Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
1.
Proc Natl Acad Sci U S A ; 119(41): e2207344119, 2022 10 11.
Article in English | MEDLINE | ID: mdl-36191214

ABSTRACT

Acyl-coenzyme A (CoA)-binding protein (ACBP), also known as diazepam-binding inhibitor (DBI), is an extracellular feedback regulator of autophagy. Here, we report that injection of a monoclonal antibody neutralizing ACBP/DBI (α-DBI) protects the murine liver against ischemia/reperfusion damage, intoxication by acetaminophen and concanavalin A, and nonalcoholic steatohepatitis caused by methionine/choline-deficient diet as well as against liver fibrosis induced by bile duct ligation or carbon tetrachloride. α-DBI downregulated proinflammatory and profibrotic genes and upregulated antioxidant defenses and fatty acid oxidation in the liver. The hepatoprotective effects of α-DBI were mimicked by the induction of ACBP/DBI-specific autoantibodies, an inducible Acbp/Dbi knockout or a constitutive Gabrg2F77I mutation that abolishes ACBP/DBI binding to the GABAA receptor. Liver-protective α-DBI effects were lost when autophagy was pharmacologically blocked or genetically inhibited by knockout of Atg4b. Of note, α-DBI also reduced myocardium infarction and lung fibrosis, supporting the contention that it mediates broad organ-protective effects against multiple insults.


Subject(s)
Diazepam Binding Inhibitor , Receptors, GABA-A , Animals , Mice , Acetaminophen , Antibodies, Monoclonal/metabolism , Antioxidants , Autoantibodies/metabolism , Autophagy , Carbon Tetrachloride , Carrier Proteins/genetics , Choline , Coenzyme A/metabolism , Concanavalin A/metabolism , Diazepam , Diazepam Binding Inhibitor/metabolism , Fatty Acids/metabolism , Fibrosis , Inflammation , Methionine
2.
Int J Mol Sci ; 24(7)2023 Mar 28.
Article in English | MEDLINE | ID: mdl-37047342

ABSTRACT

The B-cell lymphoma 2 (Bcl-2) family of proteins is the main regulator of apoptosis. However, multiple emerging evidence has revealed that Bcl-2 family proteins are also involved in cellular senescence. On the one hand, the different expression of these proteins determines the entry into senescence. On the other hand, entry into senescence modulates the expression of these proteins, generally conferring resistance to apoptosis. With some exceptions, senescent cells are characterized by the upregulation of antiapoptotic proteins and downregulation of proapoptotic proteins. Under physiological conditions, freshly formed tetraploid cells die by apoptosis due to the tetraploidy checkpoint. However, suppression of Bcl-2 associated x protein (Bax), as well as overexpression of Bcl-2, favors the appearance and survival of tetraploid cells. Furthermore, it is noteworthy that our laboratory has shown that the joint absence of Bax and Bcl-2 antagonist/killer (Bak) favors the entry into senescence of tetraploid cells. Certain microtubule inhibitory chemotherapies, such as taxanes and vinca alkaloids, induce the generation of tetraploid cells. Moreover, the combined use of inhibitors of antiapoptotic proteins of the Bcl-2 family with microtubule inhibitors increases their efficacy. In this review, we aim to shed light on the involvement of the Bcl-2 family of proteins in the senescence program activated after tetraploidization and the possibility of using this knowledge to create a new therapeutic strategy targeting cancer cells.


Subject(s)
Lymphoma, B-Cell , Proto-Oncogene Proteins c-bcl-2 , Humans , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , bcl-2 Homologous Antagonist-Killer Protein/genetics , Tetraploidy , Apoptosis Regulatory Proteins/metabolism , Lymphoma, B-Cell/metabolism , Apoptosis/physiology , bcl-X Protein/metabolism
3.
Mol Cell ; 53(5): 710-25, 2014 Mar 06.
Article in English | MEDLINE | ID: mdl-24560926

ABSTRACT

Acetyl-coenzyme A (AcCoA) is a major integrator of the nutritional status at the crossroads of fat, sugar, and protein catabolism. Here we show that nutrient starvation causes rapid depletion of AcCoA. AcCoA depletion entailed the commensurate reduction in the overall acetylation of cytoplasmic proteins, as well as the induction of autophagy, a homeostatic process of self-digestion. Multiple distinct manipulations designed to increase or reduce cytosolic AcCoA led to the suppression or induction of autophagy, respectively, both in cultured human cells and in mice. Moreover, maintenance of high AcCoA levels inhibited maladaptive autophagy in a model of cardiac pressure overload. Depletion of AcCoA reduced the activity of the acetyltransferase EP300, and EP300 was required for the suppression of autophagy by high AcCoA levels. Altogether, our results indicate that cytosolic AcCoA functions as a central metabolic regulator of autophagy, thus delineating AcCoA-centered pharmacological strategies that allow for the therapeutic manipulation of autophagy.


Subject(s)
Acetyl Coenzyme A/chemistry , Autophagy , Cytosol/enzymology , Gene Expression Regulation, Enzymologic , Adenosine Triphosphate/chemistry , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , Cytoplasm/metabolism , Cytosol/metabolism , E1A-Associated p300 Protein/chemistry , Green Fluorescent Proteins/metabolism , HCT116 Cells , HeLa Cells , Humans , Ketoglutaric Acids/chemistry , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Mitochondria/metabolism , RNA, Small Interfering/metabolism
4.
Immunol Rev ; 280(1): 165-174, 2017 11.
Article in English | MEDLINE | ID: mdl-29027230

ABSTRACT

Cancer cells are subjected to constant selection by the immune system, meaning that tumors that become clinically manifest have managed to subvert or hide from immunosurveillance. Immune control can be facilitated by induction of autophagy, as well as by polyploidization of cancer cells. While autophagy causes the release of ATP, a chemotactic signal for myeloid cells, polyploidization can trigger endoplasmic reticulum stress with consequent exposure of the "eat-me" signal calreticulin on the cell surface, thereby facilitating the transfer of tumor antigens into dendritic cells. Hence, both autophagy and polyploidization cause the emission of adjuvant signals that ultimately elicit immune control by CD8+ T lymphocytes. We investigated the possibility that autophagy and polyploidization might also affect the antigenicity of cancer cells by altering the immunopeptidome. Mass spectrometry led to the identification of peptides that were presented on major histocompatibility complex (MHC) class I molecules in an autophagy-dependent fashion or that were specifically exposed on the surface of polyploid cells, yet lost upon passage of such cells through immunocompetent (but not immunodeficient) mice. However, the preferential recognition of autophagy-competent and polyploid cells by the innate and cellular immune systems did not correlate with the preferential recognition of such peptides in vivo. Moreover, vaccination with such peptides was unable to elicit tumor growth-inhibitory responses in vivo. We conclude that autophagy and polyploidy increase the immunogenicity of cancer cells mostly by affecting their adjuvanticity rather than their antigenicity.


Subject(s)
Adjuvants, Immunologic , Antigens, Neoplasm/immunology , Cell Death , Immunologic Surveillance , Neoplasms/immunology , Adenosine Triphosphate/metabolism , Animals , Endoplasmic Reticulum Stress , Humans , Mice , Monitoring, Immunologic , Signal Transduction
5.
Mol Cell ; 48(5): 667-80, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-23084476

ABSTRACT

In a screen designed to identify novel inducers of autophagy, we discovered that STAT3 inhibitors potently stimulate the autophagic flux. Accordingly, genetic inhibition of STAT3 stimulated autophagy in vitro and in vivo, while overexpression of STAT3 variants, encompassing wild-type, nonphosphorylatable, and extranuclear STAT3, inhibited starvation-induced autophagy. The SH2 domain of STAT3 was found to interact with the catalytic domain of the eIF2α kinase 2 EIF2AK2, best known as protein kinase R (PKR). Pharmacological and genetic inhibition of STAT3 stimulated the activating phosphorylation of PKR and consequent eIF2α hyperphosphorylation. Moreover, PKR depletion inhibited autophagy as initiated by chemical STAT3 inhibitors or free fatty acids like palmitate. STAT3-targeting chemicals and palmitate caused the disruption of inhibitory STAT3-PKR interactions, followed by PKR-dependent eIF2α phosphorylation, which facilitates autophagy induction. These results unravel an unsuspected mechanism of autophagy control that involves STAT3 and PKR as interacting partners.


Subject(s)
Autophagy , Cytoplasm/enzymology , Eukaryotic Initiation Factor-2/metabolism , STAT3 Transcription Factor/metabolism , eIF-2 Kinase/metabolism , Animals , Autophagy/drug effects , Catalytic Domain , Cell Line, Tumor , Enzyme Activation , Eukaryotic Initiation Factor-2/deficiency , Eukaryotic Initiation Factor-2/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Docking Simulation , Palmitic Acid/pharmacology , Phosphorylation , Protein Conformation , Protein Interaction Domains and Motifs , Protein Interaction Mapping , RNA Interference , Recombinant Fusion Proteins/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/chemistry , STAT3 Transcription Factor/deficiency , STAT3 Transcription Factor/genetics , Signal Transduction , Time Factors , Transfection , eIF-2 Kinase/chemistry , eIF-2 Kinase/genetics , src Homology Domains
6.
Phys Chem Chem Phys ; 19(34): 22777-22784, 2017 Aug 30.
Article in English | MEDLINE | ID: mdl-28828426

ABSTRACT

The macroscopic and high temperature properties of lithium borophosphate glasses were determined in this contribution. Our data, obtained on 50Li2O-xB2O3-(50-x)P2O5 glasses, confirm a continuous and linear increase of the glass transition temperature with the B/P substitution but show a two-domain evolution of the kinetic fragility with a steep decrease in the low B2O3 region (0 ≤ x ≤ 10) followed by a moderate increase for higher B2O3 contents. In order to understand this different behaviour, the glass structure was investigated in detail using 1D and 2D 11B/31P correlation solid state nuclear magnetic resonance. The local and medium orders of borate units were determined by 1D MAS-NMR, 2D 11B DQSQ- and 11B(31P) D-HMQC NMR experiments. The latter NMR technique was also used to deeply interpret the 1D 31P MAS-NMR spectra. Altogether the data allow (i) highlighting of the presence of four borate and seven phosphate units, (ii) evaluation of the number of homopolar POP and mixed POB linkages, and (iii) contribute to a better understanding of the Tg and kinetic fragility evolution.

7.
Proc Natl Acad Sci U S A ; 111(8): 3020-5, 2014 Feb 25.
Article in English | MEDLINE | ID: mdl-24516128

ABSTRACT

Tetraploidy constitutes a genomically metastable state that can lead to aneuploidy and genomic instability. Tetraploid cells are frequently found in preneoplastic lesions, including intestinal cancers arising due to the inactivation of the tumor suppressor adenomatous polyposis coli (APC). Using a phenotypic screen, we identified resveratrol as an agent that selectively reduces the fitness of tetraploid cells by slowing down their cell cycle progression and by stimulating the intrinsic pathway of apoptosis. Selective killing of tetraploid cells was observed for a series of additional agents that indirectly or directly stimulate AMP-activated protein kinase (AMPK) including salicylate, whose chemopreventive action has been established by epidemiological studies and clinical trials. Both resveratrol and salicylate reduced the formation of tetraploid or higher-order polyploid cells resulting from the culture of human colon carcinoma cell lines or primary mouse epithelial cells lacking tumor protein p53 (TP53, best known as p53) in the presence of antimitotic agents, as determined by cytofluorometric and videomicroscopic assays. Moreover, oral treatment with either resveratrol or aspirin, the prodrug of salicylate, repressed the accumulation of tetraploid intestinal epithelial cells in the Apc(Min/+) mouse model of colon cancer. Collectively, our results suggest that the chemopreventive action of resveratrol and aspirin involves the elimination of tetraploid cancer cell precursors.


Subject(s)
Adenomatous Polyposis Coli/prevention & control , Aspirin/therapeutic use , Cell Death/drug effects , Epithelial Cells/drug effects , Stilbenes/therapeutic use , Tetraploidy , Animals , Aspirin/pharmacology , Cell Line, Tumor , Epithelial Cells/chemistry , Flow Cytometry , Image Processing, Computer-Assisted , In Situ Hybridization, Fluorescence , Mice , Mice, Inbred C57BL , Microscopy, Video , Resveratrol , Stilbenes/pharmacology
8.
Semin Cancer Biol ; 33: 86-92, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25749194

ABSTRACT

Cancer cells exposed to some forms of chemotherapy and radiotherapy die while eliciting an adaptive immune response. Such a functionally peculiar variant of apoptosis has been dubbed immunogenic cell death (ICD). One of the central events in the course of ICD is the activation of an endoplasmic reticulum (ER) stress response. This is instrumental for cells undergoing ICD to emit all the signals that are required for their demise to be perceived as immunogenic by the host, and culminates with the phosphorylation of eukaryotic translation initiation factor 2α (eIF2α). In particular, eIF2α phosphorylation is required for the pre-apoptotic exposure of the ER chaperone calreticulin (CALR) on the cell surface, which is a central determinant of ICD. Importantly, phosphorylated eIF2α can be quantified in both preclinical and clinical samples by immunoblotting or immunohistochemistry using phosphoneoepitope-specific monoclonal antibodies. Of note, the phosphorylation of eIF2α and CALR exposure do not necessarily correlate with each other, and neither of these parameters is sufficient for cell death to be perceived as immunogenic. Nonetheless, accumulating data indicate that assessing the degree of phosphorylation of eIF2α provides a convenient parameter to monitor ICD. Here, we discuss the role of the ER stress response in ICD and the potential value of eIF2α phosphorylation as a biomarker for this clinically relevant variant of apoptosis.


Subject(s)
Biomarkers/metabolism , Cell Death , Eukaryotic Initiation Factor-2/metabolism , Animals , Antigen-Presenting Cells/cytology , Apoptosis , Calreticulin/metabolism , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Epitopes/chemistry , Heat-Shock Proteins/metabolism , Humans , Mice , Phosphorylation , Protein Biosynthesis , Signal Transduction/genetics
9.
Trends Immunol ; 34(10): 471-81, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23891238

ABSTRACT

The immune system is intimately involved in the pathophysiology of several human disorders. Thus, excessive or chronic inflammation initiated by numerous insults exacerbates tissue damage and - at least in some settings - promotes oncogenesis. Nevertheless, immunosurveillance, the process whereby the immune system eliminates damaged, senescent and (pre-)malignant cells, appears to exert major homeostatic functions. Accumulating evidence indicates that defects in the molecular and cellular circuitries that underpin immune responses accelerate the course of chronic diseases, including hepatic cirrhosis and cancer. Along similar lines, the re-establishment of tissue homeostasis upon acute pathological insults such as ischemia appears to be delayed when normal immunological functions are naturally or experimentally compromised. Here, we propose that immunosurveillance is a key regulator of tissue homeostasis.


Subject(s)
Homeostasis/immunology , Immunologic Surveillance/physiology , Aging/immunology , Animals , Humans , Neoplasms/immunology
10.
EMBO J ; 29(7): 1272-84, 2010 Apr 07.
Article in English | MEDLINE | ID: mdl-20186124

ABSTRACT

Tetraploidy can constitute a metastable intermediate between normal diploidy and oncogenic aneuploidy. Here, we show that the absence of p53 is not only permissive for the survival but also for multipolar asymmetric divisions of tetraploid cells, which lead to the generation of aneuploid cells with a near-to-diploid chromosome content. Multipolar mitoses (which reduce the tetraploid genome to a sub-tetraploid state) are more frequent when p53 is downregulated and the product of the Mos oncogene is upregulated. Mos inhibits the coalescence of supernumerary centrosomes that allow for normal bipolar mitoses of tetraploid cells. In the absence of p53, Mos knockdown prevents multipolar mitoses and exerts genome-stabilizing effects. These results elucidate the mechanisms through which asymmetric cell division drives chromosomal instability in tetraploid cells.


Subject(s)
Carcinoma/metabolism , Colonic Neoplasms/metabolism , Genes, mos , Mitosis , Polyploidy , Tumor Suppressor Protein p53/metabolism , Aneuploidy , Animals , Carcinoma/genetics , Cell Line, Tumor , Centrosome/metabolism , Chromosomal Instability , Colonic Neoplasms/genetics , Female , Gene Deletion , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Nude , Tumor Suppressor Protein p53/genetics
11.
EMBO J ; 29(3): 619-31, 2010 Feb 03.
Article in English | MEDLINE | ID: mdl-19959994

ABSTRACT

In response to stress, cells start transcriptional and transcription-independent programs that can lead to adaptation or death. Here, we show that multiple inducers of autophagy, including nutrient depletion, trigger the activation of the IKK (IkappaB kinase) complex that is best known for its essential role in the activation of the transcription factor NF-kappaB by stress. Constitutively active IKK subunits stimulated autophagy and transduced multiple signals that operate in starvation-induced autophagy, including the phosphorylation of AMPK and JNK1. Genetic inhibition of the nuclear translocation of NF-kappaB or ablation of the p65/RelA NF-kappaB subunit failed to suppress IKK-induced autophagy, indicating that IKK can promote the autophagic pathway in an NF-kappaB-independent manner. In murine and human cells, knockout and/or knockdown of IKK subunits (but not that of p65) prevented the induction of autophagy in response to multiple stimuli. Moreover, the knockout of IKK-beta suppressed the activation of autophagy by food deprivation or rapamycin injections in vivo, in mice. Altogether, these results indicate that IKK has a cardinal role in the stimulation of autophagy by physiological and pharmacological stimuli.


Subject(s)
Autophagy/physiology , I-kappa B Kinase/physiology , Animals , Autophagy/genetics , Cells, Cultured , HeLa Cells , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Multiprotein Complexes/physiology , NF-kappa B/genetics , NF-kappa B/metabolism , NIH 3T3 Cells , Signal Transduction/physiology
12.
Front Oncol ; 13: 1292046, 2023.
Article in English | MEDLINE | ID: mdl-38169859

ABSTRACT

Breast cancer is the most common malignant neoplasm in women. Despite progress to date, 700,000 women worldwide died of this disease in 2020. Apparently, the prognostic markers currently used in the clinic are not sufficient to determine the most appropriate treatment. For this reason, great efforts have been made in recent years to identify new molecular biomarkers that will allow more precise and personalized therapeutic decisions in both primary and recurrent breast cancers. These molecular biomarkers include genetic and post-transcriptional alterations, changes in protein expression, as well as metabolic, immunological or microbial changes identified by multiple omics technologies (e.g., genomics, epigenomics, transcriptomics, proteomics, glycomics, metabolomics, lipidomics, immunomics and microbiomics). This review summarizes studies based on omics analysis that have identified new biomarkers for diagnosis, patient stratification, differentiation between stages of tumor development (initiation, progression, and metastasis/recurrence), and their relevance for treatment selection. Furthermore, this review highlights the importance of clinical trials based on multiomics studies and the need to advance in this direction in order to establish personalized therapies and prolong disease-free survival of these patients in the future.

13.
Methods Mol Biol ; 2279: 199-212, 2021.
Article in English | MEDLINE | ID: mdl-33683696

ABSTRACT

The success of anticancer interventions relies on their ability to ignite an anticancer immune response and to reinstate cancer immunosurveillance. Thus, high dose crizotinib can induce immunogenic cell death (ICD) in cancer cells. If combined with cisplatin, crizotinib sensitizes non-small cell lung cancers (NSCLC) to subsequent (but not simultaneous) immunotherapy with PD-1 immune checkpoint blockade, facilitating the cure of more than 90% of established orthotopic cancers in mice. Here, we detail protocols for the establishment and monitoring of transplantable orthotopic NSCLCs in syngeneic immunocompetent animals. Indeed, TC1 cells establish lung cancer upon their intravenous injection into the tail vein, while Lewis lung carcinoma (LLC) cells can be implanted intrathoracically to generate lung cancers. If transduced with luciferase, both TC1 and LLC cells form tumors that can be conveniently monitored by chemiluminescence. This type of NSCLC model is highly useful for the development of novel curative anticancer therapies.


Subject(s)
Carcinoma, Lewis Lung , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Neoplasm Transplantation , Optical Imaging , Animals , Carcinoma, Lewis Lung/diagnostic imaging , Carcinoma, Lewis Lung/metabolism , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Carcinoma, Non-Small-Cell Lung/metabolism , Humans , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/metabolism , Mice
14.
Methods Mol Biol ; 2267: 217-226, 2021.
Article in English | MEDLINE | ID: mdl-33786795

ABSTRACT

Mitotic catastrophe is an oncosuppressive mechanism that drives cells toward senescence or death when an error occurs during mitosis. Eukaryotic cells have developed adaptive signaling pathways to cope with stress. The phosphorylation on serine 51 of the eukaryotic translation initiation factor (eIF2α) is a highly conserved event in stress responses, including the one that is activated upon treatment with mitotic catastrophe inducing agents, such as microtubular poisons or actin blockers. The protocol described herein details a method to quantify the phosphorylation of eIF2α by high-throughput immunofluorescence microscopy. This method is useful to capture the 'integrated stress response', which is characterized by eIF2α phosphorylation in the context of mitotic catastrophe.


Subject(s)
Cell Death , Eukaryotic Initiation Factor-2/metabolism , Fluorescent Antibody Technique/methods , Mitosis , Phosphoproteins/metabolism , Animals , Antimitotic Agents/toxicity , Cell Line , Humans , Microscopy, Fluorescence/methods , Phosphorylation
15.
Methods Mol Biol ; 2267: 227-239, 2021.
Article in English | MEDLINE | ID: mdl-33786796

ABSTRACT

Mitotic catastrophe (MC) is a cell death modality induced by DNA damage that involves the activation of cell cycle checkpoints such as the "DNA structure checkpoint" and "spindle assembly checkpoint" (SAC) leading to aberrant mitosis. Depending on the signal, MC can drive the cell to death or to senescence. The suppression of MC favors aneuploidy. Several cancer therapies, included microtubular poisons and radiations, trigger MC. The clonogenic assay has been used to study the capacity of single cells to proliferate and to generate macroscopic colonies and to evaluate the efficacy of anticancer drugs. Nevertheless, this method cannot analyze MC events. Here, we report an improved technique based on the use of human colon cancer HCT116 stable expressing histone H2B-GFP and DsRed-centrin proteins, allowing to determine the capacity of cells to proliferate, and to determine changes in the nucleus and centrosomes.


Subject(s)
Cell Death , Cell Proliferation , Mitosis , Tumor Stem Cell Assay/methods , Antimitotic Agents/toxicity , Antineoplastic Agents/toxicity , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HCT116 Cells , Histones/genetics , Histones/metabolism , Humans
16.
Cell Death Dis ; 12(11): 978, 2021 10 21.
Article in English | MEDLINE | ID: mdl-34675191

ABSTRACT

Colorectal cancers (CRC) can be classified into four consensus molecular subtypes (CMS), among which CMS1 has the best prognosis, contrasting with CMS4 that has the worst outcome. CMS4 CRC is notoriously resistant against therapeutic interventions, as demonstrated by preclinical studies and retrospective clinical observations. Here, we report the finding that two clinically employed agents, everolimus (EVE) and plicamycin (PLI), efficiently target the prototypic CMS4 cell line MDST8. As compared to the prototypic CMS1 cell line LoVo, MDST8 cells treated with EVE or PLI demonstrated stronger cytostatic and cytotoxic effects, increased signs of apoptosis and autophagy, as well as a more pronounced inhibition of DNA-to-RNA transcription and RNA-to-protein translation. Moreover, nontoxic doses of EVE and PLI induced the shrinkage of MDST8 tumors in mice, yet had only minor tumor growth-reducing effects on LoVo tumors. Altogether, these results suggest that EVE and PLI should be evaluated for their clinical activity against CMS4 CRC.


Subject(s)
Adaptor Proteins, Signal Transducing/drug effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Cytoskeletal Proteins/drug effects , Everolimus/therapeutic use , Plicamycin/therapeutic use , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Proliferation , Colorectal Neoplasms/pathology , Everolimus/pharmacology , Humans , Mice , Plicamycin/pharmacology
17.
Cell Death Dis ; 12(11): 1039, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34725331

ABSTRACT

Pro-apoptotic multi-domain proteins of the BCL2 family such as BAX and BAK are well known for their important role in the induction of mitochondrial outer membrane permeabilization (MOMP), which is the rate-limiting step of the intrinsic pathway of apoptosis. Human or mouse cells lacking both BAX and BAK (due to a double knockout, DKO) are notoriously resistant to MOMP and cell death induction. Here we report the surprising finding that BAX/BAK DKO cells proliferate less than control cells expressing both BAX and BAK (or either BAX or BAK) when they are driven into tetraploidy by transient exposure to the microtubule inhibitor nocodazole. Mechanistically, in contrast to their BAX/BAK-sufficient controls, tetraploid DKO cells activate a senescent program, as indicated by the overexpression of several cyclin-dependent kinase inhibitors and the activation of ß-galactosidase. Moreover, DKO cells manifest alterations in ionomycin-mobilizable endoplasmic reticulum (ER) Ca2+ stores and store-operated Ca2+ entry that are affected by tetraploidization. DKO cells manifested reduced expression of endogenous sarcoplasmic/endoplasmic reticulum Ca2+ ATPase 2a (Serca2a) and transfection-enforced reintroduction of Serca2a, or reintroduction of an ER-targeted variant of BAK into DKO cells reestablished the same pattern of Ca2+ fluxes as observed in BAX/BAK-sufficient control cells. Serca2a reexpression and ER-targeted BAK also abolished the tetraploidy-induced senescence of DKO cells, placing ER Ca2+ fluxes downstream of the regulation of senescence by BAX/BAK. In conclusion, it appears that BAX/BAK prevent the induction of a tetraploidization-associated senescence program. Speculatively, this may contribute to the low incidence of cancers in BAX/BAK DKO mice and explain why human cancers rarely lose the expression of both BAX and BAK.


Subject(s)
Tetraploidy , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism , Animals , Calcium/metabolism , Calcium Signaling , Cell Line , Cellular Senescence , Clone Cells , Endoplasmic Reticulum/metabolism , Fibroblasts/metabolism , Humans , Mice, Inbred C57BL , Mice, Knockout , Microtubules/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , bcl-2 Homologous Antagonist-Killer Protein/deficiency , bcl-2-Associated X Protein/deficiency
19.
Nat Commun ; 11(1): 3819, 2020 07 30.
Article in English | MEDLINE | ID: mdl-32732875

ABSTRACT

Hormone receptor (HR)+ breast cancer (BC) causes most BC-related deaths, calling for improved therapeutic approaches. Despite expectations, immune checkpoint blockers (ICBs) are poorly active in patients with HR+ BC, in part reflecting the lack of preclinical models that recapitulate disease progression in immunocompetent hosts. We demonstrate that mammary tumors driven by medroxyprogesterone acetate (M) and 7,12-dimethylbenz[a]anthracene (D) recapitulate several key features of human luminal B HR+HER2- BC, including limited immune infiltration and poor sensitivity to ICBs. M/D-driven oncogenesis is accelerated by immune defects, demonstrating that M/D-driven tumors are under immunosurveillance. Safe nutritional measures including nicotinamide (NAM) supplementation efficiently delay M/D-driven oncogenesis by reactivating immunosurveillance. NAM also mediates immunotherapeutic effects against established M/D-driven and transplantable BC, largely reflecting increased type I interferon secretion by malignant cells and direct stimulation of immune effector cells. Our findings identify NAM as a potential strategy for the prevention and treatment of HR+ BC.


Subject(s)
Breast Neoplasms/therapy , Immunotherapy/methods , Niacinamide/administration & dosage , Receptor, ErbB-2/immunology , 9,10-Dimethyl-1,2-benzanthracene , Animals , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Carcinogenesis/drug effects , Carcinogenesis/immunology , Disease Progression , Female , Humans , Interferon Type I/immunology , Interferon Type I/metabolism , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/prevention & control , Medroxyprogesterone Acetate , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Receptor, ErbB-2/metabolism , Survival Analysis
20.
Apoptosis ; 14(4): 364-75, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19145485

ABSTRACT

It is still enigmatic under which circumstances cellular demise induces an immune response or rather remains immunologically silent. Moreover, the question remains open under which circumstances apoptotic, autophagic or necrotic cells are immunogenic or tolerogenic. Although apoptosis appears to be morphologically homogenous, recent evidence suggests that the pre-apoptotic surface-exposure of calreticulin may dictate the immune response to tumor cells that succumb to anticancer treatments. Moreover, the release of high-mobility group box 1 (HMGB1) during late apoptosis and secondary necrosis contributes to efficient antigen presentation and cytotoxic T-cell activation because HMGB1 can bind to Toll like receptor 4 on dendritic cells, thereby stimulating optimal antigen processing. Cell death accompanied by autophagy also may facilitate cross priming events. Apoptosis, necrosis and autophagy are closely intertwined processes. Often, cells manifest autophagy before they undergo apoptosis or necrosis, and apoptosis is generally followed by secondary necrosis. Whereas apoptosis and necrosis irreversibly lead to cell death, autophagy can clear cells from stress factors and thus facilitate cellular survival. We surmise that the response to cellular stress like chemotherapy or ionizing irradiation, dictates the immunological response to dying cells and that this immune response in turn determines the clinical outcome of anticancer therapies. The purpose of this review is to summarize recent insights into the immunogenicity of dying tumor cells as a function of the cell death modality.


Subject(s)
Apoptosis/immunology , Calreticulin/immunology , Immunity/immunology , Neoplasms/immunology , Animals , Autophagy , Cell Death/immunology , Humans , Models, Biological , Necrosis , Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL