Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (133)

Search Parameters:
Keywords = phenocopy

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
19 pages, 4391 KB  
Article
Brassinosteroid Synthesis and Perception Differently Regulate Phytohormone Networks in Arabidopsis thaliana
by Yaroslava Bukhonska, Michael Derevyanchuk, Roberta Filepova, Jan Martinec, Petre Dobrev, Eric Ruelland and Volodymyr Kravets
Int. J. Mol. Sci. 2025, 26(19), 9644; https://doi.org/10.3390/ijms26199644 - 2 Oct 2025
Viewed by 428
Abstract
Brassinosteroids (BRs) are essential regulators of plant development and stress responses, but the distinct contributions of BR biosynthesis and signaling to hormonal crosstalk remain poorly defined. Here, we investigated the effects of the BR biosynthesis inhibitor brassinazole (BRZ) and the BR-insensitive mutant bri1-6 [...] Read more.
Brassinosteroids (BRs) are essential regulators of plant development and stress responses, but the distinct contributions of BR biosynthesis and signaling to hormonal crosstalk remain poorly defined. Here, we investigated the effects of the BR biosynthesis inhibitor brassinazole (BRZ) and the BR-insensitive mutant bri1-6 on endogenous phytohormone profiles in Arabidopsis thaliana. Using multivariate analysis and targeted hormone quantification, we show that BRZ treatment and BRI1 disruption alter hormone balance through partially overlapping but mechanistically distinct pathways. Principal component analysis (PCA) and hierarchical clustering revealed that BRZ and the bri1-6 mutation do not phenocopy each other and that BRZ still alters hormone profiles even in the bri1-6 mutant, suggesting potential BRI1-independent effects. Both BRZ treatment and the bri1-6 mutation tend to influence cytokinins and auxin conjugates divergently. On the contrary, their effects on stress-related hormones converge: BRZ decreases salicylic acid (SA), jasmonic acid (JA), and abscisic acid (ABA) in the WT leaves; similarly, bri1-6 mutants show reduced SA, JA, and ABA. These results indicate that BR biosynthesis and BRI1-mediated perception may contribute independently to hormonal reprogramming, with BRZ eliciting additional effects, possibly via metabolic feedback, compensatory signaling, or off-target action. Hormone correlation analyses revealed conserved co-regulation clusters that reflect underlying regulatory modules. Altogether, our findings provide evidence for a partial uncoupling of BR levels and BR signaling and illustrate how BR pathways intersect with broader hormone networks to coordinate growth and stress responses. Full article
(This article belongs to the Special Issue Emerging Insights into Phytohormone Signaling in Plants)
Show Figures

Figure 1

15 pages, 1654 KB  
Article
Genetic, Clinical, and Sociodemographic Profile of Individuals with Diagnosis or Family History of Hypertrophic Cardiomyopathy: Insights from a Prospective Cohort
by Emerson de Santana Santos, Gabriel da Costa Kuhn, Antônio Guilherme Cunha de Almeida, João Victor Andrade Pimentel, Newton Vital Figueiredo Neto, Larissa Rebeca da Silva Tavares, Bárbara Letícia Lima dos Santos, Ana Beatriz Leite Aragão, Beatriz Carolina de Araujo Pereira, Caio da Silva Ferreira, Willian Moreira Leão e Silva, Cardiogenetics Research Group of Sergipe, Enaldo Vieira de Melo, Irlaneide da Silva Tavares, Antônio Carlos Sobral Sousa and Joselina Luzia Menezes Oliveira
Genes 2025, 16(9), 1100; https://doi.org/10.3390/genes16091100 - 17 Sep 2025
Viewed by 526
Abstract
Background: Hypertrophic cardiomyopathy (HCM) is a genetic cardiac disorder characterized by left ventricular hypertrophy in the absence of secondary causes. This study aimed to investigate the genetic, clinical, and epidemiological profile of individuals with clinical HCM or a family history of sudden [...] Read more.
Background: Hypertrophic cardiomyopathy (HCM) is a genetic cardiac disorder characterized by left ventricular hypertrophy in the absence of secondary causes. This study aimed to investigate the genetic, clinical, and epidemiological profile of individuals with clinical HCM or a family history of sudden cardiac death (SCD). Methods: A total of 200 participants (58% male, median age 52 years) underwent genetic testing using a 19-gene panel associated with HCM and phenocopies. Variants were classified as pathogenic/likely pathogenic (P/LP), variants of uncertain significance (VUS), or negative. Clinical and imaging data were correlated with genetic findings. Results: P/LP variants were identified in 31% of individuals, while 40.5% carried VUS, and 28.5% tested negative. A positive genotype was more frequent among patients with clinical HCM (37.7%) than among those with only a family history (18.6%, p = 0.006). Sarcomeric mutations represented 77.4% of positive results, while 22.6% involved phenocopy genes, notably TTR (amyloidosis). Positive genotypes were significantly associated with a family history of SCD (68% vs. 46%, p = 0.004) and with greater interventricular septal thickness (17 mm vs. 15 mm, p < 0.001). Conclusions: Septal thickness >17 mm and family history of SCD were strong predictors of positive genetic results. These findings emphasize the importance of genetic screening and counseling in high-risk individuals and highlight the value of integrating genetic testing into clinical practice for diagnosis, risk stratification, and family management. Full article
(This article belongs to the Section Human Genomics and Genetic Diseases)
Show Figures

