Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Biochemistry (Mosc) ; 89(5): 839-852, 2024 May.
Article in English | MEDLINE | ID: mdl-38880645

ABSTRACT

Tumor-associated macrophages (TAMs) are an important component of the tumor microenvironment (TME) and the most abundant population of immune cells infiltrating a tumor. TAMs can largely determine direction of anti-tumor immune response by promoting it or, conversely, contribute to formation of an immunosuppressive TME that allows tumors to evade immune control. Through interactions with tumor cells or other cells in the microenvironment and, as a result of action of anti-cancer therapy, macrophages can enter senescence. In this review, we have attempted to summarize information available in the literature on the role of senescent macrophages in tumors. With the recent development of senolytic therapeutic strategies aimed at removing senescent cells from an organism, it seems important to discuss functions of the senescent macrophages and potential role of the senolytic drugs in reprogramming TAMs to enhance anti-tumor immune response and improve efficacy of cancer treatment.


Subject(s)
Cellular Senescence , Neoplasms , Tumor Microenvironment , Tumor-Associated Macrophages , Tumor Microenvironment/immunology , Humans , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/metabolism , Neoplasms/drug therapy , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Animals , Macrophages/immunology , Macrophages/metabolism , Biomarkers, Tumor/metabolism
2.
Int J Mol Sci ; 25(7)2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38612842

ABSTRACT

The tumor microenvironment (TME) plays an essential role in tumor progression and in modulating tumor response to anticancer therapy. Cellular senescence leads to a switch in the cell secretome, characterized by the senescence-associated secretory phenotype (SASP), which may regulate tumorigenesis. Senolytic therapy is considered a novel anticancer strategy that eliminates the deleterious effects of senescent cells in the TME. Here, we show that two different types of senolytic drugs, despite efficiently depleting senescent cells, have opposite effects on cancer-associated fibroblasts (CAFs) and their ability to regulate epithelial-mesenchymal transition (EMT). We found that senolytic drugs, navitoclax and the combination of dasatinib/quercetin, reduced the number of spontaneously senescent and TNF-induced senescent CAFs. Despite the depletion of senescent cells, the combination of dasatinib/quercetin versus navitoclax increased the secretion of the SASP pro-inflammatory cytokine IL-6. This differential effect correlated with the promotion of enhanced migration and EMT in MC38 colorectal cancer cells. Our results demonstrate that some senolytics may have side effects unrelated to their senolytic activity and may promote tumorigenesis. We argue for more careful and extensive studies of the effects of senolytics on various aspects of tumor progression and tumor resistance to therapy before the senolytic strategy is implemented in the clinic.


Subject(s)
Aniline Compounds , Cancer-Associated Fibroblasts , Senotherapeutics , Sulfonamides , Humans , Dasatinib/pharmacology , Quercetin/pharmacology , Carcinogenesis , Cell Transformation, Neoplastic , Epithelial-Mesenchymal Transition , Cytokines , Tumor Microenvironment
3.
Cell Death Discov ; 9(1): 345, 2023 Sep 19.
Article in English | MEDLINE | ID: mdl-37726289

ABSTRACT

Progress in the development of new sequencing techniques with wider accessibility and higher sensitivity of the protocol of deciphering genome particularities led to the discovery of a new phenomenon - clonal haematopoiesis. It is characterized by the presence in the bloodstream of elderly people a minor clonal population of cells with mutations in certain genes, but without any sign of disease related to the hematopoietic system. Here we will review this recent advancement in the field of clonal haematopoiesis and how it may affect the disease's development in old age.

4.
Nat Cell Biol ; 25(9): 1265-1278, 2023 09.
Article in English | MEDLINE | ID: mdl-37652981

ABSTRACT

Despite advances in four-factor (4F)-induced reprogramming (4FR) in vitro and in vivo, how 4FR interconnects with senescence remains largely under investigated. Here, using genetic and chemical approaches to manipulate senescent cells, we show that removal of p16High cells resulted in the 4FR of somatic cells into totipotent-like stem cells. These cells expressed markers of both pluripotency and the two-cell embryonic state, readily formed implantation-competent blastoids and, following morula aggregation, contributed to embryonic and extraembryonic lineages. We identified senescence-dependent regulation of nicotinamide N-methyltransferase as a key mechanism controlling the S-adenosyl-L-methionine levels during 4FR that was required for expression of the two-cell genes and acquisition of an extraembryonic potential. Importantly, a partial 4F epigenetic reprogramming in old mice was able to reverse several markers of liver aging only in conjunction with the depletion of p16High cells. Our results show that the presence of p16High senescent cells limits cell plasticity, whereas their depletion can promote a totipotent-like state and histopathological tissue rejuvenation during 4F reprogramming.


Subject(s)
Cell Plasticity , Cellular Reprogramming , Animals , Mice , Cellular Reprogramming/genetics , Aging/genetics , Embryo Implantation , Epigenomics
5.
Int J Mol Sci ; 23(19)2022 Sep 27.
Article in English | MEDLINE | ID: mdl-36232714

ABSTRACT

Acute myeloid leukemia (AML) is a rapidly progressing heterogeneous disease with a high mortality rate, which is characterized by hyperproliferation of atypical immature myeloid cells. The number of AML patients is expected to increase in the near future, due to the old-age-associated nature of AML and increased longevity in the human population. RUNX1 and CEBPA, key transcription factors (TFs) of hematopoiesis, are frequently and independently mutated in AML. RUNX1 and CEBPA can bind TET2 demethylase and attract it to their binding sites (TFBS) in cell lines, leading to DNA demethylation of the regions nearby. Since TET2 does not have a DNA-binding domain, TFs are crucial for its guidance to target genomic locations. In this paper, we show that RUNX1 and CEBPA mutations in AML patients affect the methylation of important regulatory sites that resulted in the silencing of several RUNX1 and CEBPA target genes, most likely in a TET2-dependent manner. We demonstrated that hypermethylation of TFBS in AML cells with RUNX1 mutations was associated with resistance to anticancer chemotherapy. Demethylation therapy restored expression of the RUNX1 target gene, BIK, and increased sensitivity of AML cells to chemotherapy. If our results are confirmed, mutations in RUNX1 could be an indication for prescribing the combination of cytotoxic and demethylation therapies.


Subject(s)
CCAAT-Enhancer-Binding Proteins , Core Binding Factor Alpha 2 Subunit , Leukemia, Myeloid, Acute , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , DNA/genetics , DNA/metabolism , DNA Methylation/genetics , Demethylation/drug effects , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mutation
6.
Nat Commun ; 12(1): 3622, 2021 06 15.
Article in English | MEDLINE | ID: mdl-34131120

ABSTRACT

PPM1D/Wip1 is a negative regulator of the tumor suppressor p53 and is overexpressed in several human solid tumors. Recent reports associate gain-of-function mutations of PPM1D in immune cells with worse outcomes for several human cancers. Here we show that mice with genetic knockout of Ppm1d or with conditional knockout of Ppm1d in the hematopoietic system, in myeloid cells, or in neutrophils all display significantly reduced growth of syngeneic melanoma or lung carcinoma tumors. Ppm1d knockout neutrophils infiltrate tumors extensively. Chemical inhibition of Wip1 in human or mouse neutrophils increases anti-tumor phenotypes, p53-dependent expression of co-stimulatory ligands, and proliferation of co-cultured cytotoxic T cells. These results suggest that inhibition of Wip1 in neutrophils enhances immune anti-tumor responses.


