Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
2.
J Virol ; 97(7): e0065223, 2023 07 27.
Article in English | MEDLINE | ID: mdl-37310263

ABSTRACT

HIV-1 (HIV) infects CD4+ T cells, the gradual depletion of which can lead to AIDS in the absence of antiretroviral therapy (ART). Some cells, however, survive HIV infection and persist as part of the latently infected reservoir that causes recurrent viremia after ART cessation. Improved understanding of the mechanisms of HIV-mediated cell death could lead to a way to clear the latent reservoir. Death induced by survival gene elimination (DISE), an RNA interference (RNAi)-based mechanism, kills cells through short RNAs (sRNAs) with toxic 6-mer seeds (positions 2 to 7 of sRNA). These toxic seeds target the 3' untranslated region (UTR) of mRNAs, decreasing the expression of hundreds of genes critical for cell survival. In most cells under normal conditions, highly expressed cell-encoded nontoxic microRNAs (miRNAs) block access of toxic sRNAs to the RNA-induced silencing complex (RISC) that mediates RNAi, promoting cell survival. HIV has been shown to inhibit the biogenesis of host miRNAs in multiple ways. We now report that HIV infection of cells deficient in miRNA expression or function results in enhanced RISC loading of an HIV-encoded miRNA HIV-miR-TAR-3p, which can kill cells by DISE through a noncanonical (positions 3 to 8) 6-mer seed. In addition, cellular RISC-bound sRNAs shift to lower seed viability. This also occurs after latent HIV provirus reactivation in J-Lat cells, suggesting independence of permissiveness of cells to viral infection. More precise targeting of the balance between protective and cytotoxic sRNAs could provide new avenues to explore novel cell death mechanisms that could be used to kill latent HIV. IMPORTANCE Several mechanisms by which initial HIV infection is cytotoxic to infected cells have been reported and involve various forms of cell death. Characterizing the mechanisms underlying the long-term survival of certain T cells that become persistent provirus reservoirs is critical to developing a cure. We recently discovered death induced by survival gene elimination (DISE), an RNAi-based mechanism of cell death whereby toxic short RNAs (sRNAs) containing 6-mer seed sequences (exerting 6-mer seed toxicity) targeting essential survival genes are loaded into RNA-induced silencing complex (RISC) complexes, resulting in inescapable cell death. We now report that HIV infection in cells with low miRNA expression causes a shift of mostly cellular RISC-bound sRNAs to more toxic seeds. This could prime cells to DISE and is further enhanced by the viral microRNA (miRNA) HIV-miR-TAR-3p, which carries a toxic noncanonical 6-mer seed. Our data provide multiple new avenues to explore novel cell death mechanisms that could be used to kill latent HIV.


Subject(s)
HIV Infections , HIV-1 , MicroRNAs , Humans , HIV-1/physiology , Virus Latency/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , RNA-Induced Silencing Complex/metabolism
3.
Biochem Soc Trans ; 51(1): 21-29, 2023 02 27.
Article in English | MEDLINE | ID: mdl-36629505

ABSTRACT

The role of CD95/Fas ligand (CD95L/FasL) in the induction of CD95-mediated extrinsic apoptosis is well characterized. Trimerized, membrane-bound CD95L ligates the CD95 receptor activating downstream signaling resulting in the execution of cells by caspase proteins. However, the expression of CD95L has been reported to induce cell death in contexts in which this pathway is unlikely to be activated, such as in cell autonomous activation induced cell death (AICD) and in CD95-resistant cancer cell lines. Recent data suggests that the CD95L mRNA exerts toxicity through death induced by survival gene elimination (DISE). DISE results from the targeting of networks of survival genes by toxic short RNA (sRNA)s in the RNA-induced silencing complex (RISC). CD95L mRNA contributes to this death directly, through the processing of its mRNA into toxic sRNAs that are loaded into the RISC, and indirectly, by promoting the loading of other toxic sRNAs. Interestingly, CD95L is not the only mRNA that is processed and loaded into the RISC. Protein-coding mRNAs involved in protein translation are also selectively loaded. We propose a model in which networks of mRNA-derived sRNAs modulate DISE, with networks of genes providing non-toxic RISC substrate sRNAs that protect against DISE, and opposing networks of stress-activated genes that produce toxic RISC substrate sRNAs that promote DISE.


Subject(s)
Apoptosis , fas Receptor , Fas Ligand Protein/genetics , Fas Ligand Protein/metabolism , fas Receptor/metabolism , Apoptosis/physiology , Caspases , RNA, Messenger/genetics
4.
PLoS Comput Biol ; 18(3): e1010022, 2022 03.
Article in English | MEDLINE | ID: mdl-35358200

ABSTRACT

microRNAs (miRNAs) are (18-22nt long) noncoding short (s)RNAs that suppress gene expression by targeting the 3' untranslated region of target mRNAs. This occurs through the seed sequence located in position 2-7/8 of the miRNA guide strand, once it is loaded into the RNA induced silencing complex (RISC). G-rich 6mer seed sequences can kill cells by targeting C-rich 6mer seed matches located in genes that are critical for cell survival. This results in induction of Death Induced by Survival gene Elimination (DISE), through a mechanism we have called 6mer seed toxicity. miRNAs are often quantified in cells by aligning the reads from small (sm)RNA sequencing to the genome. However, the analysis of any smRNA Seq data set for predicted 6mer seed toxicity requires an alternative workflow, solely based on the exact position 2-7 of any short (s)RNA that can enter the RISC. Therefore, we developed SPOROS, a semi-automated pipeline that produces multiple useful outputs to predict and compare 6mer seed toxicity of cellular sRNAs, regardless of their nature, between different samples. We provide two examples to illustrate the capabilities of SPOROS: Example one involves the analysis of RISC-bound sRNAs in a cancer cell line (either wild-type or two mutant lines unable to produce most miRNAs). Example two is based on a publicly available smRNA Seq data set from postmortem brains (either from normal or Alzheimer's patients). Our methods (found at https://github.com/ebartom/SPOROS and at Code Ocean: https://doi.org/10.24433/CO.1732496.v1) are designed to be used to analyze a variety of smRNA Seq data in various normal and disease settings.


Subject(s)
MicroRNAs , 3' Untranslated Regions , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Messenger/genetics , Seeds/genetics , Sequence Analysis, RNA/methods
5.
Genes Dev ; 29(7): 732-45, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25838542

ABSTRACT

Glioblastoma multiforme (GBM) is a lethal, therapy-resistant brain cancer consisting of numerous tumor cell subpopulations, including stem-like glioma-initiating cells (GICs), which contribute to tumor recurrence following initial response to therapy. Here, we identified miR-182 as a regulator of apoptosis, growth, and differentiation programs whose expression level is correlated with GBM patient survival. Repression of Bcl2-like12 (Bcl2L12), c-Met, and hypoxia-inducible factor 2α (HIF2A) is of central importance to miR-182 anti-tumor activity, as it results in enhanced therapy susceptibility, decreased GIC sphere size, expansion, and stemness in vitro. To evaluate the tumor-suppressive function of miR-182 in vivo, we synthesized miR-182-based spherical nucleic acids (182-SNAs); i.e., gold nanoparticles covalently functionalized with mature miR-182 duplexes. Intravenously administered 182-SNAs penetrated the blood-brain/blood-tumor barriers (BBB/BTB) in orthotopic GBM xenografts and selectively disseminated throughout extravascular glioma parenchyma, causing reduced tumor burden and increased animal survival. Our results indicate that harnessing the anti-tumor activities of miR-182 via safe and robust delivery of 182-SNAs represents a novel strategy for therapeutic intervention in GBM.


Subject(s)
Apoptosis/genetics , Cell Differentiation/genetics , Glioblastoma/genetics , MicroRNAs/metabolism , Animals , Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/physiopathology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Glioblastoma/drug therapy , Glioblastoma/physiopathology , Humans , Mice , Mice, SCID , MicroRNAs/administration & dosage , MicroRNAs/genetics , Muscle Proteins/genetics , Muscle Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Survival Analysis
6.
Genes Dev ; 28(11): 1191-203, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24840202

ABSTRACT

Tumor metastasis remains the major cause of cancer-related death, but its molecular basis is still not well understood. Here we uncovered a splicing-mediated pathway that is essential for breast cancer metastasis. We show that the RNA-binding protein heterogeneous nuclear ribonucleoprotein M (hnRNPM) promotes breast cancer metastasis by activating the switch of alternative splicing that occurs during epithelial-mesenchymal transition (EMT). Genome-wide deep sequencing analysis suggests that hnRNPM potentiates TGFß signaling and identifies CD44 as a key downstream target of hnRNPM. hnRNPM ablation prevents TGFß-induced EMT and inhibits breast cancer metastasis in mice, whereas enforced expression of the specific CD44 standard (CD44s) splice isoform overrides the loss of hnRNPM and permits EMT and metastasis. Mechanistically, we demonstrate that the ubiquitously expressed hnRNPM acts in a mesenchymal-specific manner to precisely control CD44 splice isoform switching during EMT. This restricted cell-type activity of hnRNPM is achieved by competition with ESRP1, an epithelial splicing regulator that binds to the same cis-regulatory RNA elements as hnRNPM and is repressed during EMT. Importantly, hnRNPM is associated with aggressive breast cancer and correlates with increased CD44s in patient specimens. These findings demonstrate a novel molecular mechanism through which tumor metastasis is endowed by the hnRNPM-mediated splicing program.


Subject(s)
Alternative Splicing , Breast Neoplasms/genetics , Breast Neoplasms/physiopathology , Heterogeneous-Nuclear Ribonucleoprotein Group M/metabolism , Neoplasm Metastasis/physiopathology , Animals , Breast Neoplasms/secondary , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , Heterogeneous-Nuclear Ribonucleoprotein Group M/genetics , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Mice , Neoplasm Metastasis/genetics , Protein Isoforms/metabolism , Signal Transduction , Transforming Growth Factor beta1/metabolism
8.
EMBO Rep ; 19(3)2018 03.
Article in English | MEDLINE | ID: mdl-29440125

ABSTRACT

Trinucleotide repeat (TNR) expansions in the genome cause a number of degenerative diseases. A prominent TNR expansion involves the triplet CAG in the huntingtin (HTT) gene responsible for Huntington's disease (HD). Pathology is caused by protein and RNA generated from the TNR regions including small siRNA-sized repeat fragments. An inverse correlation between the length of the repeats in HTT and cancer incidence has been reported for HD patients. We now show that siRNAs based on the CAG TNR are toxic to cancer cells by targeting genes that contain long reverse complementary TNRs in their open reading frames. Of the 60 siRNAs based on the different TNRs, the six members in the CAG/CUG family of related TNRs are the most toxic to both human and mouse cancer cells. siCAG/CUG TNR-based siRNAs induce cell death in vitro in all tested cancer cell lines and slow down tumor growth in a preclinical mouse model of ovarian cancer with no signs of toxicity to the mice. We propose to explore TNR-based siRNAs as a novel form of anticancer reagents.


Subject(s)
Huntingtin Protein/genetics , Neoplasms/genetics , RNA, Small Interfering/pharmacology , Trinucleotide Repeats/genetics , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Disease Models, Animal , Humans , Huntingtin Protein/antagonists & inhibitors , Huntington Disease/genetics , Huntington Disease/pathology , Mice , Neoplasms/pathology , Neoplasms/therapy , Open Reading Frames , RNA, Small Interfering/genetics , Trinucleotide Repeat Expansion/genetics , Trinucleotide Repeats/drug effects
9.
Mol Cell ; 38(6): 908-15, 2010 Jun 25.
Article in English | MEDLINE | ID: mdl-20620960

ABSTRACT

Tumor progression shares many characteristics with the process of epithelial-to-mesenchymal transition (EMT). Cells that have undergone an EMT are known to have an increased resistance to apoptosis. CD95/Fas is an apoptosis-inducing receptor expressed on many tissues and tumor cells. During tumor progression CD95 is frequently downregulated, and tumor cells lose apoptosis sensitivity. miR-200 microRNAs repress both the EMT-inducing ZEB1 and ZEB2 transcription factors. We now demonstrate that miR-200c sensitizes cells to apoptosis mediated by CD95. We have identified the apoptosis inhibitor FAP-1 as a target for miR-200c. FAP-1 was demonstrated to be responsible for the reduced sensitivity to CD95-mediated apoptosis in cells with inhibited miR-200. The identification of FAP-1 as an miR-200c target provides a molecular mechanism to explain both the downregulation of CD95 expression and the reduction in sensitivity of cells to CD95-mediated apoptosis that is observed in the context of reduced miR-200 expression during tumor progression.


Subject(s)
Apoptosis , MicroRNAs/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 13/metabolism , fas Receptor/metabolism , Cell Line, Tumor , Humans
10.
Immunity ; 29(1): 1-2, 2008 Jul 18.
Article in English | MEDLINE | ID: mdl-18631448

ABSTRACT

Necrotic cells release HMGB1 as a danger signal to activate the immune system. In this issue of Immunity, Kazama et al. (2008) identify a mechanism that determines whether HMGB1 is tolerogenic or immunogenic.


Subject(s)
HMGB1 Protein/immunology , Immune Tolerance , Reactive Oxygen Species/immunology , Animals , Apoptosis/immunology , Dendritic Cells/immunology , Humans , Necrosis/immunology
11.
Nature ; 465(7297): 492-6, 2010 May 27.
Article in English | MEDLINE | ID: mdl-20505730

ABSTRACT

CD95 (also called Fas and APO-1) is a prototypical death receptor that regulates tissue homeostasis mainly in the immune system through the induction of apoptosis. During cancer progression CD95 is frequently downregulated or cells are rendered apoptosis resistant, raising the possibility that loss of CD95 is part of a mechanism for tumour evasion. However, complete loss of CD95 is rarely seen in human cancers and many cancer cells express large quantities of CD95 and are highly sensitive to CD95-mediated apoptosis in vitro. Furthermore, cancer patients frequently have elevated levels of the physiological ligand for CD95, CD95L. These data raise the possibility that CD95 could actually promote the growth of tumours through its non-apoptotic activities. Here we show that cancer cells in general, regardless of their CD95 apoptosis sensitivity, depend on constitutive activity of CD95, stimulated by cancer-produced CD95L, for optimal growth. Consistently, loss of CD95 in mouse models of ovarian cancer and liver cancer reduces cancer incidence as well as the size of the tumours. The tumorigenic activity of CD95 is mediated by a pathway involving JNK and Jun. These results demonstrate that CD95 has a growth-promoting role during tumorigenesis and indicate that efforts to inhibit its activity rather than to enhance it should be considered during cancer therapy.


Subject(s)
Neoplasms/metabolism , Neoplasms/pathology , fas Receptor/metabolism , Animals , Apoptosis , Carcinoma, Endometrioid/metabolism , Carcinoma, Endometrioid/pathology , Cell Line, Tumor , Cell Proliferation , Fas Ligand Protein/antagonists & inhibitors , Fas Ligand Protein/immunology , Fas Ligand Protein/metabolism , Female , Gene Expression Regulation, Neoplastic , Hepatocytes/enzymology , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mitogen-Activated Protein Kinase 8/deficiency , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 8/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , fas Receptor/deficiency , fas Receptor/genetics
12.
Nat Commun ; 15(1): 264, 2024 Jan 18.
Article in English | MEDLINE | ID: mdl-38238311

ABSTRACT

Alzheimer's disease (AD) is characterized by progressive neurodegeneration, but the specific events that cause cell death remain poorly understood. Death Induced by Survival gene Elimination (DISE) is a cell death mechanism mediated by short (s) RNAs acting through the RNA-induced silencing complex (RISC). DISE is thus a form of RNA interference, in which G-rich 6mer seed sequences in the sRNAs (position 2-7) target hundreds of C-rich 6mer seed matches in genes essential for cell survival, resulting in the activation of cell death pathways. Here, using Argonaute precipitation and RNAseq (Ago-RP-Seq), we analyze RISC-bound sRNAs to quantify 6mer seed toxicity in several model systems. In mouse AD models and aging brain, in induced pluripotent stem cell-derived neurons from AD patients, and in cells exposed to Aß42 oligomers, RISC-bound sRNAs show a shift to more toxic 6mer seeds compared to controls. In contrast, in brains of "SuperAgers", humans over age 80 who have superior memory performance, RISC-bound sRNAs are shifted to more nontoxic 6mer seeds. Cells depleted of nontoxic sRNAs are sensitized to Aß42-induced cell death, and reintroducing nontoxic RNAs is protective. Altogether, the correlation between DISE and Aß42 toxicity suggests that increasing the levels of nontoxic miRNAs in the brain or blocking the activity of toxic RISC-bound sRNAs could ameliorate neurodegeneration.


Subject(s)
Alzheimer Disease , MicroRNAs , Mice , Animals , Humans , Aged, 80 and over , Alzheimer Disease/genetics , MicroRNAs/genetics , RNA-Induced Silencing Complex/genetics , RNA Interference , Aging/genetics , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/toxicity
13.
BMC Genomics ; 14: 179, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23497354

ABSTRACT

BACKGROUND: Based on their function in cancer micro(mi)RNAs are often grouped as either tumor suppressors or oncogenes. However, miRNAs regulate multiple tumor relevant signaling pathways raising the question whether two oncogenic miRNAs could be functional antagonists by promoting different steps in tumor progression. We recently developed a method to connect miRNAs to biological function by comparing miRNA and gene array expression data from the NCI60 cell lines without using miRNA target predictions (miRConnect). RESULTS: We have now extended this analysis to three primary human cancers (ovarian cancer, glioblastoma multiforme, and kidney renal clear cell carcinoma) available at the Cancer Genome Atlas (TCGA), and have correlated the expression of the clustered miRNAs with 158 oncogenic signatures (miRConnect 2.0). We have identified functionally antagonistic groups of miRNAs. One group (the agonists), which contains many of the members of the miR-17 family, correlated with c-Myc induced genes and E2F gene signatures. A group that was directly antagonistic to the agonists in all three primary cancers contains miR-221 and miR-222. Since both miR-17 ~ 92 and miR-221/222 are considered to be oncogenic this points to a functional antagonism of different oncogenic miRNAs. Analysis of patient data revealed that in certain patients agonistic miRNAs predominated, whereas in other patients antagonists predominated. In glioblastoma a high ratio of miR-17 to miR-221/222 was predictive of better overall survival suggesting that high miR-221/222 expression is more adverse for patients than high miR-17 expression. CONCLUSION: miRConnect 2.0 is useful for identifying activities of miRNAs that are relevant to primary cancers. The new correlation data on miRNAs and mRNAs deregulated in three primary cancers are available at miRConnect.org.


Subject(s)
Carcinoma, Renal Cell/genetics , Gene Expression Profiling/methods , Glioblastoma/genetics , Kidney Neoplasms/genetics , MicroRNAs/genetics , Ovarian Neoplasms/genetics , Carcinoma, Renal Cell/physiopathology , Cell Line, Tumor , Cluster Analysis , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Glioblastoma/physiopathology , Humans , Kidney Neoplasms/physiopathology , MicroRNAs/physiology , Neoplasms/genetics , Oncogenes/physiology , Ovarian Neoplasms/physiopathology , RNA, Messenger/genetics , RNA, Messenger/metabolism
14.
Mol Biomed ; 4(1): 11, 2023 Apr 15.
Article in English | MEDLINE | ID: mdl-37059938

ABSTRACT

CD95/Fas ligand (CD95L) induces apoptosis through protein binding to the CD95 receptor. However, CD95L mRNA also induces toxicity in the absence of CD95 through induction of DISE (Death Induced by Survival Gene Elimination), a form of cell death mediated by RNA interference (RNAi). We now report that CD95L mRNA processing generates a short (s)RNA nearly identical to shL3, a commercial CD95L-targeting shRNA that led to the discovery of DISE. Neither of the miRNA biogenesis proteins Drosha nor Dicer are required for this processing. Interestingly, CD95L toxicity depends on the core component of the RISC, Ago2, in some cell lines, but not in others. In the HCT116 colon cancer cell line, Ago 1-4 appear to function redundantly in RNAi. In fact, Ago 1/2/3 knockout cells retain sensitivity to CD95L mRNA toxicity. Toxicity was only blocked by mutation of all in-frame start codons in the CD95L ORF. Dying cells exhibited an enrichment of RISC bound (R)-sRNAs with toxic 6mer seed sequences, while expression of the non-toxic CD95L mutant enriched for loading of R-sRNAs with nontoxic 6mer seeds. However, CD95L is not the only source of these R-sRNAs. We find that CD95L mRNA may induce DISE directly and indirectly, and that alternate mechanisms may underlie CD95L mRNA processing and toxicity.

15.
Int J Cancer ; 130(8): 1787-97, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-21618519

ABSTRACT

Ovarian cancer patients frequently develop resistance to chemotherapy regiments using Taxol and carboplatin. One of the resistance factors that protects cancer cells from Taxol-based therapy is multidrug resistance 1 (MDR1). micro(mi)RNAs are small noncoding RNAs that negatively regulate protein expression. Members of the let-7 family of miRNAs are downregulated in many human cancers, and low let-7 expression has been correlated with resistance to microtubule targeting drugs (Taxanes), although little is known that would explain this activity. We now provide evidence that, although let-7 is not a universal sensitizer of cancer cells to Taxanes, it affects acquired resistance of cells to this class of drugs by targeting IMP-1, resulting in destabilization of the mRNA of MDR1. Introducing let-7g into ADR-RES cells expressing both IMP-1 and MDR1 reduced expression of both proteins rendering the cells more sensitive to treatment with either Taxol or vinblastine without affecting the sensitivity of the cells to carboplatin, a non-MDR1 substrate. This effect could be reversed by reintroducing IMP-1 into let-7g high/MDR1 low cells causing MDR1 to again become stabilized. Consistently, many relapsed ovarian cancer patients tested before and after chemotherapy were found to downregulate let-7 and to co-upregulate IMP-1 and MDR1, and the increase in the expression levels of both proteins after chemotherapy negatively correlated with disease-free time before recurrence. Our data point at IMP-1 and MDR1 as indicators for response to therapy, and at IMP-1 as a novel therapeutic target for overcoming multidrug resistance of ovarian cancer.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , MicroRNAs/genetics , Ovarian Neoplasms/genetics , RNA-Binding Proteins/genetics , Taxoids/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Blotting, Western , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease-Free Survival , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , HEK293 Cells , HeLa Cells , Humans , Immunohistochemistry , In Situ Hybridization , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Taxoids/therapeutic use
16.
Curr Opin Cell Biol ; 17(6): 610-6, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16226446

ABSTRACT

Death receptors (DRs) are surface receptors that when triggered have the capacity to induce apoptosis in cells by forming the death-inducing signaling complex (DISC). The first protein recruited to form the DISC is the adaptor protein FADD/Mort1. Some members of the DR family, CD95 and the TRAIL receptors DR4 and DR5, directly bind FADD, whereas others, such as TNF receptor I and DR3, initially bind another adaptor protein, TRADD, which then recruits FADD. While all DRs can activate both apoptotic and non-apoptotic pathways, it has been widely assumed that the main physiological role of FADD-binding death receptors is to trigger apoptosis. However, recent work has ascribed multiple non-apoptotic activities to these receptors and/or the signaling components of the DISC.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis , Signal Transduction/physiology , fas Receptor/metabolism , Animals , CASP8 and FADD-Like Apoptosis Regulating Protein , Caspase 8 , Caspases/physiology , Cell Cycle Proteins/physiology , Fas-Associated Death Domain Protein , Humans , Intracellular Signaling Peptides and Proteins/physiology , Models, Biological
18.
Sci Rep ; 12(1): 5130, 2022 03 24.
Article in English | MEDLINE | ID: mdl-35332222

ABSTRACT

6mer seed toxicity is a novel cell death mechanism that kills cancer cells by triggering death induced by survival gene elimination (DISE). It is based on si- or shRNAs with a specific G-rich nucleotide composition in position 2-7 of their guide strand. An arrayed screen of 4096 6mer seeds on two human and two mouse cell lines identified G-rich 6mers as the most toxic seeds. We have now tested two additional cell lines, one human and one mouse, identifying the GGGGGC consensus as the most toxic average 6mer seed for human cancer cells while slightly less significant for mouse cancer cells. RNA Seq and bioinformatics analyses suggested that an siRNA containing the GGGGGC seed (siGGGGGC) is toxic to cancer cells by targeting GCCCCC seed matches located predominantly in the 3' UTR of a set of genes critical for cell survival. We have identified several genes targeted by this seed and demonstrate direct and specific targeting of GCCCCC seed matches, which is attenuated upon mutation of the GCCCCC seed matches in these 3' UTRs. Our data show that siGGGGGC kills cancer cells through its miRNA-like activity and points at artificial miRNAs, si- or shRNAs containing this seed as a potential new cancer therapeutics.


Subject(s)
MicroRNAs , Neoplasms , 3' Untranslated Regions , Animals , Consensus , Humans , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasms/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Seeds/genetics , Seeds/metabolism
19.
Cell Death Dis ; 13(12): 1078, 2022 12 30.
Article in English | MEDLINE | ID: mdl-36585400

ABSTRACT

Extended CAG trinucleotide repeats (TNR) in the genes huntingtin (HTT) and androgen receptor (AR) are the cause of two progressive neurodegenerative disorders: Huntington's disease (HD) and Spinal and Bulbar Muscular Atrophy (SBMA), respectively. Anyone who inherits the mutant gene in the complete penetrance range (>39 repeats for HD and 44 for SBMA) will develop the disease. An inverse correlation exists between the length of the CAG repeat and the severity and age of onset of the diseases. Growing evidence suggests that it is the length of uninterrupted CAG repeats in the mRNA rather than the length of poly glutamine (polyQ) in mutant (m)HTT protein that determines disease progression. One variant of mHTT (loss of inhibition; LOI) causes a 25 year earlier onset of HD when compared to a reference sequence, despite both coding for a protein that contains an identical number of glutamines. Short 21-22 nt CAG repeat (sCAGs)-containing RNAs can cause disease through RNA interference (RNAi). RNA hairpins (HPs) forming at the CAG TNRs are stabilized by adjacent CCG (in HD) or CUG repeats (in SBMA) making them better substrates for Dicer, the enzyme that processes CAG HPs into sCAGs. We now show that cells deficient in Dicer or unable to mediate RNAi are resistant to the toxicity of the HTT and AR derived HPs. Expression of a small HP that mimics the HD LOI variant is more stable and more toxic than a reference HP. We report that the LOI HP is processed by Dicer, loaded into the RISC more efficiently, and gives rise to a higher quantity of RISC-bound 22 nt sCAGs. Our data support the notion that RNAi contributes to the cell death seen in HD and SBMA and provide an explanation for the dramatically reduced onset of disease in HD patients that carry the LOI variant.


Subject(s)
Huntington Disease , Trinucleotide Repeat Expansion , Humans , RNA Interference , Trinucleotide Repeat Expansion/genetics , Trinucleotide Repeats/genetics , Huntington Disease/genetics , Huntington Disease/metabolism , RNA, Messenger/metabolism , Huntingtin Protein/genetics , Huntingtin Protein/metabolism
20.
Int J Cancer ; 128(6): 1327-34, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-20473948

ABSTRACT

MicroRNAs (miRNA) are small noncoding RNAs commonly deregulated in cancer. The miR-200 family (miR-200a, -200b, -200c, -141 and -429) and miR-205 are frequently silenced in advanced cancer and have been implicated in epithelial to mesenchymal transition (EMT) and tumor invasion by targeting the transcriptional repressors of E-cadherin, ZEB1 and ZEB2. ZEB1 is also known to repress miR-200c-141 transcription in a negative feedback loop, but otherwise little is known about the transcriptional regulation of the miR-200 family and miR-205. Recently, miR-200 silencing was also reported in cancer stem cells, implying that miR-200 deregulation is a key event in multiple levels of tumor biology. However, what prevents miR-200 expression remains largely unanswered. Here we report concerted transcriptional regulation of the miR-200 and miR-205 loci in bladder tumors and bladder cell lines. Using a combination of miRNA expression arrays, qPCR assays and mass spectrometry DNA methylation analyses, we show that the miR-200 and miR-205 loci are specifically silenced and gain promoter hypermethylation and repressive chromatin marks in muscle invasive bladder tumors and undifferentiated bladder cell lines. Moreover, we report that miR-200c expression is significantly correlated with early stage T1 bladder tumor progression, and propose miR-200 and miR-205 silencing and DNA hypermethylation as possible prognostic markers in bladder cancer. In addition, we observe that the mesoderm transcription factor TWIST1 and miR-200 expression are inversely correlated in bladder tumor samples and cell lines. TWIST1 associates directly with the miR-200 and miR-205 promoters, and may act as a repressor of miR-200 and miR-205 expression.


Subject(s)
Epigenomics , MicroRNAs/genetics , Urinary Bladder Neoplasms/genetics , Cells, Cultured , DNA Methylation , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Invasiveness , Polymerase Chain Reaction , Urinary Bladder/metabolism , Urinary Bladder/pathology , Urinary Bladder Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL