Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Int J Mol Sci ; 24(3)2023 Jan 23.
Article in English | MEDLINE | ID: mdl-36768557

ABSTRACT

Cancer is characterized by persistent cell proliferation driven by aberrant cell cycle regulation and stimulation of cyclin-dependent kinases (CDKs). A very intriguing and potential approach for the development of antitumor medicines is the suppression of CDKs that lead to induction of apoptosis and cell cycle arrest. The shift of the cell cycle from the G0/G1 phase to the S phase, which is characterized by active transcription and synthesis, depends on the development of the cyclin D-CDK4/6 complex. A precise balance between anticancer activity and general toxicity is demonstrated by CDK inhibitors, which can specifically block CDK4/6 and control the cell cycle by reducing the G1 to S phase transition. CDK4/6 inhibitors have recently been reported to exhibit significant cell growth inhibition via modulating the tumour microenvironment in cancerous cells. One significant new understanding is that these inhibitors serve important functions in the interaction among tumour cells and the host immune system in addition to being cytostatic. Herein, we discuss the biological significance of CDK4/6 inhibitors in cancer therapeutics, as well as their biological impact on T cells and other important immune cells. Furthermore, we explore the integration of preclinical findings of these pharmaceuticals' ability to enhance antitumor immunity.


Subject(s)
Neoplasms , Tumor Microenvironment , Humans , Cell Cycle/physiology , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 4/immunology , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Cyclin-Dependent Kinase 6/immunology , Neoplasms/drug therapy , Neoplasms/immunology , Protein Serine-Threonine Kinases/metabolism , Tumor Microenvironment/immunology
2.
Cell Mol Life Sci ; 74(8): 1553-1566, 2017 04.
Article in English | MEDLINE | ID: mdl-27915416

ABSTRACT

G1 phase cell cycle proteins, such as cyclin-dependent kinase 6 (Cdk6) and its activating partners, the D-type cyclins, are important regulators of T-cell development and function. An F-box protein, called F-box only protein 7 (Fbxo7), acts as a cell cycle regulator by enhancing cyclin D-Cdk6 complex formation and stabilising levels of p27, a cyclin-dependent kinase inhibitor. We generated a murine model of reduced Fbxo7 expression to test its physiological role in multiple tissues and found that these mice displayed a pronounced thymic hypoplasia. Further analysis revealed that Fbxo7 differentially affected proliferation and apoptosis of thymocytes at various stages of differentiation in the thymus and also mature T-cell function and proliferation in the periphery. Paradoxically, Fbxo7-deficient immature thymocytes failed to undergo expansion in the thymus due to a lack of Cdk6 activity, while mature T cells showed enhanced proliferative capacity upon T-cell receptor engagement due to reduced p27 levels. Our studies reveal differential cell cycle regulation by Fbxo7 at different stages in T-cell development.


Subject(s)
Cyclin-Dependent Kinase 6/immunology , Cyclin-Dependent Kinase Inhibitor p27/immunology , F-Box Proteins/immunology , T-Lymphocytes/cytology , Animals , Apoptosis , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Cycle , Cell Differentiation , Cell Proliferation , Down-Regulation , F-Box Proteins/genetics , Female , Gene Deletion , Lymphocyte Activation , Male , Mice, Inbred C57BL , Mutation , T-Lymphocytes/immunology , Thymus Gland/cytology , Thymus Gland/immunology
3.
Clin Exp Rheumatol ; 29(2 Suppl 65): S6-9, 2011.
Article in English | MEDLINE | ID: mdl-21586211

ABSTRACT

INTRODUCTION: Accumulating evidences show that shared autoimmunity is critical for the pathogenesis of many inflammatory rheumatic conditions. Specific phenotype could arise from specific genes, and/or combination of genetic factors and environment. Systemic sclerosis (SSc) belongs to connective tissue disorders and recent data have highlighted strong associations with some autoimmunity genes shared with other autoimmune diseases. OBJECTIVES: To determine whether novel risk loci associated with rheumatoid arthritis (RA) may confer susceptibility to SSc. Single nucleotide polymorphism from CCL21, CD244 and CDK6 were tested for association. METHODS: SNPs harbouring association with RA, CCL21-rs2812378, CDK6-rs42041 and CD244-rs6682654 were genotyped in a cohort of 1031 SSc patients and 1014 controls. All individuals were of European Caucasian origin. RESULTS: The three polymorphisms were in Hardy-Weinberg equilibrium in the control population and allelic frequencies were similar to those expected in European populations. Allelic and genotypic frequencies for these three polymorphisms were found to be similar in SSc patients and controls. Moreover, sub-phenotype analyses in particular for subgroups having diffuse subcutaneous subtype, specific auto-antibodies or fibrosing alveolitis did not detect any difference between SSc patients and controls. CONCLUSIONS: These results obtained through a large cohort of European Caucasian SSc patients do not support the implication of CCL21, CD244 and CDK6 genes in the pathogenesis of SSc although these genes were recently identified as RA susceptibility genes.


Subject(s)
Antigens, CD , Arthritis, Rheumatoid/genetics , Autoimmunity/genetics , Chemokine CCL21 , Cyclin-Dependent Kinase 6 , Receptors, Immunologic , Scleroderma, Systemic/genetics , Adult , Antigens, CD/genetics , Antigens, CD/immunology , Arthritis, Rheumatoid/complications , Arthritis, Rheumatoid/immunology , Chemokine CCL21/genetics , Chemokine CCL21/immunology , Cyclin-Dependent Kinase 6/genetics , Cyclin-Dependent Kinase 6/immunology , Female , Gene Frequency , Genetic Association Studies , Genetic Predisposition to Disease , Genotype , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Scleroderma, Systemic/etiology , Scleroderma, Systemic/immunology , Signaling Lymphocytic Activation Molecule Family , White People
4.
Front Immunol ; 12: 650977, 2021.
Article in English | MEDLINE | ID: mdl-34248938

ABSTRACT

The cyclin-dependent kinase 6 (CDK6) regulates the transition through the G1-phase of the cell cycle, but also acts as a transcriptional regulator. As such CDK6 regulates cell survival or cytokine secretion together with STATs, AP-1 or NF-κB. In the hematopoietic system, CDK6 regulates T cell development and promotes leukemia and lymphoma. CDK4/6 kinase inhibitors are FDA approved for treatment of breast cancer patients and have been reported to enhance T cell-mediated anti-tumor immunity. The involvement of CDK6 in T cell functions remains enigmatic. We here investigated the role of CDK6 in CD8+ T cells, using previously generated CDK6 knockout (Cdk6-/-) and kinase-dead mutant CDK6 (Cdk6K43M) knock-in mice. RNA-seq analysis indicated a role of CDK6 in T cell metabolism and interferon (IFN) signaling. To investigate whether these CDK6 functions are T cell-intrinsic, we generated a T cell-specific CDK6 knockout mouse model (Cdk6fl/fl CD4-Cre). T cell-intrinsic loss of CDK6 enhanced mitochondrial respiration in CD8+ T cells, but did not impact on cytotoxicity and production of the effector cytokines IFN-γ and TNF-α by CD8+ T cells in vitro. Loss of CDK6 in peripheral T cells did not affect tumor surveillance of MC38 tumors in vivo. Similarly, while we observed an impaired induction of early responses to type I IFN in CDK6-deficient CD8+ T cells, we failed to observe any differences in the response to LCMV infection upon T cell-intrinsic loss of CDK6 in vivo. This apparent contradiction might at least partially be explained by the reduced expression of Socs1, a negative regulator of IFN signaling, in CDK6-deficient CD8+ T cells. Therefore, our data are in line with a dual role of CDK6 in IFN signaling; while CDK6 promotes early IFN responses, it is also involved in the induction of a negative feedback loop. These data assign CDK6 a role in the fine-tuning of cytokine responses.


Subject(s)
Antiviral Agents/immunology , CD8-Positive T-Lymphocytes/immunology , Cyclin-Dependent Kinase 6/immunology , Cytotoxicity, Immunologic/immunology , Interferons/immunology , Neoplasms, Experimental/immunology , Animals , Antiviral Agents/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cell Line , Cell Line, Tumor , Cyclin-Dependent Kinase 6/genetics , Cyclin-Dependent Kinase 6/metabolism , Humans , Interferons/metabolism , Lymphocytic choriomeningitis virus/immunology , Lymphocytic choriomeningitis virus/physiology , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neoplasms, Experimental/metabolism , Signal Transduction/immunology
5.
Front Immunol ; 12: 799171, 2021.
Article in English | MEDLINE | ID: mdl-35095879

ABSTRACT

Background: Ovarian cancer (OC) is one of the most lethal gynecologic cancers. Growing evidence has proven that CDK4/6 plays a key role in tumor immunity and the prognosis of many cancers. However, the expression and function of CDK4/6 in OC remain unclear. Therefore, we aimed to explore the influence of CDK4/6 in OC, especially on immunity. Methods: We analyzed CDK4/6 expression and prognosis using The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO) and Genotype Tissue Expression (GTEx) data. Subsequently, we used the cytoHubba plug-in of Cytoscape software and starBase to identify the noncoding RNAs (ncRNAs) regulating CDK4/6. Finally, we verified the effect of CDK4/6 on immunity in OC cell lines and animal models. Results: CDK4/6 expression was higher in OC tissues than in normal ovarian tissues, and the high expression levels of CDK4/6 contributed to the immunosuppressive state of OC and were thus related to the poor prognosis of OC patients. This was also in general agreement with the results of OC cell line and animal experiments. Mechanistically, the CDK4/6 inhibitor palbociclib increased the secretion of interferon (IFN)-γ and the interferon-stimulated gene (ISG) response, thereby upregulating the expression of antigen-presenting molecules; this effect was partly dependent on the STING pathway and thus activated immunity in OC. Additionally, according to public data, the LRRC75A-AS1-hsa-miR-330-5p axis could inhibit the immune response of OC patients by upregulating CDK4/6, leading to a poor prognosis. Conclusion: CDK4/6 affects the immune microenvironment of OC and correlates with the prognosis of OC patients.


Subject(s)
Cyclin-Dependent Kinase 4/immunology , Cyclin-Dependent Kinase 6/immunology , Gene Expression Regulation, Neoplastic/immunology , Ovarian Neoplasms/immunology , Transcriptome/immunology , Tumor Microenvironment/immunology , Animals , Cell Line, Tumor , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/genetics , Cyclin-Dependent Kinase 6/metabolism , Female , Gene Ontology , Humans , Kaplan-Meier Estimate , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice, Inbred C57BL , MicroRNAs/genetics , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/immunology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Piperazines/pharmacology , Prognosis , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Signal Transduction/genetics , Tumor Burden/drug effects , Tumor Burden/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics
6.
Immunotherapy ; 12(12): 861-867, 2020 08.
Article in English | MEDLINE | ID: mdl-32640856

ABSTRACT

Background: Despite the profound effect that checkpoint inhibitors and BRAF/MEK inhibitors have had on survival in patients with metastatic melanoma, treatment options remain limited for those who demonstrate poor response or develop resistance to these modalities. The prospect of tumor sensitization to these treatments is therefore an attractive one. Results: We describe the case of a patient who developed a sustained response to trametinib and pembrolizumab, despite prior resistance to both these therapies, after receiving treatment with a CDK4/6 inhibitor. Discussion: We further outline the preclinical data supporting a possible role for the use of CDK4/6 inhibitors in tumor sensitization to immunotherapy.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , Imidazoles/therapeutic use , Immunotherapy/methods , Melanoma/drug therapy , Oximes/therapeutic use , Pyridones/therapeutic use , Pyrimidinones/therapeutic use , Antibodies, Monoclonal, Humanized/immunology , Antineoplastic Agents/immunology , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 4/immunology , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Cyclin-Dependent Kinase 6/immunology , Humans , Imidazoles/immunology , Male , Melanoma/immunology , Middle Aged , Oximes/immunology , Pyridones/immunology , Pyrimidinones/immunology , Treatment Outcome
7.
Mol Immunol ; 44(4): 488-93, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16584776

ABSTRACT

Cigarette smoking causes profound suppression of pulmonary T cell responses, which is associated with increased susceptibility to respiratory tract infections and decreased tumor surveillance. We previously demonstrated that the phenolic compounds in cigarette tar inhibit blastogenesis and interfere with human T cell cycle progression. To identify the mechanism by which cell cycle arrest occurs, we examined the effects of these compounds on cyclin-dependent kinases (Cdk) that control the G0/G1 transition. We found that hydroquinone inhibited induction of Cdk4 and Cdk6 kinase activities by >80%, while catechol and phenol were markedly less potent. HQ did not affect mitogenic induction of the Cdk6 protein, but inhibited expression of cyclin D3 by >90% resulting in a dramatic reduction in proper Cdk6/Cyclin D3 complex formation.


Subject(s)
Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Smoking/adverse effects , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tars/toxicity , Cell Cycle/drug effects , Cell Cycle/immunology , Cell Proliferation/drug effects , Cells, Cultured , Cyclin-Dependent Kinase 4/immunology , Cyclin-Dependent Kinase 6/immunology , Enzyme Induction/drug effects , G1 Phase , Humans , Lymphocyte Activation/drug effects , Phenols/toxicity , Resting Phase, Cell Cycle , T-Lymphocytes/cytology
8.
Mol Cancer Res ; 16(10): 1454-1457, 2018 10.
Article in English | MEDLINE | ID: mdl-29934327

ABSTRACT

Inhibitors of the cyclin-dependent kinases 4 and 6 (CDK4/6) were originally designed to block proliferation and cell cycle progression of cancer cells in which the activity of these kinases is dysregulated. CDK4/6 inhibitors have already been FDA approved for the treatment of estrogen receptor (ER)-positive breast cancer and are being tested in numerous other cancer types. However, several recent studies have identified novel effects of CDK4/6 inhibitors on tumor growth, most notably an indirect effect resulting from the activation of immune surveillance. This Perspective discusses these recent observations, including the effects that CDK4/6 inhibitors may have on immune cells themselves. It is likely that CDK4/6 inhibitors will have a broader impact than their expected induction of cell cycle arrest in the treatment of human cancers. Mol Cancer Res; 16(10); 1454-7. ©2018 AACR.


Subject(s)
Breast Neoplasms/drug therapy , Cyclin-Dependent Kinase 4/immunology , Cyclin-Dependent Kinase 6/immunology , Protein Kinase Inhibitors/therapeutic use , Animals , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Breast Neoplasms/immunology , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/immunology , Cell Proliferation/drug effects , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Estrogen Receptor alpha/immunology , Female , Humans , MCF-7 Cells , Mice , Protein Kinase Inhibitors/immunology , Xenograft Model Antitumor Assays
9.
J Clin Invest ; 126(11): 4076-4087, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27701148

ABSTRACT

Radiotherapy causes dose-limiting toxicity and long-term complications in rapidly renewing tissues, including the gastrointestinal tract. Currently, there is no FDA-approved agent for the prevention or treatment of radiation-induced intestinal injury. In this study, we have shown that PD 0332991 (PD), an FDA-approved selective inhibitor of cyclin-dependent kinase 4/6 (CDK4/6), prevents radiation-induced lethal intestinal injury in mice. Treating mice with PD or a structurally distinct CDK4/6 inhibitor prior to radiation blocked proliferation and crypt apoptosis and improved crypt regeneration. PD treatment also enhanced LGR5+ stem cell survival and regeneration after radiation. PD was an on-target inhibitor of RB phosphorylation and blocked G1/S transition in the intestinal crypts. PD treatment strongly but reversibly inhibited radiation-induced p53 activation, which blocked p53-upregulated modulator of apoptosis-dependent (PUMA-dependent) apoptosis without affecting p21-dependent suppression of DNA damage accumulation, with a repair bias toward nonhomologous end joining. Further, deletion of PUMA synergized with PD treatment for even greater intestinal radioprotection. Our results demonstrate that the cell cycle critically regulates the DNA damage response and survival of intestinal stem cells and support the concept that pharmacological quiescence is a potentially highly effective and selective strategy for intestinal radioprotection.


Subject(s)
Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Intestinal Diseases/prevention & control , Piperazines/pharmacology , Pyridines/pharmacology , Radiation Injuries, Experimental/prevention & control , Stem Cells/immunology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/immunology , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/immunology , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Cycle/immunology , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/immunology , Cyclin-Dependent Kinase 6/genetics , Cyclin-Dependent Kinase 6/immunology , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/immunology , DNA Damage/genetics , DNA Damage/immunology , Intestinal Diseases/genetics , Intestinal Diseases/immunology , Mice , Mice, Knockout , Radiation Injuries, Experimental/genetics , Radiation Injuries, Experimental/immunology , Radiation Injuries, Experimental/pathology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/immunology , Stem Cells/pathology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/immunology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/immunology
11.
Int Immunopharmacol ; 9(7-8): 978-83, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19375524

ABSTRACT

In the present study, we aimed at examining the immunosuppressive activity of saikosaponin a, a triterpene saponin derived from Bupleurum falcatum L. (Umbelliferae), and the underlying mechanisms. Saikosaponin a significantly inhibited the proliferation and activation of T cells activated by concanavalin A (Con A) in a concentration-dependent manner. Additionally, it potently suppressed Con A-stimulated IL-2, IFN-gamma and TNF-alpha production in mouse T cells. Saikosaponin a also caused G0/G1 arrest of activated T cells through down-regulating the protein levels of CDK6 and Cyclin D3 and up-regulating the protein level of p27(kip). Furthermore, the compound dose-dependently induced apoptosis of Con A-activated T cells rather than those non-activated, as determined by Annexin V/PI staining. Besides, it induced a remarkable collapse of mitochondrial membrane potential and caused significant release of cytochrome c from mitochondria to cytosol. In summary, these results suggest that the G0/G1 arrest as well as the induction of apoptosis via mitochondrial pathway are involved in the immunosuppressive activity of saikosaponin a against activated T cells. This may herald a novel approach for further studies of saikosaponin a as a candidate for the treatment of inflammatory and autoimmune diseases.


Subject(s)
Bupleurum , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cyclins/metabolism , Oleanolic Acid/analogs & derivatives , Saponins/pharmacology , T-Lymphocytes/metabolism , Animals , Apoptosis/drug effects , Apoptosis/immunology , Autoimmune Diseases/drug therapy , Cell Cycle/drug effects , Cell Cycle/immunology , Cell Proliferation/drug effects , Cyclin D3 , Cyclin-Dependent Kinase 6/genetics , Cyclin-Dependent Kinase 6/immunology , Cyclin-Dependent Kinase 6/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/immunology , Cyclins/genetics , Cyclins/immunology , Cytochromes c/metabolism , Dose-Response Relationship, Drug , Down-Regulation , Female , Immunosuppression Therapy , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukin-2/genetics , Interleukin-2/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/immunology , Mice , Mice, Inbred BALB C , Oleanolic Acid/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL