Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26.081
Filter
1.
PLoS One ; 19(7): e0304194, 2024.
Article in English | MEDLINE | ID: mdl-38968186

ABSTRACT

Deficiencies in the ING4 tumor suppressor are associated with advanced stage tumors and poor patient survival in cancer. ING4 was shown to inhibit NF-kB in several cancers. As NF-kB is a key mediator of immune response, the ING4/NF-kB axis is likely to manifest in tumor-immune modulation but has not been investigated. To characterize the tumor immune microenvironment associated with ING4-deficient tumors, three approaches were employed in this study: First, tissue microarrays composed of 246 primary breast tumors including 97 ING4-deficient tumors were evaluated for the presence of selective immune markers, CD68, CD4, CD8, and PD-1, using immunohistochemical staining. Second, an immune-competent mouse model of ING4-deficient breast cancer was devised utilizing CRISPR-mediated deletion of Ing4 in a Tp53 deletion-derived mammary tumor cell line; mammary tumors were evaluated for immune markers using flow cytometry. Lastly, the METABRIC gene expression dataset was evaluated for patient survival related to the immune markers associated with Ing4-deleted tumors. The results showed that CD68, CD4, CD8, or PD-1, was not significantly associated with ING4-deficient breast tumors, indicating no enrichment of macrophages, T cells, or exhausted T cell types. In mice, Ing4-deleted mammary tumors had a growth rate comparable to Ing4-intact tumors but showed increased tumor penetrance and metastasis. Immune marker analyses of Ing4-deleted tumors revealed a significant increase in tumor-associated macrophages (Gr-1loCD11b+F4/80+) and a decrease in granzyme B-positive (GzmB+) CD4+ T cells, indicating a suppressive and/or less tumoricidal immune microenvironment. The METABRIC data analyses showed that low expression of GZMB was significantly associated with poor patient survival, as was ING4-low expression, in the basal subtype of breast cancer. Patients with GZMB-low/ING4-low tumors had the worst survival outcomes (HR = 2.80, 95% CI 1.36-5.75, p = 0.0004), supportive of the idea that the GZMB-low immune environment contributes to ING4-deficient tumor progression. Collectively, the study results demonstrate that ING4-deficient tumors harbor a microenvironment that contributes to immune evasion and metastasis.


Subject(s)
Breast Neoplasms , Cell Cycle Proteins , Homeodomain Proteins , Tumor Microenvironment , Tumor Suppressor Proteins , Tumor Microenvironment/immunology , Animals , Breast Neoplasms/pathology , Breast Neoplasms/immunology , Breast Neoplasms/genetics , Female , Humans , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/metabolism , Mice , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Cell Line, Tumor , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/deficiency , Disease Progression , Neoplasm Metastasis
2.
Biochemistry (Mosc) ; 89(6): 987-1001, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38981695

ABSTRACT

The evolution of major taxa is often associated with the emergence of new gene families. In all multicellular animals except sponges and comb jellies, the genomes contain Hox genes, which are crucial regulators of development. The canonical function of Hox genes involves colinear patterning of body parts in bilateral animals. This general function is implemented through complex, precisely coordinated mechanisms, not all of which are evolutionarily conserved and fully understood. We suggest that the emergence of this regulatory complexity was preceded by a stage of cooperation between more ancient morphogenetic programs or their individual elements. Footprints of these programs may be present in modern animals to execute non-canonical Hox functions. Non-canonical functions of Hox genes are involved in maintaining terminal nerve cell specificity, autophagy, oogenesis, pre-gastrulation embryogenesis, vertical signaling, and a number of general biological processes. These functions are realized by the basic properties of homeodomain protein and could have triggered the evolution of ParaHoxozoa and Nephrozoa subsequently. Some of these non-canonical Hox functions are discussed in our review.


