families in whom MODY/NIDDM cosegregated with markers for MODY3 and found 10 different mutations in the TCF1 gene, all of which cosegregated with diabetes (see 142410.0003 and 142410.0004). In these families, they found no obvious relationships between the nature of the mutations observed (i.e., frameshift, nonsense, or missense), or their location in the gene, with clinical features of diabetes (e.g., age at onset, severity). The authors stated that the mechanisms by which mutations in the TCF1 gene caused diabetes mellitus were unclear, but might include abnormal pancreatic islet development during fetal life, as well as impaired transcriptional regulation of genes that play a key role in normal pancreatic beta-cell function.

Urhammer et al. (1997) found a variety of variations in the TCF1 gene in 245 Danish NIDDM patients and 242 age-matched controls. The frequencies of the variants were similar in the 2 groups except that an arg583-to-gln mutation was found in 2 of the 245 NIDDM patients and in none of the control subjects. The authors concluded that genetic variation in the TCF1 gene is not a common factor contributing to NIDDM susceptibility in white subjects of Danish ancestry. Urhammer et al. (1998) studied the frequent amino acid polymorphisms ile27 to leu and ser487 to asn of the TCF1 gene to determine whether they were associated with alterations in glucose-induced serum C-peptide and serum insulin responses among Caucasian glucose-tolerant first-degree relatives of NIDDM patients. The authors concluded that these polymorphisms had no major impact on the pancreatic beta-cell function, as estimated during an oral and intravenous glucose challenge.

Urhammer et al. (1998) studied the TCF1 ala98-to-val polymorphism in glucose-tolerant first-degree relatives of type 2 diabetic patients of the same ethnic origin. All participants, 231 glucose-tolerant offspring of 62 type 2 diabetic probands, underwent an oral glucose tolerance test (OGTT) with measurements of plasma glucose, serum insulin, and serum C-peptide during the test. Thirty-three heterozygous carriers of the ala98-to-val variant were identified, whereas no subjects had the variant in its homozygous form. Carriers of ala98 to val had a 20% reduction in serum C-peptide at 30 minutes during the OGTT compared to wildtype carriers. No significant differences in serum insulin levels during the OGTT were observed between carriers of the variant and ala98 homozygotes.

In a study of 15 UK MODY families for mutations in the TCF1 gene, Frayling et al. (1997) found 8 different mutations in 11 families (73%). A previously reported mutation, the insertion of a C in the C tract encoding the sequence 289-pro-pro-pro-291 (142410.0001), was present in 4 of the families. A screen of a further 32 probands with early-onset (less than 40 years of age) NIDDM showed the mutation in 2 additional families. This common mutation was present on at least 3 different haplotypes, suggesting that its high frequency is due to recurrent mutation rather than founder effect. Thus, Frayling et al. (1997) concluded that TSF1 mutations are a common cause of MODY in UK families and result in early-onset NIDDM with a progressive clinical course. Hansen et al. (1997) sequenced the coding region and intron-exon boundaries of the TCF1 gene in 9 unrelated Danish Caucasian subjects with MODY and found mutations in 5. These 5 mutations were found in neither 84 NIDDM patients nor in 84 control subjects.

MODY3 is characterized by a severe insulin secretory defect, compared with MODY2 (125851), a glucokinase-deficient diabetes. Because of the rapid progress to overt diabetes and the high prevalence of the requirement for insulin treatment in patients with MODY3, Yamada et al. (1997) screened the HNF1A gene for mutations in Japanese subjects with insulin-dependent diabetes mellitus (IDDM; 222100). Mutations were identified in 3 (5.5%) of the 55 unrelated subjects with IDDM (e.g., 142410.0001, 142410.0005, and 142410.0006). None of these mutations was found in 200 normal chromosomes from nondiabetic subjects. The results indicated that mutation in the HNF1A gene can lead to development not only of early-onset NIDDM but also of IDDM. In a subclassification of IDDM, the HNF1A-deficient type should be distinguished from the classic type of autoimmune-based IDDM in Japanese. All of these mutations were heterozygous.

Ellard (2000) stated that 65 different mutations in the TCF1 gene had been found to cause MODY3 in a total of 116 families worldwide. They noted that diagnostic and predictive genetic testing is possible for the majority of patients with MODY, opening new avenues for the classification, prediction, and perhaps eventually the prevention of diabetes in these families.

Fajans et al. (2001) reported that mutations in the HNF1A gene have been identified in all racial and ethnic backgrounds, including European, Chinese, Japanese, African, and American Indian. Mutations in the HNF1A gene appear to be the most common cause of MODY among adults seen in diabetic clinics.

Bluteau et al. (2002) found germline TCF1 mutations in 2 individuals who had previously had a liver-tumor resection and had familial diabetes. One of these individuals had a hepatocellular carcinoma that had developed in an adenoma and had a gly574-to-ser mutation (142410.0013). This mutation was described by Collet et al. (2002) as frequent in Africans with diabetes. These results suggested that germline mutations of TCF1 may predispose to benign liver tumor development, and may explain the previously described cosegregation of liver adenoma with diabetes mellitus in a large family (Foster et al., 1978). Bluteau et al. (2002) suggested the results of their studies indicated that individuals with MODY could benefit from liver monitoring to detect early tumor occurrence, and individuals with liver adenomas, especially those with a family history of the same condition, should be tested for diabetes.

To elucidate the function of a molecular hotspot, Chi et al. (2002) cocrystallized human HNF1A amino acids 83 to 279 with a high-affinity promoter and solved the structure of the complex. Two identical protein molecules were bound to the promoter. Each contained a homeodomain (POU-H) and a second domain structurally similar to POU-specific (POU-S) domains that was not predicted on the basis of amino acid sequence. Atypical elements in both domains created a stable interface that further distinguished HNF1A from other flexible POU-homeodomain proteins. Chi et al. (2002) determined that 76% of MODY3-associated missense mutations in HNF1A occur in the region encompassing amino acids 98 to 272, which includes the POU-H and POU-S domains and a nuclear localization signal. They subdivided these mutations according to functional classes predicted to affect DNA binding, POU-S/POU-H domain interactions, protein stability, and nuclear localization. The largest class affected DNA binding, either through direct interactions or indirectly by perturbing local environment.

To estimate the prevalence of MODY3 in Norwegian diabetic pedigrees, Bjorkhaug et al. (2003) screened a total of 130 families for HNF1A mutations; 42 families with clinical MODY, 75 with suspected MODY, and 13 pedigrees with multiplex type 1 diabetes (IDDM). Twenty-two families with clinical MODY, 15 families with suspected MODY, and 1 family with type 1 diabetes multiplex harbored HNF1A mutations. Thus, in about half of Norwegian families with clinical MODY, mutations in the HNF1A gene could be detected. Eight of the 18 different mutations identified were novel. Haplotypes were determined for recurrent mutations, indicating a founder effect in Norway for the hotspot mutation P291fsinsC (142410.0001) and possibly also for P112L (142410.0015) and R131W (142410.0016). Two mutant HNF1A proteins were unable to bind DNA and at least 5 mutants showed defective nuclear translocation. Transcriptional activation was reduced for most of the MODY3-associated mutants. Accordingly, the functional studies of HNF1A mutants indicated that beta-cell dysfunction in MODY3 is caused by loss-of-function mechanisms like reduced DNA binding, impaired transcriptional activation, and defects in subcellular localization.

Johansen et al. (2005) examined the prevalence and nature of mutations in the 3 common MODY genes HNF4A (600281), GCK (138079), and TCF1 in Danish patients with a clinical diagnosis of MODY and determined metabolic differences in probands with and without mutations in HNF4A, GCK, and TCF1. They identified 29 different mutations in 38 MODY families. Fifteen of the mutations were novel. The variants segregated with diabetes within the families, and none of the variants were found in 100 normal Danish chromosomes. Their findings suggested a relative prevalence of 3% of MODY1 (125850) (2 different mutations in 2 families), 10% of MODY2 (7 in 8), and 36% of MODY3 (21 in 28) among Danish kindred clinically diagnosed as MODY. No significant differences in biochemical and anthropometric measurements were observed at baseline examinations. Forty-nine percent of the families carried mutations in the 3 examined MODY genes.

Rebouissou et al. (2005) screened 35 renal neoplasms for HNF1A and HNF1B (189907) inactivation. In 2 of 13 clear cell renal carcinomas, the authors found a monoallelic germline mutation (142410.0001 and 142410.0022) of HNF1A with no associated suppression of target mRNA expression. In normal and tumor renal tissues, there was a network of transcription factors differentially regulated in tumor subtypes. There was a related cluster of coregulated genes associating HNF1A, HNF4A, FABP1 (134650), and UGT2B7 (600068). Rebouissou et al. (2005) suggested that germline mutation of HNF1A may predispose to renal tumors.

Ridker et al. (2008) performed a multistage genomewide association study of CRP (123260) levels and found significant association with 7 loci, 1 of which was HNF1A. Reiner et al. (2008) reported an association between common variants of the HNF1A gene and plasma CRP concentrations in 2 independent populations of older adults.

Penetrance of HNF1A Mutations in Diabetes

Mirshahi et al. (2022) comprehensively assessed the penetrance and prevalence of pathogenic variants in HNF1A, HNF4A, and GCK that account for more than 80% of monogenic diabetes. Mirshahi et al. (2022) analyzed clinical and genetic data from 1,742 clinically referred probands, 2,194 family members, clinically unselected individuals from a US health system-based cohort of 132,194 individuals, and a UK population-based cohort of 198,748 individuals, and found that 1 in 1,500 individuals harbor a pathogenic variant in one of these genes. The penetrance of pathogenic GCK variants was similar (89 to 97%) across all cohorts. The penetrance of diabetes for HNF1A and HNF4A pathogenic variants was substantially lower in the clinically unselected individuals compared to clinically referred probands and was dependent on the setting (32% in the population, 49% in the health system cohort, 86% in a family member, and 98% in probands for HNF1A). The relative risk of diabetes was similar across the clinically unselected cohorts, highlighting the role of environment/ other genetic factors. The authors suggested that for HNF1A and HNF4A, genetic interpretation and counseling should be tailored to the setting in which a pathogenic monogenic variant was identified. GCK is an exception with near-complete penetrance in all settings.


Cytogenetics

Liver adenomas are benign tumors at risk of malignant transformation. In a genomewide search for loss of heterozygosity (LOH) associated with liver adenomas, Bluteau et al. (2002) found a deletion in 12q in 5 of 10 adenomas. In most cases, LOH at 12q was the only recurrent genetic alteration observed, suggesting the presence of a tumor-suppressor gene in that region. A minimal common region of deletion was defined in 12q24 that included the TCF1 gene. Bluteau et al. (2002) found biallelic inactivation of TCF1 in 10 of 16 screened adenomas, and heterozygous germline mutations were present in 3 affected individuals. Furthermore, 2 well-differentiated hepatocellular carcinomas occurring in normal liver, out of 30 HCCs screened, contained somatic biallelic mutations. These results indicated that inactivation of TCF1, whether sporadic or associated with MODY3, is an important genetic event in the occurrence of human liver adenomas, and may be an early step in the development of some hepatocellular carcinomas.


Animal Model

Gonzalez et al. (1990) found that newborn mice homozygous for a 1.2-cM deletion of chromosome 7 do not show the increased activity of CYP2E (124040), which is regulated by the transcription factor Hnf1. They suggested that the deleted region of chromosome 7 contains a gene encoding a transacting factor that is epistatic in a regulatory cascade that includes Hnf1 gene expression.

Pontoglio et al. (1996) found that mice with inactivation of the Hnf1 gene through homologous recombination failed to thrive and died around weaning after a progressive wasting syndrome with marked liver enlargement. The transcription rate of genes such as albumin and alpha-1-antitrypsin was reduced, while the gene coding the phenylalanine hydroxylase (612349) was totally silent, giving rise to phenylketonuria. Mutant mice also suffered from severe Fanconi syndrome (see 227650) caused by renal proximal tubular dysfunction. The resulting massive urinary glucose lost led to energy and water wasting. Pontoglio et al. (1996) commented that Hnf1-deficient mice may provide a model for human renal Fanconi syndrome.

Shih et al. (2001) explored the molecular basis for the hypercholesterolemia of Tcf1 -/- mice using oligonucleotide microchip expression analysis. Shih et al. (2001) demonstrated that Tcf1 -/- mice have a defect in bile acid transport, increased bile acid and liver cholesterol synthesis, and impaired high-density lipoprotein (HDL) metabolism. Tcf1 -/- liver has decreased expression of the basolateral membrane bile acid transporters Slc10a1 (182396), Slc21a3 (602883), and Slc21a5, leading to impaired portal bile acid uptake and elevated plasma bile acid concentrations. In intestine and kidneys, Tcf1 -/- mice lack expression of the ileal bile acid transporter (Slc10a2; 601295), resulting in increased fecal and urinary bile acid excretion. Tcf1 protein also regulates transcription of Nr1h4 (603826), encoding the farnesoid X receptor-1 (Fxr1), thereby leading to reduced expression of small heterodimer partner-1 (Shp1; 604630) and repression of Cyp7a1 (118455), the rate-limiting enzyme in the classic bile acid biosynthesis pathway. In addition, hepatocyte bile acid storage protein is absent from Tcf1 -/- mice. Increased plasma cholesterol of Tcf1 -/- mice resides predominantly in large buoyant HDL particles. This is most likely due to reduced activity of the HDL-catabolic enzyme hepatic lipase (151670) and increased expression of HDL-cholesterol esterifying enzyme lecithin:cholesterol acyltransferase (LCAT; 606967). Shih et al. (2001) concluded that TCF1, in addition to being an important regulator of insulin secretion, is an essential transcriptional regulator of bile acid in HDL-cholesterol metabolism.

Hiraiwa et al. (2001) investigated whether there is a molecular link between HNF1A deficiency and function of the G6Pase (602671) system. Transactivation studies revealed that HNF1A is required for transcription of the G6PT gene. Hepatic G6PT mRNA levels and microsomal G6P transport activity are also markedly reduced in Hnf1a -/- mice as compared with Hnf1a +/+ and Hnf1a +/- littermates. On the other hand, hepatic G6Pase mRNA expression and activity are upregulated in Hnf1a -/- mice, consistent with observations that G6Pase expression is increased in diabetic animals. Taken together, these results strongly suggest that metabolic abnormalities in Hnf1a-null mice are caused in part by G6PT deficiency and by perturbations of the G6Pase system.

Huang et al. (2011) demonstrated the direct induction of functional hepatocyte-like (induced hepatocyte, iHep) cells from mouse tail-tip fibroblasts by transduction of Gata4 (600576), Hnf1-alpha, and Foxa3 (602295) and inactivation of p19(Arf) (600160). iHep cells showed typical epithelial morphology, expressed hepatic genes, and acquired hepatocyte functions. Notably, transplanted iHep cells repopulated the livers of fumarylacetoacetate hydrolase-deficient (Fah-null; see 613871) mice and rescued almost half of recipients from death by restoring liver functions.


Nomenclature

Although the symbol TCF1 (transcription factor-1) is used in the literature for this gene, its official designation is HNF1A. It should not be confused with the TCF7 gene (189908), which has also been referred to as TCF1 (T cell-specific transcription factor-1) in the literature.


ALLELIC VARIANTS ( 22 Selected Examples):

.0001 MATURITY-ONSET DIABETES OF THE YOUNG, TYPE 3

HEPATIC ADENOMA, SOMATIC, INCLUDED
RENAL CELL CARCINOMA, CLEAR CELL, INCLUDED
TYPE 1 DIABETES MELLITUS 20, INCLUDED
HNF1A, 1-BP INS, 872C
  
RCV000016062...

Maturity-Onset Diabetes of the Young, Type 3

In a patient from an Edinburgh pedigree with MODY3 (600496), Yamagata et al. (1996) found in exon 4 of the TCF1 gene an insertion of a cytosine at codon 291 (pro), resulting in a frameshift and synthesis of a truncated mutant protein of 315 amino acids. This mutation was present in all affected members and no unaffected members of this family. It was not found on screening 55 healthy nondiabetic white subjects.

Using a rapid screening PCR method for frameshift mutations in the HNF1A gene in the screening of 60 MODY probands defined according to strict diagnostic criteria, Frayling et al. (1997) detected mutations in 11 (18%); the insertion mutation accounted for 13% of the MODY cases.

Ellard (2000) stated that the C insertion in the poly(C) tract of exon 4 had been reported in 22 of the 116 families with MODY3 worldwide who were identified by the finding of a mutation in the TCF1 gene. The total number of different mutations described was 65.

Bjorkhaug et al. (2003) found the P291fsinsC mutation in 9 families, 8 of Norwegian origin. Microsatellite analysis data suggested that in 7 of these families the mutant allele had a common origin.

Hepatic Adenoma

In studies of hepatic adenomas (142330) demonstrating biallelic inactivation of TCF1, Bluteau et al. (2002) observed the pro291fsX316 frameshift mutation (142410.0001) in heterozygous state in the tumor tissue of 2 individuals, one with multiple adenoma and the other with hepatocellular carcinoma (114550).

Renal Cell Carcinoma

In a 78-year-old man with clear cell renal carcinoma (see 144700), Rebouissou et al. (2005) identified heterozygosity for the 872insC mutation. Mutation screening of a tumor sample detected the germline mutation without mutation/deletion of the second allele. The man was diagnosed with diabetes mellitus in his sixth decade that was controlled by diet and oral hypoglycemic agents. No relatives had a diagnosis of renal carcinoma or diabetes.

Type 1 Diabetes Mellitus 20

In a Japanese subject with type 1 diabetes mellitus (T1D20; 612520) in whom insulin treatment was begun when hyperglycemia and ketonuria were noticed at 15 years of age, Yamada et al. (1997) identified a heterozygous frameshift mutation of codon pro291 resulting from insertion of a C in a poly(C) tract. (They designated this mutation P291fsinsC.) The mutation was predicted to result in a mutant truncated protein of 340 amino acids. The same mutation had been observed in British, German, and Finnish MODY families (Byrne et al., 1996; Yamagata et al., 1996; Kaisaki et al., 1997). Thus, Yamada et al. (1997) concluded that this site in exon 4 of the HNF1A gene appears to be a mutation hotspot.


.0002 MATURITY-ONSET DIABETES OF THE YOUNG, TYPE 3

HNF1A, PRO447LEU
  
RCV000016065...

In their family A, Yamagata et al. (1996) found that MODY3 (600496) was associated with a single amino acid substitution in exon 7 of the TCF1 gene: codon 447 was changed from CCG (pro) to CTG (leu).

Hansen et al. (1997) found this mutation in a glucose-tolerant lean male who had relatives with MODY. He showed a low insulin secretion rate during oral glucose tolerance test (OGTT), but a 2-fold increase in pancreatic beta-cell response after intravenous glucose and a 2.5- to 4-fold increase in beta-cell response after either intravenous tolbutamide or intravenous glucagon loads. Hansen et al. (1997) concluded that early stages in the pathogenesis of MODY3 caused by the P447L mutation may be characterized by hyperexcitability of beta-cells to intravenous secretagogues.


.0003 MATURITY-ONSET DIABETES OF THE YOUNG, TYPE 3

HNF1A, 1-BP DEL
  
RCV000016066

In a family in which 4 members of 3 generations had MODY3 (600496), Vaxillaire et al. (1997) found deletion of a guanine from codon glycine-292 (G292fsdelG) resulting in frameshift in the TCF1 gene.


.0004 MATURITY-ONSET DIABETES OF THE YOUNG, TYPE 3

HNF1A, TYR122CYS
  
RCV000016067...

In a family with multiple members with MODY3 (600496) in 3 generations, Vaxillaire et al. (1997) found a TAC-to-TGC transition in codon 122 of the TCF1 gene, predicted to cause an amino acid change from tyrosine to cysteine (Y122C).


.0005 TYPE 1 DIABETES MELLITUS 20

HNF1A, ARG272HIS
  
RCV000016068...

In a Japanese subject who developed type 1 diabetes mellitus (T1D20; 612520) 1 year after the diagnosis of T2D at 8 years of age, Yamada et al. (1997) identified heterozygosity for an arg272-to-his (R272H) mutation in the DNA binding domain of the HNF1A gene.


.0006 TYPE 1 DIABETES MELLITUS 20

HNF1A, ARG583GLY
  
RCV000016069...

In a Japanese patient with sudden-onset type 1 diabetes (T1D20; 612520) at 20 years of age, Yamada et al. (1997) identified heterozygosity for an arg583-to-gly (R583G) mutation in the transactivation domain of HNF1A. When first diagnosed there was marked hyperglycemia and 'absolute' insulin deficiency, prompting the initiation of insulin therapy. Control of blood glucose levels by exogenous insulin was poor, and complications of diabetes (proliferative retinopathy, cataracts, and sensorimotor neuropathy) developed.


.0007 MATURITY-ONSET DIABETES OF THE YOUNG, TYPE 3

HNF1A, A-C, -58, PROMOTER
  
RCV000016070...

Gragnoli et al. (1997) found an A-to-C substitution at nucleotide -58 of the promoter region of the HNF1A gene that cosegregated with MODY3 (600496). This mutation is located in a highly conserved region of the promoter and disrupted the binding site for the transcription factor HNF-4-alpha (600281), mutations in the gene encoding HNF-4-alpha being another cause of MODY (MODY1; 125850). This result demonstrated that decreased levels of HNF1-alpha per se can cause MODY. Moreover, it indicated that both the promoter and the coding regions of the HNF1A gene should be screened for mutations in subjects thought to have MODY.


.0008 TYPE 2 DIABETES MELLITUS, SUSCEPTIBILITY TO

HNF1A, GLY319SER
  
RCV000016071...

Hegele et al. (1999) identified a gly319-to-ser (G319S) variant in the HNF1A gene in Ontario Oji-Cree with early-onset type 2 diabetes (125853). G319S is in the proline II-rich domain of the trans-activation site of HNF1A and alters a glycine residue that is conserved throughout evolution. S319 was absent from 990 alleles from 6 other ethnic groups, suggesting that it is private for Oji-Cree. The S319 allele was more prevalent in diabetic than in nondiabetic Oji-Cree (0.209 vs 0.087; P = 0.000001). S319/S319 homozygotes and S319/G319 heterozygotes, respectively, had odds ratios for type 2 diabetes of 4.00 (95% CI, 2.65-6.03) and 1.97 (95% CI, 1.44-2.70) compared with G319/G319 homozygotes. There was a significant difference in the mean age of onset of type 2 diabetes, with G319/G319, S319/G319, and S319/S319 subjects being affected in the fifth, fourth, and third decades of life, respectively. Among nondiabetic subjects, S319/G319 heterozygotes had significantly lower plasma insulin than G319/G319 homozygotes. The authors concluded that the G319S variant is associated with a distinct form of type 2 diabetes, characterized by onset at an earlier age, lower body mass, and a higher postchallenge plasma glucose.

That the majority of Oji-Cree subjects with diabetes did not have the HNF1A S319 variant suggested to Hegele et al. (2000) that there might be other genetic determinants of diabetes susceptibility. In the course of sequencing candidate genes in diabetic subjects who were homozygous for HNF1A G319/G319, they found that some subjects had the PPARG A12 variant (601487.0002). PPARG A12 was strongly associated with type 2 diabetes in women, but not in men. The authors concluded that, when taken together with the previously reported association of diabetes with HNF1A in both men and women, the gender-specific association with PPARG A12 confirms that type 2 diabetes is etiologically complex in the Oji-Cree and that at least 2 genes are involved in determining susceptibility to the disease in this population.

Triggs-Raine et al. (2002) stated that Oji-Cree type 2 diabetes does not resemble MODY, because affected Oji-Cree subjects are obese and insulin-resistant with elevated plasma insulin concentrations, which clearly were insufficient to prevent diabetes onset. They evaluated the in vitro function of HNF1A G319S both to confirm that the mutation had a functional effect and to determine whether this effect was distinct from those of the complete loss-of-function or dominant-negative mutations seen in the MODY3 phenotype. They also evaluated the impact of the HNF1A G319S mutation on the dynamics of type 2 diabetes onset in the whole Sandy Lake Oji-Cree community. They found that the G319S mutation reduced the in vitro ability of HNF1-alpha to activate transcription by approximately 50%, with no effect on DNA binding or protein stability. There was no evidence of a dominant-negative effect of the mutant protein. Disease onset showed significant differences according to G319S genotype when gauged by the age at which half the subjects had become diabetic. Each dose of G319S accelerated median disease onset by approximately 7 years. Thus, the transactivation-deficient HNF1A G319S mutation affects the dynamics of disease onset. The demonstration of a functional consequence for the G319S mutation provided a mechanistic basis for its strong association with Oji-Cree type 2 diabetes and its unparalleled specificity for diabetes prediction in these people, in whom diabetes presents a significant public health problem. The finding also showed that HNF1A mutations can be associated with typical adult-onset insulin-resistant obesity-related diabetes in addition to maturity-onset diabetes of the young. Triggs-Raine et al. (2002) stated that in the Oji-Cree, HNF1A G319S behaves as a susceptibility allele for type 2 diabetes. Among nondiabetic Oji-Cree, fasting plasma insulin concentration was reduced significantly in HNF1A G319S carriers, suggesting that the partial impairment of function is tolerated when there is no insulin resistance. However, among Oji-Cree with type 2 diabetes, both carriers and noncarriers of the mutation had elevated plasma insulin concentration compared with nondiabetic Oji-Cree. The stress of obesity-induced insulin resistance seemed to expose the partial defect in HNF1A G319S carriers, causing expression of the disease.


.0009 MATURITY-ONSET DIABETES OF THE YOUNG, TYPE 3

HNF1A, THR620ILE
  
RCV000016072...

Miedzybrodzka et al. (1999) described a family in which a thr620-to-ile substitution in transcription factor-1 was found in all members affected by MODY (600496). The mutation was not fully penetrant, as 2 family members aged 87 and 46 had the mutation but did not have diabetes. The severity and age at diagnosis of diabetes varied widely within the family, and most presented over the age of 25. Miedzybrodzka et al. (1999) suggested that TCF1 mutation screening should be considered in any family with autosomal dominant inheritance of diabetes where one member has presented with diabetes before the age of 25.


.0010 MATURITY-ONSET DIABETES OF THE YOUNG, TYPE 3

HNF1A, 1-BP DEL, -119G, PROMOTER
  
RCV000016073

Godart et al. (2000) observed a promoter mutation, -119delG, of the TCF1 gene that segregated with MODY3 (600496) in a diabetic family.


.0011 INSULIN RESISTANCE, SUSCEPTIBILITY TO

SERUM HDL CHOLESTEROL LEVEL, MODIFIER OF, INCLUDED
HNF1A, ILE27LEU
  
RCV000016074...

Chiu et al. (2000) examined the relationship between the ile27-to-leu (I27L) polymorphism of HNF1-alpha and insulin sensitivity (see 125853) and beta-cell function assessed by a hyperglycemic clamp. This study included 52 healthy glucose-tolerant and normotensive subjects (age, 19 to 40 years; body mass index, 17.58-35.61 kg/m2; waist/hip ratio, 0.65-1.03). Chiu et al. (2000) identified 19 LL subjects, 24 IL subjects, and 9 II subjects. The LL group had the highest postchallenge insulin levels at 30 and 90 min (P = 0.038 and P = 0.015, respectively) and also the highest insulin area under curve (P = 0.009) among the 3 genotypes. The LL group was more insulin resistant than the IL and II groups (P = 0.042 for insulin sensitivity index). After adjusting for age, gender, obesity, and ethnicity, the I27L polymorphism was an independent determinant of the insulin sensitivity index (P = 0.001). However, it had no impact on either the first or second phase insulin response. The authors concluded that the I27L polymorphism is associated with insulin resistance, but not beta-cell function. The mechanism of this association is unclear, but HNF1-alpha may play a role in regulating hepatic glucose metabolism.

Babaya et al. (2003) studied the relationship of the HNF1A gene polymorphism I27L with lipid parameters, in particular with serum HDL cholesterol level, in 356 unrelated Japanese men. Though no significant difference was observed in total cholesterol and triglyceride levels among the 3 genotypes, the serum HDL cholesterol level was significantly associated with the genotype (P less than 0.01). Subjects with the II genotype had low serum HDL cholesterol levels, and those with the LL genotype had high serum HDL cholesterol levels. The authors concluded that the HNF1A gene locus is associated with serum HDL cholesterol level and suggested that the I27 allele is a risk marker for atherosclerosis.


.0012 TYPE 1 DIABETES MELLITUS 20

HNF1A, 1-BP DEL, 142G
  
RCV000016076...

Yoshiuchi et al. (2001) identified a 142delG frameshift mutation in the TCF1 gene in a family with a strong history of type 1 diabetes (T1D20; 612520). The expression of the mutant protein was not detected in COS-7 cells by Western blot analysis after transfection of the mutant cDNA. According to the authors, this was the first case of an unstable mutant HNF1-alpha protein. Reporter gene analysis indicated that the mutant protein had no transactivation activity in HeLa and other cells. Haploinsufficiency for the TCF1 gene may lead to severe forms of diabetes resembling type I diabetes.


.0013 MATURITY-ONSET DIABETES OF THE YOUNG, TYPE 3

HNF1A, GLY574SER
  
RCV000016077...

This mutation was found by Collet et al. (2002) to be prevalent in African individuals with diabetes (600496).

In an individual who had had a liver-tumor resection and had familial diabetes, Bluteau et al. (2002) found a gly574-to-ser (G574S) mutation in the TCF1 gene. The hepatocellular carcinoma in this case had developed in an adenoma (142330).


.0014 MATURITY-ONSET DIABETES OF THE YOUNG, TYPE 3

HNF1A, ARG583GLN
  
RCV000016078...

Bluteau et al. (2002) found a heterozygous germline mutation, arg583 to gln (R583Q), in an individual with a hyperplastic liver tumor (142330) who had had liver-tumor resection and had familial diabetes (600496). The substitution involved a highly conserved amino acid.


.0015 MATURITY-ONSET DIABETES OF THE YOUNG, TYPE 3

HNF1A, PRO112LEU
  
RCV000016080...

In a 3-generation Norwegian family with MODY3 (600496), Bjorkhaug et al. (2000) found a C-to-T transition at nucleotide 358 in exon 2 of the HNF1A gene, leading to a pro112-to-leu (P112L) amino acid substitution, in all 3 affected members. The phenotype in this family was mild with mild fasting and postprandial hyperglycemia easily controlled by diet only. Diabetes-associated late complications were not observed. P112L mutant protein demonstrated a significantly reduced ability to bind a high affinity HNF1 binding site and to activate transcription. Immunolocalization studies in HeLa cells showed that P112L mutant protein was correctly targeted to the nucleus. Bjorkhaug et al. (2000) concluded that the P112L mutation seems to impair pancreatic beta-cell function by loss-of-function mechanisms.

Xu et al. (2002) found the HNF1A P112L mutation in a southern Chinese MODY family.

Bjorkhaug et al. (2003) found evidence for possible founder effect of the P112L mutation in the Norwegian population.


.0016 MATURITY-ONSET DIABETES OF THE YOUNG, TYPE 3

HNF1A, ARG131TRP
  
RCV000016081...

Bjorkhaug et al. (2003) found a C-to-T transition in exon 2 of the HNF1A gene, resulting in an arg131-to-trp (R131W) amino acid substitution, in 5 Norwegian families with MODY3 (600496). This mutation had been reported in families from North America and Great Britain. Haplotype analysis indicated possible founder effect for the Norwegian families. Immunofluorescence studies demonstrated incorrect localization and accumulation of protein in both cytoplasm and nucleus. The R131W mutant protein displayed 10 to 15% of binding activity of wildtype and levels of transcription activation close to 50% of wildtype.


.0017 MATURITY-ONSET DIABETES OF THE YOUNG, TYPE 3

HNF1A, 4-BP DEL
  
RCV000016082

In a Norwegian family with MODY3 (600496), Bjorkhaug et al. (2003) detected a novel 4-bp deletion in exon 3 of the HNF1A gene (T196fsdelCCAA).


.0018 MATURITY-ONSET DIABETES OF THE YOUNG, TYPE 3

HNF1A, IVS3, G-A, -1
  
RCV000016079...

In a Norwegian proband with MODY3 (600496), Bjorkhaug et al. (2003) found a novel splice site mutation in intron 3 of the HNF1A gene, IVS3-1G-A.


.0019 MATURITY-ONSET DIABETES OF THE YOUNG, TYPE 3

HNF1A, ALA276ASP
  
RCV000016083...

In a Norwegian proband with MODY3 (600496), Bjorkhaug et al. (2003) detected a novel C-to-A transversion in exon 4 of the HNF1A gene resulting in an arg276-to-asp (A276D) amino acid substitution. Mutant protein was targeted to both nucleus and cytoplasm of transfected cells in immunofluorescence assays. DNA binding ability of 30 to 40% of wildtype could be detected; no significant reduction in transcription activation could be demonstrated.


.0020 MATURITY-ONSET DIABETES OF THE YOUNG, TYPE 3

HNF1A, 2-BP DEL, AG
  
RCV000016084

In 3 members of a Norwegian family with MODY3 (600496), Bjorkhaug et al. (2003) found a novel 2-bp deletion in exon 7 of the HNF1A gene (S445fsdelAG). Mutant protein was targeted to both nucleus and cytoplasm of transfected cells in immunofluorescence assays. DNA binding ability of 30 to 40% of wildtype could be detected; no significant reduction in transcription activation could be demonstrated.


.0021 MATURITY-ONSET DIABETES OF THE YOUNG, TYPE 3

HNF1A, SER531THR
  
RCV000016085...

In a Norwegian proband with MODY3 (600496), Bjorkhaug et al. (2003) detected a novel G-to-C transversion in exon 8 of the HNF1A gene, leading to a ser531-to-thr (S531T) amino acid substitution.


.0022 MATURITY-ONSET DIABETES OF THE YOUNG, TYPE 3

RENAL CELL CARCINOMA, CLEAR CELL, INCLUDED
RENAL CELL CARCINOMA, CHROMOPHOBE, INCLUDED
HNF1A, GLY92ASP
  
RCV000016086...

Maturity-Onset Diabetes of the Young, Type 3

In a French family with MODY3 (600496), Chevre et al. (1998) identified heterozygosity for a 92G-A transition in exon 1 of the HNF1A gene, resulting in a gly31-to-asp (G31D) substitution in the dimerization domain of the protein.

Renal Cell Carcinoma

In a 76-year-old woman with both clear cell and chromophobe renal carcinomas (see 144700), Rebouissou et al. (2005) identified heterozygosity for the G31D mutation. Mutation screening of the tumor samples detected only the germline G31D mutation. The renal carcinomas manifested in the same kidney with a single renal cyst in the other kidney. None of her relatives had a history of diabetes or renal carcinoma.


REFERENCES

  1. Babaya, N., Ikegami, H., Fujisawa, T., Nojima, K., Itoi-Babaya, M., Inoue, K., Nakura, J., Abe, M., Yamamoto, M., Jin, J. J., Wu, Z., Miki, T., Fukuda, M., Ogihara, T. Association of I27L polymorphism of hepatocyte nuclear factor-1-alpha gene with high-density lipoprotein cholesterol level. J. Clin. Endocr. Metab. 88: 2548-2551, 2003. [PubMed: 12788852, related citations] [Full Text]

  2. Bach, I., Galcheva-Gargova, Z., Mattei, M.-G., Simon-Chazottes, D., Guenet, J.-L., Cereghini, S., Yaniv, M. Cloning of human hepatic nuclear factor 1 (HNF1) and chromosomal localization of its gene in man and mouse. Genomics 8: 155-164, 1990. [PubMed: 1707031, related citations] [Full Text]

  3. Bjorkhaug, L., Sagen, J. V., Thorsby, P., Sovik, O., Molven, A., Njolstad, P. R. Hepatocyte nuclear factor-1a gene mutations and diabetes in Norway. J. Clin. Endocr. Metab. 88: 920-931, 2003. [PubMed: 12574234, related citations] [Full Text]

  4. Bjorkhaug, L., Ye, H., Horikawa, Y., Sovik, O., Molven, A., Njolstad, P. R. MODY associated with two novel hepatocyte nuclear factor-1-alpha loss-of-function mutations (P112L and Q466X). Biochem. Biophys. Res. Commun. 279: 792-798, 2000. [PubMed: 11162430, related citations] [Full Text]

  5. Bluteau, O., Jeannot, E., Bioulac-Sage, P., Marques, J. M., Blanc, J.-F., Bui, H., Beaudoin, J.-C., Franco, D., Balabaud, C., Laurent-Puig, P., Zucman-Rossi, J. Bi-allelic inactivation of TCF1 in hepatic adenomas. Nature Genet. 32: 312-315, 2002. [PubMed: 12355088, related citations] [Full Text]

  6. 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., Bell, G. I., Polonsky, K. S. Altered insulin secretory responses to glucose in diabetic and nondiabetic subjects with mutations in the diabetes susceptibility gene MODY3 on chromosome 12. Diabetes 45: 1503-1510, 1996. [PubMed: 8866553, related citations] [Full Text]

  7. Chevre, J. C., Hani, E. H., Boutin, P., Vaxillaire, M., Blanche, H., Vionnet, N., Pardini, V. C., Timsit, J., Larger, E., Charpentier, G., Beckers, D., Maes, M., Bellanne-Chantelot, C., Velho, C., Froguel, P. Mutation screening in 18 Caucasian families suggest the existence of other MODY genes. Diabetologia 41: 1017-1023, 1998. [PubMed: 9754819, related citations] [Full Text]

  8. Chi, Y.-I., Frantz, J. D., Oh, B.-C., Hansen, L., Dhe-Paganon, S., Shoelson, S. E. Diabetes mutations delineate an atypical POU domain in HNF-1-alpha. Molec. Cell 10: 1129-1137, 2002. [PubMed: 12453420, related citations] [Full Text]

  9. Chiu, K. C., Chuang, L.-M., Ryu, J. M., Tsai, G. P., Saad, M. F. The I27L amino acid polymorphism of hepatic nuclear factor-1-alpha is associated with insulin resistance. J. Clin. Endocr. Metab. 85: 2178-2183, 2000. [PubMed: 10852449, related citations] [Full Text]

  10. Collet, C., Ducorps, M, Mayaudon, H., Dupuy, O., Ceppa, F., Boutin, P., Froguel, P., Bauduceau, B. Prevalence of the missense mutation Gly574Ser in the hepatocyte nuclear factor-1-alpha in Africans with diabetes. Diabetes Metab. 28: 39-44, 2002. [PubMed: 11938027, related citations]

  11. Courtois, G., Morgan, J. G., Campbell, L. A., Fourel, G., Crabtree, G. R. Interaction of a liver-specific nuclear factor with the fibrinogen and alpha-1-antitrypsin promoters. Science 238: 688-692, 1987. [PubMed: 3499668, related citations] [Full Text]

  12. De Simone, V., De Magistris, L., Lazzaro, D., Gerstner, J., Monaci, P., Nicosia, A., Cortese, R. LFB3, a heterodimer-forming homeoprotein of the LFB1 family, is expressed in specialized epithelia. EMBO J. 10: 1435-1443, 1991. [PubMed: 1673925, related citations] [Full Text]

  13. Ellard, S. Hepatocyte nuclear factor 1 alpha (HNF-1-alpha) mutations in maturity-onset diabetes of the young. Hum. Mutat. 16: 377-385, 2000. [PubMed: 11058894, related citations] [Full Text]

  14. Fajans, S. S., Bell, G. I., Polonsky, K. S. Molecular mechanisms and clinical pathophysiology of maturity-onset diabetes of the young. New Eng. J. Med. 345: 971-980, 2001. [PubMed: 11575290, related citations] [Full Text]

  15. Foster, J. H., Donohue, T. A., Berman, M. M. Familial liver-cell adenomas and diabetes mellitus. New Eng. J. Med. 299: 239-241, 1978. [PubMed: 207987, related citations] [Full Text]

  16. Frayling, T. M., Bulman, M. P., Appleton, M., Hattersley, A. T., Ellard, S. A rapid screening method for hepatocyte nuclear factor 1 alpha frameshift mutations; prevalence in maturity-onset diabetes of the young and late-onset non-insulin dependent diabetes. Hum. Genet. 101: 351-354, 1997. [PubMed: 9439666, related citations] [Full Text]

  17. Frayling, T. M., Bulman, M. P., Ellard, S., Appleton, M., Dronsfield, M. J., Mackie, A. D. R., Baird, J. D., Kaisaki, P. J., Yamagata, K., Bell, G. I., Bain, S. C., Hattersley, A. T. Mutations in the hepatocyte nuclear factor-1-alpha gene are a common cause of maturity-onset diabetes of the young in the U.K. Diabetes 46: 720-725, 1997. [PubMed: 9075818, related citations] [Full Text]

  18. Godart, F., Bellanne-Chantelot, C., Clauin, S., Gragnoli, C., Abderrahmani, A., Blanche, H., Boutin, P., Chevre, J. C., Froguel, P., Bailleul, B. Identification of seven novel nucleotide variants in the hepatocyte nuclear factor-1-alpha (TCF1) promoter region in MODY patients. Hum. Mutat. 15: 173-180, 2000. [PubMed: 10649494, related citations] [Full Text]

  19. Gonzalez, F. J., Liu, S.-Y., Kozak, C. A., Nebert, D. W. Decreased Hnf-1 gene expression in mice homozygous for a 1.2-centimorgan deletion on chromosome 7. DNA Cell Biol. 9: 771-776, 1990. [PubMed: 2264930, related citations] [Full Text]

  20. Gragnoli, C., Lindner, T., Cockburn, B. N., Kaisaki, P. J., Gragnoli, F., Marozzi, G., Bell, G. I. Maturity-onset diabetes of the young due to a mutation in the hepatocyte nuclear factor-4-alpha binding site in the promoter of the hepatocyte nuclear factor-1-alpha gene. Diabetes 46: 1648-1651, 1997. [PubMed: 9313764, related citations] [Full Text]

  21. Hansen, T., Eiberg, H., Rouard, M., Vaxillaire, M., Moller, A. M., Rasmussen, S. K., Fridberg, M., Urhammer, S. A., Holst, J. J., Almind, K., Echwald, S. M., Hansen, L., Bell, G. I., Pedersen, O. Novel MODY3 mutations in the hepatocyte nuclear factor-1-alpha gene: evidence for a hyperexcitability of pancreatic beta-cells to intravenous secretagogues in a glucose-tolerant carrier of a P447L mutation. Diabetes 46: 726-730, 1997. [PubMed: 9075819, related citations] [Full Text]

  22. Hegele, R. A., Cao, H., Harris, S. B., Hanley, A. J. G., Zinman, B. The hepatic nuclear factor-1-alpha G319S variant is associated with early-onset type 2 diabetes in Canadian Oji-Cree. J. Clin. Endocr. Metab. 84: 1077-1082, 1999. [PubMed: 10084598, related citations] [Full Text]

  23. Hegele, R. A., Cao, H., Harris, S. B., Zinman, B., Hanley, A. J. G., Anderson, C. M. Peroxisome proliferator-activated receptor-gamma-2 P12A and type 2 diabetes in Canadian Oji-Cree. J. Clin. Endocr. Metab. 85: 2014-2019, 2000. [PubMed: 10843190, related citations] [Full Text]

  24. Hiraiwa, H., Pan, C.-J., Lin, B., Akiyama, T. E., Gonzalez, F. J., Chou, J. Y. A molecular link between the common phenotypes of type 1 glycogen storage disease and HNF1-alpha-null mice. J. Biol. Chem. 276: 7963-7967, 2001. [PubMed: 11121425, related citations] [Full Text]

  25. Hua, Q.-X., Zhao, M., Narayana, N., Nakagawa, S. H., Jia, W., Weiss, M. A. Diabetes-associated mutations in a beta-cell transcription factor destabilize an antiparallel 'mini-zipper' in a dimerization interface. Proc. Nat. Acad. Sci. 97: 1999-2004, 2000. Note: Erratum: Proc. Nat. Acad. Sci. 98: 13472 only, 2001. [PubMed: 10696112, images, related citations] [Full Text]

  26. Huang, P., He, Z., Ji, S., Sun, H., Xiang, D., Liu, C., Hu, Y., Wang, X., Hui, L. Induction of functional hepatocyte-like cells from mouse fibroblasts by defined factors. Nature 475: 386-389, 2011. [PubMed: 21562492, related citations] [Full Text]

  27. Johansen, A., Ek, J., Mortensen, H. B., Pedersen, O., Hansen, T. Half of clinically defined maturity-onset diabetes of the young patients in Denmark do not have mutations in HNF4A, GCK, and TCF1. J. Clin. Endocr. Metab. 90: 4607-4614, 2005. [PubMed: 15928245, related citations] [Full Text]

  28. Kaisaki, P. J., Menzel, S., Lindner, T., Oda, N., Rjasanowski, I., Sahm, J., Meincke, G., Schulze, J., Schmechel, H., Petzold, C., Ledermann, H. M., Sachse, G., Boriraj, V. V., Menzel, R., Kerner, W., Turner, R. C., Yamagata, K., Bell, G. I. Mutations in the hepatocyte nuclear factor-1-alpha gene in MODY and early-onset NIDDM: evidence for a mutational hotspot in exon 4. Diabetes 46: 528-535, 1997. Note: Erratum: Diabetes 46: 1239 only, 1997. [PubMed: 9032114, related citations] [Full Text]

  29. Kuo, C. J., Conley, P. B., Hsieh, C.-L., Francke, U., Crabtree, G. R. Molecular cloning, functional expression, and chromosomal localization of mouse hepatocyte nuclear factor 1. Proc. Nat. Acad. Sci. 87: 9838-9842, 1990. [PubMed: 2263635, related citations] [Full Text]

  30. Mendel, D. B., Hansen, L. P., Graves, M. K., Conley, P. B., Crabtree, G. R. HNF-1-alpha and HNF-1-beta (vHNF-1) share dimerization and homeo domains, but not activation domains, and form heterodimers in vitro. Genes Dev. 5: 1042-1056, 1991. [PubMed: 2044952, related citations] [Full Text]

  31. Mendel, D. B., Khavari, P. A., Conley, P. B., Graves, M. K., Hansen, L. P., Admon, A., Crabtree, G. R. Characterization of a cofactor that regulates dimerization of a mammalian homeodomain protein. Science 254: 1762-1767, 1991. [PubMed: 1763325, related citations] [Full Text]

  32. Miedzybrodzka, Z., Hattersley, A. T., Ellard, S., Pearson, D., de Silva, D., Harvey, R., Haites, N. Non-penetrance in a MODY 3 family with a mutation in the hepatic nuclear factor 1a gene: implications for predictive testing. Europ. J. Hum. Genet. 7: 729-732, 1999. [PubMed: 10482964, related citations] [Full Text]

  33. Mirshahi, U. L., Colclough, K., Wright, C. F., Wood, A. R., Beaumont, R. N., Tyrrell, J., Laver, T. W., Stahl, R., Golden, A., Goehringer, J. M, Geisinger-Regeneron DiscovEHR Collaboration, Frayling, T. F., Hattersley, A. T., Carey, D. J., Weedon, M. N., Patel, K. A. Reduced penetrance of MODY-associated HNF1A/HNF4A variants but not GCK variants in clinically unselected cohorts. Am. J. Hum. Genet. 109: 2018-2028, 2022. [PubMed: 36257325, images, related citations] [Full Text]

  34. Odom, D. T., Dowell, R. D., Jacobsen, E. S., Gordon, W., Danford, T. W., MacIsaac, K. D., Rolfe, P. A., Conboy, C. M., Gifford, D. K., Fraenkel, E. Tissue-specific transcriptional regulation has diverged significantly between human and mouse. Nature Genet. 39: 730-732, 2007. [PubMed: 17529977, images, related citations] [Full Text]

  35. 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., Fraenkel, E., Bell, G. I., Young, R. A. Control of pancreas and liver gene expression by HNF transcription factors. Science 303: 1378-1381, 2004. [PubMed: 14988562, images, related citations] [Full Text]

  36. 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 84: 575-585, 1996. [PubMed: 8598044, related citations] [Full Text]

  37. Rebouissou, S., Vasiliu, V., Thomas, C., Bellanne-Chantelot, C., Bui, H., Chretien, Y., Timsit, J., Rosty, C., Laurent-Puig, P., Chauveau, D., Zucman-Rossi, J. Germline hepatocyte nuclear factor 1-alpha and 1-beta mutations in renal cell carcinomas. Hum. Molec. Genet. 14: 603-614, 2005. [PubMed: 15649945, related citations] [Full Text]

  38. Reiner, A. P., Barber, M. J., Guan, Y., Ridker, P. M., Lange, L. A., Chasman, D. I., Walston, J. D., Cooper, G. M., Jenny, N. S., Rieder, M. J., Durda, J. P., Smith, J. D., Novembre, J., Tracy, R. P., Rotter, J. I., Stephens, M., Nickerson, D. A., Krauss, R. M. Polymorphisms of the HNF1A gene encoding hepatocyte nuclear factor-1-alpha are associated with C-reactive protein. Am. J. Hum. Genet. 82: 1193-1201, 2008. [PubMed: 18439552, images, related citations] [Full Text]

  39. Rey-Campos, J., Chouard, T., Yaniv, M., Cereghini, S. vHNF1 is a homeoprotein that activates transcription and forms heterodimers with HNF1. EMBO J. 10: 1445-1457, 1991. [PubMed: 1673926, related citations] [Full Text]

  40. Ridker, P. M., Pare, G., Parker, A., Zee, R. Y. L., Danik, J. S., Buring, J. E., Kwiatkowski, D., Cook, N. R., Miletich, J. P., Chasman, D. I. Loci related to metabolic syndrome pathways including LEPR, HNF1A, IL6R, and GCKR associate with plasma C-reactive protein: the Women's Genome Health Study. Am. J. Hum. Genet. 82: 1185-1192, 2008. [PubMed: 18439548, images, related citations] [Full Text]

  41. Shih, D. Q., Bussen, M., Sehayek, E., Ananthanarayanan, M., Shneider, B. L., Suchy, F. J., Shefer, S., Bollileni, J. S., Gonzalez, F. J., Breslow, J. L., Stoffel, M. Hepatocyte nuclear factor-1-alpha is an essential regulator of bile acid and plasma cholesterol metabolism. Nature Genet. 27: 375-382, 2001. [PubMed: 11279518, related citations] [Full Text]

  42. Szpirer, C., Riviere, M., Cortese, R., Nakamura, T., Islam, M. Q., Levan, G., Szpirer, J. Chromosomal localization in man and rat of the genes encoding the liver-enriched transcription factors C/EBP, DBP, and HNF1/LFB-1 (CEBP, DBP, and transcription factor 1, TCF1, respectively) and of the hepatocyte growth factor/scatter factor gene (HGF). Genomics 13: 293-300, 1992. [PubMed: 1535333, related citations] [Full Text]

  43. Triggs-Raine, B. L., Kirkpatrick, R. D., Kelly, S. L., Norquay, L. D., Cattini, P. A., Yamagata, K., Hanley, A. J. G., Zinman, B., Harris, S. B., Barrett, P. H., Hegele, R. A. HNF1-alpha G319S, a transactivation-deficient mutant, is associated with altered dynamics of diabetes onset in an Oji-Cree community. Proc. Nat. Acad. Sci. 99: 4614-4619, 2002. [PubMed: 11904371, images, related citations] [Full Text]

  44. Urhammer, S. A., Hansen, T., Ekstrom, C. T., Eiberg, H., Pederson, O. The Ala/Val98 polymorphism of the hepatocyte nuclear factor-1-alpha gene contributes to the interindividual variation in serum C-peptide response during an oral glucose tolerance test: evidence from studies of 231 glucose-tolerant first degree relatives of type 2 diabetic probands. J. Clin. Endocr. Metab. 83: 4506-4509, 1998. [PubMed: 9851800, related citations] [Full Text]

  45. Urhammer, S. A., Moller, A. M., Nyholm, B., Ekstrom, C. T., Eiberg, H., Clausen, J. O., Hansen, T., Pedersen, O., Schmitz, O. The effect of two frequent amino acid variants of the hepatocyte nuclear factor-1-alpha gene on estimates of the pancreatic beta-cell function in Caucasian glucose-tolerant first-degree relatives of type 2 diabetic patients. J. Clin. Endocr. Metab. 83: 3992-3995, 1998. [PubMed: 9814481, related citations] [Full Text]

  46. Urhammer, S. A., Rasmussen, S. K., Kaisaki, P. J., Oda, N., Yamagata, K., Moller, A. M., Fridberg, M., Hansen, L., Hansen, T., Bell, G. I., Pedersen, O. Genetic variation in the hepatocyte nuclear factor-1-alpha gene in Danish Caucasians with late-onset NIDDM. Diabetologia 40: 473-475, 1997. [PubMed: 9112026, related citations] [Full Text]

  47. van Wering, H. M., Huibregtse, I. L., van der Zwan, S. M., de Bie, M. S., Dowling, L. N., Boudreau, F., Rings, E. H. H. M., Grand, R. J., Krasinski, S. D. Physical interaction between GATA-5 and hepatocyte nuclear factor-1-alpha results in synergistic activation of the human lactase-phlorizin hydrolase promoter. J. Biol. Chem. 277: 27659-27667, 2002. [PubMed: 12011060, related citations] [Full Text]

  48. Vaxillaire, M., Rouard, M., Yamagata, K., Oda, N., Kaisaki, P. J., Boriraj, V. V., Chevre, J.-C., Boccio, V., Cox, R. D., Lathrop, G. M., Dussoix, P., Philippe, J., Timsit, J., Charpentier, G., Velho, G., Bell, G. I., Froguel, P. Identification of nine novel mutations in the hepatocyte nuclear factor 1 alpha gene associated with maturity-onset diabetes of the young (MODY3). Hum. Molec. Genet. 6: 583-586, 1997. [PubMed: 9097962, related citations] [Full Text]

  49. Xu, J. Y., Chan, V., Zhang, W. Y., Wat, N. M. S., Lam, K. S. L. Mutations in the hepatocyte nuclear factor-1-alpha gene in Chinese MODY families: prevalence and functional analysis. Diabetologia 45: 744-746, 2002. [PubMed: 12107757, related citations] [Full Text]

  50. Yamada, S., Nishigori, H., Onda, H., Utsugi, T., Yanagawa, T., Maruyama, T., Onigata, K., Nagashima, K., Nagai, R., Morikawa, A., Takeuchi, T., Takeda, J. Identification of mutations in the hepatocyte nuclear factor (HNF)-1-alpha gene in Japanese subjects with IDDM. Diabetes 46: 1643-1647, 1997. [PubMed: 9313763, related citations] [Full Text]

  51. Yamagata, K., Oda, N., Kalsaki, P. J., Menzel, S., Furuta, H., Vaxillaire, M., Southam, L., Cox, R. D., Lathrop, G. M., Borhaj, V. V., Chen, X., Cox, N. J., Oda, Y., Yano, H., Le Beau, M. M., Yamada, S., Nishigori, H., Takeda, J., Fajans, S. S., Hattersley, A. T., Iwasaki, N., Hansen, T., Pedersen, O., Polonsky, K. S., Turner, R. C., Velho, G., Chevre, J.-C., Froguel, P., Bell, G. I. Mutations in the hepatocyte nuclear factor-1-alpha gene in maturity-onset diabetes of the young (MODY3). Nature 384: 455-457, 1996. [PubMed: 8945470, related citations] [Full Text]

  52. Yoshiuchi, I., Yamagata, K., Yoshimoto, M., Zhu, Q., Yang, Q., Nammo, T., Uenaka, R., Kinoshita, E., Hanafusa, T., Miyagawa, J., Matsuzawa, Y. Analysis of a non-functional HNF-1-alpha (TCF1) mutation in Japanese subjects with familial type 1 diabetes. Hum. Mutat. 18: 345-351, 2001. [PubMed: 11668618, related citations] [Full Text]


Ada Hamosh - updated : 01/17/2023
Ada Hamosh - updated : 8/4/2011
Marla J. F. O'Neill - updated : 4/19/2010
Marla J. F. O'Neill - updated : 6/10/2008
George E. Tiller - updated : 2/5/2008
Patricia A. Hartz - updated : 10/3/2007
Patricia A. Hartz - updated : 8/3/2007
John A. Phillips, III - updated : 10/19/2006
Ada Hamosh - updated : 6/10/2004
John A. Phillips, III - updated : 2/10/2004
John A. Phillips, III - updated : 8/21/2003
Stylianos E. Antonarakis - updated : 5/5/2003
Victor A. McKusick - updated : 9/25/2002
Victor A. McKusick - updated : 9/25/2002
Victor A. McKusick - updated : 2/15/2002
Ada Hamosh - updated : 10/18/2001
John A. Phillips, III - updated : 9/25/2001
Ada Hamosh - updated : 4/23/2001
Ada Hamosh - updated : 3/28/2001
John A. Phillips, III - updated : 2/12/2001
Victor A. McKusick - updated : 11/29/2000
John A. Phillips, III - updated : 8/9/2000
Victor A. McKusick - updated : 2/22/2000
Victor A. McKusick - updated : 11/8/1999
John A. Phillips, III - updated : 9/29/1999
John A. Phillips, III - updated : 3/24/1999
Victor A. McKusick - updated : 2/13/1998
Victor A. McKusick - updated : 2/11/1998
Victor A. McKusick - updated : 8/12/1997
Victor A. McKusick - updated : 8/7/1997
Victor A. McKusick - updated : 4/24/1997
Creation Date:
Victor A. McKusick : 11/13/1987
alopez : 01/17/2023
carol : 09/16/2021
carol : 09/04/2020
carol : 09/03/2020
carol : 09/03/2020
alopez : 12/11/2017
carol : 10/03/2017
carol : 08/04/2016
carol : 05/17/2016
terry : 11/9/2012
alopez : 8/3/2012
carol : 3/29/2012
carol : 3/29/2012
mgross : 10/4/2011
terry : 10/4/2011
alopez : 8/15/2011
terry : 8/4/2011
alopez : 4/21/2010
terry : 4/19/2010
alopez : 4/16/2010
alopez : 4/16/2010
carol : 4/14/2009
terry : 2/19/2009
mgross : 10/21/2008
carol : 6/11/2008
terry : 6/10/2008
wwang : 2/7/2008
terry : 2/5/2008
wwang : 10/25/2007
mgross : 10/3/2007
mgross : 10/3/2007
alopez : 8/3/2007
alopez : 10/19/2006
carol : 11/18/2005
alopez : 3/30/2005
alopez : 6/15/2004
alopez : 6/15/2004
terry : 6/10/2004
alopez : 2/10/2004
alopez : 8/21/2003
mgross : 5/5/2003
mgross : 5/5/2003
tkritzer : 11/19/2002
alopez : 9/25/2002
alopez : 9/25/2002
tkritzer : 9/23/2002
ckniffin : 5/29/2002
carol : 4/30/2002
cwells : 2/25/2002
cwells : 2/20/2002
terry : 2/15/2002
carol : 1/4/2002
carol : 10/18/2001
cwells : 9/28/2001
cwells : 9/25/2001
alopez : 4/30/2001
terry : 4/23/2001
alopez : 3/29/2001
terry : 3/28/2001
terry : 2/12/2001
carol : 12/18/2000
mcapotos : 12/14/2000
terry : 11/29/2000
mgross : 8/9/2000
mcapotos : 4/12/2000
alopez : 3/23/2000
mcapotos : 3/22/2000
mcapotos : 3/14/2000
terry : 2/22/2000
alopez : 11/12/1999
psherman : 11/9/1999
terry : 11/8/1999
alopez : 11/5/1999
alopez : 9/29/1999
mgross : 4/7/1999
mgross : 3/24/1999
dkim : 12/9/1998
dkim : 7/21/1998
alopez : 5/14/1998
mark : 2/22/1998
terry : 2/13/1998
alopez : 2/11/1998
dholmes : 2/4/1998
jenny : 8/20/1997
terry : 8/12/1997
terry : 8/11/1997
terry : 8/7/1997
terry : 8/7/1997
alopez : 4/30/1997
alopez : 4/24/1997
terry : 4/24/1997
mark : 3/7/1997
mark : 12/11/1996
mark : 12/4/1996
terry : 12/3/1996
mark : 3/25/1996
terry : 3/18/1996
carol : 1/5/1995
mimadm : 4/14/1994
carol : 10/22/1992
carol : 6/26/1992
carol : 6/24/1992
supermim : 3/16/1992

* 142410

HNF1 HOMEOBOX A; HNF1A


Alternative titles; symbols

TRANSCRIPTION FACTOR 1; TCF1
HEPATOCYTE NUCLEAR FACTOR-1-ALPHA
HEPATOCYTE NUCLEAR FACTOR 1; HNF1
HEPATIC NUCLEAR FACTOR-1-ALPHA
ALBUMIN PROXIMAL FACTOR


HGNC Approved Gene Symbol: HNF1A

SNOMEDCT: 1187306007, 128667008, 254915003, 41607009, 609570008, 733471003;  


Cytogenetic location: 12q24.31     Genomic coordinates (GRCh38): 12:120,978,543-121,002,512 (from NCBI)


Gene-Phenotype Relationships

Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
12q24.31 {Diabetes mellitus, insulin-dependent} 222100 Autosomal recessive 3
{Diabetes mellitus, noninsulin-dependent, 2} 125853 Autosomal dominant 3
Diabetes mellitus, insulin-dependent, 20 612520 3
Hepatic adenoma, somatic 142330 3
MODY, type III 600496 Autosomal dominant 3
Renal cell carcinoma 144700 3

TEXT

Cloning and Expression

Using a rat Hnf1 cDNA-derived probe, Bach et al. (1990) isolated HNF1 clones from a human liver cDNA library. The deduced 631-amino acid human HNF1 protein contains a homeodomain in its N-terminal half and shares close similarity with the 628-amino acid rat protein.

The amino acid sequence of HNF1 displays distant sequence homology to the homeodomains of homeotic genes (see 142950) (Courtois et al., 1987).


Gene Function

The orderly and sequential activation of genes during development is thought to be related to the selective expression of groups of regulatory proteins acting primarily at the level of transcription. Courtois et al. (1987) found a nuclear protein in hepatocytes, but not in other cell types, that binds to a sequence required for hepatocyte-specific transcription of the genes for the alpha and beta chains of fibrinogen (134820, 134830) and alpha-1-antitrypsin (107400). This protein, called hepatocyte nuclear factor-1 (HNF1) by them, interacts with sequences required for optimal promoter function of the genes mentioned. The promoter or enhancer regions for several viral and cellular genes not expressed in the liver did not compete for binding to these sequences. HNF1 is predominantly expressed in liver and kidney. The restricted expression of HNF1 and its selective interaction with the control regions of several liver-specific genes suggested to Courtois et al. (1987) that it is involved in developmentally regulated gene expression in the liver. HNF1 binds to the promoters of a variety of genes that are expressed exclusively in the liver, e.g., fibrinogen-alpha and -beta, albumin (103600), alpha-fetoprotein (104150), alpha-1-antitrypsin, liver-type pyruvate kinase (609712), transthyretin (176300), aldolase B (612724), and hepatitis B virus large surface protein.

Dimerization among transcription factors is a frequent finding in the regulation of eukaryotic gene expression. HNF1-alpha functions as a dimer. Mendel et al. (1991) identified DCOH (126090), a dimerization cofactor of HNF1-alpha, which displayed a restricted tissue distribution and did not bind to DNA but, rather, selectively stabilized HNF1-alpha dimers. Hua et al. (2000) showed that the dimerization motif of HNF1-alpha forms an intermolecular 4-helix bundle. The bundle is destabilized by a subset of mutations associated with maturity-onset diabetes of the young (MODY; 606391). Impaired dimerization of the beta-cell transcription factor thus provides a molecular mechanism of metabolic deregulation in diabetes mellitus.

Van Wering et al. (2002) showed that mouse Gata5 (611496) and Hnf1-alpha interacted in vitro and in transfected COS-7 cells. The interaction required the C-terminal zinc finger and basic region of Gata5 and the homeodomain of Hnf1-alpha. Physical association of GATA5 and HNF1-alpha was required for synergistic activation of the human lactase-phlorizin hydrolase (LCT; 603202) promoter. Deletion of the HNF1-alpha activation domains or interruption of the HNF1-binding sites in the LCT promoter resulted in complete loss of transcriptional activity, whereas deletion of the GATA5 activation domains or interruption of the GATA-binding sites reduced, but did not eliminate, transcriptional activity.

To gain insight into the transcriptional regulatory networks that specify and maintain human tissue diversity, Odom et al. (2004) used chromatin immunoprecipitation combined with promoter microarrays to identify systematically the genes occupied by the transcriptional regulators HNF1-alpha, HNF4-alpha (600281), and HNF6 (604164), together with RNA polymerase II (see 180660), in human liver and pancreatic islets. Odom et al. (2004) identified tissue-specific regulatory circuits formed by HNF1-alpha, HNF4-alpha, and HNF6 with other transcription factors, revealing how these factors function as master regulators of hepatocyte and islet transcription. Odom et al. (2004) concluded that their results suggested how misregulation of HNF4-alpha can contribute to type 2 diabetes (125853). They found that HNF1-alpha is bound to at least 222 target genes in hepatocytes. HNF1-alpha occupied the promoter regions of 106 genes within pancreatic islets, 30% of which were also bound by HNF1-alpha in hepatocytes. In islets, fewer chaperones and enzymes were bound by HNF1-alpha than in hepatocytes, and the receptors and signal transduction machinery regulated by HNF1-alpha varied between the 2 tissues. Odom et al. (2004) found that HNF4-alpha bound to the promoters of about 12% of hepatocyte islet genes represented on the microarray. HNF4-alpha acted in a much larger number of hepatocyte and beta-cell genes than did HNF1-alpha, suggesting that HNF4-alpha has broad activities in these 2 tissues.

Odom et al. (2007) analyzed the binding of FOXA2 (600288), HNF1A, HNF4A, and HNF6 to 4,000 orthologous gene pairs in hepatocytes purified from human and mouse livers. Despite the conserved function of these factors, 41 to 89% of the binding events seemed to be species-specific. Importantly, the binding sites varied widely between species in ways that could not be predicted from human-mouse sequence alignments alone.


Mapping

Bach et al. (1990) assigned the human HNF1 gene to chromosome 12q24.3 by in situ hybridization, and the mouse gene to 5F by RFLP analysis of interspecific mouse backcrosses. One other gene, that for short chain acyl-CoA-dehydrogenase (606885), had also been assigned to chromosomes 12 and 5 in man and mouse, respectively. Kuo et al. (1990) also assigned the HNF1 gene to 12q22-qter in the human and to chromosome 5 in the mouse. By means of somatic cell hybrids segregating either human or rat chromosomes, Szpirer et al. (1992) independently assigned the TCF1 gene to human chromosome 12 and found that it was located also on rat chromosome 12, thereby defining a new segment of homology between the 2 species (and a segment of mouse chromosome 5).


Molecular Genetics

MODY, a single-gene disorder responsible for 2 to 5% of noninsulin-dependent (type II) diabetes mellitus (NIDDM; 125853), is characterized by autosomal dominant inheritance and an age of onset of 25 years or younger. Yamagata et al. (1996) narrowed the localization of a form of MODY (MODY3; 600496) to chromosome 12q24.2 by a combination of genetic linkage and fluorescence in situ hybridization. To identify the nature of the MODY3 gene, Yamagata et al. (1996) used a combination of approaches, including testing genes known to be on 12q to see if they mapped into the contig to which MODY3 mapped, exon trapping, and cDNA selection, for which human pancreatic islet cDNA was used (insulin secretion is abnormal in MODY3 patients, making islets a likely site of expression of MODY3 mRNA and protein). They identified 14 genes encoding known proteins, 12 known expressed sequence tags (ESTs), and 9 new ESTs. They found mutations in the gene encoding hepatocyte nuclear factor-1-alpha, a transcription factor that helps in the tissue-specific regulation of the expression of several liver genes and also functions as a weak transactivator of the rat insulin-I gene. In all, Yamagata et al. (1996) identified 6 different mutations which were associated with MODY3 (e.g., 142410.0001). In several pedigrees individuals were found who inherited the mutant allele and the at-risk chromosome 12 haplotype but were nondiabetic or only showed evidence of impaired glucose tolerance or diabetes during pregnancy. These individuals were expected eventually to develop diabetes mellitus. In one member of a family NIDDM was diagnosed at the age of 65 years, at which time he was mildly obese, suggesting that he had late-onset NIDDM rather than MODY.

Vaxillaire et al. (1997) examined 10 unrelated Caucasian families in whom MODY/NIDDM cosegregated with markers for MODY3 and found 10 different mutations in the TCF1 gene, all of which cosegregated with diabetes (see 142410.0003 and 142410.0004). In these families, they found no obvious relationships between the nature of the mutations observed (i.e., frameshift, nonsense, or missense), or their location in the gene, with clinical features of diabetes (e.g., age at onset, severity). The authors stated that the mechanisms by which mutations in the TCF1 gene caused diabetes mellitus were unclear, but might include abnormal pancreatic islet development during fetal life, as well as impaired transcriptional regulation of genes that play a key role in normal pancreatic beta-cell function.

Urhammer et al. (1997) found a variety of variations in the TCF1 gene in 245 Danish NIDDM patients and 242 age-matched controls. The frequencies of the variants were similar in the 2 groups except that an arg583-to-gln mutation was found in 2 of the 245 NIDDM patients and in none of the control subjects. The authors concluded that genetic variation in the TCF1 gene is not a common factor contributing to NIDDM susceptibility in white subjects of Danish ancestry. Urhammer et al. (1998) studied the frequent amino acid polymorphisms ile27 to leu and ser487 to asn of the TCF1 gene to determine whether they were associated with alterations in glucose-induced serum C-peptide and serum insulin responses among Caucasian glucose-tolerant first-degree relatives of NIDDM patients. The authors concluded that these polymorphisms had no major impact on the pancreatic beta-cell function, as estimated during an oral and intravenous glucose challenge.

Urhammer et al. (1998) studied the TCF1 ala98-to-val polymorphism in glucose-tolerant first-degree relatives of type 2 diabetic patients of the same ethnic origin. All participants, 231 glucose-tolerant offspring of 62 type 2 diabetic probands, underwent an oral glucose tolerance test (OGTT) with measurements of plasma glucose, serum insulin, and serum C-peptide during the test. Thirty-three heterozygous carriers of the ala98-to-val variant were identified, whereas no subjects had the variant in its homozygous form. Carriers of ala98 to val had a 20% reduction in serum C-peptide at 30 minutes during the OGTT compared to wildtype carriers. No significant differences in serum insulin levels during the OGTT were observed between carriers of the variant and ala98 homozygotes.

In a study of 15 UK MODY families for mutations in the TCF1 gene, Frayling et al. (1997) found 8 different mutations in 11 families (73%). A previously reported mutation, the insertion of a C in the C tract encoding the sequence 289-pro-pro-pro-291 (142410.0001), was present in 4 of the families. A screen of a further 32 probands with early-onset (less than 40 years of age) NIDDM showed the mutation in 2 additional families. This common mutation was present on at least 3 different haplotypes, suggesting that its high frequency is due to recurrent mutation rather than founder effect. Thus, Frayling et al. (1997) concluded that TSF1 mutations are a common cause of MODY in UK families and result in early-onset NIDDM with a progressive clinical course. Hansen et al. (1997) sequenced the coding region and intron-exon boundaries of the TCF1 gene in 9 unrelated Danish Caucasian subjects with MODY and found mutations in 5. These 5 mutations were found in neither 84 NIDDM patients nor in 84 control subjects.

MODY3 is characterized by a severe insulin secretory defect, compared with MODY2 (125851), a glucokinase-deficient diabetes. Because of the rapid progress to overt diabetes and the high prevalence of the requirement for insulin treatment in patients with MODY3, Yamada et al. (1997) screened the HNF1A gene for mutations in Japanese subjects with insulin-dependent diabetes mellitus (IDDM; 222100). Mutations were identified in 3 (5.5%) of the 55 unrelated subjects with IDDM (e.g., 142410.0001, 142410.0005, and 142410.0006). None of these mutations was found in 200 normal chromosomes from nondiabetic subjects. The results indicated that mutation in the HNF1A gene can lead to development not only of early-onset NIDDM but also of IDDM. In a subclassification of IDDM, the HNF1A-deficient type should be distinguished from the classic type of autoimmune-based IDDM in Japanese. All of these mutations were heterozygous.

Ellard (2000) stated that 65 different mutations in the TCF1 gene had been found to cause MODY3 in a total of 116 families worldwide. They noted that diagnostic and predictive genetic testing is possible for the majority of patients with MODY, opening new avenues for the classification, prediction, and perhaps eventually the prevention of diabetes in these families.

Fajans et al. (2001) reported that mutations in the HNF1A gene have been identified in all racial and ethnic backgrounds, including European, Chinese, Japanese, African, and American Indian. Mutations in the HNF1A gene appear to be the most common cause of MODY among adults seen in diabetic clinics.

Bluteau et al. (2002) found germline TCF1 mutations in 2 individuals who had previously had a liver-tumor resection and had familial diabetes. One of these individuals had a hepatocellular carcinoma that had developed in an adenoma and had a gly574-to-ser mutation (142410.0013). This mutation was described by Collet et al. (2002) as frequent in Africans with diabetes. These results suggested that germline mutations of TCF1 may predispose to benign liver tumor development, and may explain the previously described cosegregation of liver adenoma with diabetes mellitus in a large family (Foster et al., 1978). Bluteau et al. (2002) suggested the results of their studies indicated that individuals with MODY could benefit from liver monitoring to detect early tumor occurrence, and individuals with liver adenomas, especially those with a family history of the same condition, should be tested for diabetes.

To elucidate the function of a molecular hotspot, Chi et al. (2002) cocrystallized human HNF1A amino acids 83 to 279 with a high-affinity promoter and solved the structure of the complex. Two identical protein molecules were bound to the promoter. Each contained a homeodomain (POU-H) and a second domain structurally similar to POU-specific (POU-S) domains that was not predicted on the basis of amino acid sequence. Atypical elements in both domains created a stable interface that further distinguished HNF1A from other flexible POU-homeodomain proteins. Chi et al. (2002) determined that 76% of MODY3-associated missense mutations in HNF1A occur in the region encompassing amino acids 98 to 272, which includes the POU-H and POU-S domains and a nuclear localization signal. They subdivided these mutations according to functional classes predicted to affect DNA binding, POU-S/POU-H domain interactions, protein stability, and nuclear localization. The largest class affected DNA binding, either through direct interactions or indirectly by perturbing local environment.

To estimate the prevalence of MODY3 in Norwegian diabetic pedigrees, Bjorkhaug et al. (2003) screened a total of 130 families for HNF1A mutations; 42 families with clinical MODY, 75 with suspected MODY, and 13 pedigrees with multiplex type 1 diabetes (IDDM). Twenty-two families with clinical MODY, 15 families with suspected MODY, and 1 family with type 1 diabetes multiplex harbored HNF1A mutations. Thus, in about half of Norwegian families with clinical MODY, mutations in the HNF1A gene could be detected. Eight of the 18 different mutations identified were novel. Haplotypes were determined for recurrent mutations, indicating a founder effect in Norway for the hotspot mutation P291fsinsC (142410.0001) and possibly also for P112L (142410.0015) and R131W (142410.0016). Two mutant HNF1A proteins were unable to bind DNA and at least 5 mutants showed defective nuclear translocation. Transcriptional activation was reduced for most of the MODY3-associated mutants. Accordingly, the functional studies of HNF1A mutants indicated that beta-cell dysfunction in MODY3 is caused by loss-of-function mechanisms like reduced DNA binding, impaired transcriptional activation, and defects in subcellular localization.

Johansen et al. (2005) examined the prevalence and nature of mutations in the 3 common MODY genes HNF4A (600281), GCK (138079), and TCF1 in Danish patients with a clinical diagnosis of MODY and determined metabolic differences in probands with and without mutations in HNF4A, GCK, and TCF1. They identified 29 different mutations in 38 MODY families. Fifteen of the mutations were novel. The variants segregated with diabetes within the families, and none of the variants were found in 100 normal Danish chromosomes. Their findings suggested a relative prevalence of 3% of MODY1 (125850) (2 different mutations in 2 families), 10% of MODY2 (7 in 8), and 36% of MODY3 (21 in 28) among Danish kindred clinically diagnosed as MODY. No significant differences in biochemical and anthropometric measurements were observed at baseline examinations. Forty-nine percent of the families carried mutations in the 3 examined MODY genes.

Rebouissou et al. (2005) screened 35 renal neoplasms for HNF1A and HNF1B (189907) inactivation. In 2 of 13 clear cell renal carcinomas, the authors found a monoallelic germline mutation (142410.0001 and 142410.0022) of HNF1A with no associated suppression of target mRNA expression. In normal and tumor renal tissues, there was a network of transcription factors differentially regulated in tumor subtypes. There was a related cluster of coregulated genes associating HNF1A, HNF4A, FABP1 (134650), and UGT2B7 (600068). Rebouissou et al. (2005) suggested that germline mutation of HNF1A may predispose to renal tumors.

Ridker et al. (2008) performed a multistage genomewide association study of CRP (123260) levels and found significant association with 7 loci, 1 of which was HNF1A. Reiner et al. (2008) reported an association between common variants of the HNF1A gene and plasma CRP concentrations in 2 independent populations of older adults.

Penetrance of HNF1A Mutations in Diabetes

Mirshahi et al. (2022) comprehensively assessed the penetrance and prevalence of pathogenic variants in HNF1A, HNF4A, and GCK that account for more than 80% of monogenic diabetes. Mirshahi et al. (2022) analyzed clinical and genetic data from 1,742 clinically referred probands, 2,194 family members, clinically unselected individuals from a US health system-based cohort of 132,194 individuals, and a UK population-based cohort of 198,748 individuals, and found that 1 in 1,500 individuals harbor a pathogenic variant in one of these genes. The penetrance of pathogenic GCK variants was similar (89 to 97%) across all cohorts. The penetrance of diabetes for HNF1A and HNF4A pathogenic variants was substantially lower in the clinically unselected individuals compared to clinically referred probands and was dependent on the setting (32% in the population, 49% in the health system cohort, 86% in a family member, and 98% in probands for HNF1A). The relative risk of diabetes was similar across the clinically unselected cohorts, highlighting the role of environment/ other genetic factors. The authors suggested that for HNF1A and HNF4A, genetic interpretation and counseling should be tailored to the setting in which a pathogenic monogenic variant was identified. GCK is an exception with near-complete penetrance in all settings.


Cytogenetics

Liver adenomas are benign tumors at risk of malignant transformation. In a genomewide search for loss of heterozygosity (LOH) associated with liver adenomas, Bluteau et al. (2002) found a deletion in 12q in 5 of 10 adenomas. In most cases, LOH at 12q was the only recurrent genetic alteration observed, suggesting the presence of a tumor-suppressor gene in that region. A minimal common region of deletion was defined in 12q24 that included the TCF1 gene. Bluteau et al. (2002) found biallelic inactivation of TCF1 in 10 of 16 screened adenomas, and heterozygous germline mutations were present in 3 affected individuals. Furthermore, 2 well-differentiated hepatocellular carcinomas occurring in normal liver, out of 30 HCCs screened, contained somatic biallelic mutations. These results indicated that inactivation of TCF1, whether sporadic or associated with MODY3, is an important genetic event in the occurrence of human liver adenomas, and may be an early step in the development of some hepatocellular carcinomas.


Animal Model

Gonzalez et al. (1990) found that newborn mice homozygous for a 1.2-cM deletion of chromosome 7 do not show the increased activity of CYP2E (124040), which is regulated by the transcription factor Hnf1. They suggested that the deleted region of chromosome 7 contains a gene encoding a transacting factor that is epistatic in a regulatory cascade that includes Hnf1 gene expression.

Pontoglio et al. (1996) found that mice with inactivation of the Hnf1 gene through homologous recombination failed to thrive and died around weaning after a progressive wasting syndrome with marked liver enlargement. The transcription rate of genes such as albumin and alpha-1-antitrypsin was reduced, while the gene coding the phenylalanine hydroxylase (612349) was totally silent, giving rise to phenylketonuria. Mutant mice also suffered from severe Fanconi syndrome (see 227650) caused by renal proximal tubular dysfunction. The resulting massive urinary glucose lost led to energy and water wasting. Pontoglio et al. (1996) commented that Hnf1-deficient mice may provide a model for human renal Fanconi syndrome.

Shih et al. (2001) explored the molecular basis for the hypercholesterolemia of Tcf1 -/- mice using oligonucleotide microchip expression analysis. Shih et al. (2001) demonstrated that Tcf1 -/- mice have a defect in bile acid transport, increased bile acid and liver cholesterol synthesis, and impaired high-density lipoprotein (HDL) metabolism. Tcf1 -/- liver has decreased expression of the basolateral membrane bile acid transporters Slc10a1 (182396), Slc21a3 (602883), and Slc21a5, leading to impaired portal bile acid uptake and elevated plasma bile acid concentrations. In intestine and kidneys, Tcf1 -/- mice lack expression of the ileal bile acid transporter (Slc10a2; 601295), resulting in increased fecal and urinary bile acid excretion. Tcf1 protein also regulates transcription of Nr1h4 (603826), encoding the farnesoid X receptor-1 (Fxr1), thereby leading to reduced expression of small heterodimer partner-1 (Shp1; 604630) and repression of Cyp7a1 (118455), the rate-limiting enzyme in the classic bile acid biosynthesis pathway. In addition, hepatocyte bile acid storage protein is absent from Tcf1 -/- mice. Increased plasma cholesterol of Tcf1 -/- mice resides predominantly in large buoyant HDL particles. This is most likely due to reduced activity of the HDL-catabolic enzyme hepatic lipase (151670) and increased expression of HDL-cholesterol esterifying enzyme lecithin:cholesterol acyltransferase (LCAT; 606967). Shih et al. (2001) concluded that TCF1, in addition to being an important regulator of insulin secretion, is an essential transcriptional regulator of bile acid in HDL-cholesterol metabolism.

Hiraiwa et al. (2001) investigated whether there is a molecular link between HNF1A deficiency and function of the G6Pase (602671) system. Transactivation studies revealed that HNF1A is required for transcription of the G6PT gene. Hepatic G6PT mRNA levels and microsomal G6P transport activity are also markedly reduced in Hnf1a -/- mice as compared with Hnf1a +/+ and Hnf1a +/- littermates. On the other hand, hepatic G6Pase mRNA expression and activity are upregulated in Hnf1a -/- mice, consistent with observations that G6Pase expression is increased in diabetic animals. Taken together, these results strongly suggest that metabolic abnormalities in Hnf1a-null mice are caused in part by G6PT deficiency and by perturbations of the G6Pase system.

Huang et al. (2011) demonstrated the direct induction of functional hepatocyte-like (induced hepatocyte, iHep) cells from mouse tail-tip fibroblasts by transduction of Gata4 (600576), Hnf1-alpha, and Foxa3 (602295) and inactivation of p19(Arf) (600160). iHep cells showed typical epithelial morphology, expressed hepatic genes, and acquired hepatocyte functions. Notably, transplanted iHep cells repopulated the livers of fumarylacetoacetate hydrolase-deficient (Fah-null; see 613871) mice and rescued almost half of recipients from death by restoring liver functions.


Nomenclature

Although the symbol TCF1 (transcription factor-1) is used in the literature for this gene, its official designation is HNF1A. It should not be confused with the TCF7 gene (189908), which has also been referred to as TCF1 (T cell-specific transcription factor-1) in the literature.


ALLELIC VARIANTS 22 Selected Examples):

.0001   MATURITY-ONSET DIABETES OF THE YOUNG, TYPE 3

HEPATIC ADENOMA, SOMATIC, INCLUDED
RENAL CELL CARCINOMA, CLEAR CELL, INCLUDED
TYPE 1 DIABETES MELLITUS 20, INCLUDED
HNF1A, 1-BP INS, 872C
SNP: rs587776825, ClinVar: RCV000016062, RCV000016063, RCV000022617, RCV000117225, RCV000490055, RCV001255183, RCV001506982, RCV002221997, RCV003445070

Maturity-Onset Diabetes of the Young, Type 3

In a patient from an Edinburgh pedigree with MODY3 (600496), Yamagata et al. (1996) found in exon 4 of the TCF1 gene an insertion of a cytosine at codon 291 (pro), resulting in a frameshift and synthesis of a truncated mutant protein of 315 amino acids. This mutation was present in all affected members and no unaffected members of this family. It was not found on screening 55 healthy nondiabetic white subjects.

Using a rapid screening PCR method for frameshift mutations in the HNF1A gene in the screening of 60 MODY probands defined according to strict diagnostic criteria, Frayling et al. (1997) detected mutations in 11 (18%); the insertion mutation accounted for 13% of the MODY cases.

Ellard (2000) stated that the C insertion in the poly(C) tract of exon 4 had been reported in 22 of the 116 families with MODY3 worldwide who were identified by the finding of a mutation in the TCF1 gene. The total number of different mutations described was 65.

Bjorkhaug et al. (2003) found the P291fsinsC mutation in 9 families, 8 of Norwegian origin. Microsatellite analysis data suggested that in 7 of these families the mutant allele had a common origin.

Hepatic Adenoma

In studies of hepatic adenomas (142330) demonstrating biallelic inactivation of TCF1, Bluteau et al. (2002) observed the pro291fsX316 frameshift mutation (142410.0001) in heterozygous state in the tumor tissue of 2 individuals, one with multiple adenoma and the other with hepatocellular carcinoma (114550).

Renal Cell Carcinoma

In a 78-year-old man with clear cell renal carcinoma (see 144700), Rebouissou et al. (2005) identified heterozygosity for the 872insC mutation. Mutation screening of a tumor sample detected the germline mutation without mutation/deletion of the second allele. The man was diagnosed with diabetes mellitus in his sixth decade that was controlled by diet and oral hypoglycemic agents. No relatives had a diagnosis of renal carcinoma or diabetes.

Type 1 Diabetes Mellitus 20

In a Japanese subject with type 1 diabetes mellitus (T1D20; 612520) in whom insulin treatment was begun when hyperglycemia and ketonuria were noticed at 15 years of age, Yamada et al. (1997) identified a heterozygous frameshift mutation of codon pro291 resulting from insertion of a C in a poly(C) tract. (They designated this mutation P291fsinsC.) The mutation was predicted to result in a mutant truncated protein of 340 amino acids. The same mutation had been observed in British, German, and Finnish MODY families (Byrne et al., 1996; Yamagata et al., 1996; Kaisaki et al., 1997). Thus, Yamada et al. (1997) concluded that this site in exon 4 of the HNF1A gene appears to be a mutation hotspot.


.0002   MATURITY-ONSET DIABETES OF THE YOUNG, TYPE 3

HNF1A, PRO447LEU
SNP: rs137853236, gnomAD: rs137853236, ClinVar: RCV000016065, RCV000713784, RCV000762891, RCV001526899, RCV001810399

In their family A, Yamagata et al. (1996) found that MODY3 (600496) was associated with a single amino acid substitution in exon 7 of the TCF1 gene: codon 447 was changed from CCG (pro) to CTG (leu).

Hansen et al. (1997) found this mutation in a glucose-tolerant lean male who had relatives with MODY. He showed a low insulin secretion rate during oral glucose tolerance test (OGTT), but a 2-fold increase in pancreatic beta-cell response after intravenous glucose and a 2.5- to 4-fold increase in beta-cell response after either intravenous tolbutamide or intravenous glucagon loads. Hansen et al. (1997) concluded that early stages in the pathogenesis of MODY3 caused by the P447L mutation may be characterized by hyperexcitability of beta-cells to intravenous secretagogues.


.0003   MATURITY-ONSET DIABETES OF THE YOUNG, TYPE 3

HNF1A, 1-BP DEL
SNP: rs2135842335, ClinVar: RCV000016066

In a family in which 4 members of 3 generations had MODY3 (600496), Vaxillaire et al. (1997) found deletion of a guanine from codon glycine-292 (G292fsdelG) resulting in frameshift in the TCF1 gene.


.0004   MATURITY-ONSET DIABETES OF THE YOUNG, TYPE 3

HNF1A, TYR122CYS
SNP: rs137853237, ClinVar: RCV000016067, RCV001384610, RCV001794449, RCV002464065

In a family with multiple members with MODY3 (600496) in 3 generations, Vaxillaire et al. (1997) found a TAC-to-TGC transition in codon 122 of the TCF1 gene, predicted to cause an amino acid change from tyrosine to cysteine (Y122C).


.0005   TYPE 1 DIABETES MELLITUS 20

HNF1A, ARG272HIS
SNP: rs137853238, ClinVar: RCV000016068, RCV000255916, RCV000445525, RCV002051786, RCV002288493

In a Japanese subject who developed type 1 diabetes mellitus (T1D20; 612520) 1 year after the diagnosis of T2D at 8 years of age, Yamada et al. (1997) identified heterozygosity for an arg272-to-his (R272H) mutation in the DNA binding domain of the HNF1A gene.


.0006   TYPE 1 DIABETES MELLITUS 20

HNF1A, ARG583GLY
SNP: rs137853239, gnomAD: rs137853239, ClinVar: RCV000016069, RCV000030492, RCV002514104

In a Japanese patient with sudden-onset type 1 diabetes (T1D20; 612520) at 20 years of age, Yamada et al. (1997) identified heterozygosity for an arg583-to-gly (R583G) mutation in the transactivation domain of HNF1A. When first diagnosed there was marked hyperglycemia and 'absolute' insulin deficiency, prompting the initiation of insulin therapy. Control of blood glucose levels by exogenous insulin was poor, and complications of diabetes (proliferative retinopathy, cataracts, and sensorimotor neuropathy) developed.


.0007   MATURITY-ONSET DIABETES OF THE YOUNG, TYPE 3

HNF1A, A-C, -58, PROMOTER
SNP: rs2135818776, ClinVar: RCV000016070, RCV001794450, RCV002225265

Gragnoli et al. (1997) found an A-to-C substitution at nucleotide -58 of the promoter region of the HNF1A gene that cosegregated with MODY3 (600496). This mutation is located in a highly conserved region of the promoter and disrupted the binding site for the transcription factor HNF-4-alpha (600281), mutations in the gene encoding HNF-4-alpha being another cause of MODY (MODY1; 125850). This result demonstrated that decreased levels of HNF1-alpha per se can cause MODY. Moreover, it indicated that both the promoter and the coding regions of the HNF1A gene should be screened for mutations in subjects thought to have MODY.


.0008   TYPE 2 DIABETES MELLITUS, SUSCEPTIBILITY TO

HNF1A, GLY319SER
SNP: rs137853240, gnomAD: rs137853240, ClinVar: RCV000016071, RCV003230366

Hegele et al. (1999) identified a gly319-to-ser (G319S) variant in the HNF1A gene in Ontario Oji-Cree with early-onset type 2 diabetes (125853). G319S is in the proline II-rich domain of the trans-activation site of HNF1A and alters a glycine residue that is conserved throughout evolution. S319 was absent from 990 alleles from 6 other ethnic groups, suggesting that it is private for Oji-Cree. The S319 allele was more prevalent in diabetic than in nondiabetic Oji-Cree (0.209 vs 0.087; P = 0.000001). S319/S319 homozygotes and S319/G319 heterozygotes, respectively, had odds ratios for type 2 diabetes of 4.00 (95% CI, 2.65-6.03) and 1.97 (95% CI, 1.44-2.70) compared with G319/G319 homozygotes. There was a significant difference in the mean age of onset of type 2 diabetes, with G319/G319, S319/G319, and S319/S319 subjects being affected in the fifth, fourth, and third decades of life, respectively. Among nondiabetic subjects, S319/G319 heterozygotes had significantly lower plasma insulin than G319/G319 homozygotes. The authors concluded that the G319S variant is associated with a distinct form of type 2 diabetes, characterized by onset at an earlier age, lower body mass, and a higher postchallenge plasma glucose.

That the majority of Oji-Cree subjects with diabetes did not have the HNF1A S319 variant suggested to Hegele et al. (2000) that there might be other genetic determinants of diabetes susceptibility. In the course of sequencing candidate genes in diabetic subjects who were homozygous for HNF1A G319/G319, they found that some subjects had the PPARG A12 variant (601487.0002). PPARG A12 was strongly associated with type 2 diabetes in women, but not in men. The authors concluded that, when taken together with the previously reported association of diabetes with HNF1A in both men and women, the gender-specific association with PPARG A12 confirms that type 2 diabetes is etiologically complex in the Oji-Cree and that at least 2 genes are involved in determining susceptibility to the disease in this population.

Triggs-Raine et al. (2002) stated that Oji-Cree type 2 diabetes does not resemble MODY, because affected Oji-Cree subjects are obese and insulin-resistant with elevated plasma insulin concentrations, which clearly were insufficient to prevent diabetes onset. They evaluated the in vitro function of HNF1A G319S both to confirm that the mutation had a functional effect and to determine whether this effect was distinct from those of the complete loss-of-function or dominant-negative mutations seen in the MODY3 phenotype. They also evaluated the impact of the HNF1A G319S mutation on the dynamics of type 2 diabetes onset in the whole Sandy Lake Oji-Cree community. They found that the G319S mutation reduced the in vitro ability of HNF1-alpha to activate transcription by approximately 50%, with no effect on DNA binding or protein stability. There was no evidence of a dominant-negative effect of the mutant protein. Disease onset showed significant differences according to G319S genotype when gauged by the age at which half the subjects had become diabetic. Each dose of G319S accelerated median disease onset by approximately 7 years. Thus, the transactivation-deficient HNF1A G319S mutation affects the dynamics of disease onset. The demonstration of a functional consequence for the G319S mutation provided a mechanistic basis for its strong association with Oji-Cree type 2 diabetes and its unparalleled specificity for diabetes prediction in these people, in whom diabetes presents a significant public health problem. The finding also showed that HNF1A mutations can be associated with typical adult-onset insulin-resistant obesity-related diabetes in addition to maturity-onset diabetes of the young. Triggs-Raine et al. (2002) stated that in the Oji-Cree, HNF1A G319S behaves as a susceptibility allele for type 2 diabetes. Among nondiabetic Oji-Cree, fasting plasma insulin concentration was reduced significantly in HNF1A G319S carriers, suggesting that the partial impairment of function is tolerated when there is no insulin resistance. However, among Oji-Cree with type 2 diabetes, both carriers and noncarriers of the mutation had elevated plasma insulin concentration compared with nondiabetic Oji-Cree. The stress of obesity-induced insulin resistance seemed to expose the partial defect in HNF1A G319S carriers, causing expression of the disease.


.0009   MATURITY-ONSET DIABETES OF THE YOUNG, TYPE 3

HNF1A, THR620ILE
SNP: rs137853241, ClinVar: RCV000016072, RCV002250352, RCV002298445

Miedzybrodzka et al. (1999) described a family in which a thr620-to-ile substitution in transcription factor-1 was found in all members affected by MODY (600496). The mutation was not fully penetrant, as 2 family members aged 87 and 46 had the mutation but did not have diabetes. The severity and age at diagnosis of diabetes varied widely within the family, and most presented over the age of 25. Miedzybrodzka et al. (1999) suggested that TCF1 mutation screening should be considered in any family with autosomal dominant inheritance of diabetes where one member has presented with diabetes before the age of 25.


.0010   MATURITY-ONSET DIABETES OF THE YOUNG, TYPE 3

HNF1A, 1-BP DEL, -119G, PROMOTER
SNP: rs754470733, gnomAD: rs754470733, ClinVar: RCV000016073

Godart et al. (2000) observed a promoter mutation, -119delG, of the TCF1 gene that segregated with MODY3 (600496) in a diabetic family.


.0011   INSULIN RESISTANCE, SUSCEPTIBILITY TO

SERUM HDL CHOLESTEROL LEVEL, MODIFIER OF, INCLUDED
HNF1A, ILE27LEU
SNP: rs1169288, gnomAD: rs1169288, ClinVar: RCV000016074, RCV000016075, RCV000117233, RCV000710268, RCV000988920, RCV002226650, RCV002415417, RCV003315502

Chiu et al. (2000) examined the relationship between the ile27-to-leu (I27L) polymorphism of HNF1-alpha and insulin sensitivity (see 125853) and beta-cell function assessed by a hyperglycemic clamp. This study included 52 healthy glucose-tolerant and normotensive subjects (age, 19 to 40 years; body mass index, 17.58-35.61 kg/m2; waist/hip ratio, 0.65-1.03). Chiu et al. (2000) identified 19 LL subjects, 24 IL subjects, and 9 II subjects. The LL group had the highest postchallenge insulin levels at 30 and 90 min (P = 0.038 and P = 0.015, respectively) and also the highest insulin area under curve (P = 0.009) among the 3 genotypes. The LL group was more insulin resistant than the IL and II groups (P = 0.042 for insulin sensitivity index). After adjusting for age, gender, obesity, and ethnicity, the I27L polymorphism was an independent determinant of the insulin sensitivity index (P = 0.001). However, it had no impact on either the first or second phase insulin response. The authors concluded that the I27L polymorphism is associated with insulin resistance, but not beta-cell function. The mechanism of this association is unclear, but HNF1-alpha may play a role in regulating hepatic glucose metabolism.

Babaya et al. (2003) studied the relationship of the HNF1A gene polymorphism I27L with lipid parameters, in particular with serum HDL cholesterol level, in 356 unrelated Japanese men. Though no significant difference was observed in total cholesterol and triglyceride levels among the 3 genotypes, the serum HDL cholesterol level was significantly associated with the genotype (P less than 0.01). Subjects with the II genotype had low serum HDL cholesterol levels, and those with the LL genotype had high serum HDL cholesterol levels. The authors concluded that the HNF1A gene locus is associated with serum HDL cholesterol level and suggested that the I27 allele is a risk marker for atherosclerosis.


.0012   TYPE 1 DIABETES MELLITUS 20

HNF1A, 1-BP DEL, 142G
SNP: rs1566092470, ClinVar: RCV000016076, RCV002221998, RCV002326678

Yoshiuchi et al. (2001) identified a 142delG frameshift mutation in the TCF1 gene in a family with a strong history of type 1 diabetes (T1D20; 612520). The expression of the mutant protein was not detected in COS-7 cells by Western blot analysis after transfection of the mutant cDNA. According to the authors, this was the first case of an unstable mutant HNF1-alpha protein. Reporter gene analysis indicated that the mutant protein had no transactivation activity in HeLa and other cells. Haploinsufficiency for the TCF1 gene may lead to severe forms of diabetes resembling type I diabetes.


.0013   MATURITY-ONSET DIABETES OF THE YOUNG, TYPE 3

HNF1A, GLY574SER
SNP: rs1169305, gnomAD: rs1169305, ClinVar: RCV000016077, RCV000438436, RCV000861653, RCV003894808

This mutation was found by Collet et al. (2002) to be prevalent in African individuals with diabetes (600496).

In an individual who had had a liver-tumor resection and had familial diabetes, Bluteau et al. (2002) found a gly574-to-ser (G574S) mutation in the TCF1 gene. The hepatocellular carcinoma in this case had developed in an adenoma (142330).


.0014   MATURITY-ONSET DIABETES OF THE YOUNG, TYPE 3

HNF1A, ARG583GLN
SNP: rs137853242, gnomAD: rs137853242, ClinVar: RCV000016078, RCV000455019, RCV000658670, RCV002408468, RCV002467498, RCV003924838

Bluteau et al. (2002) found a heterozygous germline mutation, arg583 to gln (R583Q), in an individual with a hyperplastic liver tumor (142330) who had had liver-tumor resection and had familial diabetes (600496). The substitution involved a highly conserved amino acid.


.0015   MATURITY-ONSET DIABETES OF THE YOUNG, TYPE 3

HNF1A, PRO112LEU
SNP: rs137853243, gnomAD: rs137853243, ClinVar: RCV000016080, RCV000517471, RCV002221999

In a 3-generation Norwegian family with MODY3 (600496), Bjorkhaug et al. (2000) found a C-to-T transition at nucleotide 358 in exon 2 of the HNF1A gene, leading to a pro112-to-leu (P112L) amino acid substitution, in all 3 affected members. The phenotype in this family was mild with mild fasting and postprandial hyperglycemia easily controlled by diet only. Diabetes-associated late complications were not observed. P112L mutant protein demonstrated a significantly reduced ability to bind a high affinity HNF1 binding site and to activate transcription. Immunolocalization studies in HeLa cells showed that P112L mutant protein was correctly targeted to the nucleus. Bjorkhaug et al. (2000) concluded that the P112L mutation seems to impair pancreatic beta-cell function by loss-of-function mechanisms.

Xu et al. (2002) found the HNF1A P112L mutation in a southern Chinese MODY family.

Bjorkhaug et al. (2003) found evidence for possible founder effect of the P112L mutation in the Norwegian population.


.0016   MATURITY-ONSET DIABETES OF THE YOUNG, TYPE 3

HNF1A, ARG131TRP
SNP: rs137853244, ClinVar: RCV000016081, RCV000441594, RCV001794451, RCV002464066

Bjorkhaug et al. (2003) found a C-to-T transition in exon 2 of the HNF1A gene, resulting in an arg131-to-trp (R131W) amino acid substitution, in 5 Norwegian families with MODY3 (600496). This mutation had been reported in families from North America and Great Britain. Haplotype analysis indicated possible founder effect for the Norwegian families. Immunofluorescence studies demonstrated incorrect localization and accumulation of protein in both cytoplasm and nucleus. The R131W mutant protein displayed 10 to 15% of binding activity of wildtype and levels of transcription activation close to 50% of wildtype.


.0017   MATURITY-ONSET DIABETES OF THE YOUNG, TYPE 3

HNF1A, 4-BP DEL
SNP: rs2135839114, ClinVar: RCV000016082

In a Norwegian family with MODY3 (600496), Bjorkhaug et al. (2003) detected a novel 4-bp deletion in exon 3 of the HNF1A gene (T196fsdelCCAA).


.0018   MATURITY-ONSET DIABETES OF THE YOUNG, TYPE 3

HNF1A, IVS3, G-A, -1
SNP: rs1463923467, gnomAD: rs1463923467, ClinVar: RCV000016079, RCV002463589

In a Norwegian proband with MODY3 (600496), Bjorkhaug et al. (2003) found a novel splice site mutation in intron 3 of the HNF1A gene, IVS3-1G-A.


.0019   MATURITY-ONSET DIABETES OF THE YOUNG, TYPE 3

HNF1A, ALA276ASP
SNP: rs137853245, gnomAD: rs137853245, ClinVar: RCV000016083, RCV000521190, RCV001248962, RCV002254266

In a Norwegian proband with MODY3 (600496), Bjorkhaug et al. (2003) detected a novel C-to-A transversion in exon 4 of the HNF1A gene resulting in an arg276-to-asp (A276D) amino acid substitution. Mutant protein was targeted to both nucleus and cytoplasm of transfected cells in immunofluorescence assays. DNA binding ability of 30 to 40% of wildtype could be detected; no significant reduction in transcription activation could be demonstrated.


.0020   MATURITY-ONSET DIABETES OF THE YOUNG, TYPE 3

HNF1A, 2-BP DEL, AG
SNP: rs2135847417, ClinVar: RCV000016084

In 3 members of a Norwegian family with MODY3 (600496), Bjorkhaug et al. (2003) found a novel 2-bp deletion in exon 7 of the HNF1A gene (S445fsdelAG). Mutant protein was targeted to both nucleus and cytoplasm of transfected cells in immunofluorescence assays. DNA binding ability of 30 to 40% of wildtype could be detected; no significant reduction in transcription activation could be demonstrated.


.0021   MATURITY-ONSET DIABETES OF THE YOUNG, TYPE 3

HNF1A, SER531THR
SNP: rs137853246, gnomAD: rs137853246, ClinVar: RCV000016085, RCV001810400

In a Norwegian proband with MODY3 (600496), Bjorkhaug et al. (2003) detected a novel G-to-C transversion in exon 8 of the HNF1A gene, leading to a ser531-to-thr (S531T) amino acid substitution.


.0022   MATURITY-ONSET DIABETES OF THE YOUNG, TYPE 3

RENAL CELL CARCINOMA, CLEAR CELL, INCLUDED
RENAL CELL CARCINOMA, CHROMOPHOBE, INCLUDED
HNF1A, GLY92ASP
SNP: rs137853247, gnomAD: rs137853247, ClinVar: RCV000016086, RCV000016087, RCV000016088, RCV000503110, RCV000763797, RCV000835061, RCV001810401, RCV002371772

Maturity-Onset Diabetes of the Young, Type 3

In a French family with MODY3 (600496), Chevre et al. (1998) identified heterozygosity for a 92G-A transition in exon 1 of the HNF1A gene, resulting in a gly31-to-asp (G31D) substitution in the dimerization domain of the protein.

Renal Cell Carcinoma

In a 76-year-old woman with both clear cell and chromophobe renal carcinomas (see 144700), Rebouissou et al. (2005) identified heterozygosity for the G31D mutation. Mutation screening of the tumor samples detected only the germline G31D mutation. The renal carcinomas manifested in the same kidney with a single renal cyst in the other kidney. None of her relatives had a history of diabetes or renal carcinoma.


See Also:

De Simone et al. (1991); Mendel et al. (1991); Rey-Campos et al. (1991)

REFERENCES

  1. Babaya, N., Ikegami, H., Fujisawa, T., Nojima, K., Itoi-Babaya, M., Inoue, K., Nakura, J., Abe, M., Yamamoto, M., Jin, J. J., Wu, Z., Miki, T., Fukuda, M., Ogihara, T. Association of I27L polymorphism of hepatocyte nuclear factor-1-alpha gene with high-density lipoprotein cholesterol level. J. Clin. Endocr. Metab. 88: 2548-2551, 2003. [PubMed: 12788852] [Full Text: https://doi.org/10.1210/jc.2002-021891]

  2. Bach, I., Galcheva-Gargova, Z., Mattei, M.-G., Simon-Chazottes, D., Guenet, J.-L., Cereghini, S., Yaniv, M. Cloning of human hepatic nuclear factor 1 (HNF1) and chromosomal localization of its gene in man and mouse. Genomics 8: 155-164, 1990. [PubMed: 1707031] [Full Text: https://doi.org/10.1016/0888-7543(90)90238-p]

  3. Bjorkhaug, L., Sagen, J. V., Thorsby, P., Sovik, O., Molven, A., Njolstad, P. R. Hepatocyte nuclear factor-1a gene mutations and diabetes in Norway. J. Clin. Endocr. Metab. 88: 920-931, 2003. [PubMed: 12574234] [Full Text: https://doi.org/10.1210/jc.2002-020945]

  4. Bjorkhaug, L., Ye, H., Horikawa, Y., Sovik, O., Molven, A., Njolstad, P. R. MODY associated with two novel hepatocyte nuclear factor-1-alpha loss-of-function mutations (P112L and Q466X). Biochem. Biophys. Res. Commun. 279: 792-798, 2000. [PubMed: 11162430] [Full Text: https://doi.org/10.1006/bbrc.2000.4024]

  5. Bluteau, O., Jeannot, E., Bioulac-Sage, P., Marques, J. M., Blanc, J.-F., Bui, H., Beaudoin, J.-C., Franco, D., Balabaud, C., Laurent-Puig, P., Zucman-Rossi, J. Bi-allelic inactivation of TCF1 in hepatic adenomas. Nature Genet. 32: 312-315, 2002. [PubMed: 12355088] [Full Text: https://doi.org/10.1038/ng1001]

  6. 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., Bell, G. I., Polonsky, K. S. Altered insulin secretory responses to glucose in diabetic and nondiabetic subjects with mutations in the diabetes susceptibility gene MODY3 on chromosome 12. Diabetes 45: 1503-1510, 1996. [PubMed: 8866553] [Full Text: https://doi.org/10.2337/diab.45.11.1503]

  7. Chevre, J. C., Hani, E. H., Boutin, P., Vaxillaire, M., Blanche, H., Vionnet, N., Pardini, V. C., Timsit, J., Larger, E., Charpentier, G., Beckers, D., Maes, M., Bellanne-Chantelot, C., Velho, C., Froguel, P. Mutation screening in 18 Caucasian families suggest the existence of other MODY genes. Diabetologia 41: 1017-1023, 1998. [PubMed: 9754819] [Full Text: https://doi.org/10.1007/s001250051025]

  8. Chi, Y.-I., Frantz, J. D., Oh, B.-C., Hansen, L., Dhe-Paganon, S., Shoelson, S. E. Diabetes mutations delineate an atypical POU domain in HNF-1-alpha. Molec. Cell 10: 1129-1137, 2002. [PubMed: 12453420] [Full Text: https://doi.org/10.1016/s1097-2765(02)00704-9]

  9. Chiu, K. C., Chuang, L.-M., Ryu, J. M., Tsai, G. P., Saad, M. F. The I27L amino acid polymorphism of hepatic nuclear factor-1-alpha is associated with insulin resistance. J. Clin. Endocr. Metab. 85: 2178-2183, 2000. [PubMed: 10852449] [Full Text: https://doi.org/10.1210/jcem.85.6.6618]

  10. Collet, C., Ducorps, M, Mayaudon, H., Dupuy, O., Ceppa, F., Boutin, P., Froguel, P., Bauduceau, B. Prevalence of the missense mutation Gly574Ser in the hepatocyte nuclear factor-1-alpha in Africans with diabetes. Diabetes Metab. 28: 39-44, 2002. [PubMed: 11938027]

  11. Courtois, G., Morgan, J. G., Campbell, L. A., Fourel, G., Crabtree, G. R. Interaction of a liver-specific nuclear factor with the fibrinogen and alpha-1-antitrypsin promoters. Science 238: 688-692, 1987. [PubMed: 3499668] [Full Text: https://doi.org/10.1126/science.3499668]

  12. De Simone, V., De Magistris, L., Lazzaro, D., Gerstner, J., Monaci, P., Nicosia, A., Cortese, R. LFB3, a heterodimer-forming homeoprotein of the LFB1 family, is expressed in specialized epithelia. EMBO J. 10: 1435-1443, 1991. [PubMed: 1673925] [Full Text: https://doi.org/10.1002/j.1460-2075.1991.tb07664.x]

  13. Ellard, S. Hepatocyte nuclear factor 1 alpha (HNF-1-alpha) mutations in maturity-onset diabetes of the young. Hum. Mutat. 16: 377-385, 2000. [PubMed: 11058894] [Full Text: https://doi.org/10.1002/1098-1004(200011)16:5<377::AID-HUMU1>3.0.CO;2-2]

  14. Fajans, S. S., Bell, G. I., Polonsky, K. S. Molecular mechanisms and clinical pathophysiology of maturity-onset diabetes of the young. New Eng. J. Med. 345: 971-980, 2001. [PubMed: 11575290] [Full Text: https://doi.org/10.1056/NEJMra002168]

  15. Foster, J. H., Donohue, T. A., Berman, M. M. Familial liver-cell adenomas and diabetes mellitus. New Eng. J. Med. 299: 239-241, 1978. [PubMed: 207987] [Full Text: https://doi.org/10.1056/NEJM197808032990508]

  16. Frayling, T. M., Bulman, M. P., Appleton, M., Hattersley, A. T., Ellard, S. A rapid screening method for hepatocyte nuclear factor 1 alpha frameshift mutations; prevalence in maturity-onset diabetes of the young and late-onset non-insulin dependent diabetes. Hum. Genet. 101: 351-354, 1997. [PubMed: 9439666] [Full Text: https://doi.org/10.1007/s004390050640]

  17. Frayling, T. M., Bulman, M. P., Ellard, S., Appleton, M., Dronsfield, M. J., Mackie, A. D. R., Baird, J. D., Kaisaki, P. J., Yamagata, K., Bell, G. I., Bain, S. C., Hattersley, A. T. Mutations in the hepatocyte nuclear factor-1-alpha gene are a common cause of maturity-onset diabetes of the young in the U.K. Diabetes 46: 720-725, 1997. [PubMed: 9075818] [Full Text: https://doi.org/10.2337/diab.46.4.720]

  18. Godart, F., Bellanne-Chantelot, C., Clauin, S., Gragnoli, C., Abderrahmani, A., Blanche, H., Boutin, P., Chevre, J. C., Froguel, P., Bailleul, B. Identification of seven novel nucleotide variants in the hepatocyte nuclear factor-1-alpha (TCF1) promoter region in MODY patients. Hum. Mutat. 15: 173-180, 2000. [PubMed: 10649494] [Full Text: https://doi.org/10.1002/(SICI)1098-1004(200002)15:2<173::AID-HUMU6>3.0.CO;2-W]

  19. Gonzalez, F. J., Liu, S.-Y., Kozak, C. A., Nebert, D. W. Decreased Hnf-1 gene expression in mice homozygous for a 1.2-centimorgan deletion on chromosome 7. DNA Cell Biol. 9: 771-776, 1990. [PubMed: 2264930] [Full Text: https://doi.org/10.1089/dna.1990.9.771]

  20. Gragnoli, C., Lindner, T., Cockburn, B. N., Kaisaki, P. J., Gragnoli, F., Marozzi, G., Bell, G. I. Maturity-onset diabetes of the young due to a mutation in the hepatocyte nuclear factor-4-alpha binding site in the promoter of the hepatocyte nuclear factor-1-alpha gene. Diabetes 46: 1648-1651, 1997. [PubMed: 9313764] [Full Text: https://doi.org/10.2337/diacare.46.10.1648]

  21. Hansen, T., Eiberg, H., Rouard, M., Vaxillaire, M., Moller, A. M., Rasmussen, S. K., Fridberg, M., Urhammer, S. A., Holst, J. J., Almind, K., Echwald, S. M., Hansen, L., Bell, G. I., Pedersen, O. Novel MODY3 mutations in the hepatocyte nuclear factor-1-alpha gene: evidence for a hyperexcitability of pancreatic beta-cells to intravenous secretagogues in a glucose-tolerant carrier of a P447L mutation. Diabetes 46: 726-730, 1997. [PubMed: 9075819] [Full Text: https://doi.org/10.2337/diab.46.4.726]

  22. Hegele, R. A., Cao, H., Harris, S. B., Hanley, A. J. G., Zinman, B. The hepatic nuclear factor-1-alpha G319S variant is associated with early-onset type 2 diabetes in Canadian Oji-Cree. J. Clin. Endocr. Metab. 84: 1077-1082, 1999. [PubMed: 10084598] [Full Text: https://doi.org/10.1210/jcem.84.3.5528]

  23. Hegele, R. A., Cao, H., Harris, S. B., Zinman, B., Hanley, A. J. G., Anderson, C. M. Peroxisome proliferator-activated receptor-gamma-2 P12A and type 2 diabetes in Canadian Oji-Cree. J. Clin. Endocr. Metab. 85: 2014-2019, 2000. [PubMed: 10843190] [Full Text: https://doi.org/10.1210/jcem.85.5.6610]

  24. Hiraiwa, H., Pan, C.-J., Lin, B., Akiyama, T. E., Gonzalez, F. J., Chou, J. Y. A molecular link between the common phenotypes of type 1 glycogen storage disease and HNF1-alpha-null mice. J. Biol. Chem. 276: 7963-7967, 2001. [PubMed: 11121425] [Full Text: https://doi.org/10.1074/jbc.M010523200]

  25. Hua, Q.-X., Zhao, M., Narayana, N., Nakagawa, S. H., Jia, W., Weiss, M. A. Diabetes-associated mutations in a beta-cell transcription factor destabilize an antiparallel 'mini-zipper' in a dimerization interface. Proc. Nat. Acad. Sci. 97: 1999-2004, 2000. Note: Erratum: Proc. Nat. Acad. Sci. 98: 13472 only, 2001. [PubMed: 10696112] [Full Text: https://doi.org/10.1073/pnas.97.5.1999]

  26. Huang, P., He, Z., Ji, S., Sun, H., Xiang, D., Liu, C., Hu, Y., Wang, X., Hui, L. Induction of functional hepatocyte-like cells from mouse fibroblasts by defined factors. Nature 475: 386-389, 2011. [PubMed: 21562492] [Full Text: https://doi.org/10.1038/nature10116]

  27. Johansen, A., Ek, J., Mortensen, H. B., Pedersen, O., Hansen, T. Half of clinically defined maturity-onset diabetes of the young patients in Denmark do not have mutations in HNF4A, GCK, and TCF1. J. Clin. Endocr. Metab. 90: 4607-4614, 2005. [PubMed: 15928245] [Full Text: https://doi.org/10.1210/jc.2005-0196]

  28. Kaisaki, P. J., Menzel, S., Lindner, T., Oda, N., Rjasanowski, I., Sahm, J., Meincke, G., Schulze, J., Schmechel, H., Petzold, C., Ledermann, H. M., Sachse, G., Boriraj, V. V., Menzel, R., Kerner, W., Turner, R. C., Yamagata, K., Bell, G. I. Mutations in the hepatocyte nuclear factor-1-alpha gene in MODY and early-onset NIDDM: evidence for a mutational hotspot in exon 4. Diabetes 46: 528-535, 1997. Note: Erratum: Diabetes 46: 1239 only, 1997. [PubMed: 9032114] [Full Text: https://doi.org/10.2337/diab.46.3.528]

  29. Kuo, C. J., Conley, P. B., Hsieh, C.-L., Francke, U., Crabtree, G. R. Molecular cloning, functional expression, and chromosomal localization of mouse hepatocyte nuclear factor 1. Proc. Nat. Acad. Sci. 87: 9838-9842, 1990. [PubMed: 2263635] [Full Text: https://doi.org/10.1073/pnas.87.24.9838]

  30. Mendel, D. B., Hansen, L. P., Graves, M. K., Conley, P. B., Crabtree, G. R. HNF-1-alpha and HNF-1-beta (vHNF-1) share dimerization and homeo domains, but not activation domains, and form heterodimers in vitro. Genes Dev. 5: 1042-1056, 1991. [PubMed: 2044952] [Full Text: https://doi.org/10.1101/gad.5.6.1042]

  31. Mendel, D. B., Khavari, P. A., Conley, P. B., Graves, M. K., Hansen, L. P., Admon, A., Crabtree, G. R. Characterization of a cofactor that regulates dimerization of a mammalian homeodomain protein. Science 254: 1762-1767, 1991. [PubMed: 1763325] [Full Text: https://doi.org/10.1126/science.1763325]

  32. Miedzybrodzka, Z., Hattersley, A. T., Ellard, S., Pearson, D., de Silva, D., Harvey, R., Haites, N. Non-penetrance in a MODY 3 family with a mutation in the hepatic nuclear factor 1a gene: implications for predictive testing. Europ. J. Hum. Genet. 7: 729-732, 1999. [PubMed: 10482964] [Full Text: https://doi.org/10.1038/sj.ejhg.5200358]

  33. Mirshahi, U. L., Colclough, K., Wright, C. F., Wood, A. R., Beaumont, R. N., Tyrrell, J., Laver, T. W., Stahl, R., Golden, A., Goehringer, J. M, Geisinger-Regeneron DiscovEHR Collaboration, Frayling, T. F., Hattersley, A. T., Carey, D. J., Weedon, M. N., Patel, K. A. Reduced penetrance of MODY-associated HNF1A/HNF4A variants but not GCK variants in clinically unselected cohorts. Am. J. Hum. Genet. 109: 2018-2028, 2022. [PubMed: 36257325] [Full Text: https://doi.org/10.1016/j.ajhg.2022.09.014]

  34. Odom, D. T., Dowell, R. D., Jacobsen, E. S., Gordon, W., Danford, T. W., MacIsaac, K. D., Rolfe, P. A., Conboy, C. M., Gifford, D. K., Fraenkel, E. Tissue-specific transcriptional regulation has diverged significantly between human and mouse. Nature Genet. 39: 730-732, 2007. [PubMed: 17529977] [Full Text: https://doi.org/10.1038/ng2047]

  35. 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., Fraenkel, E., Bell, G. I., Young, R. A. Control of pancreas and liver gene expression by HNF transcription factors. Science 303: 1378-1381, 2004. [PubMed: 14988562] [Full Text: https://doi.org/10.1126/science.1089769]

  36. 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 84: 575-585, 1996. [PubMed: 8598044] [Full Text: https://doi.org/10.1016/s0092-8674(00)81033-8]

  37. Rebouissou, S., Vasiliu, V., Thomas, C., Bellanne-Chantelot, C., Bui, H., Chretien, Y., Timsit, J., Rosty, C., Laurent-Puig, P., Chauveau, D., Zucman-Rossi, J. Germline hepatocyte nuclear factor 1-alpha and 1-beta mutations in renal cell carcinomas. Hum. Molec. Genet. 14: 603-614, 2005. [PubMed: 15649945] [Full Text: https://doi.org/10.1093/hmg/ddi057]

  38. Reiner, A. P., Barber, M. J., Guan, Y., Ridker, P. M., Lange, L. A., Chasman, D. I., Walston, J. D., Cooper, G. M., Jenny, N. S., Rieder, M. J., Durda, J. P., Smith, J. D., Novembre, J., Tracy, R. P., Rotter, J. I., Stephens, M., Nickerson, D. A., Krauss, R. M. Polymorphisms of the HNF1A gene encoding hepatocyte nuclear factor-1-alpha are associated with C-reactive protein. Am. J. Hum. Genet. 82: 1193-1201, 2008. [PubMed: 18439552] [Full Text: https://doi.org/10.1016/j.ajhg.2008.03.017]

  39. Rey-Campos, J., Chouard, T., Yaniv, M., Cereghini, S. vHNF1 is a homeoprotein that activates transcription and forms heterodimers with HNF1. EMBO J. 10: 1445-1457, 1991. [PubMed: 1673926] [Full Text: https://doi.org/10.1002/j.1460-2075.1991.tb07665.x]

  40. Ridker, P. M., Pare, G., Parker, A., Zee, R. Y. L., Danik, J. S., Buring, J. E., Kwiatkowski, D., Cook, N. R., Miletich, J. P., Chasman, D. I. Loci related to metabolic syndrome pathways including LEPR, HNF1A, IL6R, and GCKR associate with plasma C-reactive protein: the Women's Genome Health Study. Am. J. Hum. Genet. 82: 1185-1192, 2008. [PubMed: 18439548] [Full Text: https://doi.org/10.1016/j.ajhg.2008.03.015]

  41. Shih, D. Q., Bussen, M., Sehayek, E., Ananthanarayanan, M., Shneider, B. L., Suchy, F. J., Shefer, S., Bollileni, J. S., Gonzalez, F. J., Breslow, J. L., Stoffel, M. Hepatocyte nuclear factor-1-alpha is an essential regulator of bile acid and plasma cholesterol metabolism. Nature Genet. 27: 375-382, 2001. [PubMed: 11279518] [Full Text: https://doi.org/10.1038/86871]

  42. Szpirer, C., Riviere, M., Cortese, R., Nakamura, T., Islam, M. Q., Levan, G., Szpirer, J. Chromosomal localization in man and rat of the genes encoding the liver-enriched transcription factors C/EBP, DBP, and HNF1/LFB-1 (CEBP, DBP, and transcription factor 1, TCF1, respectively) and of the hepatocyte growth factor/scatter factor gene (HGF). Genomics 13: 293-300, 1992. [PubMed: 1535333] [Full Text: https://doi.org/10.1016/0888-7543(92)90245-n]

  43. Triggs-Raine, B. L., Kirkpatrick, R. D., Kelly, S. L., Norquay, L. D., Cattini, P. A., Yamagata, K., Hanley, A. J. G., Zinman, B., Harris, S. B., Barrett, P. H., Hegele, R. A. HNF1-alpha G319S, a transactivation-deficient mutant, is associated with altered dynamics of diabetes onset in an Oji-Cree community. Proc. Nat. Acad. Sci. 99: 4614-4619, 2002. [PubMed: 11904371] [Full Text: https://doi.org/10.1073/pnas.062059799]

  44. Urhammer, S. A., Hansen, T., Ekstrom, C. T., Eiberg, H., Pederson, O. The Ala/Val98 polymorphism of the hepatocyte nuclear factor-1-alpha gene contributes to the interindividual variation in serum C-peptide response during an oral glucose tolerance test: evidence from studies of 231 glucose-tolerant first degree relatives of type 2 diabetic probands. J. Clin. Endocr. Metab. 83: 4506-4509, 1998. [PubMed: 9851800] [Full Text: https://doi.org/10.1210/jcem.83.12.5359]

  45. Urhammer, S. A., Moller, A. M., Nyholm, B., Ekstrom, C. T., Eiberg, H., Clausen, J. O., Hansen, T., Pedersen, O., Schmitz, O. The effect of two frequent amino acid variants of the hepatocyte nuclear factor-1-alpha gene on estimates of the pancreatic beta-cell function in Caucasian glucose-tolerant first-degree relatives of type 2 diabetic patients. J. Clin. Endocr. Metab. 83: 3992-3995, 1998. [PubMed: 9814481] [Full Text: https://doi.org/10.1210/jcem.83.11.5228]

  46. Urhammer, S. A., Rasmussen, S. K., Kaisaki, P. J., Oda, N., Yamagata, K., Moller, A. M., Fridberg, M., Hansen, L., Hansen, T., Bell, G. I., Pedersen, O. Genetic variation in the hepatocyte nuclear factor-1-alpha gene in Danish Caucasians with late-onset NIDDM. Diabetologia 40: 473-475, 1997. [PubMed: 9112026] [Full Text: https://doi.org/10.1007/s001250050703]

  47. van Wering, H. M., Huibregtse, I. L., van der Zwan, S. M., de Bie, M. S., Dowling, L. N., Boudreau, F., Rings, E. H. H. M., Grand, R. J., Krasinski, S. D. Physical interaction between GATA-5 and hepatocyte nuclear factor-1-alpha results in synergistic activation of the human lactase-phlorizin hydrolase promoter. J. Biol. Chem. 277: 27659-27667, 2002. [PubMed: 12011060] [Full Text: https://doi.org/10.1074/jbc.M203645200]

  48. Vaxillaire, M., Rouard, M., Yamagata, K., Oda, N., Kaisaki, P. J., Boriraj, V. V., Chevre, J.-C., Boccio, V., Cox, R. D., Lathrop, G. M., Dussoix, P., Philippe, J., Timsit, J., Charpentier, G., Velho, G., Bell, G. I., Froguel, P. Identification of nine novel mutations in the hepatocyte nuclear factor 1 alpha gene associated with maturity-onset diabetes of the young (MODY3). Hum. Molec. Genet. 6: 583-586, 1997. [PubMed: 9097962] [Full Text: https://doi.org/10.1093/hmg/6.4.583]

  49. Xu, J. Y., Chan, V., Zhang, W. Y., Wat, N. M. S., Lam, K. S. L. Mutations in the hepatocyte nuclear factor-1-alpha gene in Chinese MODY families: prevalence and functional analysis. Diabetologia 45: 744-746, 2002. [PubMed: 12107757] [Full Text: https://doi.org/10.1007/s00125-002-0814-9]

  50. Yamada, S., Nishigori, H., Onda, H., Utsugi, T., Yanagawa, T., Maruyama, T., Onigata, K., Nagashima, K., Nagai, R., Morikawa, A., Takeuchi, T., Takeda, J. Identification of mutations in the hepatocyte nuclear factor (HNF)-1-alpha gene in Japanese subjects with IDDM. Diabetes 46: 1643-1647, 1997. [PubMed: 9313763] [Full Text: https://doi.org/10.2337/diacare.46.10.1643]

  51. Yamagata, K., Oda, N., Kalsaki, P. J., Menzel, S., Furuta, H., Vaxillaire, M., Southam, L., Cox, R. D., Lathrop, G. M., Borhaj, V. V., Chen, X., Cox, N. J., Oda, Y., Yano, H., Le Beau, M. M., Yamada, S., Nishigori, H., Takeda, J., Fajans, S. S., Hattersley, A. T., Iwasaki, N., Hansen, T., Pedersen, O., Polonsky, K. S., Turner, R. C., Velho, G., Chevre, J.-C., Froguel, P., Bell, G. I. Mutations in the hepatocyte nuclear factor-1-alpha gene in maturity-onset diabetes of the young (MODY3). Nature 384: 455-457, 1996. [PubMed: 8945470] [Full Text: https://doi.org/10.1038/384455a0]

  52. Yoshiuchi, I., Yamagata, K., Yoshimoto, M., Zhu, Q., Yang, Q., Nammo, T., Uenaka, R., Kinoshita, E., Hanafusa, T., Miyagawa, J., Matsuzawa, Y. Analysis of a non-functional HNF-1-alpha (TCF1) mutation in Japanese subjects with familial type 1 diabetes. Hum. Mutat. 18: 345-351, 2001. [PubMed: 11668618] [Full Text: https://doi.org/10.1002/humu.1196]


Contributors:
Ada Hamosh - updated : 01/17/2023
Ada Hamosh - updated : 8/4/2011
Marla J. F. O'Neill - updated : 4/19/2010
Marla J. F. O'Neill - updated : 6/10/2008
George E. Tiller - updated : 2/5/2008
Patricia A. Hartz - updated : 10/3/2007
Patricia A. Hartz - updated : 8/3/2007
John A. Phillips, III - updated : 10/19/2006
Ada Hamosh - updated : 6/10/2004
John A. Phillips, III - updated : 2/10/2004
John A. Phillips, III - updated : 8/21/2003
Stylianos E. Antonarakis - updated : 5/5/2003
Victor A. McKusick - updated : 9/25/2002
Victor A. McKusick - updated : 9/25/2002
Victor A. McKusick - updated : 2/15/2002
Ada Hamosh - updated : 10/18/2001
John A. Phillips, III - updated : 9/25/2001
Ada Hamosh - updated : 4/23/2001
Ada Hamosh - updated : 3/28/2001
John A. Phillips, III - updated : 2/12/2001
Victor A. McKusick - updated : 11/29/2000
John A. Phillips, III - updated : 8/9/2000
Victor A. McKusick - updated : 2/22/2000
Victor A. McKusick - updated : 11/8/1999
John A. Phillips, III - updated : 9/29/1999
John A. Phillips, III - updated : 3/24/1999
Victor A. McKusick - updated : 2/13/1998
Victor A. McKusick - updated : 2/11/1998
Victor A. McKusick - updated : 8/12/1997
Victor A. McKusick - updated : 8/7/1997
Victor A. McKusick - updated : 4/24/1997

Creation Date:
Victor A. McKusick : 11/13/1987

Edit History:
alopez : 01/17/2023
carol : 09/16/2021
carol : 09/04/2020
carol : 09/03/2020
carol : 09/03/2020
alopez : 12/11/2017
carol : 10/03/2017
carol : 08/04/2016
carol : 05/17/2016
terry : 11/9/2012
alopez : 8/3/2012
carol : 3/29/2012
carol : 3/29/2012
mgross : 10/4/2011
terry : 10/4/2011
alopez : 8/15/2011
terry : 8/4/2011
alopez : 4/21/2010
terry : 4/19/2010
alopez : 4/16/2010
alopez : 4/16/2010
carol : 4/14/2009
terry : 2/19/2009
mgross : 10/21/2008
carol : 6/11/2008
terry : 6/10/2008
wwang : 2/7/2008
terry : 2/5/2008
wwang : 10/25/2007
mgross : 10/3/2007
mgross : 10/3/2007
alopez : 8/3/2007
alopez : 10/19/2006
carol : 11/18/2005
alopez : 3/30/2005
alopez : 6/15/2004
alopez : 6/15/2004
terry : 6/10/2004
alopez : 2/10/2004
alopez : 8/21/2003
mgross : 5/5/2003
mgross : 5/5/2003
tkritzer : 11/19/2002
alopez : 9/25/2002
alopez : 9/25/2002
tkritzer : 9/23/2002
ckniffin : 5/29/2002
carol : 4/30/2002
cwells : 2/25/2002
cwells : 2/20/2002
terry : 2/15/2002
carol : 1/4/2002
carol : 10/18/2001
cwells : 9/28/2001
cwells : 9/25/2001
alopez : 4/30/2001
terry : 4/23/2001
alopez : 3/29/2001
terry : 3/28/2001
terry : 2/12/2001
carol : 12/18/2000
mcapotos : 12/14/2000
terry : 11/29/2000
mgross : 8/9/2000
mcapotos : 4/12/2000
alopez : 3/23/2000
mcapotos : 3/22/2000
mcapotos : 3/14/2000
terry : 2/22/2000
alopez : 11/12/1999
psherman : 11/9/1999
terry : 11/8/1999
alopez : 11/5/1999
alopez : 9/29/1999
mgross : 4/7/1999
mgross : 3/24/1999
dkim : 12/9/1998
dkim : 7/21/1998
alopez : 5/14/1998
mark : 2/22/1998
terry : 2/13/1998
alopez : 2/11/1998
dholmes : 2/4/1998
jenny : 8/20/1997
terry : 8/12/1997
terry : 8/11/1997
terry : 8/7/1997
terry : 8/7/1997
alopez : 4/30/1997
alopez : 4/24/1997
terry : 4/24/1997
mark : 3/7/1997
mark : 12/11/1996
mark : 12/4/1996
terry : 12/3/1996
mark : 3/25/1996
terry : 3/18/1996
carol : 1/5/1995
mimadm : 4/14/1994
carol : 10/22/1992
carol : 6/26/1992
carol : 6/24/1992
supermim : 3/16/1992