Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters











Publication year range
1.
Nat Cardiovasc Res ; 3(9): 1049-1066, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39215106

ABSTRACT

Myocardial injury may ultimately lead to adverse ventricular remodeling and development of heart failure (HF), which is a major cause of morbidity and mortality worldwide. Given the slow pace and substantial costs of developing new therapeutics, drug repurposing is an attractive alternative. Studies of many organs, including the heart, highlight the importance of the immune system in modulating injury and repair outcomes. Glatiramer acetate (GA) is an immunomodulatory drug prescribed for patients with multiple sclerosis. Here, we report that short-term GA treatment improves cardiac function and reduces scar area in a mouse model of acute myocardial infarction and a rat model of ischemic HF. We provide mechanistic evidence indicating that, in addition to its immunomodulatory functions, GA exerts beneficial pleiotropic effects, including cardiomyocyte protection and enhanced angiogenesis. Overall, these findings highlight the potential repurposing of GA as a future therapy for a myriad of heart diseases.


Subject(s)
Disease Models, Animal , Drug Repositioning , Glatiramer Acetate , Animals , Glatiramer Acetate/therapeutic use , Glatiramer Acetate/pharmacology , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology , Mice, Inbred C57BL , Neovascularization, Physiologic/drug effects , Rats , Mice , Heart Failure/drug therapy , Ventricular Function, Left/drug effects , Rats, Sprague-Dawley , Cells, Cultured , Ventricular Remodeling/drug effects
3.
Nat Cancer ; 4(5): 629-647, 2023 05.
Article in English | MEDLINE | ID: mdl-37217651

ABSTRACT

Immunotherapy revolutionized treatment options in cancer, yet the mechanisms underlying resistance in many patients remain poorly understood. Cellular proteasomes have been implicated in modulating antitumor immunity by regulating antigen processing, antigen presentation, inflammatory signaling and immune cell activation. However, whether and how proteasome complex heterogeneity may affect tumor progression and the response to immunotherapy has not been systematically examined. Here, we show that proteasome complex composition varies substantially across cancers and impacts tumor-immune interactions and the tumor microenvironment. Through profiling of the degradation landscape of patient-derived non-small-cell lung carcinoma samples, we find that the proteasome regulator PSME4 is upregulated in tumors, alters proteasome activity, attenuates presented antigenic diversity and associates with lack of response to immunotherapy. Collectively, our approach affords a paradigm by which proteasome composition heterogeneity and function should be examined across cancer types and targeted in the context of precision oncology.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Antigen Presentation , Lung Neoplasms/pathology , Precision Medicine , Proteasome Endopeptidase Complex/metabolism , Tumor Microenvironment
4.
Nat Biotechnol ; 41(2): 239-251, 2023 02.
Article in English | MEDLINE | ID: mdl-36203013

ABSTRACT

Post-translational modification (PTM) of antigens provides an additional source of specificities targeted by immune responses to tumors or pathogens, but identifying antigen PTMs and assessing their role in shaping the immunopeptidome is challenging. Here we describe the Protein Modification Integrated Search Engine (PROMISE), an antigen discovery pipeline that enables the analysis of 29 different PTM combinations from multiple clinical cohorts and cell lines. We expanded the antigen landscape, uncovering human leukocyte antigen class I binding motifs defined by specific PTMs with haplotype-specific binding preferences and revealing disease-specific modified targets, including thousands of new cancer-specific antigens that can be shared between patients and across cancer types. Furthermore, we uncovered a subset of modified peptides that are specific to cancer tissue and driven by post-translational changes that occurred in the tumor proteome. Our findings highlight principles of PTM-driven antigenicity, which may have broad implications for T cell-mediated therapies in cancer and beyond.


Subject(s)
Neoplasms , Protein Processing, Post-Translational , Humans , Protein Processing, Post-Translational/genetics , Peptides/genetics , Antigens , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Neoplasms/genetics
5.
Nat Commun ; 12(1): 1893, 2021 03 25.
Article in English | MEDLINE | ID: mdl-33767202

ABSTRACT

B cells have essential functions in multiple sclerosis and in its mouse model, experimental autoimmune encephalomyelitis, both as drivers and suppressors of the disease. The suppressive effects are driven by a regulatory B cell (Breg) population that functions, primarily but not exclusively, via the production of IL-10. However, the mechanisms modulating IL-10-producing Breg abundance are poorly understood. Here we identify SLAMF5 for controlling IL-10+ Breg maintenance and function. In EAE, the deficiency of SLAMF5 in B cells causes accumulation of IL10+ Bregs in the central nervous system and periphery. Blocking SLAMF5 in vitro induces both human and mouse IL-10-producing Breg cells and increases their survival with a concomitant increase of a transcription factor, c-Maf. Finally, in vivo SLAMF5 blocking in EAE elevates IL-10+ Breg levels and ameliorates disease severity. Our results suggest that SLAMF5 is a negative moderator of IL-10+ Breg cells, and may serve as a therapeutic target in MS and other autoimmune diseases.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Interleukin-10/immunology , Signaling Lymphocytic Activation Molecule Family/metabolism , Animals , Cell Survival/immunology , Cells, Cultured , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Sclerosis/immunology , Signaling Lymphocytic Activation Molecule Family/antagonists & inhibitors , Signaling Lymphocytic Activation Molecule Family/genetics
6.
JCI Insight ; 6(4)2021 02 22.
Article in English | MEDLINE | ID: mdl-33465053

ABSTRACT

Multiple myeloma (MM) is characterized by an accumulation of malignant plasma cells (PCs) within the BM. The BM microenvironment supports survival of the malignant cells and is composed of cellular fractions that foster myeloma development and progression by suppression of the immune response. Despite major progress in understanding the biology and pathophysiology of MM, this disease is still incurable and requires aggressive treatment with significant side effects. CD84 is a self-binding immunoreceptor belonging to the signaling lymphocyte activation molecule (SLAM) family. Previously, we showed that CD84 bridges between chronic lymphocytic leukemia cells and their microenvironment, and it regulates T cell function. In the current study, we investigated the role of CD84 in MM. Our results show that MM cells express low levels of CD84. However, these cells secrete the cytokine macrophage migration inhibitory factor (MIF), which induces CD84 expression on cells in their microenvironment. Its activation leads to an elevation of expression of genes regulating differentiation to monocytic/granulocytic-myeloid-derived suppressor cells (M-MDSCs and G-MDSCs, respectively) and upregulation of PD-L1 expression on MDSCs, which together suppress T cell function. Downregulation of CD84 or its blocking reduce MDSC accumulation, resulting in elevated T cell activity and reduced tumor load. Our data suggest that CD84 might serve as a novel therapeutic target in MM.


Subject(s)
Multiple Myeloma/immunology , Signaling Lymphocytic Activation Molecule Family/genetics , Signaling Lymphocytic Activation Molecule Family/immunology , Signaling Lymphocytic Activation Molecule Family/metabolism , Tumor Microenvironment/immunology , Animals , B7-H1 Antigen , Cell Line, Tumor , Humans , Immunotherapy , Intramolecular Oxidoreductases/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Lymphocyte Activation , Macrophage Migration-Inhibitory Factors/metabolism , Mice , Multiple Myeloma/therapy , Myeloid-Derived Suppressor Cells/immunology , T-Lymphocytes/immunology
7.
Nat Commun ; 11(1): 409, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31964869

ABSTRACT

The Golgi is a dynamic organelle whose correct assembly is crucial for cellular homeostasis. Perturbations in Golgi structure are associated with numerous disorders from neurodegeneration to cancer. However, whether and how dispersal of the Golgi apparatus is actively regulated under stress, and the consequences of Golgi dispersal, remain unknown. Here we demonstrate that 26S proteasomes are associated with the cytosolic surface of Golgi membranes to facilitate Golgi Apparatus-Related Degradation (GARD) and degradation of GM130 in response to Golgi stress. The degradation of GM130 is dependent on p97/VCP and 26S proteasomes, and required for Golgi dispersal. Finally, we show that perturbation of Golgi homeostasis induces cell death of multiple myeloma in vitro and in vivo, offering a therapeutic strategy for this malignancy. Taken together, this work reveals a mechanism of Golgi-localized proteasomal degradation, providing a functional link between proteostasis control and Golgi architecture, which may be critical in various secretion-related pathologies.