Subject(s)
Genes, Homeobox , Animals , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Multigene Family , Humans , Evolution, Molecular , Gene Expression Regulation, Developmental
4.
FASEB J ; 38(14): e23771, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-38989564

ABSTRACT

DUX4 has been widely reported in facioscapulohumeral muscular dystrophy, but its role in Duchenne muscular dystrophy (DMD) is unclear. Dux is the mouse paralog of DUX4. In Dux-/- mdx mice, forelimb grip strength test and treadmill test were performed, and extensor digitorum longus (EDL) contraction properties were measured to assess skeletal muscle function. Pathological changes in mice were determined by serum CK and LDH levels and muscle Masson staining. Inflammatory factors, oxidative stress, and mitochondrial function indicators were detected using kits. Primary muscle satellite cells were isolated, and the antioxidant molecule Nrf2 was detected. MTT assay and Edu assay were used to evaluate proliferation and TUNEL assay for cell death. The results show that the deletion of Dux enhanced forelimb grip strength and EDL contractility, prolonged running time and distance in mdx mice. Deleting Dux also attenuated muscle fibrosis, inflammation, oxidative stress, and mitochondrial dysfunction in mdx mice. Furthermore, Dux deficiency promoted proliferation and survival of muscle satellite cells by increasing Nrf2 levels in mdx mice.


Subject(s)
Homeodomain Proteins , Mice, Inbred mdx , Muscular Dystrophy, Duchenne , NF-E2-Related Factor 2 , Oxidative Stress , Animals , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Mice , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Male , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Satellite Cells, Skeletal Muscle/metabolism , Mice, Inbred C57BL , Mice, Knockout , Gene Deletion
5.
Medicine (Baltimore) ; 103(28): e38867, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38996143

ABSTRACT

BACKGROUND: Lung adenocarcinoma (LUAD) represents the most prevalent type of lung cancer. SHOX2 and RASSF1A methylation have been identified as important biomarkers for diagnosis and prognosis of lung cancer. Bronchoalveolar lavage fluid (BALF) exhibits good specificity and sensitivity in diagnosing pulmonary diseases, but its acquisition is challenging and may cause discomfort to patients. In clinical, plasma samples are more convenient to obtain than BALF; however, there is little research on the concurrent detection of SHOX2 and RASSF1A methylation in plasma. This study aims to assess the diagnostic value of a combined promoter methylation assay for SHOX2 and RASSF1A in early-stage LUAD using plasma samples. METHODS: BALF and blood samples were obtained from 36 early-stage LUAD patients, with a control group of nineteen non-tumor individuals. The promoter methylation levels of SHOX2 and RASSF1A in all subjects were assessed using the human SHOX2 and RASSF1A gene methylation kit. RESULTS: The methylation detection rate of SHOX2 and RASSF1A in plasma was 61.11%, slightly lower than that in BALF (66.7%). The Chi-square test revealed no significant difference in the methylation rate between BALF and plasma (P > 0.05). The area under the receiver operating characteristic (ROC) curve analysis for blood was 0.806 (95% CI, 0.677 to 0.900), while for BALF it was 0.781 (95% CI, 0.649 to 0.881). Additionally, we conducted an analysis on the correlation between SHOX2 and RASSF1A methylation levels in plasma with gender, age, tumor differentiation, pathologic classification, and other clinicopathological variables; however, no significant correlations were observed. CONCLUSIONS: Measurement of SHOX2 and RASSF1A methylation for early diagnosis of LUAD can be achieved with high sensitivity and specificity by using plasma as a substitute for BALF samples.


Subject(s)
Adenocarcinoma of Lung , Biomarkers, Tumor , DNA Methylation , Early Detection of Cancer , Homeodomain Proteins , Lung Neoplasms , Promoter Regions, Genetic , Tumor Suppressor Proteins , Humans , Male , Female , Middle Aged , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/blood , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/diagnosis , Adenocarcinoma of Lung/blood , Lung Neoplasms/diagnosis , Lung Neoplasms/genetics , Lung Neoplasms/blood , Early Detection of Cancer/methods , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Aged , Homeodomain Proteins/genetics , Homeodomain Proteins/blood , Bronchoalveolar Lavage Fluid/chemistry , ROC Curve , Adult , Sensitivity and Specificity , Case-Control Studies
7.
J Physiol Pharmacol ; 75(3)2024 Jun.
Article in English | MEDLINE | ID: mdl-39042388

ABSTRACT

Allyl isothiocyanate (AITC) is the pungent ingredient of brassica species, used as a food additive and flavoring agent, including condiments such as wasabi, horseradish, and mustard. Currently, there is much evidence that AITC modulates glucose and lipids metabolism. Interestingly, AITC has been shown to improve glycaemia, and insulin action along with the induction of a deepened decline in blood insulin levels in T2DM rats. Therefore, in the present study, we characterized the role of AITC at a wide concentration range (5, 10, 25, 50, 100 µM) in controlling viability, proliferation, apoptosis, mitochondrial condition, mRNA expression of encoding pancreatic and duodenal homeobox 1 (Pdx1), and Ins1, Ins2 genes, and insulin content in INS-1E cells. The INS-1E cell line is a suitable, and well-characterized model to study beta cell functions. We demonstrate that AITC reduced the viability (p≤0.001) (also in the presence of transient receptor potential cation subfamily A member 1 (TRPA1) selective antagonist; HC-030031; p≤0.05), and proliferation of INS-1E cells (p≤0.001). AITC evoked a significant reduction of mitochondrial membrane potential (p≤0.01) and decreased the intracellular level of adenosine triphosphate (ATP) (p≤0.001) without influence on reactive oxygen species (ROS) level. Additionally, AITC inhibited the insulin mRNA expression (p≤0.001) in INS-1E cells along with insulin content (p≤0.05). Mitochondrial dysfunction is proposed to be a significant disruption mechanism of AITC in INS-1E cells, and it was independent of ROS, and the influx of external calcium.


Subject(s)
Cell Proliferation , Cell Survival , Insulin-Secreting Cells , Insulin , Isothiocyanates , Membrane Potential, Mitochondrial , Animals , Isothiocyanates/pharmacology , Rats , Insulin/metabolism , Membrane Potential, Mitochondrial/drug effects , Cell Survival/drug effects , Cell Line , Cell Proliferation/drug effects , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Apoptosis/drug effects , Reactive Oxygen Species/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , TRPA1 Cation Channel , Trans-Activators
8.
Int J Mol Sci ; 25(13)2024 Jun 26.
Article in English | MEDLINE | ID: mdl-39000102

ABSTRACT

Aberrant expression of the double homeobox 4 (DUX4) gene in skeletal muscle predominantly drives the pathogenesis of facioscapulohumeral muscular dystrophy (FSHD). We recently demonstrated that berberine, an herbal extract known for its ability to stabilize guanine-quadruplex structures, effectively downregulates DUX4 expression in FSHD patient-derived myoblasts and in mice overexpressing exogenous DUX4 after viral vector-based treatment. Here, we sought to confirm berberine's inhibitory efficacy on DUX4 in the widely used FSHD-like transgenic mouse model, ACTA1-MCM/FLExDUX4, where DUX4 is induced at pathogenic levels using tamoxifen. Animals repeatedly treated with berberine via intraperitoneal injections for 4 weeks exhibited significant reductions in both mRNA and protein levels of DUX4, and in mRNA expression of murine DUX4-related genes. This inhibition translated into improved forelimb muscle strength and positive alterations in important FSHD-relevant cellular pathways, although its impact on muscle mass and histopathology was less pronounced. Collectively, our data confirm the efficacy of berberine in downregulating DUX4 expression in the most relevant FSHD mouse model. However, further optimization of dosing regimens and new studies to enhance the bioavailability of berberine in skeletal muscle are warranted to fully leverage its therapeutic potential for FSHD treatment.


Subject(s)
Berberine , Disease Models, Animal , Homeodomain Proteins , Mice, Transgenic , Muscle, Skeletal , Muscular Dystrophy, Facioscapulohumeral , Animals , Muscular Dystrophy, Facioscapulohumeral/drug therapy , Muscular Dystrophy, Facioscapulohumeral/metabolism , Muscular Dystrophy, Facioscapulohumeral/genetics , Muscular Dystrophy, Facioscapulohumeral/pathology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Muscle, Skeletal/metabolism , Muscle, Skeletal/drug effects , Berberine/pharmacology , Actins/metabolism , Actins/genetics , Humans
9.
Development ; 151(13)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38994775

ABSTRACT

Super-enhancers (SEs) are expansive regions of genomic DNA that regulate the expression of genes involved in cell identity and cell fate. We recently identified developmental stage- and cell type-specific modules within the murine Vsx2 SE. Here, we show that the human VSX2 SE modules have similar developmental stage- and cell type-specific activity in reporter gene assays. By inserting the human sequence of one VSX2 SE module into a mouse with microphthalmia, eye size was rescued. To understand the function of these SE modules during human retinal development, we deleted individual modules in human embryonic stem cells and generated retinal organoids. Deleting one module results in small organoids, recapitulating the small-eyed phenotype of mice with microphthalmia, while deletion of the other module led to disruptions in bipolar neuron development. This prototypical SE serves as a model for understanding developmental stage- and cell type-specific effects of neurogenic transcription factors with complex expression patterns. Moreover, by elucidating the gene regulatory mechanisms, we can begin to examine how dysregulation of these mechanisms contributes to phenotypic diversity and disease.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Homeodomain Proteins , Retina , Transcription Factors , Animals , Humans , Mice , Enhancer Elements, Genetic/genetics , Evolution, Molecular , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Microphthalmos/genetics , Microphthalmos/pathology , Neurogenesis/genetics , Organoids/metabolism , Retina/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics
10.
Nat Commun ; 15(1): 5894, 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39003281

ABSTRACT

Remarkable advances in protocol development have been achieved to manufacture insulin-secreting islets from human pluripotent stem cells (hPSCs). Distinct from current approaches, we devised a tunable strategy to generate islet spheroids enriched for major islet cell types by incorporating PDX1+ cell budding morphogenesis into staged differentiation. In this process that appears to mimic normal islet morphogenesis, the differentiating islet spheroids organize with endocrine cells that are intermingled or arranged in a core-mantle architecture, accompanied with functional heterogeneity. Through in vitro modelling of human pancreas development, we illustrate the importance of PDX1 and the requirement for EphB3/4 signaling in eliciting cell budding morphogenesis. Using this new approach, we model Mitchell-Riley syndrome with RFX6 knockout hPSCs illustrating unexpected morphogenesis defects in the differentiation towards islet cells. The tunable differentiation system and stem cell-derived islet models described in this work may facilitate addressing fundamental questions in islet biology and probing human pancreas diseases.


Subject(s)
Cell Differentiation , Homeodomain Proteins , Islets of Langerhans , Morphogenesis , Pluripotent Stem Cells , Spheroids, Cellular , Trans-Activators , Humans , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism , Trans-Activators/metabolism , Trans-Activators/genetics , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Signal Transduction , Receptors, Eph Family/metabolism , Receptors, Eph Family/genetics
11.
Open Biol ; 14(7): 240071, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38955222

ABSTRACT

The enzymatic breakdown and regulation of food passage through the vertebrate antral stomach and pyloric sphincter (antropyloric region) is a trait conserved over 450 million years. Development of the structures involved is underpinned by a highly conserved signalling pathway involving the hedgehog, bone morphogenetic protein and Wingless/Int-1 (Wnt) protein families. Monotremes are one of the few vertebrate lineages where acid-based digestion has been lost, and this is consistent with the lack of genes for hydrochloric acid secretion and gastric enzymes in the genomes of the platypus (Ornithorhynchus anatinus) and short-beaked echidna (Tachyglossus aculeatus) . Furthermore, these species feature unique gastric phenotypes, both with truncated and aglandular antral stomachs and the platypus with no pylorus. Here, we explore the genetic underpinning of monotreme gastric phenotypes, investigating genes important in antropyloric development using the newest monotreme genomes (mOrnAna1.pri.v4 and mTacAcu1) together with RNA-seq data. We found that the pathway constituents are generally conserved, but surprisingly, NK3 homeobox 2 (Nkx3.2) was pseudogenized in both platypus and echidna. We speculate that the unique sequence evolution of Grem1 and Bmp4 sequences in the echidna lineage may correlate with their pyloric-like restriction and that the convergent loss of gastric acid and stomach size genotypes and phenotypes in teleost and monotreme lineages may be a result of eco-evolutionary dynamics. These findings reflect the effects of gene loss on phenotypic evolution and further elucidate the genetic control of monotreme stomach anatomy and physiology.


Subject(s)
Stomach , Animals , Stomach/anatomy & histology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Platypus/genetics , Phylogeny , Evolution, Molecular
12.
Dokl Biochem Biophys ; 517(1): 259-263, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39002012

ABSTRACT

Nucleoli form interchromosomal contacts with genes controlling differentiation and carcinogenesis. DUX4 genes specify transcription factor possessing two homeodomains. Previously, using Circular Chromosome Conformation Capture (4С) approach on population of cells, it was demonstrated that DUX4 gene clusters form frequent contacts with nucleoli. It was found also that these contacts are almost completely abolished after heat shock treatment. 4C approach as all ligation-mediated methods is capable to detect rather close interactions between chromatin loops in nuclei. In order to independently confirm the formation and the frequency of the contacts in single cells we used FISH approach. Here, we show that DUX genes in single cells form stable contacts in all tested HEK293T cells. During heat shock, DUX4 genes reversibly move 1-3 µm away from the nuclei. We conclude that interchromosomal contacts formed by nucleoli are strong, dynamic, and reversible, providing both the initiation and maintenance of a differentiated state.


Subject(s)
Cell Nucleolus , Homeodomain Proteins , Humans , Cell Nucleolus/metabolism , Cell Nucleolus/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , HEK293 Cells , Chromosomes, Human/genetics , Chromosomes, Human/metabolism , In Situ Hybridization, Fluorescence
13.
Adv Exp Med Biol ; 1459: 405-430, 2024.
Article in English | MEDLINE | ID: mdl-39017854

ABSTRACT

HOXA9, an important transcription factor (TF) in hematopoiesis, is aberrantly expressed in numerous cases of both acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) and is a strong indicator of poor prognosis in patients. HOXA9 is a proto-oncogene which is both sufficient and necessary for leukemia transformation. HOXA9 expression in leukemia correlates with patient survival outcomes and response to therapy. Chromosomal transformations (such as NUP98-HOXA9), mutations, epigenetic dysregulation (e.g., MLL- MENIN -LEDGF complex or DOT1L/KMT4), transcription factors (such as USF1/USF2), and noncoding RNA (such as HOTTIP and HOTAIR) regulate HOXA9 mRNA and protein during leukemia. HOXA9 regulates survival, self-renewal, and progenitor cell cycle through several of its downstream target TFs including LMO2, antiapoptotic BCL2, SOX4, and receptor tyrosine kinase FLT3 and STAT5. This dynamic and multilayered HOXA9 regulome provides new therapeutic opportunities, including inhibitors targeting DOT1L/KMT4, MENIN, NPM1, and ENL proteins. Recent findings also suggest that HOXA9 maintains leukemia by actively repressing myeloid differentiation genes. This chapter summarizes the recent advances understanding biochemical mechanisms underlying HOXA9-mediated leukemogenesis, the clinical significance of its abnormal expression, and pharmacological approaches to treat HOXA9-driven leukemia.


Subject(s)
Gene Expression Regulation, Leukemic , Homeodomain Proteins , Nucleophosmin , Proto-Oncogene Mas , Humans , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Gene Expression Regulation, Leukemic/drug effects , Animals , Leukemia/genetics , Leukemia/metabolism , Leukemia/drug therapy , Leukemia/pathology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology
14.
Autoimmunity ; 57(1): 2364686, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38946534

ABSTRACT

BACKGROUND: Chondrocyte viability, apoptosis, and migration are closely related to cartilage injury in osteoarthritis (OA) joints. Exosomes are identified as potential therapeutic agents for OA. OBJECTIVE: This study aimed to investigate the role of exosomes derived from osteocytes in OA, particularly focusing on their effects on cartilage repair and molecular mechanisms. METHODS: An injury cell model was established by treating chondrocytes with IL-1ß. Cartilage repair was evaluated using cell counting kit-8, flow cytometry, scratch test, and Western Blot. Molecular mechanisms were analyzed using quantitative real-time PCR, bioinformatic analysis, and Western Blot. An OA mouse model was established to explore the role of exosomal DLX2 in vivo. RESULTS: Osteocyte-released exosomes promoted cell viability and migration, and inhibited apoptosis and extracellular matrix (ECM) deposition. Moreover, exosomes upregulated DLX2 expression, and knockdown of DLX2 activated the Wnt pathway. Additionally, exosomes attenuated OA in mice by transmitting DLX2. CONCLUSION: Osteocyte-derived exosomal DLX2 alleviated IL-1ß-induced cartilage repair and inactivated the Wnt pathway, thereby alleviating OA progression. The findings suggested that osteocyte-derived exosomes may hold promise as a treatment for OA.


Subject(s)
Chondrocytes , Exosomes , Homeodomain Proteins , Osteoarthritis , Osteocytes , Transcription Factors , Wnt Signaling Pathway , Exosomes/metabolism , Animals , Osteoarthritis/metabolism , Osteoarthritis/pathology , Mice , Transcription Factors/metabolism , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Osteocytes/metabolism , Chondrocytes/metabolism , Disease Models, Animal , Humans , Interleukin-1beta/metabolism , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Apoptosis , Cartilage/metabolism , Cartilage/pathology , Male , Cell Movement , Cell Survival
15.
Adv Exp Med Biol ; 1459: 97-113, 2024.
Article in English | MEDLINE | ID: mdl-39017841

ABSTRACT

Helix-loop-helix (HLH) transcription factors (TFs) play a key role in various cellular differentiation and function through the regulation of enhancer activity. E2A, a member of the mammalian E-protein family (class I HLH protein), is well known to play an important role in hematopoiesis, especially in adaptive lymphocyte development. E2A instructs B- and T-cell lineage development through the regulation of enhancer activity for B- or T-cell signature gene expression, including Rag1 and Rag2 (Rag1/2) genes. In this chapter, we mainly focus on the function of E2A in B-cell development and on the roles of E2A in establishing the enhancer landscape through the recruitment of EP300/KAT3B, chromatin remodeling complex, mediator, cohesion, and TET proteins. Finally, we demonstrate how E2A orchestrates the assembly of the Rag1/2 gene super-enhancer (SE) formation by changing the chromatin conformation across the Rag gene locus.


Subject(s)
B-Lymphocytes , Homeodomain Proteins , Humans , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Enhancer Elements, Genetic/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Chromatin Assembly and Disassembly , Cell Differentiation/genetics , Chromatin/metabolism , Chromatin/genetics , E1A-Associated p300 Protein/metabolism , E1A-Associated p300 Protein/genetics , DNA-Binding Proteins , Nuclear Proteins
16.
Protein Sci ; 33(8): e5106, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39012010

ABSTRACT

Miniproteins constitute an excellent basis for the development of structurally demanding functional molecules. The engrailed homeodomain, a three-helix-containing miniprotein, was applied as a scaffold for constructing programmed cell death protein 1/programmed death-ligand 1 (PD-1/PD-L1) interaction inhibitors. PD-L1 binders were initially designed using the computer-aided approach and subsequently optimized iteratively. The conformational stability was assessed for each obtained miniprotein using circular dichroism spectroscopy, indicating that numerous mutations could be introduced. The formation of a sizable hydrophobic surface at the inhibitor that fits the molecular target imposed the necessity for the incorporation of additional charged amino acid residues to retain its appropriate solubility. Finally, the miniprotein effectively binding to PD-L1 (KD = 51.4 nM) that inhibits PD-1/PD-L1 interaction in cell-based studies with EC50 = 3.9 µM, was discovered.


Subject(s)
B7-H1 Antigen , Programmed Cell Death 1 Receptor , Protein Engineering , Programmed Cell Death 1 Receptor/chemistry , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/genetics , B7-H1 Antigen/chemistry , B7-H1 Antigen/metabolism , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/genetics , Humans , Protein Binding , Models, Molecular , Homeodomain Proteins/chemistry , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics
17.
Sci Rep ; 14(1): 13912, 2024 06 17.
Article in English | MEDLINE | ID: mdl-38886487

ABSTRACT

DNA methylation is an epigenetic mark that plays an important role in defining cancer phenotypes, with global hypomethylation and focal hypermethylation at CpG islands observed in tumors. These methylation marks can also be used to define tumor types and provide an avenue for biomarker identification. The homeobox gene class is one that has potential for this use, as well as other genes that are Polycomb Repressive Complex 2 targets. To begin to unravel this relationship, we performed a pan-cancer DNA methylation analysis using sixteen Illumina HM450k array datasets from TCGA, delving into cancer-specific qualities and commonalities between tumor types with a focus on homeobox genes. Our comparisons of tumor to normal samples suggest that homeobox genes commonly harbor significant hypermethylated differentially methylated regions. We identified two homeobox genes, HOXA3 and HOXD10, that are hypermethylated in all 16 cancer types. Furthermore, we identified several potential homeobox gene biomarkers from our analysis that are uniquely methylated in only one tumor type and that could be used as screening tools in the future. Overall, our study demonstrates unique patterns of DNA methylation in multiple tumor types and expands on the interplay between the homeobox gene class and oncogenesis.


Subject(s)
DNA Methylation , Homeodomain Proteins , Neoplasms , Humans , Neoplasms/genetics , Homeodomain Proteins/genetics , Genes, Homeobox , Gene Expression Regulation, Neoplastic , Polycomb-Group Proteins/genetics , Polycomb-Group Proteins/metabolism , CpG Islands , Transcription Factors/genetics , Transcription Factors/metabolism , Epigenesis, Genetic , Biomarkers, Tumor/genetics
18.
Cell Death Dis ; 15(6): 420, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38886383

ABSTRACT

The regeneration of the mammalian skeleton's craniofacial bones necessitates the action of intrinsic and extrinsic inductive factors from multiple cell types, which function hierarchically and temporally to control the differentiation of osteogenic progenitors. Single-cell transcriptomics of developing mouse calvarial suture recently identified a suture mesenchymal progenitor population with previously unappreciated tendon- or ligament-associated gene expression profile. Here, we developed a Mohawk homeobox (MkxCG; R26RtdT) reporter mouse and demonstrated that this reporter identifies an adult calvarial suture resident cell population that gives rise to calvarial osteoblasts and osteocytes during homeostatic conditions. Single-cell RNA sequencing (scRNA-Seq) data reveal that Mkx+ suture cells display a progenitor-like phenotype with expression of teno-ligamentous genes. Bone injury with Mkx+ cell ablation showed delayed bone healing. Remarkably, Mkx gene played a critical role as an osteo-inhibitory factor in calvarial suture cells, as knockdown or knockout resulted in increased osteogenic differentiation. Localized deletion of Mkx in vivo also resulted in robustly increased calvarial defect repair. We further showed that mechanical stretch dynamically regulates Mkx expression, in turn regulating calvarial cell osteogenesis. Together, we define Mkx+ cells within the suture mesenchyme as a progenitor population for adult craniofacial bone repair, and Mkx acts as a mechanoresponsive gene to prevent osteogenic differentiation within the stem cell niche.


Subject(s)
Cell Differentiation , Homeodomain Proteins , Osteogenesis , Skull , Animals , Mice , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Osteogenesis/genetics , Skull/metabolism , Osteoblasts/metabolism , Osteoblasts/cytology , Cranial Sutures/metabolism , Stem Cells/metabolism , Stem Cells/cytology , Biomarkers/metabolism
19.
Cell Rep ; 43(6): 114372, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38878289

ABSTRACT

Emerging evidence highlights the regulatory role of paired-like (PRD-like) homeobox transcription factors (TFs) in embryonic genome activation (EGA). However, the majority of PRD-like genes are lost in rodents, thus prompting an investigation into PRD-like TFs in other mammals. Here, we showed that PRD-like TFs were transiently expressed during EGA in human, monkey, and porcine fertilized embryos, yet they exhibited inadequate expression in their cloned embryos. This study, using pig as the research model, identified LEUTX as a key PRD-like activator of porcine EGA through genomic profiling and found that LEUTX overexpression restored EGA failure and improved preimplantation development and cloning efficiency in porcine cloned embryos. Mechanistically, LEUTX opened EGA-related genomic regions and established histone acetylation via recruiting acetyltransferases p300 and KAT2A. These findings reveal the regulatory mechanism of LEUTX to govern EGA in pigs, which may provide valuable insights into the study of early embryo development for other non-rodent mammals.


Subject(s)
Genome , Nuclear Transfer Techniques , Animals , Swine , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Embryonic Development/genetics , Embryo, Mammalian/metabolism , Humans , Transcription Factors/metabolism , Transcription Factors/genetics , Acetylation , Cloning, Organism/methods , Histones/metabolism , Blastocyst/metabolism
20.
Planta ; 260(1): 30, 2024 Jun 16.
Article in English | MEDLINE | ID: mdl-38879830

ABSTRACT

MAIN CONCLUSION: Ectopic expression of OsWOX9A induces narrow adaxially rolled rice leaves with larger bulliform cells and fewer large veins, probably through regulating the expression of auxin-related and expansin genes. The WUSCHEL-related homeobox (WOX) family plays a pivotal role in plant development by regulating genes involved in various aspects of growth and differentiation. OsWOX9A (DWT1) has been linked to tiller growth, uniform plant growth, and flower meristem activity. However, its impact on leaf growth and development in rice has not been studied. In this study, we investigated the biological role of OsWOX9A in rice growth and development using transgenic plants. Overexpression of OsWOX9A conferred narrow adaxially rolled rice leaves and altered plant architecture. These plants exhibited larger bulliform cells and fewer larger veins compared to wild-type plants. OsWOX9A overexpression also reduced plant height, tiller number, and seed-setting rate. Comparative transcriptome analysis revealed several differentially expressed auxin-related and expansin genes in OsWOX9A overexpressing plants, consistent with their roles in leaf and plant development. These results indicate that the ectopic expression of OsWOX9A may have multiple effects on the development and growth of rice, providing a more comprehensive picture of how the WOX9 subfamily contributes to leaf development and plant architecture.


Subject(s)
Ectopic Gene Expression , Gene Expression Regulation, Plant , Oryza , Plant Leaves , Plant Proteins , Plants, Genetically Modified , Oryza/genetics , Oryza/growth & development , Oryza/anatomy & histology , Plant Leaves/genetics , Plant Leaves/growth & development , Plant Leaves/anatomy & histology , Plant Proteins/genetics , Plant Proteins/metabolism , Indoleacetic Acids/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Gene Expression Profiling
SELECTION OF CITATIONS
SEARCH DETAIL
...