Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
STAR Protoc ; 4(1): 101871, 2023 03 17.
Article in English | MEDLINE | ID: mdl-36856767

ABSTRACT

Evaluation of autophagy flux could be challenging for muscle fibers due to the baseline expression of mCherry-EGFP-LC3 along the Z-line. We established a protocol to overcome this difficulty. We overexpress mChery-EGFP-LC3 in the FDB muscle of an adult mouse via electroporation. Then, we enzymatically digest FDB muscle to yield individual fibers for live cell imaging. Finally, we develop an ImageJ-based program to eliminate the baseline striation pattern and semi-automatically quantify autophagosomes (APs) and autolysosomes (ALs) for autophagy flux analysis.


Subject(s)
Autophagy , Microtubule-Associated Proteins , Mice , Animals , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Autophagy/genetics , Muscle Fibers, Skeletal/metabolism , Autophagosomes/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Coloring Agents/metabolism
2.
Cell Death Differ ; 30(5): 1366-1381, 2023 05.
Article in English | MEDLINE | ID: mdl-36899106

ABSTRACT

Gasdermin (GSDM)-mediated pyroptosis is functionally involved in multiple diseases, but Gasdermin-B (GSDMB) exhibit cell death-dependent and independent activities in several pathologies including cancer. When the GSDMB pore-forming N-terminal domain is released by Granzyme-A cleavage, it provokes cancer cell death, but uncleaved GSDMB promotes multiple pro-tumoral effects (invasion, metastasis, and drug resistance). To uncover the mechanisms of GSDMB pyroptosis, here we determined the GSDMB regions essential for cell death and described for the first time a differential role of the four translated GSDMB isoforms (GSDMB1-4, that differ in the alternative usage of exons 6-7) in this process. Accordingly, we here prove that exon 6 translation is essential for GSDMB mediated pyroptosis, and therefore, GSDMB isoforms lacking this exon (GSDMB1-2) cannot provoke cancer cell death. Consistently, in breast carcinomas the expression of GSDMB2, and not exon 6-containing variants (GSDMB3-4), associates with unfavourable clinical-pathological parameters. Mechanistically, we show that GSDMB N-terminal constructs containing exon-6 provoke cell membrane lysis and a concomitant mitochondrial damage. Moreover, we have identified specific residues within exon 6 and other regions of the N-terminal domain that are important for GSDMB-triggered cell death as well as for mitochondrial impairment. Additionally, we demonstrated that GSDMB cleavage by specific proteases (Granzyme-A, Neutrophil Elastase and caspases) have different effects on pyroptosis regulation. Thus, immunocyte-derived Granzyme-A can cleave all GSDMB isoforms, but in only those containing exon 6, this processing results in pyroptosis induction. By contrast, the cleavage of GSDMB isoforms by Neutrophil Elastase or caspases produces short N-terminal fragments with no cytotoxic activity, thus suggesting that these proteases act as inhibitory mechanisms of pyroptosis. Summarizing, our results have important implications for understanding the complex roles of GSDMB isoforms in cancer or other pathologies and for the future design of GSDMB-targeted therapies.


Subject(s)
Breast Neoplasms , Pyroptosis , Humans , Female , Granzymes/genetics , Granzymes/metabolism , Peptide Hydrolases/metabolism , Leukocyte Elastase/metabolism , Gasdermins , Neoplasm Proteins/metabolism , Caspases/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Breast Neoplasms/genetics , Pore Forming Cytotoxic Proteins/metabolism
3.
J Cell Mol Med ; 26(7): 1886-1895, 2022 04.
Article in English | MEDLINE | ID: mdl-35199443

ABSTRACT

Nitrogen mustard (NM) is an alkylating vesicant that causes severe pulmonary injury. Currently, there are no effective means to counteract vesicant-induced lung injury. MG53 is a vital component of cell membrane repair and lung protection. Here, we show that mice with ablation of MG53 are more susceptible to NM-induced lung injury than the wild-type mice. Treatment of wild-type mice with exogenous recombinant human MG53 (rhMG53) protein ameliorates NM-induced lung injury by restoring arterial blood oxygen level, by improving dynamic lung compliance and by reducing airway resistance. Exposure of lung epithelial and endothelial cells to NM leads to intracellular oxidative stress that compromises the intrinsic cell membrane repair function of MG53. Exogenous rhMG53 protein applied to the culture medium protects lung epithelial and endothelial cells from NM-induced membrane injury and oxidative stress, and enhances survival of the cells. Additionally, we show that loss of MG53 leads to increased vulnerability of macrophages to vesicant-induced cell death. Overall, these findings support the therapeutic potential of rhMG53 to counteract vesicant-induced lung injury.


Subject(s)
Acute Lung Injury , Mechlorethamine , Acute Lung Injury/chemically induced , Acute Lung Injury/drug therapy , Acute Lung Injury/genetics , Animals , Endothelial Cells/metabolism , Lung/metabolism , Mechlorethamine/therapeutic use , Mechlorethamine/toxicity , Membrane Proteins/metabolism , Mice , Recombinant Proteins/metabolism
4.
Acta Pharmacol Sin ; 41(11): 1427-1432, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32555510

ABSTRACT

Induced pluripotent stem cells (iPSCs) have become an essential research platform to study different human diseases once being discovered by Dr. Shinya Yamanaka in 2006. Another breakthrough in biomedical research is the application of CRISPR/Cas9 system for genome editing in mammalian cells. Although numerous studies have been done to develop methods for gene editing in iPSCs, the current approaches suffer from several limitations, including time and labor consuming, low editing efficiency, and potential off-target effects. In the current study, we report an electroporation-mediated plasmid CRISPR/Cas9 delivery approach for genome editing in iPSCs. With this approach, an edited iPSC cell line could be obtained within 2 weeks. In addition, the transit introducing of CRISPR/Cas9 machinery could minimize genomic integration of Cas9 gene, which avoided potential long-term side effects of Cas9 enzyme. We showed that CRISPR/Cas9-mediated genomic editing did not affect pluripotency and differentiation ability of iPSCs. With the quickly evolving of both iPSC and CRISPR/Cas9-mediated genome editing research fields, we believe that our method can significantly facilitate the application of genome editing in iPSCs research.


Subject(s)
CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems , Gene Editing/methods , Induced Pluripotent Stem Cells/metabolism , Base Sequence , Cell Differentiation/genetics , Gene Knockout Techniques/methods , Humans , Plasmids , Ubiquitin Thiolesterase/genetics
5.
FASEB J ; 25(8): 2638-49, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21518849

ABSTRACT

We describe a novel transgenic system for tissue-specific and inducible control of gene expression in mice. The system employs a tetracycline-responsive CMV promoter that controls transcription of a short-hairpin RNA (shRNA) that remains nonfunctional until an interrupting reporter cassette is excised by Cre recombinase. Insertion of Dicer and Drosha RNase processing sites within the shRNA allows generation of siRNA to knock down a target gene efficiently. Tissue-specific shRNA expression is achieved through the use of appropriate inducer mice with tissue-specific expression of Cre. We applied this system to regulate expression of junctophilins (JPs), genes essential for maintenance of membrane ultrastructure and Ca(2+) signaling in muscle. Transgenic mice with skeletal muscle-specific expression of shRNA against JP mRNAs displayed no basal change of JP expression before treatment with doxycycline (Dox), while inducible and reversible knockdown of JPs was achieved by feeding mice with Dox-containing water. Dox-induced knockdown of JPs led to abnormal junctional membrane structure and Ca(2+) signaling in adult muscle fibers, consistent with essential roles of JPs in muscle development and function. This transgenic approach can be applied for inducible and reversible gene knockdown or gene overexpression in many different tissues, thus providing a versatile system for elucidating the physiological gene function in viable animal models.


Subject(s)
Genetic Techniques , Mice, Transgenic/genetics , Plasmids/genetics , Animals , Base Sequence , CHO Cells , Cricetinae , Cricetulus , Gene Expression , Gene Knockdown Techniques , Genes, Reporter , HEK293 Cells , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Muscle, Skeletal/metabolism , Muscle, Skeletal/ultrastructure , RNA Interference , RNA, Small Interfering/genetics , Tissue Distribution
6.
J Biol Chem ; 286(15): 12820-4, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21343302

ABSTRACT

Plasma membrane repair is an essential process for maintenance of homeostasis at the cellular and tissue levels, whereas compromised repair capacity contributes to degenerative human diseases. Our recent studies show that MG53 is essential for muscle membrane repair, and defects in MG53 function are linked to muscular dystrophy and cardiac dysfunction. Here we report that polymerase I and transcript release factor (PTRF), a gene known to regulate caveolae membrane structure, is an indispensable component of the membrane repair machinery. PTRF acts as a docking protein for MG53 during membrane repair potentially by binding exposed membrane cholesterol at the injury site. Cells lacking expression of endogenous PTRF show defective trafficking of MG53 to membrane injury sites. A mutation in PTRF associated with human disease results in aberrant nuclear localization of PTRF and disrupts MG53 function in membrane resealing. Although RNAi silencing of PTRF leads to defective muscle membrane repair, overexpression of PTRF can rescue membrane repair defects in dystrophic muscle. Our data suggest that membrane-delimited interaction between MG53 and PTRF contributes to initiation of cell membrane repair, which can be an attractive target for treatment or prevention of tissue injury in human diseases.