Subject(s)
Golgi Apparatus/metabolism , Ionophores/therapeutic use , Multiple Myeloma/drug therapy , Proteasome Endopeptidase Complex/metabolism , Proteostasis/physiology , Animals , Apoptosis/drug effects , Autoantigens/metabolism , Cell Line, Tumor/transplantation , Disease Models, Animal , Golgi Apparatus/drug effects , HEK293 Cells , Humans , Intracellular Membranes/metabolism , Ionophores/pharmacology , Membrane Proteins/metabolism , Mice , Monensin/pharmacology , Monensin/therapeutic use , Multiple Myeloma/pathology , Proteolysis/drug effects , Proteostasis/drug effects , Ubiquitination/drug effects , Valosin Containing Protein/metabolism
8.
Front Immunol ; 10: 141, 2019.
Article in English | MEDLINE | ID: mdl-30833945

ABSTRACT

Antigen presentation on HLA molecules is a major mechanism by which the immune system monitors self and non-self-recognition. Importantly, HLA-I presentation has gained much attention through its role in eliciting anti-tumor immunity. Several determinants controlling the peptides presented on HLA have been uncovered, mainly through the study of model substrates and large-scale immunopeptidome analyses. These determinants include the relative abundances of proteins in the cell, the stability or turnover rate of these proteins and the binding affinities of a given peptide to the HLA haplotypes found in a cell. However, the regulatory principles involved in selection and regulation of specific antigens in response to tumor pro-inflammatory signals remain largely unknown. Here, we chose to examine the effect that TNFα and IFNγ stimulation may exert on the immunopeptidome landscape of lung cancer cells. We show that the expression of many of the proteins involved in the class I antigen presentation pathway are changed by pro-inflammatory cytokines. Further, we could show that increased expression of the HLA-B allomorph drives a significant change in HLA-bound antigens, independently of the significant changes observed in the cellular proteome. Finally, we observed increased HLA-B levels in correlation with tumor infiltration across the TCGA lung cancer cohorts. Taken together, our results suggest that the immunopeptidome landscape should be examined in the context of anti-tumor immunity whereby signals in the microenvironment may be critical in shaping and modulating this important aspect of host-tumor interactions.


Subject(s)
HLA-B Antigens/immunology , Interferon-gamma/pharmacology , Lung Neoplasms/immunology , Peptides/immunology , Tumor Necrosis Factor-alpha/pharmacology , A549 Cells , Humans , Proteome
9.
J Clin Invest ; 128(12): 5465-5478, 2018 12 03.
Article in English | MEDLINE | ID: mdl-30277471

ABSTRACT

Chronic lymphocytic leukemia (CLL) is characterized by clonal proliferation and progressive accumulation of mature B lymphocytes in the peripheral blood, lymphoid tissues, and bone marrow. CLL is characterized by profound immune defects leading to severe infectious complications. T cells are numerically, phenotypically, and functionally highly abnormal in CLL, with only limited ability to exert antitumor immune responses. Exhaustion of T cells has also been suggested to play an important role in antitumor responses. CLL-mediated T cell exhaustion is achieved by the aberrant expression of several inhibitory molecules on CLL cells and their microenvironment, prominently the programmed cell death ligand 1/programmed cell death 1 (PD-L1/PD-1) receptors. Previously, we showed that CD84, a member of the SLAM family of receptors, bridges between CLL cells and their microenvironment. In the current study, we followed CD84 regulation of T cell function. We showed that cell-cell interaction mediated through human and mouse CD84 upregulates PD-L1 expression on CLL cells and in their microenvironment and PD-1 expression on T cells. This resulted in suppression of T cell responses and activity in vitro and in vivo. Thus, our results demonstrate a role for CD84 in the regulation of immune checkpoints by leukemia cells and identify CD84 blockade as a therapeutic strategy to reverse tumor-induced immune suppression.


Subject(s)
B7-H1 Antigen/immunology , Gene Expression Regulation, Leukemic/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Neoplasm Proteins/immunology , Programmed Cell Death 1 Receptor/immunology , Signaling Lymphocytic Activation Molecule Family/immunology , Animals , B7-H1 Antigen/genetics , Gene Expression Regulation, Leukemic/genetics , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Programmed Cell Death 1 Receptor/genetics , Signaling Lymphocytic Activation Molecule Family/genetics
10.
Nat Biotechnol ; 2018 Oct 22.
Article in English | MEDLINE | ID: mdl-30346940

ABSTRACT

Cellular function is critically regulated through degradation of substrates by the proteasome. To enable direct analysis of naturally cleaved proteasomal peptides under physiological conditions, we developed mass spectrometry analysis of proteolytic peptides (MAPP), a method for proteasomal footprinting that allows for capture, isolation and analysis of proteasome-cleaved peptides. Application of MAPP to cancer cell lines as well as primary immune cells revealed dynamic modulation of the cellular degradome in response to various stimuli, such as proinflammatory signals. Further, we performed analysis of minute amounts of clinical samples by studying cells from the peripheral blood of patients with systemic lupus erythematosus (SLE). We found increased degradation of histones in patient immune cells, thereby suggesting a role of aberrant proteasomal degradation in the pathophysiology of SLE. Thus, MAPP offers a broadly applicable method to facilitate the study of the cellular-degradation landscape in various cellular conditions and diseases involving changes in proteasomal degradation, including protein aggregation diseases, autoimmunity and cancer.

11.
J Immunol ; 199(8): 2745-2757, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28904129

ABSTRACT

The control of lymphoid homeostasis is the result of a very fine balance between lymphocyte production, proliferation, and apoptosis. In this study, we focused on the role of T cells in the maintenance/survival of the mature naive peripheral B cell population. We show that naive B and T cells interact via the signaling lymphocyte activation molecule (SLAM) family receptor, SLAMF6. This interaction induces cell type-specific signals in both cell types, mediated by the SLAM-associated protein (SAP) family of adaptors. This signaling results in an upregulation of the expression of the cytokine migration inhibitory factor in the T cells and augmented expression of its receptor CD74 on the B cell counterparts, consequently enhancing B cell survival. Furthermore, in X-linked lymphoproliferative disease patients, SAP deficiency reduces CD74 expression, resulting in the perturbation of B cell maintenance from the naive stage. Thus, naive T cells regulate B cell survival in a SLAMF6- and SAP-dependent manner.


Subject(s)
B-Lymphocyte Subsets/physiology , B-Lymphocytes/physiology , Blood Cells/physiology , Lymphoproliferative Disorders/immunology , Signaling Lymphocytic Activation Molecule Associated Protein/metabolism , Signaling Lymphocytic Activation Molecule Family/metabolism , T-Lymphocytes/physiology , Animals , Antibodies, Blocking/administration & dosage , Cell Communication , Cell Differentiation , Cell Proliferation , Cell Survival , Cells, Cultured , Homeostasis , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Small Interfering/genetics , Signaling Lymphocytic Activation Molecule Associated Protein/genetics , Signaling Lymphocytic Activation Molecule Family/genetics
12.
J Immunol ; 198(12): 4659-4671, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28507030

ABSTRACT

Chemokines and chemokine receptors establish a complex network modulating immune cell migration and localization. These molecules were also suggested to mediate the differentiation of leukocytes; however, their intrinsic, direct regulation of lymphocyte fate remained unclear. CCR2 is the main chemokine receptor inducing macrophage and monocyte recruitment to sites of inflammation, and it is also expressed on T cells. To assess whether CCR2 directly regulates T cell responses, we followed the fates of CCR2-/- T cells in T cell-specific inflammatory models. Our in vitro and in vivo results show that CCR2 intrinsically mediates the expression of inflammatory T cell cytokines, and its absence on T cells results in attenuated colitis progression. Moreover, CCR2 deficiency in T cells promoted a program inducing the accumulation of Foxp3+ regulatory T cells, while decreasing the levels of Th17 cells in vivo, indicating that CCR2 regulates the immune response by modulating the effector/regulatory T ratio.


Subject(s)
Immunity, Cellular , Receptors, CCR2/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Cell Movement , Colitis/immunology , Cytokines/genetics , Cytokines/immunology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Macrophages/immunology , Mice , Receptors, CCR2/deficiency , Receptors, CCR2/genetics , Receptors, CCR2/immunology , Receptors, CCR5/immunology , Receptors, CCR5/metabolism , Th17 Cells/immunology , Th17 Cells/physiology
13.
Proc Natl Acad Sci U S A ; 114(3): 562-567, 2017 01 17.
Article in English | MEDLINE | ID: mdl-28031488

ABSTRACT

CD74 is a cell-surface receptor for the cytokine macrophage migration inhibitory factor. Macrophage migration inhibitory factor binding to CD74 induces its intramembrane cleavage and the release of its cytosolic intracellular domain (CD74-ICD), which regulates cell survival. In the present study, we characterized the transcriptional activity of CD74-ICD in chronic lymphocytic B cells. We show that following CD74 activation, CD74-ICD interacts with the transcription factors RUNX (Runt related transcription factor) and NF-κB and binds to proximal and distal regulatory sites enriched for genes involved in apoptosis, immune response, and cell migration. This process leads to regulation of expression of these genes. Our results suggest that identifying targets of CD74 will help in understanding of essential pathways regulating B-cell survival in health and disease.

14.
PLoS One ; 9(12): e115683, 2014.
Article in English | MEDLINE | ID: mdl-25531780

ABSTRACT

The naturally occurring triterpenoid betulinic acid (BA) shows pronounced polypharmacology ranging from anti-inflammatory to anti-lipogenic activities. Recent evidence suggests that rather diverse cellular signaling events may be attributed to the same common upstream switch in cellular metabolism. In this study we therefore examined the metabolic changes induced by BA (10 µM) administration, with focus on cellular glucose metabolism. We demonstrate that BA elevates the rates of cellular glucose uptake and aerobic glycolysis in mouse embryonic fibroblasts with concomitant reduction of glucose oxidation. Without eliciting signs of obvious cell death BA leads to compromised mitochondrial function, increased expression of mitochondrial uncoupling proteins (UCP) 1 and 2, and liver kinase B1 (LKB1)-dependent activation AMP-activated protein kinase. AMPK activation accounts for the increased glucose uptake and glycolysis which in turn are indispensable for cell viability upon BA treatment. Overall, we show for the first time a significant impact of BA on cellular bioenergetics which may be a central mediator of the pleiotropic actions of BA.


Subject(s)
AMP-Activated Protein Kinases/physiology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Glucose/metabolism , Glycolysis/drug effects , Protein Serine-Threonine Kinases/physiology , Triterpenes/pharmacology , Animals , Blotting, Western , Cell Proliferation/drug effects , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Energy Metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice , Mice, Knockout , Mitochondria/drug effects , Mitochondria/metabolism , Oxidation-Reduction , Pentacyclic Triterpenes , Phosphorylation/drug effects , RNA, Messenger/genetics , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Betulinic Acid
15.
Biochim Biophys Acta ; 1830(10): 4813-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23811337

ABSTRACT

BACKGROUND: Peroxisome proliferator-activated receptor gamma (PPARγ) agonists are clinically used to counteract hyperglycemia. However, so far experienced unwanted side effects, such as weight gain, promote the search for new PPARγ activators. METHODS: We used a combination of in silico, in vitro, cell-based and in vivo models to identify and validate natural products as promising leads for partial novel PPARγ agonists. RESULTS: The natural product honokiol from the traditional Chinese herbal drug Magnolia bark was in silico predicted to bind into the PPARγ ligand binding pocket as dimer. Honokiol indeed directly bound to purified PPARγ ligand-binding domain (LBD) and acted as partial agonist in a PPARγ-mediated luciferase reporter assay. Honokiol was then directly compared to the clinically used full agonist pioglitazone with regard to stimulation of glucose uptake in adipocytes as well as adipogenic differentiation in 3T3-L1 pre-adipocytes and mouse embryonic fibroblasts. While honokiol stimulated basal glucose uptake to a similar extent as pioglitazone, it did not induce adipogenesis in contrast to pioglitazone. In diabetic KKAy mice oral application of honokiol prevented hyperglycemia and suppressed weight gain. CONCLUSION: We identified honokiol as a partial non-adipogenic PPARγ agonist in vitro which prevented hyperglycemia and weight gain in vivo. GENERAL SIGNIFICANCE: This observed activity profile suggests honokiol as promising new pharmaceutical lead or dietary supplement to combat metabolic disease, and provides a molecular explanation for the use of Magnolia in traditional medicine.


Subject(s)
Biological Products/pharmacology , Biphenyl Compounds/pharmacology , Lignans/pharmacology , PPAR gamma/agonists , 3T3-L1 Cells , Adipose Tissue/cytology , Adipose Tissue/drug effects , Animals , Biological Products/isolation & purification , Biphenyl Compounds/isolation & purification , Cell Differentiation/drug effects , Diabetes Mellitus, Experimental/physiopathology , HEK293 Cells , Humans , Lignans/isolation & purification , Mice , Molecular Docking Simulation
16.
PLoS One ; 8(4): e61755, 2013.
Article in English | MEDLINE | ID: mdl-23630612

ABSTRACT

Peroxisome proliferator-activated receptor gamma (PPARγ) is a key regulator of glucose and lipid metabolism and therefore an important pharmacological target to combat metabolic diseases. Since the currently used full PPARγ agonists display serious side effects, identification of novel ligands, particularly partial agonists, is highly relevant. Searching for new active compounds, we investigated extracts of the underground parts of Notopterygium incisum, a medicinal plant used in traditional Chinese medicine, and observed significant PPARγ activation using a PPARγ-driven luciferase reporter model. Activity-guided fractionation of the dichloromethane extract led to the isolation of six polyacetylenes, which displayed properties of selective partial PPARγ agonists in the luciferase reporter model. Since PPARγ activation by this class of compounds has so far not been reported, we have chosen the prototypical polyacetylene falcarindiol for further investigation. The effect of falcarindiol (10 µM) in the luciferase reporter model was blocked upon co-treatment with the PPARγ antagonist T0070907 (1 µM). Falcarindiol bound to the purified human PPARγ receptor with a Ki of 3.07 µM. In silico docking studies suggested a binding mode within the ligand binding site, where hydrogen bonds to Cys285 and Glu295 are predicted to be formed in addition to extensive hydrophobic interactions. Furthermore, falcarindiol further induced 3T3-L1 preadipocyte differentiation and enhanced the insulin-induced glucose uptake in differentiated 3T3-L1 adipocytes confirming effectiveness in cell models with endogenous PPARγ expression. In conclusion, we identified falcarindiol-type polyacetylenes as a novel class of natural partial PPARγ agonists, having potential to be further explored as pharmaceutical leads or dietary supplements.


Subject(s)
Apiaceae/chemistry , Diynes/pharmacology , Fatty Alcohols/pharmacology , PPAR gamma/agonists , Plant Extracts/pharmacology , 3T3-L1 Cells , Adipogenesis , Animals , Binding Sites , Deoxyglucose/metabolism , Diynes/chemistry , Diynes/isolation & purification , Fatty Alcohols/chemistry , Fatty Alcohols/isolation & purification , Genes, Reporter , HEK293 Cells , Humans , Luciferases/biosynthesis , Luciferases/genetics , Mice , Molecular Docking Simulation , PPAR gamma/chemistry , PPAR gamma/metabolism , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Polyynes/chemistry , Polyynes/isolation & purification , Polyynes/pharmacology , Protein Binding , Transcriptional Activation/drug effects
17.
Future Med Chem ; 4(4): 395-424, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22416772

ABSTRACT

Malignant cells in chronic lymphocytic leukemia (CLL) and related diseases are heterogeneous and consist primarily of long-lived resting cells in the periphery and a minor subset of dividing cells in proliferating centers. Both cell populations have different molecular signatures that play a major role in determining their sensitivity to therapy. Contemporary approaches to treating CLL are heavily reliant on cytotoxic chemotherapeutics. However, none of the current treatment regimens can be considered curative. Pharmacological CDK inhibitors have extended the repertoire of potential drugs for CLL. Multi-targeted CDK inhibitors affect CDKs involved in regulating both cell cycle progression and transcription. Their interference with transcriptional elongation represses anti-apoptotic proteins and, thus, promotes the induction of apoptosis. Importantly, there is evidence that treatment with CDK inhibitors can overcome resistance to therapy. The pharmacological CDK inhibitors have great potential for use in combination with other therapeutics and represent promising tools for the development of new curative treatments for CLL.


Subject(s)
Apoptosis/drug effects , Cyclin-Dependent Kinases/antagonists & inhibitors , Forkhead Transcription Factors/physiology , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Protein Kinase Inhibitors/pharmacology , Repressor Proteins/physiology , Small Molecule Libraries/pharmacology , Cell Cycle/drug effects , Clinical Trials as Topic , Cyclin-Dependent Kinases/metabolism , DNA-Binding Proteins/metabolism , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , MicroRNAs/metabolism , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-bcl-6 , Small Molecule Libraries/therapeutic use , Tumor Microenvironment
18.
Expert Opin Investig Drugs ; 20(12): 1611-28, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22017180

ABSTRACT

INTRODUCTION: The progression of the mammalian cell cycle is driven by the transient activation of complexes consisting of cyclins and cyclin-dependent kinases (CDKs). Loss of control over the cell cycle results in accelerated cell division and malignant transformation and can be caused by the upregulation of cyclins, the aberrant activation of CDKs or the inactivation of cellular CDK inhibitors. For these reasons, cell cycle regulators are regarded as very promising therapeutic targets for the treatment of human malignancies. AREAS COVERED: This review covers the structures and anti-breast cancer activity of selected pharmacological pan-specific CDK inhibitors. Multi-targeted CDK inhibitors affect CDKs involved in the regulation of both cell cycle progression and transcriptional control. The inhibition of CDK7/CDK9 has a serious impact on the activity of RNA polymerase II; when its carboxy-terminal domain is unphosphorylated, it is unable to recruit the cofactors required for transcriptional elongation, resulting in a global transcriptional block. Multi-targeted inhibition of CDKs represses anti-apoptotic proteins and thus promotes the induction of apoptosis. Moreover, the inhibition of CDK7 in estrogen receptor (ER)-positive breast cancer cells prevents activating phosphorylation of ER-α. EXPERT OPINION: These diverse modes of action make multi-targeted CDK inhibitors promising drugs for the treatment of breast cancers.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/physiopathology , Cell Cycle/physiology , Cyclin-Dependent Kinase Inhibitor Proteins/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Division/drug effects , Cell Line, Tumor , Female , Humans , Molecular Targeted Therapy , Receptors, Estrogen/metabolism
19.
Ann N Y Acad Sci ; 1171: 257-63, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19723063

ABSTRACT

Expression of the human papillomavirus-encoded oncoproteins E6 and E7 in human HeLa cervical carcinoma cells results in their escape from the proper control of the cell cycle progression. Therefore, their susceptibility to agents modulating cell cycle differs from that in cells in which the control of cell cycle regulation is intact. Recently, a number of experimental studies revealed that polyphenols, especially resveratrol, could exert a strong antiproliferative effect. Polyphenols (e.g., resveratrol or epicatechins), potent antioxidant agents, are abundant components of our diet and, therefore, may not only affect the proliferation of healthy cells in the organism but also modulate the action of distinct anticancer drugs. Indeed, it has been shown that resveratrol enhances the antimitotic effect exerted by roscovitine (ROSC), a potent cyclin-dependent kinase inhibitor, on human MCF-7 breast cancer cells. In the present contribution the action of resveratrol alone and in combination with ROSC on human HeLa cells was determined. Resveratrol inhibited proliferation of exponentially growing HeLa cells. Exposure of HeLa cells to 50 micromol/L resveratrol blocked cells in the S phase in a time-dependent manner. After 12 h the population of G(2)/M-phase cells completely disappeared, and during a further 12 h the frequency of S-phase cells markedly increased and reached approximately 90%. Thus, resveratrol synchronized HeLa cells in the S phase. After removal of resveratrol, synchronized HeLa cells rapidly progressed through the cell cycle. Four hours after medium change, more than 70% of cells moved into the G(2)/M phase. Moreover, resveratrol combined with ROSC enhanced the antiproliferative action of resveratrol.


Subject(s)
Cell Cycle/drug effects , Cell Proliferation/drug effects , Purines/pharmacology , Stilbenes/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Cell Survival/drug effects , Cyclin-Dependent Kinases/antagonists & inhibitors , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Dose-Response Relationship, Drug , Drug Synergism , Flow Cytometry , HeLa Cells , Humans , Protein Kinase Inhibitors/pharmacology , Resveratrol , Ribonucleotide Reductases/antagonists & inhibitors , Roscovitine , Time Factors
20.
J Cell Physiol ; 219(2): 459-69, 2009 May.
Article in English | MEDLINE | ID: mdl-19170070

ABSTRACT

Data on the biological effects of some overexpressed oncogenes and their cooperation with cellular factors are, at least partially, contradictory. There are reports on the strong pro-apoptotic action of temperature-sensitive (ts) p53(135val) in transformed cells at permissive temperature. However, in our experience very high levels of p53(135val) induce in transformed rat cells at permissive temperature cell cycle arrest but not apoptosis. Comparison of the experimental protocols reveals that cells used for transfection strongly differ. Therefore, we decided to explore the impact of primary cells used for generation of cell clones on the biological effects evoked by p53 and c-Ha-Ras. In the present study, we used primary rat cells (RECs) isolated from rat embryos of different age: at 13.5 gd (y) and 15.5 gd (o). We immortalized rat cells using ts p53(135val) mutant and additionally generated transformed cells after co-transfection with oncogenic Ras. The RECs were transfected with a constitutively activated Ha-Ras protein, a mutation that is found in a wide variety of human tumors. The ts p53(135Val) mutant, switching between wild-type (wt) and mutant conformation, offers the possibility to study the escape from p53-mediated cell cycle control in a model of malignant transformation in cells with the same genetic background. Surprisingly, the kinetics of cell proliferation at non-permissive temperature and that of cell cycle arrest at 32 degrees C strongly differed between cell clones established from yRECs and oRECs, thereby indicating that overexpression of genes such as ts p53(135Val) mutant and oncogenic-Ha-Ras does not fully override the intrinsic cellular program.


Subject(s)
Cell Cycle/physiology , Environment , Proto-Oncogene Proteins p21(ras)/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line, Transformed , Cell Proliferation , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/physiology , Female , Gestational Age , Humans , Mice , Pregnancy , Proto-Oncogene Proteins p21(ras)/genetics , Rats , Temperature , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL