Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Mol Cancer ; 23(1): 45, 2024 02 29.
Article in English | MEDLINE | ID: mdl-38424542

ABSTRACT

BACKGROUND: In the myeloid compartment of the tumor microenvironment, CD244 signaling has been implicated in immunosuppressive phenotype of monocytes. However, the precise molecular mechanism and contribution of CD244 to tumor immunity in monocytes/macrophages remains elusive due to the co-existing lymphoid cells expressing CD244. METHODS: To directly assess the role of CD244 in tumor-associated macrophages, monocyte-lineage-specific CD244-deficient mice were generated using cre-lox recombination and challenged with B16F10 melanoma. The phenotype and function of tumor-infiltrating macrophages along with antigen-specific CD8 T cells were analyzed by flow cytometry and single cell RNA sequencing data analysis, and the molecular mechanism underlying anti-tumorigenic macrophage differentiation, antigen presentation, phagocytosis was investigated ex vivo. Finally, the clinical feasibility of CD244-negative monocytes as a therapeutic modality in melanoma was confirmed by adoptive transfer experiments. RESULTS: CD244fl/flLysMcre mice demonstrated a significant reduction in tumor volume (61% relative to that of the CD244fl/fl control group) 14 days after tumor implantation. Within tumor mass, CD244fl/flLysMcre mice also showed higher percentages of Ly6Clow macrophages, along with elevated gp100+IFN-γ+ CD8 T cells. Flow cytometry and RNA sequencing data demonstrated that ER stress resulted in increased CD244 expression on monocytes. This, in turn, impeded the generation of anti-tumorigenic Ly6Clow macrophages, phagocytosis and MHC-I antigen presentation by suppressing autophagy pathways. Combining anti-PD-L1 antibody with CD244-/- bone marrow-derived macrophages markedly improved tumor rejection compared to the anti-PD-L1 antibody alone or in combination with wild-type macrophages. Consistent with the murine data, transcriptome analysis of human melanoma tissue single-cell RNA-sequencing dataset revealed close association between CD244 and the inhibition of macrophage maturation and function. Furthermore, the presence of CD244-negative monocytes/macrophages significantly increased patient survival in primary and metastatic tumors. CONCLUSION: Our study highlights the novel role of CD244 on monocytes/macrophages in restraining anti-tumorigenic macrophage generation and tumor antigen-specific T cell response in melanoma. Importantly, our findings suggest that CD244-deficient macrophages could potentially be used as a therapeutic agent in combination with immune checkpoint inhibitors. Furthermore, CD244 expression in monocyte-lineage cells serve as a prognostic marker in cancer patients.


Subject(s)
Melanoma , Monocytes , Humans , Animals , Mice , Monocytes/metabolism , Melanoma/drug therapy , Melanoma/genetics , Melanoma/metabolism , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Macrophages/metabolism , CD8-Positive T-Lymphocytes , Carcinogenesis/metabolism , Tumor Microenvironment , Signaling Lymphocytic Activation Molecule Family/metabolism
2.
Cells ; 12(12)2023 06 11.
Article in English | MEDLINE | ID: mdl-37371075

ABSTRACT

Adoptive cell therapy using chimeric antigen receptor (CAR) technology is one of the most advanced engineering platforms for cancer immunotherapy. CAR-T cells have shown remarkable efficacy in the treatment of hematological malignancies. However, their limitations in solid tumors include an immunosuppressive tumor microenvironment (TME), insufficient tumor infiltration, toxicity, and the absence of tumor-specific antigens. Although recent advances in CAR-T cell design-such as the incorporation of co-stimulatory domains and the development of armored CAR-T cells-have shown promising results in treating solid tumors, there are still challenges that need to be addressed. To overcome these limitations, other immune cells, such as natural killer (NK) cells and macrophages (M), have been developed as attractive options for efficient cancer immunotherapy of solid tumors. CAR-NK cells exhibit substantial clinical improvements with "off-the-shelf" availability and low toxicity. CAR-M cells have promising therapeutic potential because macrophages can infiltrate the TME of solid tumors. Here, we review the recent advances and future perspectives associated with engineered immune cell-based cancer immunotherapies for solid tumors. We also summarize ongoing clinical trials investigating the safety and efficacy of engineered immune cells, such as CAR-T, CAR-NK, and CAR-M, for targeting solid tumors.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , Humans , Receptors, Chimeric Antigen/metabolism , Immunotherapy, Adoptive/methods , Neoplasms/pathology , Immunotherapy/methods , T-Lymphocytes , Antigens, Neoplasm/metabolism , Tumor Microenvironment
3.
Biomed Pharmacother ; 154: 113553, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35994815

ABSTRACT

To overcome the hurdles of immunotherapy, we investigated whether calcipotriol, a synthetic vitamin D analog, could overcome the immune evasion of glioblastoma multiforme (GBM) by modulating immune responses and the immunosuppressive tumor microenvironment. Administration of calcipotriol considerably reduced tumor growth. Both in vivo and in vitro studies revealed that CD8+T and natural killer (NK) cell gene signatures were enriched and activated, producing high levels of IFN-γ and granzyme B. In contrast, regulatory T cells (Treg) were significantly reduced in the calcipotriol-treated group. The expression of CD127, the receptor for thymic stromal lymphopoietin (TSLP), is elevated in CD4+T cells and potentially supports T-cell priming. Depleting CD4+T cells, but not NK or CD8+T cells, completely abrogated the antitumor efficacy of calcipotriol. These data highlight that the calcipotriol/TSLP/CD4+T axis can activate CD8+T and NK cells with a concomitant reduction in the number of Tregs in GBM. Therefore, calcipotriol can be a novel therapeutic modality to overcome the immune resistance of GBM by converting immunologically "cold" tumors into "hot" tumors. DATA AVAILABILITY: Data are available upon reasonable request. The RNA-seq dataset comparing the transcriptomes of control and calcipotriol-treated GL261 tumors is available from the corresponding author upon request.


Subject(s)
Glioblastoma , Vitamin D , CD8-Positive T-Lymphocytes , Calcitriol/analogs & derivatives , Glioblastoma/metabolism , Humans , Killer Cells, Natural , Lymphocyte Activation , Tumor Microenvironment , Vitamin D/metabolism
4.
Cancers (Basel) ; 13(8)2021 Apr 15.
Article in English | MEDLINE | ID: mdl-33920906

ABSTRACT

NK cells are the predominant innate lymphocyte subsets specialized to kill malignant tumor cells. In patients with advanced cancer, hypoxic stress shapes NK cells toward tumor-resistant and immunosuppressive phenotypes, hence a strategy to restore NK function is critical for successful tumor immunotherapy. Here, we present evidence that pre-activation and subsequent HIF-1α-dependent metabolic shift of NK cells from oxidative phosphorylation into glycolysis are keys to overcome hypoxia-mediated impairment in NK cell survival, proliferation, and tumor cytotoxicity. Specifically, exposing NK cells to 7-9 days of normoxic culture followed by a pO2 of 1.5% hypoxia led to a highly potent effector phenotype via HIF-1α stabilization and upregulation of its target genes, BNIP3, PDK1, VEGF, PKM2, and LDHA. RNA sequencing and network analyses revealed that concomitant reduction of p21/p53 apoptotic pathways along with upregulation of cell cycle-promoting genes, CCNE1, CDC6, CDC20, and downregulation of cell cycle-arrest genes, CDKN1A, GADD45A, and MDM2 were accountable for superior expansion of NK cells via ERK/STAT3 activation. Furthermore, HIF-1α-dependent upregulation of the NKp44 receptor in hypoxia-exposed NK cells resulted in increased killing against K562, CEM, and A375 tumor targets both in-vitro and in-vivo tumor clearance assays. Therefore, hypoxic exposure on pre-activated proliferating NK cells triggered HIF-1α-dependent pathways to initiate coordinated regulation of cell cycle, apoptosis, and cytotoxicity at the global gene transcription level. Our results uncover a previously unidentified role of HIF-1α-mediated metabolic reprogramming that can reverse impaired NK effector phenotypes to generate requisite numbers of functionally robust NK cells for adoptive cellular therapy for clinical evaluation.

5.
Infect Immun ; 89(4)2021 03 17.
Article in English | MEDLINE | ID: mdl-33468581

ABSTRACT

Thymosin beta-4 (Tß4) is an actin-sequestering peptide that plays important roles in regeneration and remodeling of injured tissues. However, its function in a naturally occurring pathogenic bacterial infection model has remained elusive. We adopted Tß4-overexpressing transgenic (Tg) mice to investigate the role of Tß4 in acute pulmonary infection and systemic sepsis caused by Legionella pneumophila Upon infection, Tß4-Tg mice demonstrated significantly lower bacterial loads in the lung, less hyaline membranes and necrotic abscess, with lower interstitial infiltration of neutrophils, CD4+, and CD8+ T cells. Bronchoalveolar lavage fluid of Tß4-Tg mice possessed higher bactericidal activity against exogenously added L. pneumophila, suggesting that constitutive expression of Tß4 could efficiently control L. pneumophila Furthermore, qPCR analysis of lung homogenates demonstrated significant reduction of interleukin 1 beta (IL-1ß) and tumor necrosis factor alpha (TNF-α), which primarily originate from lung macrophages, in Tß4-Tg mice after pulmonary infection. Upon L. pneumophila challenge of bone marrow-derived macrophages (BMDM) in vitro, secretion of IL-1ß and TNF-α proteins was also reduced in Tß4-Tg macrophages, without affecting their survival. The anti-inflammatory effects of BMDM in Tß4-Tg mice on each cytokine were affected when triggering with tlr2, tlr4, tlr5, or tlr9 ligands, suggesting that anti-inflammatory effects of Tß4 are likely mediated by the reduced activation of Toll-like receptors (TLR). Finally, Tß4-Tg mice in a systemic sepsis model were protected from L. pneumophila-induced lethality compared to wild-type controls. Therefore, Tß4 confers effective resistance against L. pneumophila via two pathways, a bactericidal and an anti-inflammatory pathway, which can be harnessed to treat acute pneumonia and septic conditions caused by L. pneumophila in humans.


Subject(s)
Disease Resistance/genetics , Ectopic Gene Expression , Legionella pneumophila/physiology , Legionnaires' Disease/genetics , Legionnaires' Disease/microbiology , Pneumonia, Bacterial/genetics , Pneumonia, Bacterial/microbiology , Thymosin/genetics , Animals , Cytokines/metabolism , Disease Models, Animal , Host-Pathogen Interactions/genetics , Humans , Immunohistochemistry , Immunophenotyping , Legionnaires' Disease/pathology , Ligands , Male , Mice , Mice, Transgenic , Pneumonia, Bacterial/pathology , Sepsis/genetics , Sepsis/microbiology , Sepsis/pathology , Toll-Like Receptors/metabolism
6.
Immune Netw ; 20(2): e14, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32395366

ABSTRACT

With the development of technologies that can transform immune cells into therapeutic modalities, immunotherapy has remarkably changed the current paradigm of cancer treatment in recent years. NK cells are components of the innate immune system that act as key regulators and exhibit a potent tumor cytolytic function. Unlike T cells, NK cells exhibit tumor cytotoxicity by recognizing non-self, without deliberate immunization or activation. Currently, researchers have developed various approaches to improve the number and anti-tumor function of NK cells. These approaches include the use of cytokines and Abs to stimulate the efficacy of NK cell function, adoptive transfer of autologous or allogeneic ex vivo expanded NK cells, establishment of homogeneous NK cell lines using the NK cells of patients with cancer or healthy donors, derivation of NK cells from induced pluripotent stem cells (iPSCs), and modification of NK cells with cutting-edge genetic engineering technologies to generate chimeric Ag receptor (CAR)-NK cells. Such NK cell-based immunotherapies are currently reported as being promising anti-tumor strategies that have shown enhanced functional specificity in several clinical trials investigating malignant tumors. Here, we summarize the recent advances in NK cell-based cancer immunotherapies that have focused on providing improved function through the use of the latest genetic engineering technologies. We also discuss the different types of NK cells developed for cancer immunotherapy and present the clinical trials being conducted to test their safety and efficacy.

7.
Int J Mol Sci ; 21(7)2020 Apr 05.
Article in English | MEDLINE | ID: mdl-32260561

ABSTRACT

The recent development of high-throughput genomics has revolutionized personalized medicine by identifying key pathways and molecular targets controlling tumor progression and survival. Mitogen-activated protein kinase (MAPK) pathways are examples of such targets, and inhibitors against these pathways have shown promising clinical responses in patients with melanoma, non-small-cell lung cancer, colorectal cancer, pancreatic cancer, and thyroid cancer. Although MAPK pathway-targeted therapies have resulted in significant clinical responses in a large proportion of cancer patients, the rate of tumor recurrence is high due to the development of resistance. Conversely, immunotherapies have shown limited clinical responses, but have led to durable tumor regression in patients, and complete responses. Recent evidence indicates that MAPK-targeted therapies may synergize with immune cells, thus providing rationale for the development of combination therapies. Here, we review the current status of ongoing clinical trials investigating MAPK pathway inhibitors, such as BRAF and MAPK/ERK kinase (MEK) inhibitors, in combination with checkpoint inhibitors targeting programmed death protein 1 (PD-1), programmed death-ligand 1 (PD-L1), and cytotoxic T cell associated antigen-4 (CTLA-4). A better understanding of an individual drug's mechanism of action, patterns of acquired resistance, and the influence on immune cells will be critical for the development of novel combination therapies.


Subject(s)
Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy/methods , Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Animals , Clinical Trials as Topic , Combined Modality Therapy , Humans , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Neoplasms/therapy
8.
Nat Commun ; 10(1): 3258, 2019 07 22.
Article in English | MEDLINE | ID: mdl-31332204

ABSTRACT

CD160 and BTLA both bind to herpes virus entry mediator. Although a negative regulatory function of BTLA in natural killer T (NKT) cell activation has been reported, whether CD160 is also involved is unclear. By analyzing CD160-/- mice and mixed bone marrow chimeras, we show that CD160 is not essential for NKT cell development. However, CD160-/- mice exhibit severe liver injury after in vivo challenge with α-galactosylceramide (α-GalCer). Moreover, CD160-/- mice are more susceptible to Concanavalin A challenge, and display elevated serum AST and ALT levels, hyperactivation of NKT cells, and enhanced IFN-γ, TNF, and IL-4 production. Lastly, inhibition of BTLA by anti-BTLA mAb aggravates α-GalCer-induced hepatic injury in CD160-/- mice, suggesting that both CD160 and BTLA serve as non-overlapping negative regulators of NKT cells. Our data thus implicate CD160 as a co-inhibitory receptor that delivers antigen-dependent signals in NKT cells to dampen cytokine production during early innate immune activation.


Subject(s)
Antigens, CD/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Liver/metabolism , Natural Killer T-Cells/metabolism , Receptors, Immunologic/metabolism , Animals , Antigens, CD/genetics , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/genetics , Concanavalin A/administration & dosage , Concanavalin A/toxicity , Cytokines/metabolism , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Galactosylceramides/administration & dosage , Galactosylceramides/toxicity , Liver/drug effects , Liver/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice, Inbred C57BL , Mice, Knockout , Natural Killer T-Cells/immunology , Receptors, Immunologic/genetics , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Survival Analysis
9.
Front Immunol ; 10: 496, 2019.
Article in English | MEDLINE | ID: mdl-31024520

ABSTRACT

Tumor-infiltrating lymphocytes (TILs), found in patients with advanced pancreatic ductal adenocarcinoma (PDAC), are shown to correlate with overall survival (OS) rate. Although majority of TILs consist of CD8+/CD4+ T cells, the presence of NK cells and their role in the pathogenesis of PDAC remains elusive. We performed comprehensive analyses of TIL, PBMC, and autologous tumor cells from 80 enrolled resectable PDAC patients to comprehend the NK cell defects within PDAC. Extremely low frequencies of NK cells (<0.5%) were found within PDAC tumors, which was attributable not to the low expression of tumor chemokines, but to the lack of chemokine receptor, CXCR2. Forced expression of CXCR2 in patients' NK cells rendered them capable of trafficking into PDAC. Furthermore, NK cells exhibited impaired cell-mediated killing of autologous PDAC cells, primarily due to insufficient ligation of NKG2D and DNAM-1, and failed to proliferate within the hypoxic tumor microenvironment. Importantly, these defects could be overcome by ex-vivo stimulation of NK cells from such patients. Importantly, when the proliferative capacity of NK cells in vitro was used to stratify patients on the basis of cell expansion, patients whose NK cells proliferated <250-fold experienced significantly lower DFS and OS than those with ≥250-fold. Ex-vivo activation of NK cells restored tumor trafficking and reactivity, hence provided a therapeutic modality while their fold expansion could be a potentially significant prognostic indicator of OS and DFS in such patients.


Subject(s)
Killer Cells, Natural/immunology , Pancreatic Neoplasms/immunology , Tumor Escape/immunology , Adenocarcinoma/immunology , Aged , Carcinoma, Pancreatic Ductal/immunology , Cell Line, Tumor , Cell Proliferation/physiology , Female , Humans , K562 Cells , Lymphocytes, Tumor-Infiltrating/immunology , Male , Pancreatic Neoplasms/pathology , Prognosis , Tumor Microenvironment/immunology
10.
Stem Cell Reports ; 8(6): 1630-1644, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28552607

ABSTRACT

Bone marrow-derived mesenchymal stem cells (BMSCs) are proposed as the cells of origin of several subtypes of osteosarcoma (OS). However, signals that direct BMSCs to form different subtypes of OS are unclear. Here we show that the default tumor type from spontaneously transformed p53 knockout (p53_KO) BMSCs is osteoblastic OS. The development of this default tumor type caused by p53 loss can be overridden by various oncogenic signals: RAS reprograms p53_KO BMSCs into undifferentiated sarcoma, AKT enhances osteoblastic OS, while cFOS promotes chondroblastic OS formation. We focus on studying the mechanism of cFOS-induced chondroblastic OS formation. Integrated genome-wide studies reveal a regulatory mechanism whereby cFOS binds to the promoter of a key chondroblastic transcription factor, Sox9, and induces its transcription in BMSCs. Importantly, SOX9 mediates cFOS-induced cartilage formation in chondroblastic OS. In summary, oncogenes determine tumor types derived from BMSCs, and the cFOS-SOX9 axis is critical for chondroblastic OS formation.


Subject(s)
Bone Marrow Cells/cytology , Bone Neoplasms/pathology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Osteosarcoma/pathology , Proto-Oncogene Proteins c-fos/metabolism , SOX9 Transcription Factor/metabolism , Animals , Bone Neoplasms/metabolism , Bone Neoplasms/therapy , Cell Differentiation , Cellular Reprogramming , Chondrocytes/cytology , Chondrocytes/metabolism , Chondrogenesis , Humans , Mesenchymal Stem Cells/cytology , Mice , Mice, Knockout , Osteogenesis , Osteosarcoma/metabolism , Osteosarcoma/therapy , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-fos/genetics , SOX9 Transcription Factor/antagonists & inhibitors , SOX9 Transcription Factor/genetics , Signal Transduction , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , ras Proteins/metabolism
11.
PLoS Genet ; 12(2): e1005884, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26925584

ABSTRACT

The inactivation of p53 creates a major challenge for inducing apoptosis in cancer cells. An attractive strategy is to identify and subsequently target the survival signals in p53 defective cancer cells. Here we uncover a RUNX2-mediated survival signal in p53 defective cancer cells. The inhibition of this signal induces apoptosis in cancer cells but not non-transformed cells. Using the CRISPR technology, we demonstrate that p53 loss enhances the apoptosis caused by RUNX2 knockdown. Mechanistically, RUNX2 provides the survival signal partially through inducing MYC transcription. Cancer cells have high levels of activating histone marks on the MYC locus and concomitant high MYC expression. RUNX2 knockdown decreases the levels of these histone modifications and the recruitment of the Menin/MLL1 (mixed lineage leukemia 1) complex to the MYC locus. Two inhibitors of the Menin/MLL1 complex induce apoptosis in p53 defective cancer cells. Together, we identify a RUNX2-mediated epigenetic mechanism of the survival of p53 defective cancer cells and provide a proof-of-principle that the inhibition of this epigenetic axis is a promising strategy to kill p53 defective cancer cells.


Subject(s)
Bone Neoplasms/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Epigenesis, Genetic , Osteosarcoma/genetics , Tumor Suppressor Protein p53/genetics , Animals , Apoptosis/genetics , Binding Sites , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Survival/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor beta Subunit/genetics , Core Binding Factor beta Subunit/metabolism , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Genes, myc , Humans , Mice, Knockout , Osteosarcoma/pathology , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
12.
Stem Cells ; 33(4): 1304-19, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25524638

ABSTRACT

The tumor suppressor, p53, plays a critical role in suppressing osteosarcoma. Bone marrow stromal cells (BMSCs, also known as bone marrow-derived mesenchymal stem cells) have been suggested to give rise to osteosarcomas. However, the role of p53 in BMSCs has not been extensively explored. Here, we report that p53 regulates the lineage choice of mouse BMSCs (mBMSCs). Compared to mBMSCs with wild-type p53, mBMSCs deficient in p53 have enhanced osteogenic differentiation, but with similar adipogenic and chondrogenic differentiation. The role of p53 in inhibiting osteogenic lineage differentiation is mainly through the action of Runx2, a master transcription factor required for the osteogenic differentiation of mBMSCs. We find that p53 indirectly represses the expression of Runx2 by activating the microRNA-34 family, which suppresses the translation of Runx2. Since osteosarcoma may derive from BMSCs, we examined whether p53 has a role in the osteogenic differentiation of osteosarcoma cells and found that osteosarcoma cells with p53 deletion have higher levels of Runx2 and faster osteogenic differentiation than those with wild-type p53. A systems biology approach reveals that p53-deficient mBMSCs are more closely related to human osteosarcoma while mBMSCs with wild-type p53 are similar to normal human BMSCs. In summary, our results indicate that p53 activity can influence cell fate specification of mBMSCs, and provide molecular and cellular insights into the observation that p53 loss is associated with increased osteosarcoma incidence.


Subject(s)
Cell Differentiation/physiology , Mesenchymal Stem Cells/metabolism , Osteogenesis/physiology , Tumor Suppressor Protein p53/deficiency , Animals , Cell Line, Tumor , Cells, Cultured , Humans , Mice , Mice, Knockout
13.
Cell Biosci ; 3(1): 42, 2013 Oct 09.
Article in English | MEDLINE | ID: mdl-24171803

ABSTRACT

The viability and subtle developmental defects of p53 knockout mice suggest that p53 does not play major role in development. However, contradictory evidence also exists. This discrepancy mainly results from the lack of molecular and cellular mechanisms and the general fact that p53 activation requires stresses. Recent studies of p53 in mouse and human ES cells and induced pluripotent stem (iPS) cells shed new light on the mechanisms of the developmental roles of p53. This review summarizes these new studies that support the developmental roles of p53, highlights the possible underlying molecular mechanisms, and discusses the potential relationship between the developmental roles and the tumor suppressive function of p53. In summary, the molecular mechanisms underlying the developmental roles of p53 are emerging, and the developmental roles and tumor suppressive function of p53 may be closely related.

14.
Cell Cycle ; 11(9): 1796-803, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22453082

ABSTRACT

Upon genotoxic stress and during normal S phase, ATM phosphorylates the checkpoint clamp protein Rad9 in a manner that depends on Ser272. Ser272 is the only known ATM-dependent phosphorylation site in human Rad9. However, Ser272 phosphorylation is not required for survival or checkpoint activation after DNA damage. The physiological function of Ser272 remains elusive. Here, we show that ATM-dependent Rad9(Ser272) phosphorylation requires the MRN complex and controls repair pathways. Furthermore, the mutant cells accumulate large numbers of chromosome breaks and induce gross chromosomal rearrangements. Our findings establish a new and unexpected role for ATM: it phosphorylates the checkpoint clamp in order to control repair pathways, thereby maintaining genomic integrity during unperturbed cell cycle and upon DNA damage.


Subject(s)
Cell Cycle Checkpoints , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Genomic Instability , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Acid Anhydride Hydrolases , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Chromosome Breakage , DNA Breaks, Double-Stranded , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , HeLa Cells , Humans , MRE11 Homologue Protein , Morpholines/pharmacology , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Mutagenesis, Site-Directed , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oxidation-Reduction , Phosphorylation , Plasmids/genetics , Plasmids/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Pyrones/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Recombinational DNA Repair , Serine/metabolism , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics
15.
Mol Cell Biol ; 29(18): 4878-90, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19564421

ABSTRACT

In noradrenergic progenitors, Phox2a mediates cell cycle exit and neuronal differentiation by inducing p27(Kip1) transcription in response to activation of the cyclic AMP (cAMP) pathway. The mechanism of cAMP-mediated activation of Phox2a is unknown. We identified a cluster of phosphoserine-proline sites in Phox2a by mass spectrometry. Ser206 appeared to be the most prominent phosphorylation site. A phospho-Ser206 Phox2a antibody detected dephosphorylation of Phox2a that was dependent on activation of the cAMP pathway, which occurred prior to neuronal differentiation of noradrenergic CAD cells. Employing serine-to-alanine and serine-to-aspartic acid Phox2a substitution mutants expressed in inducible CAD cell lines, we demonstrated that the transcriptional activity of Phox2a is regulated by two sequential cAMP-dependent events: first, cAMP signaling promotes dephosphorylation of Phox2a in at least one site, Ser206, thereby allowing Phox2a to bind DNA and initiate p27(Kip1) transcription; second, following dephosphorylation of the phosphoserine cluster (Ser202 and Ser208), Phox2a becomes phosphorylated by protein kinase A (PKA) on Ser153, which prevents association of Phox2a with DNA and terminates p27(Kip1) transcription. This represents a novel mechanism by which the same stimulus, cAMP signaling, first activates Phox2a by dephosphorylation of Ser206 and then, after a built-in delay, inactivates Phox2a via PKA-dependent phosphorylation of Ser153, thereby modulating onset and duration of p27(Kip1) transcription.


Subject(s)
Cyclic AMP/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Homeodomain Proteins/metabolism , Signal Transduction , Transcription, Genetic , Alanine/genetics , Amino Acid Sequence , Amino Acid Substitution , Cell Differentiation , Cell Line , Chromatin Immunoprecipitation , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , DNA/metabolism , Gene Expression Regulation , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Intermediate Filament Proteins/metabolism , Mass Spectrometry , Membrane Glycoproteins/metabolism , Models, Biological , Molecular Sequence Data , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Peripherins , Phosphorylation , Phosphoserine/metabolism , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Time Factors
16.
Mol Cell Biol ; 26(23): 8826-39, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16982676

ABSTRACT

Mechanisms coordinating neural progenitor cell cycle exit and differentiation are incompletely understood. The cyclin-dependent kinase inhibitor p27(Kip1) is transcriptionally induced, switching specific neural progenitors from proliferation to differentiation. However, neuronal differentiation-specific transcription factors mediating p27(Kip1) transcription have not been identified. We demonstrate the homeodomain transcription factor Phox2a, required for central nervous system (CNS)- and neural crest (NC)-derived noradrenergic neuron differentiation, coordinates cell cycle exit and differentiation by inducing p27(Kip1) transcription. Phox2a transcription and activation in the CNS-derived CAD cell line and primary NC cells is mediated by combined cyclic AMP (cAMP) and bone morphogenetic protein 2 (BMP2) signaling. In the CAD cellular model, cAMP and BMP2 signaling initially induces proliferation of the undifferentiated precursors, followed by p27(Kip1) transcription, G(1) arrest, and neuronal differentiation. Small interfering RNA silencing of either Phox2a or p27(Kip1) suppresses p27(Kip1) transcription and neuronal differentiation, suggesting a causal link between p27(Kip1) expression and differentiation. Conversely, ectopic Phox2a expression via the Tet-off expression system promotes accelerated CAD cell neuronal differentiation and p27(Kip1) transcription only in the presence of cAMP signaling. Importantly, endogenous or ectopically expressed Phox2a activated by cAMP signaling binds homeodomain cis-acting elements of the p27(Kip1) promoter in vivo and mediates p27(Kip1)-luciferase expression in CAD and NC cells. We conclude that developmental cues of cAMP signaling causally link Phox2a activation with p27(Kip1) transcription, thereby coordinating neural progenitor cell cycle exit and differentiation.


Subject(s)
Cell Cycle/physiology , Cell Differentiation/physiology , Cyclic AMP/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Homeodomain Proteins/genetics , Neural Crest/embryology , Stem Cells/cytology , Transcription, Genetic , Animals , Cell Line , Cells, Cultured , Chromatin Immunoprecipitation , Coturnix/embryology , Cyclic AMP/physiology , Immunohistochemistry , Models, Biological , Neural Crest/cytology , Neural Crest/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Transfection
17.
Exp Mol Med ; 34(6): 401-10, 2002 Dec 31.
Article in English | MEDLINE | ID: mdl-12526081

ABSTRACT

Rat hippocampal precursor cells isolated from hippocampi of embryonic day 16.5 (E16.5) rat embryos were found to proliferate in the presence of basic fibroblast growth factor. Addition of soluble neural cell adhesion molecule (NCAM) to these precursor cells reduced cell proliferation in a dose dependent manner and enhanced the induction of precursor cells' differentiation to the neuronal lineage. Given these findings that NCAM induces the differentiation of hippocampal precursor cells, we investigated possible effects of NCAM on the expression of basic helix-loop-helix (bHLH) transcription factors during the differentiation. Soluble NCAM upregulated the transcription of bHLH transcription factors, neurogenin1 and NeuroD, but decreased HES5. Western blot analysis showed that NCAM increased the expression levels of CaMKII, p-MAPK, GluR1 and NR1 but decreased p-STAT3. These results support a role for NCAM in the inhibition of proliferation and the induction of neural differentiation of hippocampal neural precursor cells, and act as developmental regulators of the bHLH families, ultimately leading to the generation of glutamatergic neural cell types in the differentiation of hippocampal precursor cells.


Subject(s)
Cell Differentiation/drug effects , Hippocampus/cytology , Hippocampus/drug effects , Neural Cell Adhesion Molecules/pharmacology , Neurons/drug effects , Receptors, Glutamate/metabolism , Stem Cells/drug effects , Animals , Apoptosis/drug effects , Cell Division/drug effects , Cell Lineage/drug effects , Cells, Cultured , Helix-Loop-Helix Motifs , Neurons/cytology , Neurons/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Signal Transduction , Stem Cells/cytology , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...