Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Rep ; 41(11): 111796, 2022 12 13.
Article in English | MEDLINE | ID: mdl-36516755

ABSTRACT

Intestinal epithelial replenishment is fueled by continuously dividing intestinal stem cells (ISCs) resident at the crypt niche. However, the cell type(s) enabling replenishment upon damage and subsequent loss of whole crypts remain largely unclear. Using Set domain-containing protein 4 (Setd4), we identify a small population with reserve stem cell characteristics in the mouse intestine. Upon irradiation-induced injury, Setd4-expressing (Setd4+) cells survive radiation exposure and then activate to produce Sca-1-expressing cell types to restore the epithelial wall and regenerate crypts de novo via crypt fission. Setd4+ cells are confirmed to originate from the early fetal period, subsequently contributing to the development of embryonic gut and the establishment of postnatal crypts. Setd4+ cells are therefore represented as both originators and key regenerators of the intestine.


Subject(s)
Embryonic Stem Cells , Intestines , Mice , Animals , Proteins/metabolism , Intestinal Mucosa/metabolism
2.
Stem Cell Reports ; 17(9): 2081-2096, 2022 09 13.
Article in English | MEDLINE | ID: mdl-36027907

ABSTRACT

Cellular quiescence facilitates maintenance of neural stem cells (NSCs) and their subsequent regenerative functions in response to brain injury and aging. However, the specification and maintenance of NSCs in quiescence from embryo to adulthood remain largely unclear. Here, using Set domain-containing protein 4 (SETD4), an epigenetic determinant of cellular quiescence, we mark a small but long-lived NSC population in deep quiescence in the subventricular zone of adult murine brain. Genetic lineage tracing shows that SETD4+ cells appear before neuroectoderm formation and contribute to brain development. In the adult, conditional knockout of Setd4 resulted in quiescence exit of NSCs, generating newborn neurons in the olfactory bulb and contributing to damage repair. However, long period deletion of SETD4 lead to exhaustion of NSC reservoir or SETD4 overexpression caused quiescence entry of NSCs, leading to suppressed neurogenesis. This study reveals the existence of long-lived deep quiescent NSCs and their neurogenetic capacities beyond activation.


Subject(s)
Adult Stem Cells , Neural Stem Cells , Adult Stem Cells/metabolism , Animals , Lateral Ventricles , Mice , Neural Stem Cells/metabolism , Neurogenesis/genetics , Neurons
3.
Sci Rep ; 11(1): 12614, 2021 06 15.
Article in English | MEDLINE | ID: mdl-34131249

ABSTRACT

In the adult pancreas, the presence of progenitor or stem cells and their potential involvement in homeostasis and regeneration remains unclear. Here, we identify that SET domain-containing protein 4 (SETD4), a histone lysine methyltransferase, is expressed in a small cell population in the adult mouse pancreas. Genetic lineage tracing shows that during pancreatic development, descendants of SETD4+ cells make up over 70% of pancreatic cells and then contribute to each pancreatic lineage during pancreatic homeostasis. SETD4+ cells generate newborn acinar cells in response to cerulein-induced pancreatitis in acinar compartments. Ablation of SETD4+ cells compromises regeneration of acinar cells, in contrast to controls. Our findings provide a new cellular narrative for pancreatic development, homeostasis and response to injury via a small SETD4+ cell population. Potential applications may act to preserve pancreatic function in case of pancreatic disease and/or damage.


Subject(s)
Methyltransferases/genetics , Pancreas/metabolism , Pancreatitis/genetics , Regeneration/genetics , Acinar Cells/metabolism , Acinar Cells/pathology , Animals , Cell Lineage/genetics , Ceruletide/toxicity , Disease Models, Animal , Homeostasis/drug effects , Homeostasis/genetics , Humans , Mice , Pancreas/growth & development , Pancreas/injuries , Pancreas/pathology , Pancreatitis/chemically induced , Pancreatitis/pathology , Stem Cells/cytology , Stem Cells/drug effects
4.
Sci Rep ; 11(1): 11603, 2021 06 02.
Article in English | MEDLINE | ID: mdl-34079011

ABSTRACT

Blood vessels in the adult mammal exist in a highly organized and stable state. In the ischemic heart, limited expansion capacity of the myocardial vascular bed cannot satisfy demands for oxygen supply and the myocardium eventually undergoes irreversible damage. The predominant contribution of endogenous c-Kit+ cells is understood to be in the development and homeostasis of cardiac endothelial cells, which suggests potential for their targeting in treatments for cardiac ischemic injury. Quiescent cells in other tissues are known to contribute to the long-term maintenance of a cell pool, preserve proliferation capacity and, upon activation, facilitate tissue homeostasis and regeneration in response to tissue injury. Here, we present evidence of a Setd4-expressing quiescent c-Kit+ cell population in the adult mouse heart originating from embryonic stages. Conditional knock-out of Setd4 in c-Kit-CreERT2;Setd4f/f;Rosa26TdTomato mice induced an increase in vascular endothelial cells of capillaries in both neonatal and adult mice. We show that Setd4 regulates quiescence of c-Kit+ cells by the PI3K-Akt-mTOR signaling pathway via H4K20me3 catalysis. In myocardial infarction injured mice, Setd4 knock-out resulted in attenuated cardiomyocyte apoptosis, decreased infarction size and improved cardiac function. Lineage tracing in Setd4-Cre;Rosa26mT/mG mice showed that Setd4+ cells contribute to each cardiac lineage. Overall, Setd4 epigenetically controls c-Kit+ cell quiescence in the adult heart by facilitating heterochromatin formation via H4K20me3. Beyond activation, endogenous quiescent c-Kit+ cells were able to improve cardiac function in myocardial infarction injured mice via the neovascularization of capillaries.


Subject(s)
Endothelial Cells/metabolism , Epigenesis, Genetic , Methyltransferases/genetics , Myocardial Infarction/genetics , Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins c-kit/genetics , Animals , Apoptosis , Capillaries/growth & development , Cell Division , Cell Proliferation , Disease Models, Animal , Echocardiography , Endothelial Cells/cytology , Female , Histones/genetics , Histones/metabolism , Integrases/genetics , Integrases/metabolism , Methyltransferases/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/cytology , Neovascularization, Physiologic , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-kit/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
5.
Cancer Res ; 79(18): 4729-4743, 2019 Sep 15.
Article in English | MEDLINE | ID: mdl-31308046

ABSTRACT

Quiescent cancer stem cells (CSC) play important roles in tumorigenesis, relapse, and resistance to chemoradiotherapy. However, the determinants of CSC quiescence and how they sustain themselves to generate tumors and relapse beyond resistance to chemoradiotherapy remains unclear. Here, we found that SET domain-containing protein 4 (SETD4) epigenetically controls breast CSC (BCSC) quiescence by facilitating heterochromatin formation via H4K20me3 catalysis. H4K20me3 localized to the promoter regions and regulated the expression of a set of genes in quiescent BCSCs (qBCSC). SETD4-defined qBCSCs were resistant to chemoradiotherapy and promoted tumor relapse in a mouse model. Upon activation, a SETD4-defined qBCSC sustained itself in a quiescent state by asymmetric division and concurrently produced an active daughter cell that proliferated to produce a cancer cell population. Single-cell sequence analysis indicated that SETD4+ qBCSCs clustered together as a distinct cell type within the heterogeneous BCSC population. SETD4-defined quiescent CSCs were present in multiple cancer types including gastric, cervical, ovarian, liver, and lung cancers and were resistant to chemotherapy. SETD4-defined qBCSCs had a high tumorigenesis potential and correlated with malignancy and chemotherapy resistance in clinical breast cancer patients. Taken together, the results from our previous study and current study on six cancer types reveal an evolutionarily conserved mechanism of cellular quiescence epigenetically controlled by SETD4. Our findings provide insights into the mechanism of tumorigenesis and relapse promoted by SETD4-defined quiescent CSCs and have broad implications for clinical therapies. SIGNIFICANCE: These findings advance our knowledge on the epigenetic determinants of quiescence in cancer stem cell populations and pave the way for future pharmacologic developments aimed at targeting drug-resistant quiescent stem cells.


Subject(s)
Breast Neoplasms/pathology , Drug Resistance, Neoplasm , Epigenomics , Methyltransferases/metabolism , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/pathology , Resting Phase, Cell Cycle , Animals , Apoptosis , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/therapy , Carcinoma, Basal Cell/genetics , Carcinoma, Basal Cell/metabolism , Carcinoma, Basal Cell/pathology , Carcinoma, Basal Cell/therapy , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Chemoradiotherapy , Female , Humans , Methyltransferases/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/therapy , Neoplastic Stem Cells/metabolism , Prognosis , Protein Domains , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Biochem J ; 476(12): 1753-1769, 2019 06 26.
Article in English | MEDLINE | ID: mdl-31189566

ABSTRACT

To cope with harsh environments, the Artemia shrimp produces gastrula embryos in diapause, a state of obligate dormancy, having cellular quiescence and suppressed metabolism. The mechanism behind these cellular events remains largely unknown. Here, we study the regulation of cell quiescence using diapause embryos of Artemia We found that Artemia DEK (Ar-DEK), a nuclear factor protein, was down-regulated in the quiescent cells of diapause embryos and enriched in the activated cells of post-diapause embryos. Knockdown of Ar-DEK induced the production of diapause embryos whereas the control Artemia released free-swimming nuaplii. Our results indicate that Ar-DEK correlated with the termination of cellular quiescence via the increase in euchromatin and decrease in heterochromatin. The phenomena of quiescence have many implications beyond shrimp ecology. In cancer cells, for example, knockdown of DEK also induced a short period of cellular quiescence and increased resistance to environmental stress in MCF-7 and MKN45 cancer cell lines. Analysis of RNA sequences in Artemia and in MCF-7 revealed that the Wnt and AURKA signaling pathways were all down-regulated and the p53 signaling pathway was up-regulated upon inhibition of DEK expression. Our results provide insight into the functions of Ar-DEK in the activation of cellular quiescence during diapause formation in Artemia.


Subject(s)
Artemia/embryology , Arthropod Proteins/biosynthesis , Diapause/physiology , Embryo, Nonmammalian/enzymology , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Receptors, Eph Family/biosynthesis , Wnt Signaling Pathway/physiology , Animals , Artemia/genetics , Arthropod Proteins/genetics , Aurora Kinase A/genetics , Aurora Kinase A/metabolism , Humans , MCF-7 Cells , Receptors, Eph Family/genetics
SELECTION OF CITATIONS
SEARCH DETAIL