Subject(s)
Carrier Proteins/metabolism , Caveolae/metabolism , Muscle, Skeletal/metabolism , RNA-Binding Proteins/metabolism , Animals , Carrier Proteins/genetics , Caveolae/pathology , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Nucleus/pathology , HeLa Cells , Heart Diseases/genetics , Heart Diseases/metabolism , Heart Diseases/pathology , Hep G2 Cells , Humans , Membrane Proteins , Mice , Muscle, Skeletal/pathology , Muscular Dystrophies/genetics , Muscular Dystrophies/metabolism , Muscular Dystrophies/pathology , Muscular Dystrophies/therapy , Mutation , Protein Transport/genetics , RNA-Binding Proteins/genetics , Tripartite Motif Proteins
7.
J Biol Chem ; 286(11): 9038-48, 2011 Mar 18.
Article in English | MEDLINE | ID: mdl-21189256

ABSTRACT

Bcl-2 homology domain-3 (BH3) peptides are potent cancer therapeutic reagents that target regulators of apoptotic cell death in cancer cells. However, their cytotoxic effects are affected by different expression levels of Bcl-2 family proteins. We recently found that the amphipathic tail-anchoring peptide (ATAP) from Bfl-1, a bifunctional Bcl-2 family member, produced strong pro-apoptotic activity by permeabilizing the mitochondrial outer membrane. Here, we test whether the activity of ATAP requires other cellular factors and whether ATAP has an advantage over the BH3 peptides in targeting cancer cells. Confocal microscopic imaging illustrates specific targeting of ATAP to mitochondria, whereas BH3 peptides show diffuse patterns of cytosolic distribution. Although the pro-apoptotic activities of BH3 peptides are largely inhibited by either overexpression of anti-apoptotic Bcl-2 or Bcl-xL or nullification of pro-apoptotic Bax and Bak in cells, the pro-apoptotic function of ATAP is not affected by these cellular factors. Reconstitution of synthetic ATAP into liposomal membranes results in release of fluorescent molecules of the size of cytochrome c from the liposomes, suggesting that the membrane permeabilizing activity of ATAP does not require additional protein factors. Because ATAP can target to the mitochondrial membrane and its pro-apoptotic activity does not depend on the content of Bcl-2 family proteins, it represents a promising candidate for anti-cancer drugs that can potentially overcome the intrinsic apoptosis-resistant nature of cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Mitochondrial Membranes/metabolism , Neoplasms/drug therapy , Peptide Fragments/pharmacology , Proto-Oncogene Proteins c-bcl-2/pharmacology , Proto-Oncogene Proteins/pharmacology , bcl-X Protein/metabolism , Cytochromes c/metabolism , HeLa Cells , Humans , Liposomes/chemistry , Minor Histocompatibility Antigens , Neoplasms/metabolism , Permeability/drug effects
8.
J Cell Sci ; 120(Pt 16): 2912-23, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17666431

ABSTRACT

Many Bcl2 family proteins target intracellular membranes by their C-terminal tail-anchor domain. Bfl1 is a bi-functional Bcl2 family protein with both anti- and pro-apoptotic activities and contains an amphipathic tail-anchoring peptide (ATAP; residues 147-175) with unique properties. Here we show that ATAP targets specifically to mitochondria, and induces caspase-dependent apoptosis that does not require Bax or Bak. Mutagenesis studies revealed that lysine residues flanking the ATAP sequence are involved in targeting of the peptide to the mitochondrial membrane, and charged residues that contribute to the amphipathic nature of ATAP are critical for its pro-apoptotic function. The ATAP sequence is present in another tumor suppressor gene, HCCS1, which contains an additional mitochondria-targeting signal (MTS) close to the ATAP. We propose that both ATAP and MTS could be used as therapeutic peptides to induce cell death in the treatment of cancer cells.


Subject(s)
Apoptosis , Mitochondrial Membranes/metabolism , Proto-Oncogene Proteins c-bcl-2/chemistry , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism , Amino Acid Sequence , Base Sequence , Cell Line, Tumor , Conserved Sequence , Humans , Lipid Bilayers/metabolism , Membrane Potential, Mitochondrial , Minor Histocompatibility Antigens , Molecular Sequence Data , Peptides/chemistry , Permeability , Protein Sorting Signals , Protein Structure, Tertiary , Protein Transport , Proto-Oncogene Proteins c-bcl-2/genetics , Structure-Activity Relationship , Tumor Suppressor Proteins/genetics , Vesicular Transport Proteins , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism
9.
J Biol Chem ; 281(21): 14691-9, 2006 May 26.
Article in English | MEDLINE | ID: mdl-16574658

ABSTRACT

The TRAP/Mediator coactivator complex serves as a functional interface between DNA-bound transactivators and the RNA polymerase II-associated basal transcription apparatus. TRAP220/MED1 is a variably associated subunit of the complex that plays a specialized role in selectively targeting TRAP/Mediator to specific genes. Ablation of the Trap220/Med1 gene in mice impairs embryonic cell growth, yet the underlying mechanism is unknown. In this report, we identified distinct cell growth regulatory genes whose expression is affected by the loss of TRAP220/MED1 by RNA interference. Among the down-regulated genes revealed by cDNA microarray analyses, we identified Aurora-A, a centrosome kinase that plays a critical role in regulating M phase events and is frequently amplified in several types of cancer. In general, we found that TRAP220/MED1 expression is required for high basal levels of Aurora-A gene expression and that ectopic overexpression of TRAP220/MED1 coactivates transcription from the Aurora-A gene promoter. Furthermore, chromatin immunoprecipitation assays show that TRAP220/MED1-containing TRAP/Mediator complexes directly bind to the Aurora-A promoter in vivo. Finally, we present evidence suggesting that TRAP/Mediator is recruited to the Aurora-A gene via direct interactions between TRAP220/MED1 and the Ets-related transcription factor GABP. Taken together, these findings suggest that TRAP220/MED1 plays a novel coregulatory role in facilitating the recruitment of TRAP/Mediator to specific target genes involved in growth and cell cycle progression.


Subject(s)
Endodeoxyribonucleases/metabolism , Gene Expression Regulation, Enzymologic , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/genetics , Transcription Factors/metabolism , Aurora Kinase A , Aurora Kinases , Cell Proliferation , DNA/chemistry , HeLa Cells , Humans , Mediator Complex Subunit 1 , Models, Genetic , Promoter Regions, Genetic , RNA Interference , RNA, Small Interfering/metabolism , Thyroid Hormones/metabolism , Transcriptional Activation
10.
J Cell Biochem ; 91(2): 410-22, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14743399

ABSTRACT

Various chemotherapeutic agents have been shown to sensitize cancer cells to members of the tumor necrosis factor (TNF) family. However, it is unclear whether sensitization by chemotherapeutic agents involves the transcriptional regulation of apoptosis-related genes. In this study, we investigated mRNA regulation of TNF family receptors and Bcl-2 family members after treating the murine colon cancer cell line, CT26, with various apoptosis inducers. We found that treatment with cycloheximide, a protein synthesis inhibitor, remarkably increased CD40 mRNA levels by semi-quantitative RT-PCR. Other protein synthesis inhibitors, such as anisomycin and emetine, also enhanced CD40 mRNA expression, which was significantly blocked by a NF-kappaB antagonist and a p38 MAP kinase antagonist. After treatment with cycloheximide, and further cultivation in fresh medium, CD40 protein levels were found to increase by flow cytometry. Additionally, we found that cycloheximide treatment appeared to downregulate the Bcl-xL mRNA level but not the Bax mRNA level by RNase protection assay. Because the upregulation of CD40 mRNA and the downregulation of Bcl-xL correlated with CT26 cell death, our results suggest that chemotherapeutic agents, including cycloheximide, may exert their synergistic effects on the TNF family treatment of cancer cells by regulating the mRNA levels of apoptosis-related genes.


Subject(s)
Gene Expression Regulation/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Transcription, Genetic , Animals , Anisomycin/pharmacology , Apoptosis/drug effects , CD40 Antigens/metabolism , Cell Line, Tumor , Cell Survival , Cycloheximide/pharmacology , Down-Regulation , Humans , Mice , Mitogen-Activated Protein Kinases/metabolism , Protein Synthesis Inhibitors/pharmacology , Signal Transduction , p38 Mitogen-Activated Protein Kinases
11.
FEBS Lett ; 551(1-3): 29-36, 2003 Sep 11.
Article in English | MEDLINE | ID: mdl-12965200

ABSTRACT

Human Bfl-1 is an anti-apoptotic Bcl-2 family member. Here, we found that Bfl-1 was converted into a potent death-promoting protein by green fluorescent protein (GFP) fusion with its N-terminus. The transient expression of GFP-Bfl-1 induced cytochrome c release and triggered apoptosis in 293T cells, which depended on the mitochondrial localization of GFP-Bfl-1. Apoptosis induced by GFP-Bfl-1 was significantly blocked by the pan-caspase inhibitor carbobenzoxy-Val-Ala-Asp-fluoromethyl ketone, but was not blocked by either Bcl-xL or Bfl-1. Our findings provide a useful model for understanding the structural basis of Bcl-2 family proteins that act in an opposite way despite sharing structural similarity between anti-apoptotic and pro-apoptotic proteins.


Subject(s)
Apoptosis , Proto-Oncogene Proteins c-bcl-2/chemistry , Caspases/physiology , Cell Line , Cytochrome c Group/metabolism , Green Fluorescent Proteins , Humans , Luminescent Proteins/genetics , Minor Histocompatibility Antigens , Mitochondria/chemistry , Mitochondria/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/metabolism , bcl-X Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...