Figure 1

39 pages, 1987 KB  
Review
Gas Plasma Combination Therapies—Promises from Preclinical Oncology Research
by Lingyun Yu, Julia Berner, Alice Martinet, Eric Freund, Debora Singer, Thomas von Woedtke, Klaus-Dieter Weltmann, Steffen Emmert, Ramona Clemen and Sander Bekeschus
Antioxidants 2025, 14(9), 1055; https://doi.org/10.3390/antiox14091055 - 27 Aug 2025
Viewed by 1499
Abstract
The absent decline in cancer mortality rates is primarily due to moderate therapeutic efficacy and intrinsic or acquired tumor cell resistance toward treatments. Combining different oncology treatments increases therapy success and decreases the chance of refractory tumor cells. Therefore, combination cancer treatments are [...] Read more.
The absent decline in cancer mortality rates is primarily due to moderate therapeutic efficacy and intrinsic or acquired tumor cell resistance toward treatments. Combining different oncology treatments increases therapy success and decreases the chance of refractory tumor cells. Therefore, combination cancer treatments are the principal paradigm of 21st-century oncology. Physical modalities such as radiotherapy have a long-standing tradition in such combination treatments. In the last decade, another physical principle emerged as a promising anticancer agent: cold gas plasma. This partially ionized gas, operated at about body temperature, emits multiple bioactive components, including reactive oxygen and nitrogen species (ROS/RNS). This technology’s multi-ROS/RNS nature cannot be phenocopied by other means, and it capitalizes on the vulnerability of tumor cells within metabolic and redox signaling pathways. Many cancer models exposed to mono or combination gas plasma treatments have shown favorable results, and first cancer patients have benefited from cold gas plasma therapy. The main findings and proposed mechanisms of action are summarized. Considering the specific application modes, this review identifies promising gas plasma combination therapies within guideline-directed treatment schemes for several tumor entities. In conclusion, gas plasmas may become a potential (neo)adjuvant therapy to existing treatment modalities to help improve the efficacy of oncological treatments. Full article
Show Figures

Figure 1

25 pages, 4801 KB  
Article
Genotype–Phenotype Correlations in PRPH2 Retinopathies: A Comprehensive Analysis of 36 Patients from the Oxford Eye Hospital, UK
by Saoud Al-Khuzaei, Mital Shah, Arun Reginald, Edna Baba, Morag Shanks, Penny Clouston, Robert E. MacLaren, Stephanie Halford, Samantha R. De Silva and Susan M. Downes
Genes 2025, 16(9), 1016; https://doi.org/10.3390/genes16091016 - 27 Aug 2025
Viewed by 922
Abstract
Purpose: To investigate genotype–phenotype correlations in PRPH2-retinopathies in a cohort of 36 patients from the Oxford Eye Hospital and report on novel pathogenic variants. Methods: Clinical data, including best corrected visual acuities (BCVA), fundus autofluorescence (FAF), and optical coherence tomography (OCT) imaging, [...] Read more.
Purpose: To investigate genotype–phenotype correlations in PRPH2-retinopathies in a cohort of 36 patients from the Oxford Eye Hospital and report on novel pathogenic variants. Methods: Clinical data, including best corrected visual acuities (BCVA), fundus autofluorescence (FAF), and optical coherence tomography (OCT) imaging, were analysed. Genetic testing was performed using next-generation sequencing (NGS). Results: In this cohort, 26 different PRPH2 variants, including 8 novel variants, were identified. Variants were clustered in the D2 loop of the protein. A diverse range of phenotypes were observed: pseudo-Stargardt pattern dystrophy (PSPD) (47.2%), adult-onset vitelliform macular dystrophy (AVMD) (22.2%), pattern dystrophy (PD) (25.0%), atypical macular dystrophy (2.8%), and retinitis pigmentosa (RP) (2.8%). The mean age of symptom onset was 44.0 ± 14.4 years. Mean BCVA was 0.20 ± 0.54 logMAR OD and 0.14 ± 0.29 logMAR OS at baseline and 0.33 ± 0.40 logMAR OD and 0.32 ± 0.40 logMAR OS after a mean follow up duration of 6.0 ± 3.2 years (range 1–11 years). A thickened ellipsoid zone (EZ) was noted in 34/36 patients with a mean EZ thickness of 44.3 ± 11.3 µm OD and 42.7 ± 11.6 µm OS. No clear genotype–phenotype correlations were observed. Conclusions: The significant phenotypic range described in this study is consistent with the previously reported phenotypic variability in PRPH2 retinopathy and emphasises the complexity of establishing genotype–phenotype correlations in this disease. The thickness of the EZ on OCT may serve as a useful biomarker in distinguishing PRPH2 retinopathy from other phenocopies. These findings contribute to improved understanding of PRPH2 retinopathy and help inform diagnosis and genetic counselling. Full article
(This article belongs to the Section Human Genomics and Genetic Diseases)
Show Figures