Subject(s)
DNA Damage , Immunity , Neutrophils/metabolism , Protein Phosphatase 2C/genetics , Protein Phosphatase 2C/metabolism , Animals , Antineoplastic Agents , Cell Line, Tumor , Cell Proliferation , Female , Humans , Lung , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , T-Lymphocytes , Tumor Microenvironment , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
7.
J Gastroenterol ; 56(5): 442-455, 2021 05.
Article in English | MEDLINE | ID: mdl-33782752

ABSTRACT

BACKGROUND: We previously showed that supernatants of Lactobacillus biofilms induced an anti-inflammatory response by affecting the secretion of macrophage-derived cytokines, which was abrogated upon immunodepletion of the stress protein GroEL. METHODS: We purified GroEL from L. reuteri and analysed its anti-inflammatory properties in vitro in human macrophages isolated from buffy coats, ex vivo in explants from human biopsies and in vivo in a mouse model of DSS induced intestinal inflammation. As a control, we used GroEL purified (LPS-free) from E. coli. RESULTS: We found that L. reuteri GroEL (but not E. coli GroEL) inhibited pro-inflammatory M1-like macrophages markers, and favored M2-like markers. Consequently, L. reuteri GroEL inhibited pro-inflammatory cytokines (TNFα, IL-1ß, IFNγ) while favouring an anti-inflammatory secretome. In colon tissues from human biopsies, L. reuteri GroEL was also able to decrease markers of inflammation and apoptosis (caspase 3) induced by LPS. In mice, we found that rectal administration of L. reuteri GroEL (but not E. coli GroEL) inhibited all signs of haemorrhagic colitis induced by DSS including intestinal mucosa degradation, rectal bleeding and weight loss. It also decreased intestinal production of inflammatory cytokines (such as IFNγ) while increasing anti-inflammatory IL-10 and IL-13. These effects were suppressed when animals were immunodepleted in macrophages. From a mechanistic point of view, the effect of L. reuteri GroEL seemed to involve TLR4, since it was lost in TRL4-/- mice, and the activation of a non-canonical TLR4 pathway. CONCLUSIONS: L. reuteri GroEL, by affecting macrophage inflammatory features, deserves to be explored as an alternative to probiotics.


Subject(s)
Chaperonin 60/pharmacology , Colon/drug effects , Inflammation/prevention & control , Lactobacillus/metabolism , Animals , Chaperonin 60/therapeutic use , Colon/physiopathology , Disease Models, Animal , Inflammation/drug therapy , Limosilactobacillus reuteri/drug effects , Limosilactobacillus reuteri/metabolism , Mice, Inbred BALB C , Statistics, Nonparametric
8.
PLoS Biol ; 18(7): e3000564, 2020 07.
Article in English | MEDLINE | ID: mdl-32701952

ABSTRACT

Amyloids are protein aggregates with a highly ordered spatial structure giving them unique physicochemical properties. Different amyloids not only participate in the development of numerous incurable diseases but control vital functions in archaea, bacteria and eukarya. Plants are a poorly studied systematic group in the field of amyloid biology. Amyloid properties have not yet been demonstrated for plant proteins under native conditions in vivo. Here we show that seeds of garden pea Pisum sativum L. contain amyloid-like aggregates of storage proteins, the most abundant one, 7S globulin Vicilin, forms bona fide amyloids in vivo and in vitro. Full-length Vicilin contains 2 evolutionary conserved ß-barrel domains, Cupin-1.1 and Cupin-1.2, that self-assemble in vitro into amyloid fibrils with similar physicochemical properties. However, Cupin-1.2 fibrils unlike Cupin-1.1 can seed Vicilin fibrillation. In vivo, Vicilin forms amyloids in the cotyledon cells that bind amyloid-specific dyes and possess resistance to detergents and proteases. The Vicilin amyloid accumulation increases during seed maturation and wanes at germination. Amyloids of Vicilin resist digestion by gastrointestinal enzymes, persist in canned peas, and exhibit toxicity for yeast and mammalian cells. Our finding for the first time reveals involvement of amyloid formation in the accumulation of storage proteins in plant seeds.


Subject(s)
Amyloid/metabolism , Pisum sativum/metabolism , Seed Storage Proteins/metabolism , Seeds/metabolism , Amyloid/ultrastructure , Detergents/pharmacology , Escherichia coli/metabolism , Ions , Pancreatin/metabolism , Pisum sativum/drug effects , Pepsin A/metabolism , Protein Aggregates , Protein Domains , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology , Saccharomyces cerevisiae/metabolism , Seed Storage Proteins/chemistry , Seed Storage Proteins/pharmacology , Seed Storage Proteins/ultrastructure
9.
Cell Death Differ ; 27(1): 117-129, 2020 01.
Article in English | MEDLINE | ID: mdl-31068676

ABSTRACT

Pro-survival stress-inducible chaperone HSP110 is the only HSP for which a mutation has been found in a cancer. Multicenter clinical studies demonstrated a direct association between HSP110 inactivating mutation presence and excellent prognosis in colorectal cancer patients. Here, we have combined crystallographic studies on human HSP110 and in silico modeling to identify HSP110 inhibitors that could be used in colorectal cancer therapy. Two molecules (foldamers 33 and 52), binding to the same cleft of HSP110 nucleotide-binding domain, were selected from a chemical library (by co-immunoprecipitation, AlphaScreening, Interference-Biolayer, Duo-link). These molecules block HSP110 chaperone anti-aggregation activity and HSP110 association to its client protein STAT3, thereby inhibiting STAT3 phosphorylation and colorectal cancer cell growth. These effects were strongly decreased in HSP110 knockdown cells. Foldamer's 33 ability to inhibit tumor growth was confirmed in two colorectal cancer animal models. Although tumor cell death (apoptosis) was noted after treatment of the animals with foldamer 33, no apparent toxicity was observed, notably in epithelial cells from intestinal crypts. Taken together, we identified the first HSP110 inhibitor, a possible drug-candidate for colorectal cancer patients whose unfavorable outcome is associated to HSP110.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/drug therapy , HSP110 Heat-Shock Proteins/antagonists & inhibitors , Animals , Antineoplastic Agents/toxicity , Cell Proliferation , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Crystallography, X-Ray , HSP110 Heat-Shock Proteins/chemistry , HSP110 Heat-Shock Proteins/metabolism , Humans , Mice , Models, Molecular , STAT3 Transcription Factor/metabolism
10.
Oncogene ; 38(15): 2767-2777, 2019 04.
Article in English | MEDLINE | ID: mdl-30542121

ABSTRACT

A multicenter clinical study demonstrated the presence of a loss-of-function HSP110 mutation in about 15% of colorectal cancers, which resulted from an alternative splicing and was produced at the detriment of wild-type HSP110. Patients expressing low levels of wild-type HSP110 had excellent outcomes (i.e. response to an oxaliplatin-based chemotherapy). Here, we show in vitro, in vivo, and in patients' biopsies that HSP110 co-localizes with DNA damage (γ-H2AX). In colorectal cancer cells, HSP110 translocates into the nucleus upon treatment with genotoxic chemotherapy such as oxaliplatin. Furthermore, we show that HSP110 interacts with the Ku70/Ku80 heterodimer, an essential element of the non-homologous end joining (NHEJ) repair machinery. We also demonstrate by evaluating the resolved 53BP1 foci that depletion in HSP110 impairs repair steps of the NHEJ pathway, which is associated with an increase in DNA double-strand breaks and in the cells' sensitivity to oxaliplatin. HSP110-depleted cells sensitization to oxaliplatin-induced DNA damage is abolished upon re-expression of HSP110. Confirming a role for HSP110 in DNA non-homologous repair, SCR7 and NU7026, two inhibitors of the NHEJ pathway, circumvents HSP110-induced resistance to chemotherapy. In conclusion, HSP110 through its interaction with the Ku70/80 heterodimer may participate in DNA repair, thereby inducing a protection against genotoxic therapy.


Subject(s)
Cell Nucleus/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , DNA End-Joining Repair/genetics , HSP110 Heat-Shock Proteins/genetics , Mutagens/pharmacology , Translocation, Genetic/genetics , Animals , Cell Line, Tumor , Cell Nucleus/drug effects , DNA Breaks, Double-Stranded/drug effects , DNA Damage/drug effects , DNA Damage/genetics , DNA End-Joining Repair/drug effects , DNA-Binding Proteins/genetics , HCT116 Cells , Humans , Ku Autoantigen/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Oxaliplatin/pharmacology , Translocation, Genetic/drug effects
12.
Oncotarget ; 7(21): 31563-71, 2016 May 24.
Article in English | MEDLINE | ID: mdl-26883196

ABSTRACT

Cells undergoing oncogenic transformation frequently inactivate tumor suppressor pathways that could prevent their uncontrolled growth. Among those pathways p53 and p38MAPK pathways play a critical role in regulation of cell cycle, senescence and cell death in response to activation of oncogenes, stress and DNA damage. Consequently, these two pathways are important in determining the sensitivity of tumor cells to anti-cancer treatment. Wild type p53-induced phosphatase, Wip1, is involved in governance of both pathways. Recently, strategies directed to manipulation with Wip1 activity proposed to advance current day anticancer treatment and novel chemical compounds synthesized to improve specificity of manipulation with Wip1 activity. Here we reviewed the history of Wip1 studies in vitro and in vivo, in genetically modified animal models that support Wip1 role in tumorigenesis through regulation of p53 and p38MAPK pathways. Based on our knowledge we propose several recommendations for future more accurate studies of Wip1 interactions with other pathways involved in tumorigenesis using recently developed tools and for adoption of Wip1 manipulation strategies in anti-cancer therapy.


Subject(s)
Protein Phosphatase 2C/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Cycle/genetics , Cell Transformation, Neoplastic/genetics , DNA Damage , Humans , Mutation , Protein Phosphatase 2C/genetics
13.
J Clin Invest ; 124(7): 3263-73, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24911145

ABSTRACT

The number of newly formed neurons declines rapidly during aging, and this decrease in neurogenesis is associated with decreased function of neural stem/progenitor cells (NPCs). Here, we determined that a WIP1-dependent pathway regulates NPC differentiation and contributes to the age-associated decline of neurogenesis. Specifically, we found that WIP1 is expressed in NPCs of the mouse subventricular zone (SVZ) and aged animals with genetically enhanced WIP1 expression exhibited higher NPC numbers and neuronal differentiation compared with aged WT animals. Additionally, augmenting WIP1 expression in aged animals markedly improved neuron formation and rescued a functional defect in fine odor discrimination in aged mice. We identified the WNT signaling pathway inhibitor DKK3 as a key downstream target of WIP1 and found that expression of DKK3 in the SVZ is restricted to NPCs. Using murine reporter strains, we determined that DKK3 inhibits neuroblast formation by suppressing WNT signaling and Dkk3 deletion or pharmacological activation of the WNT pathway improved neuron formation and olfactory function in aged mice. We propose that WIP1 controls DKK3-dependent inhibition of neuronal differentiation during aging and suggest that regulating WIP1 levels could prevent certain aspects of functional decline of the aging brain.


Subject(s)
Aging/metabolism , Aging/pathology , Neurogenesis/physiology , Phosphoprotein Phosphatases/metabolism , Wnt Signaling Pathway , Adaptor Proteins, Signal Transducing , Adult Stem Cells/metabolism , Adult Stem Cells/pathology , Animals , Brain/metabolism , Brain/pathology , Cell Differentiation , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neurogenesis/genetics , Olfactory Perception/physiology , Phosphoprotein Phosphatases/genetics , Protein Phosphatase 2C , Tumor Suppressor Protein p53/metabolism
14.
Cell Adh Migr ; 6(4): 333-43, 2012.
Article in English | MEDLINE | ID: mdl-22983193

ABSTRACT

Dendritic spine morphology is modulated by protein kinase p38, a mitogen-activated protein (MAPK), in the hippocampus. Protein p38MAPK is a substrate of wip1, a protein phosphatase. The role of wip1 in the central nervous system (CNS) has never been explored. Here, we report a novel function of wip1 in dendritic spine morphology and memory processes. Wip1 deficiency decreases dendritic spine size and density in pyramidal neurons of the hippocampal CA1 region. Simultaneously, impairments in object recognition tasks and contextual memory occur in wip1 deficient mice, but are reversed in wip1/p38 double mutant mice. Thus, our findings demonstrate that wip1 modulates dendritic morphology and memory processes through the p38MAPK signaling pathway. In addition to the well-characterized role of the wip1/p38MAPK in cell death and differentiation, we revealed the novel contribution of wip1 to cognition and dendritic spine morphology, which may suggest new approaches to treating neurodegenerative disorders.


Subject(s)
Cell Shape , Dendritic Spines/enzymology , MAP Kinase Signaling System , Memory , Phosphoprotein Phosphatases/deficiency , Analysis of Variance , Animals , Cells, Cultured , Conditioning, Psychological , Dendritic Spines/physiology , Fear , Form Perception , Freezing Reaction, Cataleptic , Hippocampus/cytology , Hippocampus/metabolism , Hippocampus/physiology , Maze Learning , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Phosphoprotein Phosphatases/genetics , Protein Phosphatase 2C , Pyramidal Cells/enzymology , Pyramidal Cells/ultrastructure , p38 Mitogen-Activated Protein Kinases/deficiency , p38 Mitogen-Activated Protein Kinases/genetics
15.
Cancer Lett ; 325(2): 117-24, 2012 Dec 28.
Article in English | MEDLINE | ID: mdl-22750096

ABSTRACT

HSP70 is a chaperone that accumulates in the cells after many different stresses promoting cell survival in response to the adverse conditions. In contrast to normal cells, most cancer cells abundantly express HSP70 at the basal level to resist to various insults at different stages of tumorigenesis and during anti-cancer treatment. This cancer cells addiction for HSP70 is the rational for its targeting in cancer therapy. Much effort has been dedicated in the last years for the active search of HSP70 inhibitors. Additionally, the recent clinical trials on highly promising inhibitors of another stress protein, HSP90, showed compensatory increase in HSP70 levels and raised the question of necessity to combine HSP90 inhibitors with simultaneous inhibition of HSP70. Here we analyzed the recent advancement in creation of novel HSP70 inhibitors and different strategies for their use in anti-cancer therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , HSP70 Heat-Shock Proteins/antagonists & inhibitors , Molecular Targeted Therapy , Neoplasm Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Apoptosis/physiology , Autophagy/physiology , Drug Design , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , HSP70 Heat-Shock Proteins/chemistry , HSP70 Heat-Shock Proteins/physiology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Humans , Molecular Chaperones/antagonists & inhibitors , Molecular Chaperones/physiology , Neoplasm Proteins/physiology , Neoplasms/immunology , Protein Structure, Tertiary/drug effects , Stress, Physiological
16.
Cell Cycle ; 11(10): 1883-7, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22544321

ABSTRACT

Wip1 is a stress-response phosphatase that negatively regulates several tumor suppressors, including p53. In a sizeable fraction of tumors, overexpression or amplification of Wip1 compromises p53 functions; inhibition of Wip1 activity is an attractive strategy for improving treatment of these tumors. However, over half of human tumors contain mutations in the p53 gene or have lost both alleles. Recently, we observed that in cancer cells lacking wild type p53, reduction of Wip1 expression was ineffective, whereas, surprisingly, overexpression of Wip1 increased anticancer drug sensitivity. The increased sensitivity resulted from activation of the intrinsic pathway of apoptosis through increased levels of the pro-apoptotic protein Bax and decreased levels of the anti-apoptotic protein Bcl-xL. We showed that interaction of Wip1 and the transcription factor RUNX2, specifically through dephosphorylation of RUNX2 phospho-S432, resulted in increased expression of Bax. Interestingly, overexpression of Wip1 increased drug sensitivity only in the p53-negative tumor cells while protecting the wild type p53-containing normal cells from drug-induced collateral injury. Here, we provide evidence that Wip1 overexpression decreases expression of Bcl-xL through negative regulation of NFκB activity. Thus, Wip1 overexpression increases the sensitivity of p53-negative cancer cells to anticancer drugs by separately affecting Bax and Bcl-xL protein levels.


Subject(s)
Apoptosis/drug effects , Phosphoprotein Phosphatases/metabolism , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism , Antineoplastic Agents/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cisplatin/pharmacology , Core Binding Factor Alpha 1 Subunit/metabolism , Humans , NF-kappa B/metabolism , Phosphoprotein Phosphatases/genetics , Phosphorylation , Promoter Regions, Genetic , Protein Phosphatase 2C , Transcription Factor RelA/metabolism , Tumor Suppressor Protein p53/genetics , bcl-2-Associated X Protein/genetics
17.
Proc Natl Acad Sci U S A ; 109(2): E68-75, 2012 Jan 10.
Article in English | MEDLINE | ID: mdl-22065775

ABSTRACT

The inactivation of the p53 tumor suppressor pathway in many cancers often increases their resistance to anticancer therapy. Here we show that a previously proposed strategy directed to Wip1 inhibition could be ineffective in tumors lacking p53. On the contrary, Wip1 overexpression sensitized these tumors to chemotherapeutic agents. This effect was mediated through interaction between Wip1 and RUNX2 that resulted, in response to anticancer treatment, in RUNX2-dependent transcriptional induction of the proapoptotic Bax protein. The potentiating effects of Wip1 overexpression on chemotherapeutic agents were directed only to tumor cells lacking p53. The overexpression of Wip1 in normal tissues provided protection from cisplatin-induced apoptosis through decreased strength of upstream signaling to p53. Thus, Wip1 phosphatase promotes apoptosis in p53-negative tumors and protects normal tissues during treatment with anticancer agents.


Subject(s)
Antineoplastic Agents/pharmacology , Core Binding Factor Alpha 1 Subunit/metabolism , Gene Expression Regulation, Neoplastic/physiology , Neoplasms/drug therapy , Phosphoprotein Phosphatases/metabolism , Tumor Suppressor Protein p53/deficiency , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Apoptosis/physiology , Blotting, Western , Cell Line , DNA Primers/genetics , Drug Synergism , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Immunohistochemistry , Immunoprecipitation , Mice , Neoplasms/metabolism , Plasmids/genetics , Protein Phosphatase 2C , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/metabolism
18.
Cancer Res ; 71(2): 484-95, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21224349

ABSTRACT

The inhibition of heat shock protein 70 (HSP70) is an emerging strategy in cancer therapy. Unfortunately, no specific inhibitors are clinically available. By yeast two-hybrid screening, we have identified multiple peptide aptamers that bind HSP70. When expressed in human tumor cells, two among these peptide aptamers-A8 and A17-which bind to the peptide-binding and the ATP-binding domains of HSP70, respectively, specifically inhibited the chaperone activity, thereby increasing the cells' sensitivity to apoptosis induced by anticancer drugs. The 13-amino acid peptide from the variable region of A17 (called P17) retained the ability to specifically inhibit HSP70 and induced the regression of subcutaneous tumors in vivo after local or systemic injection. This antitumor effect was associated with an important recruitment of macrophages and T lymphocytes into the tumor bed. Altogether, these data indicate that peptide aptamers or peptides that target HSP70 may be considered as novel lead compounds for cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Aptamers, Peptide/pharmacology , HSP70 Heat-Shock Proteins/antagonists & inhibitors , Molecular Targeted Therapy/methods , Peptides/pharmacology , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Aptamers, Peptide/chemistry , Aptamers, Peptide/genetics , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , HeLa Cells , Humans , Melanoma, Experimental/drug therapy , Mice , Mice, Inbred C57BL , Peptides/chemistry , Peptides/genetics , Protein Structure, Tertiary , Rats , Transfection
19.
Dev Cell ; 17(1): 142-9, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19619499

ABSTRACT

Aging is a complex organismal process that is controlled by genetic, environmental, and behavioral factors. Accumulating evidence supports a role for different cell cycle inhibitors in mammalian aging. Little is known, however, about the upstream signals that induce their expression. Here, we explore the role of p38MAPK by generating a dominant-negative allele (p38(AF)) in which activating phosphorylation sites Thr180 and Tyr182 are mutated. Heterozygous p38(AF) mice show a marked attenuation of p38-dependent signaling and age-induced expression of multiple cell cycle inhibitors in different organs, including pancreatic islets. As a result, aged p38(AF/+) mice show enhanced proliferation and regeneration of islets when compared to wild-type littermates. We further find an age-related reduction in expression of the p38-specific phosphatase Wip1. Wip1-deficient mice demonstrate decreased islet proliferation, while Wip1 overexpression rescues aging-related decline in proliferation and regenerative capacity. We propose that modulation of p38MAPK activity may provide new avenues for treating certain age-related degenerative diseases.


Subject(s)
Aging/physiology , Cell Cycle/physiology , Islets of Langerhans/physiology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Enzyme Activation , Humans , Islets of Langerhans/cytology , Kidney/metabolism , Liver/metabolism , Lung/metabolism , Mice , Mice, Inbred C57BL , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/metabolism , Protein Phosphatase 2C , Repressor Proteins/genetics , Repressor Proteins/metabolism , Spleen/metabolism , p38 Mitogen-Activated Protein Kinases/genetics
20.
Stem Cells ; 27(6): 1433-42, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19489034

ABSTRACT

Continual generation of new neural cells from adult neural stem/progenitor cells (NPCs) is an important component of life-long brain plasticity. However, the intrinsic regulation of this process remains poorly defined. Here we report that Wip1 phosphatase, previously studied in oncogenesis, functions as a crucial physiological regulator in adult neural cell generation. Wip1 deficiency resulted in a 90% decrease in new cell formation in adult olfactory bulb, accompanied by aberrantly decreased NPC amplification, stem cell frequency, and self-renewal. At a cellular level, Wip1 knockout NPCs exhibit a prolonged cell cycle, an accumulation at G(2) to M phase transition, and enhanced p53 activity. Interestingly, the impaired M-phase entry and NPC amplification of Wip1-null mice can be reversed in Wip1/p53 double-null mice. Importantly, there is no difference in NPC amplification between p53-null and Wip1/p53 double-null mice. Our data demonstrate that Wip1 regulates the generation of new neural cells in adult olfactory bulb specifically through p53-dependent M-phase entry of the NPC cell cycle.


Subject(s)
Adult Stem Cells/cytology , Neurogenesis/physiology , Neurons/cytology , Olfactory Bulb/cytology , Phosphoprotein Phosphatases/metabolism , Tumor Suppressor Protein p53/physiology , Adult Stem Cells/metabolism , Animals , Apoptosis/physiology , Blotting, Western , Cell Cycle , Cell Differentiation/genetics , Flow Cytometry , Immunohistochemistry , In Situ Nick-End Labeling , Mice , Mice, Knockout , Neurons/metabolism , Olfactory Bulb/metabolism , Phosphoprotein Phosphatases/genetics , Protein Phosphatase 2C , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...