Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (145)

Search Parameters:
Keywords = insulin gene mutations

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
20 pages, 887 KiB  
Review
Epigenetics of Endometrial Cancer: The Role of Chromatin Modifications and Medicolegal Implications
by Roberto Piergentili, Enrico Marinelli, Lina De Paola, Gaspare Cucinella, Valentina Billone, Simona Zaami and Giuseppe Gullo
Int. J. Mol. Sci. 2025, 26(15), 7306; https://doi.org/10.3390/ijms26157306 - 29 Jul 2025
Viewed by 259
Abstract
Endometrial cancer (EC) is the most common gynecological malignancy in developed countries. Risk factors for EC include metabolic alterations (obesity, metabolic syndrome, insulin resistance), hormonal imbalance, age at menopause, reproductive factors, and inherited conditions, such as Lynch syndrome. For the inherited forms, several [...] Read more.
Endometrial cancer (EC) is the most common gynecological malignancy in developed countries. Risk factors for EC include metabolic alterations (obesity, metabolic syndrome, insulin resistance), hormonal imbalance, age at menopause, reproductive factors, and inherited conditions, such as Lynch syndrome. For the inherited forms, several genes had been implicated in EC occurrence and development, such as POLE, MLH1, TP53, PTEN, PIK3CA, PIK3R1, CTNNB1, ARID1A, PPP2R1A, and FBXW7, all mutated at high frequency in EC patients. However, gene function impairment is not necessarily caused by mutations in the coding sequence of these and other genes. Gene function alteration may also occur through post-transcriptional control of messenger RNA translation, frequently caused by microRNA action, but transcriptional impairment also has a profound impact. Here, we review how chromatin modifications change the expression of genes whose impaired function is directly related to EC etiopathogenesis. Chromatin modification plays a central role in EC. The modification of chromatin structure alters the accessibility of genes to transcription factors and other regulatory proteins, thus altering the intracellular protein amount. Thus, DNA structural alterations may impair gene function as profoundly as mutations in the coding sequences. Hence, its central importance is in the diagnostic and prognostic evaluation of EC patients, with the caveat that chromatin alteration is often difficult to identify and needs investigations that are specific and not broadly used in common clinical practice. The different phases of the healthy endometrium menstrual cycle are characterized by differential gene expression, which, in turn, is also regulated through epigenetic mechanisms involving DNA methylation, histone post-translational modifications, and non-coding RNA action. From a medicolegal and policy-making perspective, the implications of using epigenetics in cancer care are briefly explored as well. Epigenetics in endometrial cancer is not only a topic of biomedical interest but also a crossroads between science, ethics, law, and public health, requiring integrated approaches and careful regulation. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

20 pages, 1783 KiB  
Review
Beyond Muscle Weakness: Unraveling Endocrine and Metabolic Dysfunctions in Duchenne Muscular Dystrophy, a Narrative Review
by Giuseppe Cannalire, Giacomo Biasucci, Vanessa Sambati, Tommaso Toschetti, Arianna Maria Bellani, Anna-Mariia Shulhai, Federica Casadei, Erika Rita Di Bari, Francesca Ferraboschi, Cecilia Parenti, Maria Carmela Pera, Susanna Esposito and Maria Elisabeth Street
Biomedicines 2025, 13(7), 1613; https://doi.org/10.3390/biomedicines13071613 - 1 Jul 2025
Viewed by 908
Abstract
Background: Duchenne muscular dystrophy (DMD) is a severe X-linked neuromuscular disorder caused by mutations in the DMD gene, leading to progressive muscle degeneration, loss of ambulation, and multi-systemic complications. Beyond its impact on mobility, DMD is associated with significant endocrine and metabolic dysfunctions [...] Read more.
Background: Duchenne muscular dystrophy (DMD) is a severe X-linked neuromuscular disorder caused by mutations in the DMD gene, leading to progressive muscle degeneration, loss of ambulation, and multi-systemic complications. Beyond its impact on mobility, DMD is associated with significant endocrine and metabolic dysfunctions that develop over time. Objective: To provide a comprehensive analysis of growth disturbances, endocrine dysfunctions, and metabolic complications in DMD including bone metabolism, considering the underlying mechanisms, clinical implications, and management strategies for daily clinical guidance. Methods: In this narrative review, an evaluation of the literature was conducted by searching the Medline database via the PubMed, Scopus, and Web of Science interfaces. Results: Growth retardation is a hallmark feature of DMD, with patients exhibiting significantly shorter stature compared to their healthy peers. This is exacerbated by long-term glucocorticoid therapy, which disrupts the growth hormone/insulin-like growth factor-1 (GH/IGF-1) axis and delays puberty. Obesity prevalence follows a biphasic trend, with increased risk in early disease stages due to reduced mobility and corticosteroid use, followed by a decline in body mass index (BMI) in later stages due to muscle wasting. Metabolic complications, including insulin resistance, altered lipid metabolism, and hepatic steatosis, further characterize disease burden. Osteoporosis and increased fracture risk, primarily due to reduced mechanical loading and glucocorticoid-induced bone resorption, are major concerns, needing early screening and intervention. The RANK/RANKL/OPG signaling pathway has emerged as a critical factor in bone deterioration, providing potential therapeutic targets for improving skeletal health. Conclusions: Growth and endocrine disorders in DMD are complex and multifactorial, requiring proactive monitoring and early intervention. Addressing these issues requires a multidisciplinary approach integrating endocrine, nutritional, and bone health management. Further research is essential to refine treatment strategies that mitigate growth and metabolic disturbances while preserving overall patient well-being. Full article
(This article belongs to the Special Issue Musculoskeletal Diseases: From Molecular Basis to Therapy (Volume II))
Show Figures

Figure 1

27 pages, 912 KiB  
Review
Connecting the Dots: Mitochondrial Dysfunction, PCOS, and Insulin Resistance—Insights and Therapeutic Advances
by Samia Palat Tharayil and Pallavi Shukla
Int. J. Mol. Sci. 2025, 26(13), 6233; https://doi.org/10.3390/ijms26136233 - 28 Jun 2025
Viewed by 1159
Abstract
Insulin resistance (IR) frequently develops in women with polycystic ovary syndrome (PCOS), an endocrinological disorder typified by hyperandrogenaemia, erratic menstrual cycles, and the presence of multiple cysts in the ovaries. It results in elevated androgen production contributing to the clinical manifestations of the [...] Read more.
Insulin resistance (IR) frequently develops in women with polycystic ovary syndrome (PCOS), an endocrinological disorder typified by hyperandrogenaemia, erratic menstrual cycles, and the presence of multiple cysts in the ovaries. It results in elevated androgen production contributing to the clinical manifestations of the syndrome including associated co-morbidities such as obesity and type 2 diabetes (T2D). Mounting data suggest the involvement of free fatty acids, reactive oxygen species (ROS) signalling, and mitochondrial dysfunction with IR. In recent years, numerous reports have suggested that mitochondrial dysregulation is associated with the pathogenesis of PCOS. Increased ROS, mutations/variants in mitochondrial DNA (mtDNA), and the altered expression of nuclear-related mitochondrial genes in insulin-resistant women with PCOS provide sufficient evidence for mitochondrial dysfunction as one of the factors contributing to PCOS pathogenesis. Despite the advancements in the field of interconnecting links between mitochondrial dysfunction, IR, and PCOS, various underlying mechanisms needs to be elucidated. Advancements in therapeutic interventions showed promising results in improving mitochondrial functions and IR in PCOS pathogenesis, including evolving mitochondrial transfer approaches that may improve in vitro fertilisation (IVF) outcomes in obese and insulin-resistant women with PCOS in future. Full article
(This article belongs to the Special Issue Advances in Insulin Resistance Research: 2nd Edition)
Show Figures

Figure 1

19 pages, 5311 KiB  
Article
Antiaging Effect of 2-O-β-D-Glucopyranosyl Ascorbic Acid Derived from Lycium barbarum L. Through Modulating the IIS Pathway and Gut Microbiota in Caenorhabditis elegans
by Jiayue Fang, Wei Dong, Jingqian Zheng, Boxuan Han, Yuying Zhang, Jianing Wang and Xiaoxiong Zeng
Foods 2025, 14(11), 1875; https://doi.org/10.3390/foods14111875 - 25 May 2025
Viewed by 911
Abstract
2-O-β-D-Glucopyranosyl ascorbic acid (AA-2βG), a bioactive ascorbic acid derivative isolated from the fruits of Lycium barbarum L., exhibited significant antiaging effects in Caenorhabditis elegans. It significantly extended their lifespan, enhanced stress resistance, reduced lipofuscin accumulation, and improved their healthspan, while [...] Read more.
2-O-β-D-Glucopyranosyl ascorbic acid (AA-2βG), a bioactive ascorbic acid derivative isolated from the fruits of Lycium barbarum L., exhibited significant antiaging effects in Caenorhabditis elegans. It significantly extended their lifespan, enhanced stress resistance, reduced lipofuscin accumulation, and improved their healthspan, while strengthening antioxidant defenses. Transcriptomic analysis identified the insulin/insulin-like growth factor (IGF)-1 signaling pathway as a key regulator, with quantitative real-time polymerase chain reaction confirming the upregulation of longevity-associated genes. Functional studies showed that the transcription factors DAF-16, HSF-1, and SIR-2.1 were essential for the lifespan-extending effects of AA-2βG, as mutations in these genes abolished lifespan extension. Moreover, 16S rRNA sequencing revealed that AA-2βG modulated gut microbiota by increasing longevity-associated taxa and reducing pro-aging species, with these alterations linked to metabolic pathways. These findings suggest that AA-2βG exerts antiaging effects through the coordinated regulation of the IIS pathway and gut microbiota composition, highlighting its potential as a natural geroprotective compound. Full article
(This article belongs to the Section Nutraceuticals, Functional Foods, and Novel Foods)
Show Figures

Figure 1

19 pages, 7605 KiB  
Case Report
Genetic Insights into Severe Obesity: A Case Study of MC4R Variant Identification and Clinical Implications
by Altynay Imangaliyeva, Nurgul Sikhayeva, Aidos Bolatov, Talgat Utupov, Aliya Romanova, Ilyas Akhmetollayev and Elena Zholdybayeva
Genes 2025, 16(5), 508; https://doi.org/10.3390/genes16050508 - 28 Apr 2025
Viewed by 1288
Abstract
Background/Objectives: Severe early-onset obesity is a complex condition shaped by genetic and metabolic influences. The melanocortin 4 receptor (MC4R) gene plays a crucial role in energy balance, and pathogenic variants are associated with monogenic forms of obesity. This study aims [...] Read more.
Background/Objectives: Severe early-onset obesity is a complex condition shaped by genetic and metabolic influences. The melanocortin 4 receptor (MC4R) gene plays a crucial role in energy balance, and pathogenic variants are associated with monogenic forms of obesity. This study aims to examine the clinical, metabolic, and genetic characteristics of a patient with severe early-onset obesity and his family, to assess the contribution of an MC4R variant to the observed phenotype. Methods: A 22-year-old male with severe obesity, first recognized at age 3, underwent detailed clinical, metabolic, and genetic evaluations. Laboratory assessments included insulin, lipid profile, uric acid, and IGF-1 levels. Whole-exome sequencing (WES) was performed on the patient and selected family members to identify potential pathogenic variants associated with obesity. Results: Clinical assessment revealed a body mass index (BMI) of 44.68 kg/m2, hyperinsulinemia (98.2 µIU/mL), prediabetes (HbA1c: 5.85%), dyslipidemia, hyperuricemia (421.0 µmol/L), and elevated IGF-1 levels (646.7 ng/mL). WES identified a heterozygous MC4R:c.216C>G (p.Asn72Lys) variant present in the patient, his mother, and maternal relatives. This variant, with a population frequency of 0.0004%, is predicted as likely pathogenic by SIFT, MutationTaster, and PrimateAI. However, its segregation pattern suggests a complex inheritance mechanism rather than classical autosomal dominant or recessive inheritance. Conclusions: Early genetic testing in individuals with severe obesity is essential for guiding personalized treatment strategies. Although the MC4R:c.216C>G variant may contribute to the patient’s metabolic profile, further functional studies are required to confirm its pathogenicity and elucidate its role in obesity pathogenesis. Full article
(This article belongs to the Special Issue Genetics of Multifactorial Diseases: 2nd Edition)
Show Figures

Figure 1

18 pages, 2334 KiB  
Article
Evaluating the Impact of rs4025935, rs71748309, rs699947, and rs4646994 Genetic Determinants on Polycystic Ovary Syndrome Predisposition—A Case-Control Study
by Reema Almotairi, Rashid Mir, Kholoud S. Almasoudi, Eram Husain and Nabil Mtiraoui
Life 2025, 15(4), 558; https://doi.org/10.3390/life15040558 - 29 Mar 2025
Viewed by 933
Abstract
Background: As a complicated endocrine condition, polycystic ovarian syndrome affects around 20% of women who are of reproductive age. It is linked to an increased risk of endometrial cancer, cardiovascular diseases, mental illnesses, non-alcoholic fatty liver disease, metabolic syndrome, and Type 2 diabetes. [...] Read more.
Background: As a complicated endocrine condition, polycystic ovarian syndrome affects around 20% of women who are of reproductive age. It is linked to an increased risk of endometrial cancer, cardiovascular diseases, mental illnesses, non-alcoholic fatty liver disease, metabolic syndrome, and Type 2 diabetes. Despite numerous genetic studies identifying several susceptibility loci, these only account for approximately 10% of the hereditary factors contributing to PCOS, leaving its etiology largely unknown. While genome-wide association studies (GWAS) have been conducted on various populations to identify SNPs linked to PCOS risk, no such study has been reported in Tabuk. Thus, this study aims to investigate the association of a glutathione S-transferase M1 (GSTM1) deletion, VEGF gene (I/D) insertion/deletion, and VEGF-2578 gene polymorphism with polycystic ovarian syndrome. Methodology: In this research study (case-control), we utilized the ARMS-PCR to determine and analyze the polymorphic variants of VEGF-2578 C/A (rs699947). We employed multiplex PCR for the GSTM1 deletion and MS-PCR (mutation specific PCR) for the vascular endothelial growth factor gene insertion/deletion. Results: The findings indicated statistically significant differences in various biochemical and endocrine serum biomarkers, including lipid profiles (cholesterol, HDL, and LDL), Type 2 diabetes markers (HOMA-IR (Homeostatic Model Assessment for Insulin Resistance), free insulin fasting glucose), and hormone levels (testosterone, LH, progesterone and FSH) in PCOS patients. Specifically, regarding the GSTT1 genotype, individuals with the GSTT1-null genotype had an odds ratio (OR) of 4.16 and a relative risk (RR) of 2.14 compared to those with the GSTT1 genotype, with statistically significant differences (p = 0.0001). However, for the GSTM1 genotype, there was a statistically significant difference (p = 0.0002) in the OR and RR for the GSTM1-null genotype, which were 2.66 and 1.64, respectively. Protective effects were observed for individuals with either GSTT1 (+) GSTM1 (−) or GSTT1 (−) GSTM1 (+) genotypes, as well as for those with both null genotypes, yielding an OR of 0.41 and p < 0.003. The VEGF rs699947 C>A gene variation showed a statistically significant association between PCOS patients and controls (p < 0.020), with the A allele frequency higher among PCOS patients (0.42 vs. 0.30). Similarly, the VEGF rs4646994 I>D gene variation exhibited a statistically significant difference (p < 0.0034), with the D allele being more frequent in PCOS patients (0.52 vs. 0.35). The VEGF-A allele was strongly linked to PCOS susceptibility in the allelic model, exhibiting an OR of 1.62, RR of 1.27, and p < 0.007, while in the allelic comparison, the OR was 1.71, the RR was 1.32, and p < 0.004. Conclusions: This study concluded that null genotypes at rs4025935 and rs71748309, an insertion deletion at rs4646994, and the A allele of rs699947 were significantly associated with PCOS predisposition in our population and these could serve as potential loci for PCOS predisposition. To the best of our knowledge, it is the first study to highlight the association between these genetic variations and the predisposition of PCOS in our populations. Large-scale case-control studies in the future are required to confirm these results. Full article
(This article belongs to the Section Medical Research)
Show Figures

Figure 1

29 pages, 1297 KiB  
Review
Caveolae: Metabolic Platforms at the Crossroads of Health and Disease
by Dante Maria Stea and Alessio D’Alessio
Int. J. Mol. Sci. 2025, 26(7), 2918; https://doi.org/10.3390/ijms26072918 - 24 Mar 2025
Viewed by 1494
Abstract
Caveolae are small flask-shaped invaginations of the plasma membrane enriched in cholesterol and sphingolipids. They play a critical role in various cellular processes, including signal transduction, endocytosis, and mechanotransduction. Caveolin proteins, specifically Cav-1, Cav-2, and Cav-3, in addition to their role as structural [...] Read more.
Caveolae are small flask-shaped invaginations of the plasma membrane enriched in cholesterol and sphingolipids. They play a critical role in various cellular processes, including signal transduction, endocytosis, and mechanotransduction. Caveolin proteins, specifically Cav-1, Cav-2, and Cav-3, in addition to their role as structural components of caveolae, have been found to regulate the activity of signaling molecules. A growing body of research has highlighted the pivotal role of caveolae and caveolins in maintaining cellular metabolic homeostasis. Indeed, studies have demonstrated that caveolins interact with the key components of insulin signaling, glucose uptake, and lipid metabolism, thereby influencing energy production and storage. The dysfunction of caveolae or the altered expression of caveolins has been associated with metabolic disorders, including obesity, type 2 diabetes, and ocular diseases. Remarkably, mutations in caveolin genes can disrupt cellular energy balance, promote oxidative stress, and exacerbate metabolic dysregulation. This review examines current research on the molecular mechanisms through which caveolae and caveolins regulate cellular metabolism, explores their involvement in the pathogenesis of metabolic disorders, and discusses potential therapeutic strategies targeting caveolin function and the stabilization of caveolae to restore metabolic homeostasis. Full article
Show Figures

Figure 1

20 pages, 3749 KiB  
Review
EFR3A, an Intriguing Gene, and Protein with a Scaffolding Function
by Magdalena Trybus, Anita Hryniewicz-Jankowska, Aleksander Czogalla and Aleksander F. Sikorski
Cells 2025, 14(6), 445; https://doi.org/10.3390/cells14060445 - 17 Mar 2025
Cited by 1 | Viewed by 971
Abstract
The EFR3 (Eighty-Five Requiring 3) protein and its homologs are rather poorly understood eukaryotic plasma membrane peripheral proteins. They belong to the armadillo-like family of superhelical proteins. In higher vertebrates two paralog genes, A and B were found, each expressing at least 2–3 [...] Read more.
The EFR3 (Eighty-Five Requiring 3) protein and its homologs are rather poorly understood eukaryotic plasma membrane peripheral proteins. They belong to the armadillo-like family of superhelical proteins. In higher vertebrates two paralog genes, A and B were found, each expressing at least 2–3 protein isoforms. EFR3s are involved in several physiological functions, mostly including phosphatidyl inositide phosphates, e.g., phototransduction (insects), GPCRs, and insulin receptors regulated processes (mammals). Mutations in the EFR3A were linked to several types of human disorders, i.e., neurological, cardiovascular, and several tumors. Structural data on the atomic level indicate the extended superhelical rod-like structure of the first two-thirds of the molecule with a typical armadillo repeat motif (ARM) in the N-terminal part and a triple helical motif in its C-terminal part. EFR3s’ best-known molecular function is anchoring the giant phosphatidylinositol 4-kinase A complex to the plasma membrane crucial for cell signaling, also linked directly to the KRAS mutant oncogenic function. Another function connected to the newly uncovered interaction of EFR3A with flotillin-2 may be the participation of the former in the organization and regulation of the membrane raft domain. This review presents EFR3A as an intriguing subject of future studies. Full article
Show Figures

Figure 1

14 pages, 1078 KiB  
Article
Early Changes in the Plasma Lipidome of People at Very High Cardiovascular Risk: A New Approach to Assessing the Risk of Cardiovascular Changes
by Joanna Waś, Piotr Dobrowolski, Aleksander Prejbisz, Magdalena Niedolistek, Ilona Kowalik, Anna Drohomirecka, Dorota Sokołowska and Jolanta Krzysztoń-Russjan
Biomedicines 2025, 13(3), 643; https://doi.org/10.3390/biomedicines13030643 - 6 Mar 2025
Viewed by 764
Abstract
Background/Objectives: Cardiovascular disease (CVD) remains the leading cause of death worldwide and requires a deeper understanding of its pathogenesis for effective prevention and treatment. Familial hypercholesterolemia (FH), characterized by high levels of LDL cholesterol, is a significant risk factor for CVD. FH background [...] Read more.
Background/Objectives: Cardiovascular disease (CVD) remains the leading cause of death worldwide and requires a deeper understanding of its pathogenesis for effective prevention and treatment. Familial hypercholesterolemia (FH), characterized by high levels of LDL cholesterol, is a significant risk factor for CVD. FH background remains unexplained despite advances in genetic testing. The aim was identification early changes in the plasma lipidome of individuals at high cardiovascular risk (HCVR) using liquid chromatography coupled with mass spectrometry. Methods: The lipidomic analysis examined over 400 compounds. Twenty individuals with suspected FH, very high cardiovascular risk (VHCVR), and undetectable mutations in the LDLR, APOB, or PCSK9 genes were compared to control group in a qualitative-quantitative analysis. Results: Multivariate analyses revealed statistically significant alterations in glycerophospholipids (GC), with a notable increase in phosphatidylcholines ((O-36:0/16:0), OR (95% CI): 1.246 (1.042–1.490), p = 0.0157), phosphatidylethanolamines ((O-40:7/22:6), OR (95% CI): 1.119 (1.039–1.205), p = 0.0028), and phosphatidylglycerol ((40:8/20:4), OR (95% CI): 1.053 (1.008–1.101), p = 0.0219) only in patients with HCVR. These changes, particularly in major classes of GC, underscored their potential as biomarkers for early assessment of cardiovascular risk. Lipidomic profiling revealed associations between specific lipid species and the comorbidities of arterial hypertension, atherosclerosis, and insulin resistance, implicating their role in atherosclerotic cardiovascular disease (ASCVD). Conclusions: This study points early changes in the plasma lipidome in individuals at HCVR, underline potential biomarkers, therapeutic targets for ASCVD, and offer opportunities to improve ASCVD diagnosis, therapy, and risk management strategies through detailed personalized medical approach. Full article
(This article belongs to the Section Endocrinology and Metabolism Research)
Show Figures

Figure 1

7 pages, 178 KiB  
Case Report
Diagnosis and Treatment of Neonatal Diabetes Caused by ATP-Channel Mutations: Genetic Insights, Sulfonylurea Therapy, and Future Directions
by Michela Trada, Chiara Novara, Martina Moretto, Edoardo Burzi, Davide Tinti and Luisa De Sanctis
Children 2025, 12(2), 219; https://doi.org/10.3390/children12020219 - 12 Feb 2025
Cited by 1 | Viewed by 1287
Abstract
Background: Neonatal diabetes (NDM) is a rare genetic disorder diagnosed in infants under six months of age, characterized by persistent hyperglycemia resulting from insufficient or absent insulin production. Unlike the more common forms of diabetes, such as type 1 diabetes (T1D) and type [...] Read more.
Background: Neonatal diabetes (NDM) is a rare genetic disorder diagnosed in infants under six months of age, characterized by persistent hyperglycemia resulting from insufficient or absent insulin production. Unlike the more common forms of diabetes, such as type 1 diabetes (T1D) and type 2 diabetes (T2D), NDM is predominantly caused by monogenic mutations affecting ATP-sensitive potassium (K-ATP) channels in pancreatic beta cells. The most common mutations involved in NDM are found in the KCNJ11 and ABCC8 genes, which encode the Kir6.2 and SUR1 subunits of the K-ATP channel, respectively. These mutations prevent normal insulin secretion by disrupting the function of the K-ATP channel. While genetic advances have identified about 40 genes implicated in NDM, the KCNJ11 and ABCC8 mutations are most commonly seen. Methods: This review provides a comprehensive exploration of the genetic basis, clinical presentation, and treatment strategies for NDM including the role of sulfonylureas, which have revolutionized the management of this condition. Furthermore, it presents a detailed case study of an infant diagnosed with an ABCC8 mutation, illustrating the pivotal role of genetic testing in guiding clinical decisions. Conclusions: Finally, the article discusses challenges in management, such as the persistence of neurological impairments, and outlines potential directions for future research including genetic therapies and prenatal diagnosis. Full article
(This article belongs to the Special Issue Advances in Childhood Diabetes)
22 pages, 1456 KiB  
Article
Genetic Structure of Hereditary Forms of Diabetes Mellitus in Russia
by Ildar R. Minniakhmetov, Rita I. Khusainova, Dmitry N. Laptev, Bulat I. Yalaev, Yulia S. Karpova, Roman V. Deev, Ramil R. Salakhov, Dmitry D. Panteleev, Kirill V. Smirnov, Galina A. Melnichenko, Marina V. Shestakova and Natalia G. Mokrysheva
Int. J. Mol. Sci. 2025, 26(2), 740; https://doi.org/10.3390/ijms26020740 - 16 Jan 2025
Viewed by 1420
Abstract
Analyzing the genetic architecture of hereditary forms of diabetes in different populations is a critical step toward optimizing diagnostic and preventive algorithms. This requires consideration of regional and population-specific characteristics, including the spectrum and frequency of pathogenic variants in targeted genes. As part [...] Read more.
Analyzing the genetic architecture of hereditary forms of diabetes in different populations is a critical step toward optimizing diagnostic and preventive algorithms. This requires consideration of regional and population-specific characteristics, including the spectrum and frequency of pathogenic variants in targeted genes. As part of this study, we used a custom-designed NGS panel to screen for mutations in 28 genes associated with the pathogenesis of hereditary diabetes mellitus in 506 unrelated patients from Russia. The study identified 180 pathogenic or likely pathogenic variants across 13 genes (GCK, HNF1A, HNF1B, HNF4A, ABCC8, INS, INSR, KCNJ11, PAX4, PDX1, ZFP57, BLK, WFS1), representing 46.44% of the analyzed cohort (235 individuals). The glucokinase gene (GCK) had the highest number of identified variants, with 111 variants detected in 161 patients, 20 of which were identified for the first time. In the tissue-specific transcription factor genes HNF1A, HNF4A, and HNF1B, 34 variants were found in 38 patients, including 13 that were previously unreported. Seventeen variants were identified in the ABCC8 gene, which encodes the ATP-binding cassette transporter 8 of subfamily C, each found in a different patient; four of these were novel discoveries. Nine pathogenic or likely pathogenic variants were identified in the insulin gene (INS) and its receptor gene (INSR), including four previously unreported variants. Additionally, we identified 10 previously unreported variants in six other genes among 11 patients. Variants in the genes GCK, HNF1A, HNF1B, HNF4A, ABCC8, INS, and INSR were the main contributors to the genetic pathogenesis of hereditary diabetes mellitus in the Russian cohort. These findings enhance our understanding of the molecular mechanisms underlying the disease and provide a solid basis for future studies aimed at improving diagnostic accuracy and advancing personalized therapeutic strategies. This knowledge provides a foundation for developing region-specific genetic testing algorithms and personalized therapeutic strategies, which are critical for future initiatives in precision medicine. Full article
(This article belongs to the Special Issue Diabetes: From Molecular Basis to Therapy, 2nd Edition)
Show Figures

Figure 1

14 pages, 4085 KiB  
Article
Identification of Functional Variants Between Tong Sheep and Hu Sheep by Whole-Genome Sequencing Pools of Individuals
by Xiaoqin Tang, Shuhui Wang, Xiaohua Yi, Qi Li and Xiuzhu Sun
Int. J. Mol. Sci. 2024, 25(23), 12919; https://doi.org/10.3390/ijms252312919 - 30 Nov 2024
Cited by 1 | Viewed by 1052
Abstract
Tong sheep, known for their superior meat quality and disease resilience, face breeding challenges due to low prolificacy, unlike Hu sheep, which exhibit higher fertility and growth rates. This study identified over 700,000 genetic variants between these breeds through pooled whole-genome sequencing. Functional [...] Read more.
Tong sheep, known for their superior meat quality and disease resilience, face breeding challenges due to low prolificacy, unlike Hu sheep, which exhibit higher fertility and growth rates. This study identified over 700,000 genetic variants between these breeds through pooled whole-genome sequencing. Functional analysis reveals key differences in pathways related to fat metabolism, insulin signaling, and cell cycle regulation. Notable findings include unique microRNA variants (miR-1185-3p in Tong sheep and miR-487-5p in Hu sheep), with the miR-487-5p mutation potentially regulating KITLG, a fertility-related gene. These results suggest that non-coding RNA mutations contribute to phenotypic differences and provide a genomic foundation for molecular-assisted selection to improve Tong sheep breeding programs. Full article
(This article belongs to the Section Molecular Genetics and Genomics)
Show Figures

Figure 1

18 pages, 1782 KiB  
Review
Monogenic Defects of Beta Cell Function: From Clinical Suspicion to Genetic Diagnosis and Management of Rare Types of Diabetes
by Anastasios Serbis, Evanthia Kantza, Ekaterini Siomou, Assimina Galli-Tsinopoulou, Christina Kanaka-Gantenbein and Stelios Tigas
Int. J. Mol. Sci. 2024, 25(19), 10501; https://doi.org/10.3390/ijms251910501 - 29 Sep 2024
Cited by 3 | Viewed by 2506
Abstract
Monogenic defects of beta cell function refer to a group of rare disorders that are characterized by early-onset diabetes mellitus due to a single gene mutation affecting insulin secretion. It accounts for up to 5% of all pediatric diabetes cases and includes transient [...] Read more.
Monogenic defects of beta cell function refer to a group of rare disorders that are characterized by early-onset diabetes mellitus due to a single gene mutation affecting insulin secretion. It accounts for up to 5% of all pediatric diabetes cases and includes transient or permanent neonatal diabetes, maturity-onset diabetes of the young (MODY), and various syndromes associated with diabetes. Causative mutations have been identified in genes regulating the development or function of the pancreatic beta cells responsible for normal insulin production and/or release. To date, more than 40 monogenic diabetes subtypes have been described, with those caused by mutations in HNF1A and GCK genes being the most prevalent. Despite being caused by a single gene mutation, each type of monogenic diabetes, especially MODY, can appear with various clinical phenotypes, even among members of the same family. This clinical heterogeneity, its rarity, and the fact that it shares some features with more common types of diabetes, can make the clinical diagnosis of monogenic diabetes rather challenging. Indeed, several cases of MODY or syndromic diabetes are accurately diagnosed in adulthood, after having been mislabeled as type 1 or type 2 diabetes. The recent widespread use of more reliable sequencing techniques has improved monogenic diabetes diagnosis, which is important to guide appropriate treatment and genetic counselling. The current review aims to summarize the latest knowledge on the clinical presentation, genetic confirmation, and therapeutic approach of the various forms of monogenic defects of beta cell function, using three imaginary clinical scenarios and highlighting clinical and laboratory features that can guide the clinician in reaching the correct diagnosis. Full article
(This article belongs to the Special Issue Diabetes: From Molecular Basis to Therapy)
Show Figures

Figure 1

15 pages, 1539 KiB  
Article
Genome-Wide Search for Gene Mutations Likely Conferring Insecticide Resistance in the Common Bed Bug, Cimex lectularius
by Kouhei Toga, Fumiko Kimoto, Hiroki Fujii and Hidemasa Bono
Insects 2024, 15(10), 737; https://doi.org/10.3390/insects15100737 - 24 Sep 2024
Cited by 1 | Viewed by 4419
Abstract
Insecticide resistance in the bed bug Cimex lectularius is poorly understood due to the lack of genome sequences for resistant strains. In Japan, we identified a resistant strain of C. lectularius that exhibits a higher pyrethroid resistance ratio compared to many previously discovered [...] Read more.
Insecticide resistance in the bed bug Cimex lectularius is poorly understood due to the lack of genome sequences for resistant strains. In Japan, we identified a resistant strain of C. lectularius that exhibits a higher pyrethroid resistance ratio compared to many previously discovered strains. We sequenced the genomes of the pyrethroid-resistant and susceptible strains using long-read sequencing, resulting in the construction of highly contiguous genomes (N50 of the resistant strain: 2.1 Mb and N50 of the susceptible strain: 1.5 Mb). Gene prediction was performed by BRAKER3, and the functional annotation was performed by the Fanflow4insects workflow. Next, we compared their amino acid sequences to identify gene mutations, identifying 729 mutated transcripts that were specific to the resistant strain. Among them, those defined previously as resistance genes were included. Additionally, enrichment analysis implicated DNA damage response, cell cycle regulation, insulin metabolism, and lysosomes in the development of pyrethroid resistance. Genome editing of these genes can provide insights into the evolution and mechanisms of insecticide resistance. This study expanded the target genes to monitor allele distribution and frequency changes, which will likely contribute to the assessment of resistance levels. These findings highlight the potential of genome-wide approaches to understand insecticide resistance in bed bugs. Full article
Show Figures

Figure 1

24 pages, 22049 KiB  
Article
Deciphering Dormant Cells of Lung Adenocarcinoma: Prognostic Insights from O-glycosylation-Related Tumor Dormancy Genes Using Machine Learning
by Chenfei Dong, Yang Liu, Suli Chong, Jiayue Zeng, Ziming Bian, Xiaoming Chen and Sairong Fan
Int. J. Mol. Sci. 2024, 25(17), 9502; https://doi.org/10.3390/ijms25179502 - 31 Aug 2024
Cited by 2 | Viewed by 2653
Abstract
Lung adenocarcinoma (LUAD) poses significant challenges due to its complex biological characteristics and high recurrence rate. The high recurrence rate of LUAD is closely associated with cellular dormancy, which enhances resistance to chemotherapy and evasion of immune cell destruction. Using single-cell RNA sequencing [...] Read more.
Lung adenocarcinoma (LUAD) poses significant challenges due to its complex biological characteristics and high recurrence rate. The high recurrence rate of LUAD is closely associated with cellular dormancy, which enhances resistance to chemotherapy and evasion of immune cell destruction. Using single-cell RNA sequencing (scRNA-seq) data from LUAD patients, we categorized the cells into two subclusters: dormant and active cells. Utilizing high-density Weighted Gene Co-expression Network Analysis (hdWGCNA) and pseudo-time cell trajectory, aberrant expression of genes involved in protein O-glycosylation was detected in dormant cells, suggesting a crucial role for O-glycosylation in maintaining the dormant state. Intercellular communication analysis highlighted the interaction between fibroblasts and dormant cells, where the Insulin-like Growth Factor (IGF) signaling pathway regulated by O-glycosylation was crucial. By employing Gene Set Variation Analysis (GSVA) and machine learning, a risk score model was developed using hub genes, which showed high accuracy in determining LUAD prognosis. The model also demonstrated robust performance on the training dataset and excellent predictive capability, providing a reliable basis for predicting patient clinical outcomes. The group with a higher risk score exhibited a propensity for adverse outcomes in the tumor microenvironment (TME) and tumor mutational burden (TMB). Additionally, the 50% inhibitory concentration (IC50) values for chemotherapy exhibited significant variations among the different risk groups. In vitro experiments demonstrated that EFNB2, PTTG1IP, and TNFRSF11A were upregulated in dormant tumor cells, which also contributed greatly to the diagnosis of LUAD. In conclusion, this study highlighted the crucial role of O-glycosylation in the dormancy state of LUAD tumors and developed a predictive model for the prognosis of LUAD patients. Full article
(This article belongs to the Special Issue Biomarkers of Tumor Progression, Prognosis and Therapy: 2nd Edition)
Show Figures

Figure 1

Back to TopTop