Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 372
Filter
1.
Biochem Biophys Res Commun ; 722: 150155, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-38795454

ABSTRACT

Runt-related transcription factor (RUNX) family members play critical roles in the development of multiple organs. Mammalian RUNX family members, consisting of RUNX1, RUNX2, and RUNX3, have distinct tissue-specific expression and function. In this study, we examined the spatiotemporal expression patterns of RUNX family members in developing kidneys and analyzed the role of RUNX1 during kidney development. In the developing mouse kidney, RUNX1 protein was strongly expressed in the ureteric bud (UB) tip and weakly expressed in the distal segment of the renal vesicle (RV), comma-shaped body (CSB), and S-shaped body (SSB). In contrast, RUNX2 protein was restricted to the stroma, and RUNX3 protein was only expressed in immune cells. We also analyzed the expression of RUNX family members in the cynomolgus monkey kidney. We found that expression patterns of RUNX2 and RUNX3 were conserved between rodents and primates, whereas RUNX1 was only expressed in the UB tip, not in the RV, CSB, or SSB of cynomolgus monkeys, suggesting a species differences. We further evaluated the roles of RUNX1 using two different conditional knockout mice: Runx1f/f:HoxB7-Cre and Runx1f/f:R26-CreERT2 and found no abnormalities in the kidney. Our findings showed that RUNX1, which is mainly expressed in the UB tip, is not essential for kidney development.


Subject(s)
Core Binding Factor Alpha 2 Subunit , Kidney , Animals , Core Binding Factor Alpha 2 Subunit/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , Kidney/metabolism , Kidney/embryology , Kidney/growth & development , Mice , Macaca fascicularis , Gene Expression Regulation, Developmental , Core Binding Factor Alpha 1 Subunit/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 3 Subunit/metabolism , Core Binding Factor Alpha 3 Subunit/genetics , Core Binding Factor alpha Subunits/metabolism , Core Binding Factor alpha Subunits/genetics , Mice, Inbred C57BL , Mice, Knockout
2.
J Med Chem ; 67(11): 9165-9172, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38803164

ABSTRACT

Runt-related transcription factor (RUNX) proteins are considered to play various roles in cancer. Here, we evaluated the anticancer activity of Chb-M', a compound that specifically and covalently binds to the consensus sequence for RUNX family proteins, in p53-mutated non-small cell lung cancer cells. Chb-M' killed the cancer cells by inducing apoptosis. The compound showed an anticancer effect comparable to that of the clinically used drugs alectinib and ceritinib in vivo. Notably, Chb-M' extended the cancer-free survival of mice after ending treatment more effectively than did the other two drugs. The results presented here suggest that Chb-M' is an attractive candidate as an anticancer drug applicable to the treatment of non-small cell lung cancer and various other types of cancers.


Subject(s)
Antineoplastic Agents , Apoptosis , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Tumor Suppressor Protein p53 , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , Humans , Animals , Tumor Suppressor Protein p53/metabolism , Apoptosis/drug effects , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/therapeutic use , Mice , Cell Line, Tumor , Mutation , Cell Proliferation/drug effects , Core Binding Factor alpha Subunits/metabolism , Mice, Nude , Xenograft Model Antitumor Assays
3.
Clin Exp Med ; 24(1): 50, 2024 Mar 02.
Article in English | MEDLINE | ID: mdl-38430423

ABSTRACT

Runt-related transcription factors (RUNX) are a family of transcription factors that are essential for normal and malignant hematopoietic processes. Their most widely recognized role in malignancy is to promote the occurrence and development of acute myeloid leukemia. However, it is worth noting that during the last decade, studies of RUNX proteins in solid tumors have made considerable progress, suggesting that these proteins are directly involved in different stages of tumor development, including tumor initiation, progression, and invasion. RUNX proteins also play a role in tumor angiogenesis, the maintenance of tumor cell stemness, and resistance to antitumor drugs. These findings have led to the consideration of RUNX as a tumor biomarker. All RUNX proteins are involved in the occurrence and development of solid tumors, but the role of each RUNX protein in different tumors and the major signaling pathways involved are complicated by tumor heterogeneity and the interacting tumor microenvironment. Understanding how the dysregulation of RUNX in tumors affects normal biological processes is important to elucidate the molecular mechanisms by which RUNX affects malignant tumors.


Subject(s)
Neoplasms , Transcription Factors , Humans , Neoplasms/genetics , Core Binding Factor alpha Subunits/genetics , Core Binding Factor alpha Subunits/metabolism , Signal Transduction , Tumor Microenvironment
4.
Nat Immunol ; 24(9): 1458-1472, 2023 09.
Article in English | MEDLINE | ID: mdl-37563311

ABSTRACT

Runx factors are essential for lineage specification of various hematopoietic cells, including T lymphocytes. However, they regulate context-specific genes and occupy distinct genomic regions in different cell types. Here, we show that dynamic Runx binding shifts in mouse early T cell development are mostly not restricted by local chromatin state but regulated by Runx dosage and functional partners. Runx cofactors compete to recruit a limited pool of Runx factors in early T progenitor cells, and a modest increase in Runx protein availability at pre-commitment stages causes premature Runx occupancy at post-commitment binding sites. This increased Runx factor availability results in striking T cell lineage developmental acceleration by selectively activating T cell-identity and innate lymphoid cell programs. These programs are collectively regulated by Runx together with other, Runx-induced transcription factors that co-occupy Runx-target genes and propagate gene network changes.


Subject(s)
Gene Regulatory Networks , T-Lymphocytes , Mice , Animals , T-Lymphocytes/metabolism , Immunity, Innate/genetics , Lymphocytes/metabolism , Core Binding Factor alpha Subunits/genetics , Core Binding Factor alpha Subunits/metabolism , Cell Differentiation/genetics
5.
Cells ; 12(8)2023 04 07.
Article in English | MEDLINE | ID: mdl-37190015

ABSTRACT

The RUNX family of transcription factors, including RUNX1, RUNX2, and RUNX3, are key regulators of development and can function as either tumor suppressors or oncogenes in cancer. Emerging evidence suggests that the dysregulation of RUNX genes can promote genomic instability in both leukemia and solid cancers by impairing DNA repair mechanisms. RUNX proteins control the cellular response to DNA damage by regulating the p53, Fanconi anemia, and oxidative stress repair pathways through transcriptional or non-transcriptional mechanisms. This review highlights the importance of RUNX-dependent DNA repair regulation in human cancers.


Subject(s)
Core Binding Factor Alpha 3 Subunit , Neoplasms , Humans , Core Binding Factor Alpha 3 Subunit/genetics , Core Binding Factor Alpha 3 Subunit/metabolism , Core Binding Factor alpha Subunits/genetics , Core Binding Factor alpha Subunits/metabolism , Neoplasms/genetics , Neoplasms/pathology , DNA Repair/genetics , DNA Damage/genetics
6.
J Cancer Res Clin Oncol ; 149(11): 9409-9423, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37081242

ABSTRACT

PURPOSE: Cancer is one of the deadliest pathologies with more than 19 million new cases and 10 million cancer-related deaths across the globe. Despite development of advanced therapeutic interventions, cancer remains as a fatal pathology due to lack of early prognostic biomarkers, therapy resistance and requires identification of novel drug targets. METHODS: Runt-related transcription factors (Runx) family controls several cellular and physiological functions including osteogenesis. Recent literatures from PubMed was mined and the review was written in comprehensive manner RESULTS: Recent literature suggests that aberrant expression of Runx contributes to tumorigenesis of many organs. Conversely, cell- and tissue-specific tumor suppressor roles of Runx are also reported. In this review, we have provided the structural/functional properties of Runx isoforms and its regulation in context of human cancer. Moreover, in an urgent need to discover novel therapeutic interventions against cancer, we comprehensively discussed the reported oncogenic and tumor suppressive roles of Runx isoforms in several tumor types and discussed the discrepancies that may have risen on Runx as a driver of malignant transformation. CONCLUSION: Runx may be a novel therapeutic target against a battery of deadly human cancers.


Subject(s)
Neoplasms , Transcription Factors , Humans , Transcription Factors/metabolism , Core Binding Factor alpha Subunits/metabolism , Neoplasms/genetics , Carcinogenesis/genetics
7.
Acta Biomater ; 161: 298-308, 2023 04 15.
Article in English | MEDLINE | ID: mdl-36871775

ABSTRACT

Successful bone regeneration on titanium (Ti) surfaces is a key process in dental implant treatment. Bone marrow mesenchymal stem cells (BMSCs) are fundamental cellular components of this process, and their early recruitment, proliferation, and differentiation into bone-forming osteoblasts are crucial. A proteoglycan (PG)-rich layer has been reported to exist between Ti surfaces and bones; however, the molecules that could potentially affect the formation of this layer remain unknown. Family with sequence similarity 20 member B (FAM20B) is a newly identified kinase that regulates the synthesis of glycosaminoglycans, an important component of the PG-rich layer. Because FAM20B is also closely associated with bone development, in this study, we examined the function of FAM20B in osteogenic differentiation of BMSCs on Ti surfaces. For this, BMSC cell lines with knocked down FAM20B (shBMSCs) were cultured on Ti surfaces. The results showed that the depletion of FAM20B reduced the formation of a PG-rich layer between the Ti surfaces and cells. The shBMSCs exhibited downregulated expression of osteogenic marker genes (ALP and OCN) and decreased mineral deposition. Moreover, shBMSCs reduced the molecular levels of p-ERK1/2, which plays an important role in MSC osteogenesis. The nuclear translocation of RUNX2, an important transcription factor for osteogenic differentiation, on the Ti surfaces is inhibited by the depletion of FAM20B in BMSCs. Moreover, the depletion of FAM20B reduced the transcriptional activity of RUNX2, which is important in regulating the expression of osteogenic genes. STATEMENT OF SIGNIFICANCE: Bone healing and regeneration on implanted titanium surfaces is a cell-material interaction. Such an interaction is enabled by bone marrow mesenchymal stem cells (BMSCs), and their early recruitment, proliferation, and differentiation into bone-forming osteoblasts are essential for bone healing and osseointegration. In this study, we found that the family with sequence similarity 20-B influenced the formation of a proteoglycan rich layer between BMSCs and the titanium surface and regulated the differentiation of BMSCs into bone-forming osteoblasts. We believe that our study contributes significantly to the further exploration of bone healing and osseointegration mechanisms on implanted titanium surfaces.


Subject(s)
Bone Marrow Cells , Mesenchymal Stem Cells , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/physiology , Proteoglycans/metabolism , Titanium/chemistry , Core Binding Factor alpha Subunits , Cell Differentiation , Osteogenesis , Cells, Cultured
8.
Cells ; 12(3)2023 01 25.
Article in English | MEDLINE | ID: mdl-36766749

ABSTRACT

The runt-related transcription factors (RUNX) play prominent roles in cell cycle progression, differentiation, apoptosis, immunity and epithelial-mesenchymal transition. There are three members in the mammalian RUNX family, each with distinct tissue expression profiles. RUNX genes play unique and redundant roles during development and adult tissue homeostasis. The ability of RUNX proteins to influence signaling pathways, such as Wnt, TGFß and Hippo-YAP, suggests that they integrate signals from the environment to dictate cell fate decisions. All RUNX genes hold master regulator roles, albeit in different tissues, and all have been implicated in cancer. Paradoxically, RUNX genes exert tumor suppressive and oncogenic functions, depending on tumor type and stage. Unlike RUNX1 and 2, the role of RUNX3 in stem cells is poorly understood. A recent study using cancer-derived RUNX3 mutation R122C revealed a gatekeeper role for RUNX3 in gastric epithelial stem cell homeostasis. The corpora of RUNX3R122C/R122C mice showed a dramatic increase in proliferating stem cells as well as inhibition of differentiation. Tellingly, RUNX3R122C/R122C mice also exhibited a precancerous phenotype. This review focuses on the impact of RUNX3 dysregulation on (1) stem cell fate and (2) the molecular mechanisms underpinning early carcinogenesis.


Subject(s)
Core Binding Factor alpha Subunits , Neoplasms , Animals , Mice , Biology , Core Binding Factor alpha Subunits/genetics , Core Binding Factor alpha Subunits/metabolism , Mammals/metabolism , Mutation , Neoplasms/genetics , Signal Transduction , Humans
9.
Cells ; 12(2)2023 01 07.
Article in English | MEDLINE | ID: mdl-36672189

ABSTRACT

The DNA repair machinery exists to protect cells from daily genetic insults by orchestrating multiple intrinsic and extrinsic factors. One such factor recently identified is the Runt-related transcription factor (RUNX) family, a group of proteins that act as a master transcriptional regulator for multiple biological functions such as embryonic development, stem cell behaviors, and oncogenesis. A significant number of studies in the past decades have delineated the involvement of RUNX proteins in DNA repair. Alterations in RUNX genes cause organ failure and predisposition to cancers, as seen in patients carrying mutations in the other well-established DNA repair genes. Herein, we review the currently existing findings and provide new insights into transcriptional and non-transcriptional multifaceted regulation of DNA repair by RUNX family proteins.


Subject(s)
Core Binding Factor alpha Subunits , Neoplasms , Female , Pregnancy , Humans , Core Binding Factor alpha Subunits/genetics , Core Binding Factor alpha Subunits/metabolism , Transcription Factors/metabolism , Genome , Gene Expression Regulation , Neoplasms/genetics
10.
Int J Mol Sci ; 23(21)2022 Oct 31.
Article in English | MEDLINE | ID: mdl-36362086

ABSTRACT

RUNX proteins, such as RUNX2, regulate the proliferation and differentiation of chondrocytes and osteoblasts. Haploinsufficiency of RUNX2 causes cleidocranial dysplasia, but a detailed analysis of Runx2+/- mice has not been reported. Furthermore, CBFB is required for the stability and DNA binding of RUNX family proteins. CBFB has two isoforms, and CBFB2 plays a major role in skeletal development. The calvaria, femurs, vertebrae and ribs in Cbfb2-/- mice were analyzed after birth, and compared with those in Runx2+/- mice. Calvarial development was impaired in Runx2+/- mice but mildly delayed in Cbfb2-/- mice. In femurs, the cortical bone but not trabecular bone was reduced in Cbfb2-/- mice, whereas both the trabecular and cortical bone were reduced in Runx2+/- mice. The trabecular bone in vertebrae increased in Cbfb2-/- mice but not in Runx2+/- mice. Rib development was impaired in Cbfb2-/- mice but not in Runx2+/- mice. These differences were likely caused by differences in the indispensability of CBFB and RUNX2, the balance of bone formation and resorption, or the number and maturation stage of osteoblasts. Thus, different amounts of CBFB and RUNX2 were required among the bone tissues for proper bone development and maintenance.


Subject(s)
Core Binding Factor Alpha 1 Subunit , Osteoblasts , Animals , Mice , Cell Differentiation/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Core Binding Factor alpha Subunits/metabolism , Osteoblasts/metabolism , Osteogenesis/genetics , Ribs/metabolism , Skull/metabolism , Spine/metabolism
11.
Cells ; 11(22)2022 11 20.
Article in English | MEDLINE | ID: mdl-36429115

ABSTRACT

RUNX proteins are highly conserved in metazoans and perform critical functions during development. Dysregulation of RUNX proteins through various molecular mechanisms facilitates the development and progression of various cancers, where different RUNX proteins show tumor type-specific functions and regulate different aspects of tumorigenesis by cross-talking with different signaling pathways such as Wnt, TGF-ß, and Hippo. Molecularly, they could serve as transcription factors (TFs) to activate their direct target genes or interact with many other TFs to modulate chromatin architecture globally. Here, we review the current knowledge on the functions and regulations of RUNX proteins in different cancer types and highlight their potential role as epigenetic modulators in cancer.


Subject(s)
Core Binding Factor alpha Subunits , Neoplasms , Humans , Core Binding Factor alpha Subunits/genetics , Core Binding Factor alpha Subunits/metabolism , Neoplasms/metabolism , Epigenomics , Transcription Factors/genetics , Transcription Factors/metabolism , Epigenesis, Genetic
12.
Cells ; 11(19)2022 10 01.
Article in English | MEDLINE | ID: mdl-36231060

ABSTRACT

The tumor microenvironment (TME) is broadly implicated in tumorigenesis, as tumor cells interact with surrounding cells to influence the development and progression of the tumor. Blood vessels are a major component of the TME and are attributed to the creation of a hypoxic microenvironment, which is a common feature of advanced cancers and inflamed premalignant tissues. Runt-related transcription factor (RUNX) proteins, a transcription factor family of developmental master regulators, are involved in vital cellular processes such as differentiation, proliferation, cell lineage specification, and apoptosis. Furthermore, the RUNX family is involved in the regulation of various oncogenic processes and signaling pathways as well as tumor suppressive functions, suggesting that the RUNX family plays a strategic role in tumorigenesis. In this review, we have discussed the relevant findings that describe the crosstalk of the RUNX family with the hypoxic TME and tumor angiogenesis or with their signaling molecules in cancer development and progression.


Subject(s)
Core Binding Factor alpha Subunits , Neoplasms , Carcinogenesis , Core Binding Factor alpha Subunits/genetics , Core Binding Factor alpha Subunits/metabolism , Humans , Hypoxia , Morphogenesis , Neoplasms/genetics , Neovascularization, Pathologic , Transcription Factors/physiology , Tumor Microenvironment
13.
Cells ; 11(19)2022 10 03.
Article in English | MEDLINE | ID: mdl-36231078

ABSTRACT

The Runt-related transcription factor (RUNX) family of proteins are crucial for many developmental and immuno-physiological processes. Their importance in cellular and tissue development has been repeatedly demonstrated as they are often found mutated and implicated in tumorigenesis. Most importantly, RUNX have now emerged as critical regulators of lymphocyte function against pathogenic infections and tumorigenic cells, the latter has now revolutionized our current understandings as to how RUNX proteins contribute to control tumor pathogenicity. These multifunctional roles of RUNX in mammalian immune responses and tissue homeostasis have led us to appreciate their value in controlling anti-tumor immune responses. Here, we summarize and discuss the role of RUNX in regulating the development and function of lymphocytes responding to foreign and tumorigenic threats and highlight their key roles in anti-tumor immunity.


Subject(s)
Core Binding Factor alpha Subunits , Neoplasms , Animals , Core Binding Factor alpha Subunits/genetics , Core Binding Factor alpha Subunits/metabolism , Immunity , Lymphocytes/metabolism , Mammals/metabolism , Neoplasms/genetics , Transcription Factors
14.
Nature ; 609(7929): 1021-1028, 2022 09.
Article in English | MEDLINE | ID: mdl-36131014

ABSTRACT

Medulloblastoma (MB) comprises a group of heterogeneous paediatric embryonal neoplasms of the hindbrain with strong links to early development of the hindbrain1-4. Mutations that activate Sonic hedgehog signalling lead to Sonic hedgehog MB in the upper rhombic lip (RL) granule cell lineage5-8. By contrast, mutations that activate WNT signalling lead to WNT MB in the lower RL9,10. However, little is known about the more commonly occurring group 4 (G4) MB, which is thought to arise in the unipolar brush cell lineage3,4. Here we demonstrate that somatic mutations that cause G4 MB converge on the core binding factor alpha (CBFA) complex and mutually exclusive alterations that affect CBFA2T2, CBFA2T3, PRDM6, UTX and OTX2. CBFA2T2 is expressed early in the progenitor cells of the cerebellar RL subventricular zone in Homo sapiens, and G4 MB transcriptionally resembles these progenitors but are stalled in developmental time. Knockdown of OTX2 in model systems relieves this differentiation blockade, which allows MB cells to spontaneously proceed along normal developmental differentiation trajectories. The specific nature of the split human RL, which is destined to generate most of the neurons in the human brain, and its high level of susceptible EOMES+KI67+ unipolar brush cell progenitor cells probably predisposes our species to the development of G4 MB.


Subject(s)
Cell Differentiation , Cerebellar Neoplasms , Medulloblastoma , Metencephalon , Cell Differentiation/genetics , Cell Lineage , Cerebellar Neoplasms/classification , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/pathology , Cerebellum/embryology , Cerebellum/pathology , Core Binding Factor alpha Subunits/genetics , Hedgehog Proteins/metabolism , Histone Demethylases , Humans , Ki-67 Antigen/metabolism , Medulloblastoma/classification , Medulloblastoma/genetics , Medulloblastoma/pathology , Metencephalon/embryology , Metencephalon/pathology , Muscle Proteins , Mutation , Otx Transcription Factors/deficiency , Otx Transcription Factors/genetics , Repressor Proteins , T-Box Domain Proteins/metabolism , Transcription Factors
15.
Cells ; 10(11)2021 10 28.
Article in English | MEDLINE | ID: mdl-34831147

ABSTRACT

The Hippo-YAP signaling pathway serves roles in cell proliferation, stem cell renewal/maintenance, differentiation and apoptosis. Many of its functions are central to early development, adult tissue repair/regeneration and not surprisingly, tumorigenesis and metastasis. The Hippo pathway represses the activity of YAP and paralog TAZ by modulating cell proliferation and promoting differentiation to maintain tissue homeostasis and proper organ size. Similarly, master regulators of development RUNX transcription factors have been shown to play critical roles in proliferation, differentiation, apoptosis and cell fate determination. In this review, we discuss the multiple interactions of RUNX with the Hippo-YAP pathway, their shared collaborators in Wnt, TGFß, MYC and RB pathways, and their overlapping functions in development and tumorigenesis.


Subject(s)
Core Binding Factor alpha Subunits/metabolism , Hippo Signaling Pathway , YAP-Signaling Proteins/metabolism , Animals , Humans , Protein Binding , Transforming Growth Factor beta/metabolism , Wnt Signaling Pathway
16.
Front Immunol ; 12: 701924, 2021.
Article in English | MEDLINE | ID: mdl-34421907

ABSTRACT

Runx proteins (also known as Runt-domain transcription factors) have been studied for a long time as key regulators of cellular differentiation. RUNX2 has been described as essential for osteogenesis, whereas RUNX1 and RUNX3 are known to control blood cell development during different stages of cell lineage specification. However, recent studies show evidence of complex relationships between RUNX proteins, chromatin-modifying machinery, the cytoskeleton and different transcription factors in various non-embryonic contexts, including mature T cell homeostasis, inflammation and cancer. In this review, we discuss the diversity of Runx functions in mature T helper cells, such as production of cytokines and chemokines by different CD4 T cell populations; apoptosis; and immunologic memory acquisition. We then briefly cover recent findings about the contribution of RUNX1, RUNX2 and RUNX3 to various immunologic diseases. Finally, we discuss areas that require further study to better understand the role that Runx proteins play in inflammation and immunity.


Subject(s)
Core Binding Factor alpha Subunits/immunology , T-Lymphocytes/immunology , Thymus Gland/immunology , Animals , Humans , Immunity/immunology , Immunologic Memory/immunology , Inflammation/immunology
17.
J Allergy Clin Immunol ; 147(5): 1549-1560, 2021 05.
Article in English | MEDLINE | ID: mdl-33965092

ABSTRACT

Innate lymphoid cells (ILCs) mainly reside at barrier surfaces and regulate tissue homeostasis and immunity. ILCs are divided into 3 groups, group 1 ILCs, group 2 ILCs, and group 3 ILC3, on the basis of their similar effector programs to T cells. The development of ILCs from lymphoid progenitors in adult mouse bone marrow has been studied in detail, and multiple ILC progenitors have been characterized. ILCs are mostly tissue-resident cells that develop in the perinatal period. More recently, ILC progenitors have also been identified in peripheral tissues. In this review, we discuss the stepwise transcription factor-directed differentiation of mouse ILC progenitors into mature ILCs, the critical time windows in ILC development, and the contribution of bone marrow versus tissue ILC progenitors to the pool of mature ILCs in tissues.


Subject(s)
Lymphocytes/cytology , Lymphocytes/immunology , Animals , Core Binding Factor alpha Subunits/immunology , Cytokines/immunology , Epigenesis, Genetic , Humans , Immunity, Innate , Infections/immunology , Inflammation/immunology , Thymus Gland/immunology , Transcription Factors/immunology
18.
Dev Dyn ; 250(10): 1494-1504, 2021 10.
Article in English | MEDLINE | ID: mdl-33760336

ABSTRACT

BACKGROUND: Unlike many vertebrates with continuous dental replacement, mammals have a maximum of two dental generations. Due to the absence of dental replacement in the laboratory mouse, the mechanisms of the mammalian tooth replacement system are poorly known. In this study, we use the European rabbit as a model for mammalian tooth development and replacement. RESULTS: We provide data on some key regulators of tooth development. We detected the presence of SOX2 in both the replacement dental lamina and the rudimentary successional dental lamina of unreplaced molars, indicating that SOX2 may not be sufficient to initiate and maintain tooth replacement. We showed that Shh does not seem to be directly involved in tooth replacement. The transient presence of the rudimentary successional dental lamina in the molar allowed us to identify genes that could be essential for the initiation or the maintenance of tooth replacement. Hence, the locations of Sostdc1, RUNX2, and LEF1 vary between the deciduous premolar, the replacement premolar, and the molar, indicating possible roles in tooth replacement. CONCLUSION: According to our observations, initiation and the maintenance of tooth replacement correlate with the presence of LEF1+ cells and the absence of both mesenchymal RUNX2 and epithelial Sostdc1+ cells.


Subject(s)
Gene Expression , Odontogenesis/drug effects , SOXB1 Transcription Factors/metabolism , Tooth/growth & development , Animals , Core Binding Factor alpha Subunits/genetics , Core Binding Factor alpha Subunits/metabolism , Rabbits , SOXB1 Transcription Factors/genetics , Tooth/metabolism
19.
Immunol Rev ; 300(1): 100-124, 2021 03.
Article in English | MEDLINE | ID: mdl-33682165

ABSTRACT

Adaptive immunity to intracellular pathogens and tumors is mediated by antigen-experienced CD8 T cells. Individual naive CD8 T cells have the potential to differentiate into a diverse array of antigen-experienced subsets that exhibit distinct effector functions, life spans, anatomic positioning, and potential for regenerating an entirely new immune response during iterative pathogenic exposures. The developmental process by which activated naive cells undergo diversification involves regulation of chromatin structure and transcription but is not entirely understood. This review examines how alterations in chromatin structure, transcription factor binding, extracellular signals, and single-cell gene expression explain the differential development of distinct effector (TEFF ) and memory (TMEM ) CD8 T cell subsets. Special emphasis is placed on how Runx proteins function with additional transcription factors to pioneer changes in chromatin accessibility and drive transcriptional programs that establish the core attributes of cytotoxic T lymphocytes, subdivide circulating and non-circulating TMEM cell subsets, and govern terminal differentiation. The discussion integrates the roles of specific cytokine signals, transcriptional circuits and how regulation of individual nucleosomes and RNA polymerase II activity can contribute to the process of differentiation. A model that integrates many of these features is discussed to conceptualize how activated CD8 T cells arrive at their fates.


Subject(s)
CD8-Positive T-Lymphocytes , Core Binding Factor alpha Subunits , Cell Differentiation , Chromatin , Core Binding Factor alpha Subunits/genetics , Immunologic Memory , T-Lymphocyte Subsets
20.
FEBS J ; 288(21): 6112-6126, 2021 11.
Article in English | MEDLINE | ID: mdl-33682350

ABSTRACT

Prostate cancer (PCa) is a very complex disease that is a major cause of death in men worldwide. Currently, PCa dependence on the androgen receptor (AR) has resulted in use of AR antagonists and antiandrogen therapies that reduce endogenous steroid hormone production. However, within two to three years of receiving first-line androgen deprivation therapy, the majority of patients diagnosed with PCa progress to castration-resistant prostate cancer (CRPC). There is an urgent need for therapies that are more durable than antagonism of the AR axis. Studies of runt-related transcription factors (RUNX) and their heterodimerization partner, core-binding factor subunit b (CBFß), are revealing that the RUNX family are drivers of CRPC. In this review, we describe what is presently understood about RUNX members in PCa, including what regulates and is regulated by RUNX proteins, and the role of RUNX proteins in the tumor microenvironment and AR signaling. We discuss the implications for therapeutically targeting RUNX, the potential for RUNX as PCa biomarkers, and the current pressing questions in the field.


Subject(s)
Core Binding Factor alpha Subunits/metabolism , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms/metabolism , Transcription Factors/metabolism , Androgen Antagonists/therapeutic use , Humans , Male , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms, Castration-Resistant/drug therapy , Receptors, Androgen/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...