Figure 1

21 pages, 16819 KB  
Article
LncRNA TSPEAR-AS2 Maintains the Stemness of Gastric Cancer Stem Cells by Regulating the miR-15a-5p/CCND1 Axis
by Qiong Li, Yanan Wang, Liyang Chen, Yan Shen, Shijiao Zhang, Dengyuan Yue and Xiaowei Chen
Biomolecules 2025, 15(9), 1227; https://doi.org/10.3390/biom15091227 - 26 Aug 2025
Viewed by 777
Abstract
Cancer stem cells (CSCs), a subpopulation of tumor cells endowed with self-renewal capacity, drive cancer initiation and progression. While long non-coding RNAs (lncRNAs) are increasingly recognized as critical regulators of CSC stemness, their specific roles in gastric cancer stem cells (GCSCs) remain poorly [...] Read more.
Cancer stem cells (CSCs), a subpopulation of tumor cells endowed with self-renewal capacity, drive cancer initiation and progression. While long non-coding RNAs (lncRNAs) are increasingly recognized as critical regulators of CSC stemness, their specific roles in gastric cancer stem cells (GCSCs) remain poorly understood. This study investigates the functional significance of lncRNA TSPEAR-AS2 in modulating GCSC properties and uncovers its underlying molecular mechanisms. Through integrated whole-transcriptome sequencing, bioinformatics analysis, and validation in 48 paired gastric cancer tissues and adjacent normal tissues, TSPEAR-AS2 was identified as a differentially expressed lncRNA upregulated in both GCSCs and tumor samples. Functional experiments revealed that TSPEAR-AS2 overexpression significantly enhanced GCSC sphere-forming ability, proliferation, cell cycle progression, epithelial–mesenchymal transition (EMT), and expression of stemness markers (CD54, CD44, OCT4, NANOG, and SOX2) while suppressing apoptosis. Conversely, TSPEAR-AS2 knockdown attenuated these malignant phenotypes. In vivo tumorigenicity assays in nude mice further confirmed that TSPEAR-AS2 promotes tumor growth, with overexpression accelerating and knockdown inhibiting tumor formation. Mechanistically, bioinformatics predictions and dual-luciferase reporter assays established TSPEAR-AS2 as a competing endogenous RNA (ceRNA) that sponges miR-15a-5p, thereby derepressing the miR-15a-5p target gene CCND1. Rescue experiments demonstrated that overexpression of miR-15a-5p phenocopied TSPEAR-AS2 knockdown, reducing GCSC stemness, while miR-15a-5p inhibition rescued the effects of TSPEAR-AS2 suppression. Collectively, these findings reveal a novel TSPEAR-AS2/miR-15a-5p/CCND1 regulatory axis that sustains GCSC stemness and tumorigenicity. These results highlight TSPEAR-AS2 as a potential therapeutic target for eradicating gastric cancer stem cells and improving clinical outcomes. Full article
(This article belongs to the Section Molecular Genetics)
Show Figures

Figure 1

14 pages, 2017 KB  
Article
The S2 Glycoprotein Subunit Determines Intestinal Tropism in Infectious Bronchitis Virus
by Zhenkai Dai, Jing Zhang, Ying Huang, Benli Huang, Zhengzhong Xiao, Keyu Feng, Guanming Shao, Xinheng Zhang and Qingmei Xie
Microorganisms 2025, 13(8), 1918; https://doi.org/10.3390/microorganisms13081918 - 17 Aug 2025
Viewed by 614
Abstract
The molecular basis for the distinct intestinal tropism of infectious bronchitis virus (IBV) strains remains poorly understood. This study identifies the S2 subunit of the spike glycoprotein as the critical determinant conferring duodenal tropism to the IBV CSL strain. Comparative pathogenesis in specific-pathogen-free [...] Read more.
The molecular basis for the distinct intestinal tropism of infectious bronchitis virus (IBV) strains remains poorly understood. This study identifies the S2 subunit of the spike glycoprotein as the critical determinant conferring duodenal tropism to the IBV CSL strain. Comparative pathogenesis in specific-pathogen-free (SPF) chicks revealed that the CSL strain achieved significantly higher viral titers in the duodenum compared to strains D90, PYG QX1, and XXX QX5. This duodenal replication was associated with severe epithelial inflammation, characterized by upregulation of pro-inflammatory cytokines (IL-6, IL-17A, IL-22, TNF-α, IFN-β, IFN-γ) and disruption of barrier integrity via downregulation of tight junction proteins (Occludin, Claudin-1, ZO-1). Crucially, reverse genetics using the non-enterotropic D90 backbone demonstrated that recombinant viruses carrying the CSL-S2 gene (rD90-ΔS/CSL and rD90-ΔS2/CSL), but not those carrying CSL-S1 (rD90-ΔS1/CSL), replicated efficiently and induced inflammation in the duodenum, phenocopying wild-type CSL. In contrast, renal tropism was independent of the S2 subunit. These findings establish the S2 subunit as both necessary and sufficient for IBV duodenal tropism, uncoupling it from renal pathogenicity. This identifies S2 as a prime molecular target for developing next-generation vaccines against intestinal IBV pathotypes. Full article
(This article belongs to the Special Issue Animal Viral Infectious Diseases)
Show Figures

Figure 1

21 pages, 1699 KB  
Review
Cardiac Hypertrophy: A Comprehensive Review from Prenatal Life to Young Adulthood
by Martina Avesani, Elettra Pomiato, Sara Moscatelli, Jolanda Sabatino, Nunzia Borrelli, Leonie Luedke, Rosalba De Sarro, Sara Pavesi, Giulia Pelaia, Claudio Mastellone, Isabella Leo and Giovanni Di Salvo
Children 2025, 12(8), 989; https://doi.org/10.3390/children12080989 - 28 Jul 2025
Viewed by 1437
Abstract
Myocardial hypertrophy (MH) represents a complex and heterogeneous condition in the pediatric and young adult population. While rare in children, MH encompasses a wide spectrum of physiological and pathological entities, ranging from transient hypertrophy in the infants of diabetic mothers to progressive genetic [...] Read more.
Myocardial hypertrophy (MH) represents a complex and heterogeneous condition in the pediatric and young adult population. While rare in children, MH encompasses a wide spectrum of physiological and pathological entities, ranging from transient hypertrophy in the infants of diabetic mothers to progressive genetic hypertrophic cardiomyopathies (HCM) with significant morbidity and mortality. Differential diagnosis is critical, as many phenocopies—including metabolic, mitochondrial, and syndromic diseases—can mimic HCM. Echocardiography remains the first-line imaging modality, with cardiac magnetic resonance (CMR) and molecular diagnostics increasingly used for detailed characterization. Risk stratification tools, such as the HCM Risk-Kids model, support clinical decision-making but must be integrated with individualized assessment. Advances in prenatal screening and genetic testing have significantly improved outcomes, though long-term management requires multidisciplinary care. Understanding age-specific presentations and the underlying etiologies is essential for accurate diagnosis and targeted treatment. This review provides a comprehensive overview of cardiac hypertrophy from fetal life through young adulthood, with a focus on etiologies, diagnostic approaches, imaging modalities, and therapeutic strategies, and aims to guide clinicians through the evolving landscape of MH, emphasizing early recognition, comprehensive evaluation, and personalized care. Full article
(This article belongs to the Special Issue Evaluation and Management of Children with Congenital Heart Disease)
Show Figures

Figure 1

18 pages, 3640 KB  
Article
NsrR Represses σE-Dependent Small RNAs and Interacts with RpoE via a Noncanonical Mechanism in Escherichia coli
by Joseph I. Aubee, Jalisa Nurse, Dale Lewis, Chin-Hsien Tai and Karl M. Thompson
Int. J. Mol. Sci. 2025, 26(13), 6318; https://doi.org/10.3390/ijms26136318 - 30 Jun 2025
Viewed by 589
Abstract
The envelope stress response in Escherichia coli is primarily governed by the sigma factor RpoE (σE), which activates protective genes upon membrane perturbation. Under non-stress conditions, σE is sequestered by its anti-sigma factor RseA. In this study, we identify an [...] Read more.
The envelope stress response in Escherichia coli is primarily governed by the sigma factor RpoE (σE), which activates protective genes upon membrane perturbation. Under non-stress conditions, σE is sequestered by its anti-sigma factor RseA. In this study, we identify an unexpected role for the nitric-oxide-sensing repressor NsrR in dampening σE activity and repressing σE-dependent small RNAs, including rybB, micA, and micL. Overexpression of nsrR represses transcription from σE-dependent promoters and phenocopies σE inactivation, resulting in filamentous morphology and growth defects. Conversely, ΔnsrR de-represses σE targets, with additive effects in rseA mutants—supporting an RseA-independent regulatory role. Time-course analysis shows NsrR represses σE activity, with kinetics comparable to those of RseA. While in vitro assays failed to detect robust NsrR binding to σE target promoters, NsrR directly interacts with σE in bacterial two-hybrid assays. Structural modeling using AlphaFold3 supports a plausible NsrR–RpoE interaction interface. These findings suggest that NsrR functions as a noncanonical anti-sigma-like modulator of σE, integrating redox and envelope stress signals to maintain membrane homeostasis. Full article
Show Figures

Figure 1

20 pages, 1641 KB  
Review
Hypertrophic Cardiomyopathy and Phenocopies: New Therapies for Old Diseases—Current Evidence and Future Perspectives
by Maria Alfarano, Federico Ciccarelli, Giulia Marchionni, Federico Ballatore, Jacopo Costantino, Antonio Lattanzio, Giulia Pecci, Silvia Stavagna, Leonardo Iannelli, Gioacchino Galardo, Carlo Lavalle, Fabio Miraldi, Carmine Dario Vizza and Cristina Chimenti
J. Clin. Med. 2025, 14(12), 4228; https://doi.org/10.3390/jcm14124228 - 13 Jun 2025
Viewed by 1453
Abstract
The hypertrophic cardiomyopathy (HCM) clinical phenotype includes sarcomeric HCM, which is the most common form of inherited cardiomyopathy with a population prevalence of 1:500, and phenocopies such as cardiac amyloidosis and Anderson–Fabry disease, which are considered rare diseases. Identification of cardiac and non-cardiac [...] Read more.
The hypertrophic cardiomyopathy (HCM) clinical phenotype includes sarcomeric HCM, which is the most common form of inherited cardiomyopathy with a population prevalence of 1:500, and phenocopies such as cardiac amyloidosis and Anderson–Fabry disease, which are considered rare diseases. Identification of cardiac and non-cardiac red flags in the context of multi-organ syndrome, multimodality imaging, including echocardiography, cardiac magnetic resonance, and genetic testing, has a central role in the diagnostic pathway. Identifying the specific disease underlying the hypertrophic phenotype is very important since many disease-modifying therapies are currently available, and phase 3 trials for new treatments have been completed or are ongoing. In particular, many chemotherapy agents (alkylating agents, proteasome inhibitors, immunomodulatory drugs, and monoclonal antibodies targeting clonal cells) allowing one to treat AL amyloidosis, transthyretin stabilizers (tafamidis and acoramidis), and gene silencers (patisiran and vutrisiran) are available in transthyretin cardiac amyloidosis, and enzyme replacement therapies (agalsidase-alpha, agalsidase-beta, and pegunigalsidase-alpha) or oral chaperone therapy (migalastat) can be used in Anderson–Fabry disease. In addition, the introduction of cardiac myosin inhibitors (mavacamten and aficamten) has deeply modified the treatment of hypertrophic obstructive cardiomyopathy. The aim of this review is to describe the new disease-modifying treatments available in HCM and phenocopies in light of current scientific evidence. Full article
(This article belongs to the Special Issue What’s New in Cardiomyopathies: Diagnosis, Treatment and Management)
Show Figures

Graphical abstract

20 pages, 7340 KB  
Article
PTPA Governs Stress-Responsive Differentiation and Metabolic Homeostasis in Toxoplasma gondii
by Zhu Ying, Yuntong Wu, Yanqun Pei, Zheng Shang, Jing Liu and Qun Liu
Cells 2025, 14(11), 835; https://doi.org/10.3390/cells14110835 - 3 Jun 2025
Viewed by 855
Abstract
The protozoan parasite Toxoplasma gondii transitions between acute (tachyzoite) and chronic (bradyzoite) stages, enabling lifelong persistence in hosts. Iron depletion triggers bradyzoite differentiation, with the phosphotyrosyl phosphatase activator (PTPA) identified as a key regulator. Here, we define PTPA’s role in T. gondii pathogenesis. [...] Read more.
The protozoan parasite Toxoplasma gondii transitions between acute (tachyzoite) and chronic (bradyzoite) stages, enabling lifelong persistence in hosts. Iron depletion triggers bradyzoite differentiation, with the phosphotyrosyl phosphatase activator (PTPA) identified as a key regulator. Here, we define PTPA’s role in T. gondii pathogenesis. PTPA forms a ternary complex with PP2A A/C subunits, validated by reciprocal pull-down assays. Depleting PTPA impaired tachyzoite proliferation, invasion, and gliding motility, while stress-induced bradyzoites exhibited defective cyst formation and vacuolar swelling. Metabolic dysregulation included amylopectin accumulation and lipid droplet proliferation. The PP2A inhibitor LB-100 phenocopied PTPA depletion, suppressing tachyzoite growth and bradyzoite differentiation. TgPTPA emerges as a linchpin coordinating PP2A activity, metabolic flux, and lifecycle transitions. Its dual roles in acute virulence and chronic persistence, combined with LB-100’s efficacy, position the PTPA–PP2A axis as a promising target for antitoxoplasmosis strategies. Full article
Show Figures

Figure 1

53 pages, 1175 KB  
Review
Revisiting the Pathogenesis of X-Linked Adrenoleukodystrophy
by Pierre Bougnères and Catherine Le Stunff
Genes 2025, 16(5), 590; https://doi.org/10.3390/genes16050590 - 17 May 2025
Cited by 3 | Viewed by 3723
Abstract
Background: X-ALD is a white matter (WM) disease caused by mutations in the ABCD1 gene encoding the transporter of very-long-chain fatty acids (VLCFAs) into peroxisomes. Strikingly, the same ABCD1 mutation causes either devastating brain inflammatory demyelination during childhood or, more often, progressive spinal [...] Read more.
Background: X-ALD is a white matter (WM) disease caused by mutations in the ABCD1 gene encoding the transporter of very-long-chain fatty acids (VLCFAs) into peroxisomes. Strikingly, the same ABCD1 mutation causes either devastating brain inflammatory demyelination during childhood or, more often, progressive spinal cord axonopathy starting in middle-aged adults. The accumulation of undegraded VLCFA in glial cell membranes and myelin has long been thought to be the central mechanism of X-ALD. Methods: This review discusses studies in mouse and drosophila models that have modified our views of X-ALD pathogenesis. Results: In the Abcd1 knockout (KO) mouse that mimics the spinal cord disease, the late manifestations of axonopathy are rapidly reversed by ABCD1 gene transfer into spinal cord oligodendrocytes (OLs). In a peroxin-5 KO mouse model, the selective impairment of peroxisomal biogenesis in OLs achieves an almost perfect phenocopy of cerebral ALD. A drosophila knockout model revealed that VLCFA accumulation in glial myelinating cells causes the production of a toxic lipid able to poison axons and activate inflammatory cells. Other mouse models showed the critical role of OLs in providing energy substrates to axons. In addition, studies on microglial changing substates have improved our understanding of neuroinflammation. Conclusions: Animal models supporting a primary role of OLs and axonal pathology and a secondary role of microglia allow us to revisit of X-ALD mechanisms. Beyond ABCD1 mutations, pathogenesis depends on unidentified contributors, such as genetic background, cell-specific epigenomics, potential environmental triggers, and stochasticity of crosstalk between multiple cell types among billions of glial cells and neurons. Full article
(This article belongs to the Section Molecular Genetics and Genomics)
Show Figures

Figure 1

15 pages, 1917 KB  
Article
Clinical Relevance of IFT140 Loss-of-Function Variants in Development of Renal Cysts
by Carlotta Pia Cristalli, Sara Calabrese, Luca Caramanna, Andrea Pietra, Giulia Vitetta, Bianca De Nicolo, Elena Bonora, Giulia Severi, Soara Menabò, Simona Ferrari, Francesca Ciurli, Valeria Aiello, Irene Capelli, Andrea Pasini, Irene Alberici, Roberto Pillon, Claudio La Scola, Cesare Rossi, Francesca Montanari and Claudio Graziano
Genes 2025, 16(5), 472; https://doi.org/10.3390/genes16050472 - 22 Apr 2025
Viewed by 1082
Abstract
Background: Autosomal dominant polycystic kidney disease (ADPKD) is the most common inherited kidney disease, affecting approximately 1 in 1000 individuals. This genetically heterogeneous condition is primarily caused by monoallelic pathogenic or likely pathogenic variants in the PKD1 and PKD2 genes, accounting for 78% [...] Read more.
Background: Autosomal dominant polycystic kidney disease (ADPKD) is the most common inherited kidney disease, affecting approximately 1 in 1000 individuals. This genetically heterogeneous condition is primarily caused by monoallelic pathogenic or likely pathogenic variants in the PKD1 and PKD2 genes, accounting for 78% and 15% of typical cases, respectively. Recently, the application of NGS methods has led to the identification of additional genes associated with ADPKD, which have been incorporated into routine diagnostic testing for detecting phenocopies of the disease. Methods: In this study, targeted NGS (tNGS) analysis of the main cystogenes associated with classic and atypical ADPKD was performed in a cohort of 218 patients clinically diagnosed with cystic nephropathies. Results: Genetic testing identified variants in 175 out of 218 cases (80.3%). Among these, 133 probands (76%) harbored likely pathogenic or pathogenic variants in one or more genes of the panel, while 42 individuals (24%) had a variant of unknown significance (VUS). Specifically, one or more class 4/5 variants in PKD1, PKD2, or both were identified in 111 (83.5%) probands. Remarkably, a pathogenic variant in the IFT140 gene was identified in 14 index cases (8% of positive individuals, 6.4% of the global cohort): 10 distinct loss-of-function (LoF) variants were identified (including four frameshift variants, four nonsense variants, and two splice site defects); one individual carried a second IFT140 missense variant classified as VUS. Furthermore, five affected family members were found to carry a P/LP LoF variant in IFT140. Conclusions: Our data support that IFT140 heterozygous IFT140 LoF variants result in an atypical, mild form of ADPKD, consisting of bilateral kidney cysts and renal functional decline at older ages. Furthermore, we describe the second pediatric patient with a mild form of ADPKD due to an IFT140 variant and discuss hyperuricemia as a previously unappreciated feature of this condition. Full article
(This article belongs to the Section Human Genomics and Genetic Diseases)
Show Figures

Figure 1

18 pages, 8249 KB  
Article
Extracts from Allium pseudojaponicum Makino Target STAT3 Signaling Pathway to Overcome Cisplatin Resistance in Lung Cancer
by Soo-Bin Nam, Jung Hoon Choi, Ga-Eun Lee, Jin Young Kim, Mee-Hyun Lee, Gabsik Yang, Yong-Yeon Cho, Hye Gwang Jeong, Geul Bang and Cheol-Jung Lee
Mar. Drugs 2025, 23(4), 167; https://doi.org/10.3390/md23040167 - 14 Apr 2025
Cited by 1 | Viewed by 1089
Abstract
Lung cancer, particularly non-small-cell lung cancer (NSCLC), remains a leading cause of cancer-related mortality, with cisplatin-based chemotherapy being a standard treatment. However, the development of chemoresistance significantly limits its efficacy, necessitating alternative therapeutic approaches. Here, we demonstrate the anticancer effects of the extracts [...] Read more.
Lung cancer, particularly non-small-cell lung cancer (NSCLC), remains a leading cause of cancer-related mortality, with cisplatin-based chemotherapy being a standard treatment. However, the development of chemoresistance significantly limits its efficacy, necessitating alternative therapeutic approaches. Here, we demonstrate the anticancer effects of the extracts of Allium pseudojaponicum Makino (APE), a salt-tolerant plant, in cisplatin-resistant NSCLC. Metabolite profiling using UPLC-Q-TOF-MSE identified 13 major compounds, predominantly alkaloids (71.65%) and flavonoids (8.81%), with key bioactive constituents such as lycorine (29.81%), tazettine (17.22%), and tricetin (8.19%). APE significantly inhibited cell viability in A549 and H460 cells, reducing viability to 38.6% (A549-Ctr), 37.2% (A549-CR), 28.4% (H460-Ctr), and 30.4% (H460-CR) at 40 µg/mL after 48 h. APE also suppressed colony formation by over 90% in both 2D and soft agar assays, while showing no cytotoxicity in normal human keratinocytes up to 80 µg/mL. Flow cytometry analysis revealed APE-induced G1 phase arrest, with the G1 population increasing from 50.4% to 56.6% (A549-Ctr) and 47.5% to 58.4% (A549-CR), accompanied by reduced S phase populations. This effect was associated with the downregulation of G1/S transition regulators, including cyclin D1, CDK4, cyclin E, and CDK2. Furthermore, proteomic analysis identified STAT3 signaling as a major target of APE; APE decreased phosphorylated STAT3 and c-Myc expression, and STAT3 knockdown phenocopied the effects of APE. These findings highlight the potential of APE as a natural product-based therapeutic strategy for overcoming cisplatin resistance in NSCLC. Full article
(This article belongs to the Special Issue Marine Natural Products as Anticancer Agents, 4th Edition)
Show Figures

Graphical abstract

14 pages, 6033 KB  
Article
Ivosidenib Confers BRCAness Phenotype and Synthetic Lethality to Poly (ADP-Ribose) Polymerase Inhibition in BRCA1/2-Proficient Cancer Cells
by Danyang Zhou, Wei Liu, Yanyan Zhang and Chong Li
Biomedicines 2025, 13(4), 958; https://doi.org/10.3390/biomedicines13040958 - 14 Apr 2025
Viewed by 1196
Abstract
Background/Objectives: PARP inhibitors (PARPi) are pivotal to treating homologous recombination repair-deficient (HRD) cancers, particularly BRCA1/2-mutated ovarian and breast cancers. However, most ovarian and breast cancers harbor wild-type (WT) BRCA1/2, limiting PARPi eligibility. This study aims to identify an approved drug [...] Read more.
Background/Objectives: PARP inhibitors (PARPi) are pivotal to treating homologous recombination repair-deficient (HRD) cancers, particularly BRCA1/2-mutated ovarian and breast cancers. However, most ovarian and breast cancers harbor wild-type (WT) BRCA1/2, limiting PARPi eligibility. This study aims to identify an approved drug that could induce a BRCAness phenotype, thereby sensitizing WT BRCA cancers to PARPi. Methods: Ovarian and breast cancer cell lines with WT BRCA1/2 were treated with ivosidenib. HR repair efficiency was assessed via RAD51 foci formation and reporter assays. Synthetic lethality with PARPi was evaluated using viability and colony formation assays. Mechanistic studies included RNA-binding protein pulldown, co-immunoprecipitation, and functional analyses of DNA repair pathways. YTHDC2′s role in HR was investigated through siRNA knockdown and rescue experiments. Results: Ivosidenib significantly reduced HR repair efficiency and sensitized cells to PARPi, inducing synthetic lethality. Mechanistically, ivosidenib directly bound YTHDC2, an m6A reader critical for HR. This interaction disrupted YTHDC2′s ability to promote DNA double-strand break repair via HR, evidenced by impaired recruitment of repair proteins (e.g., BRCA1, RAD51) and accumulation of DNA damage (γH2AX foci). YTHDC2 knockdown phenocopied ivosidenib effects, while overexpression rescued HR defects. Conclusions: Ivosidenib induces BRCAness in WT BRCA ovarian and breast cancers by targeting YTHDC2, thereby suppressing HR repair and enhancing PARPi sensitivity. This uncovers a novel, metabolism-independent mechanism of ivosidenib, repositioning it as a therapeutic agent for HRD tumors. These findings propose a strategy to expand PARPi eligibility to WT BRCA cancers, addressing a critical unmet need in oncology. Full article
(This article belongs to the Section Gene and Cell Therapy)
Show Figures

Figure 1

17 pages, 3168 KB  
Article
Notch Is Required for Neural Progenitor Proliferation During Embryonic Eye Regrowth
by Dylan J. Guerin, Belen Gutierrez, Baoyi Zhang and Kelly Ai-Sun Tseng
Int. J. Mol. Sci. 2025, 26(6), 2637; https://doi.org/10.3390/ijms26062637 - 14 Mar 2025
Cited by 1 | Viewed by 1240
Abstract
The ability of an organism to regrow tissues is regulated by various signaling pathways. One such pathway that has been studied widely both in the context of regeneration and development is the Notch signaling pathway. Notch is required for the development of the [...] Read more.
The ability of an organism to regrow tissues is regulated by various signaling pathways. One such pathway that has been studied widely both in the context of regeneration and development is the Notch signaling pathway. Notch is required for the development of the eye and regeneration of tissues in multiple organisms, but it is unknown if Notch plays a role in the regulation of Xenopus laevis embryonic eye regrowth. We found that Notch1 is required for eye regrowth and regulates retinal progenitor cell proliferation. Chemical and molecular inhibition of Notch1 significantly decreased eye regrowth by reducing retinal progenitor cell proliferation without affecting retinal differentiation. Temporal inhibition studies showed that Notch function is required during the first day of regrowth. Interestingly, Notch1 loss-of-function phenocopied the effects of the inhibition of the proton pump, vacuolar-type ATPase (V-ATPase), where retinal proliferation but not differentiation was blocked during eye regrowth. Overexpression of a form of activated Notch1, the Notch intracellular domain (NICD) rescued the loss of eye regrowth due to V-ATPase inhibition. These findings highlight the importance of the Notch signaling pathway in eye regeneration and its role in inducing retinal progenitor cell proliferation in response to injury. Full article
(This article belongs to the Special Issue Notch Signaling Pathways)
Show Figures

Figure 1

Back to TopTop