Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Cells ; 13(11)2024 May 25.
Article in English | MEDLINE | ID: mdl-38891043

ABSTRACT

BAX plays an essential role in retinal ganglion cell (RGC) death induced by optic nerve injury. Recently, we developed M109S, an orally bioactive and cytoprotective small compound (CPSC) that inhibits BAX-mediated cell death. We examined whether M109S can protect RGC from optic nerve crush (ONC)-induced apoptosis. M109S was administered starting 5 h after ONC for 7 days. M109S was orally administered in two groups (5 mg/kg twice a day or 7.5 mg/kg once a day). The retina was stained with anti-BRN3A and cleaved Caspase-3 (active Caspase-3) that are the markers of RGC and apoptotic cells, respectively. ONC decreased the number of BRN3A-positive RGC and increased the number of active Caspase-3-expressing apoptotic cells. In ONC-treated retina, there were cells that were double stained with anti-BRN3A and ant-cleaved Caspase-3, indicating that apoptosis in BRN3A-positive RGCs occurred. M109S inhibited the decrease of BRN3A-positive cells whereas it inhibited the increase of active Caspase-3-positive cells in the retina of ONC-treated mice, suggesting that M109S inhibited apoptosis in RGCs. M109S did not induce detectable histological damage to the lungs or kidneys in mice, suggesting that M109S did not show toxicities in the lung or kidneys when the therapeutic dose was used. The present study suggests that M109S is effective in rescuing damaged RGCs. Since M109S is an orally bioactive small compound, M109S may become the basis for a portable patient-friendly medicine that can be used to prevent blindness by rescuing damaged optic nerve cells from death.


Subject(s)
Apoptosis , Nerve Crush , Optic Nerve Injuries , Retinal Ganglion Cells , Animals , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Retinal Ganglion Cells/metabolism , Mice , Optic Nerve Injuries/drug therapy , Optic Nerve Injuries/pathology , Apoptosis/drug effects , Male , Caspase 3/metabolism , Mice, Inbred C57BL , Cytoprotection/drug effects , Optic Nerve/drug effects , Optic Nerve/pathology
2.
J Inflamm (Lond) ; 21(1): 14, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38689261

ABSTRACT

BACKGROUND: The DNA-dependent protein kinase (DNA-PK) complex comprises a catalytic (PRKDC) and two requisite DNA-binding (Ku70/Ku80) subunits. The role of the complex in repairing double-stranded DNA breaks (DSBs) is established, but its role in inflammation, as a complex or individual subunits, remains elusive. While only ~ 1% of PRKDC is necessary for DNA repair, we reported that partial inhibition blocks asthma in mice without causing SCID. METHODS: We investigated the central role of PRKDC in inflammation and its potential association with DNA repair. We also elucidated the relationship between inflammatory cytokines (e.g., TNF-α) and PRKDC by analyzing its connections to inflammatory kinases. Human cell lines, primary human endothelial cells, and mouse fibroblasts were used to conduct the in vitro studies. For animal studies, LPS- and oxazolone-induced mouse models of acute lung injury (ALI) and delayed-type hypersensitivity (DHT) were used. Wild-type, PRKDC+/-, or Ku70+/- mice used in this study. RESULTS: A ~ 50% reduction in PRKDC markedly blocked TNF-α-induced expression of inflammatory factors (e.g., ICAM-1/VCAM-1). PRKDC regulates Th1-mediated inflammation, such as DHT and ALI, and its role is highly sensitive to inhibition achieved by gene heterozygosity or pharmacologically. In endothelial or epithelial cells, TNF-α promoted rapid PRKDC phosphorylation in a fashion resembling that induced by, but independent of, DSBs. Ku70 heterozygosity exerted little to no effect on ALI in mice, and whatever effect it had was associated with a specific increase in MCP-1 in the lungs and systemically. While Ku70 knockout blocked VP-16-induced PRKDC phosphorylation, it did not prevent TNF-α - induced phosphorylation of the kinase, suggesting Ku70 dispensability. Immunoprecipitation studies revealed that PRKDC transiently interacts with p38MAPK. Inhibition of p38MAPK blocked TNF-α-induced PRKDC phosphorylation. Direct phosphorylation of PRKDC by p38MAPK was demonstrated using a cell-free system. CONCLUSIONS: This study presents compelling evidence that PRKDC functions independently of the DNA-PK complex, emphasizing its central role in Th1-mediated inflammation. The distinct functionality of PRKDC as an individual enzyme, its remarkable sensitivity to inhibition, and its phosphorylation by p38MAPK offer promising therapeutic opportunities to mitigate inflammation while sparing DNA repair processes. These findings expand our understanding of PRKDC biology and open new avenues for targeted anti-inflammatory interventions.

3.
Aging Cell ; 23(5): e14112, 2024 May.
Article in English | MEDLINE | ID: mdl-38439206

ABSTRACT

Allogenic hematopoietic stem cell transplantation is a therapeutic procedure performed over a wide range of donor and recipient age combinations, representing natural experiments of how the age of the recipient affects aging in transplanted donor cells in vivo. We measured DNA methylation and epigenetic aging in donors and recipients and found that biological epigenetic clocks are accelerated in cells transplanted into an older body and decelerated in a younger body. This is the first evidence that the age of the circulating environment influences human epigenetic aging in vivo.


Subject(s)
Aging , Cellular Senescence , DNA Methylation , Epigenesis, Genetic , Humans , DNA Methylation/genetics , Cellular Senescence/genetics , Aging/genetics , Blood Cells/metabolism , Hematopoietic Stem Cell Transplantation/methods , Adult , Middle Aged , Male , Female
4.
iScience ; 26(10): 107916, 2023 Oct 20.
Article in English | MEDLINE | ID: mdl-37841588

ABSTRACT

We identified cytoprotective small molecules (CSMs) by a cell-based high-throughput screening of Bax inhibitors. Through a medicinal chemistry program, M109S was developed, which is orally bioactive and penetrates the blood-brain/retina barriers. M109S protected retinal cells in ocular disease mouse models. M109S directly interacted with Bax and inhibited the conformational change and mitochondrial translocation of Bax. M109S inhibited ABT-737-induced apoptosis both in Bax-only and Bak-only mouse embryonic fibroblasts. M109S also inhibited apoptosis induced by staurosporine, etoposide, and obatoclax. M109S decreased maximal mitochondrial oxygen consumption rate and reactive oxygen species production, whereas it increased glycolysis. These effects on cellular metabolism may contribute to the cytoprotective activity of M109S. M109S is a novel small molecule protecting cells from mitochondria-dependent apoptosis both in vitro and in vivo. M109S has the potential to become a research tool for studying cell death mechanisms and to develop therapeutics targeting mitochondria-dependent cell death pathway.

6.
Exp Biol Med (Maywood) ; 245(17): 1543-1551, 2020 11.
Article in English | MEDLINE | ID: mdl-32762265

ABSTRACT

IMPACT STATEMENT: Aging is associated with DNA methylation (DNAm) changes. Recent advancement of the whole-genome DNAm analysis technology allowed scientists to develop DNAm-based age estimators. A majority of these estimators use DNAm data from a single tissue type such as blood. In 2013, a multi-tissue age estimator using DNAm pattern of 353 CpGs was developed by Steve Horvath. This estimator was named "epigenetic clock", and the improved version using DNAm pattern of 391 CpGs was developed in 2018. The estimated age by epigenetic clock is named DNAmAge. DNAmAge can be used as a biomarker of aging predicting the risk of age-associated diseases and mortality. Although the DNAm-based age estimators were developed, the mechanism of epigenetic aging is still enigmatic. The biological significance of epigenetic aging is not well understood, either. This minireview discusses the current understanding of the mechanism of epigenetic aging and the future direction of aging research.


Subject(s)
Aging/genetics , Epigenesis, Genetic , Animals , Biological Clocks/genetics , Cell Hypoxia/genetics , DNA Methylation/genetics , Hematopoietic Stem Cells/metabolism , Humans
8.
Aging (Albany NY) ; 11(10): 3012-3022, 2019 05 21.
Article in English | MEDLINE | ID: mdl-31113906

ABSTRACT

Aging is associated with a genome-wide change of DNA methylation (DNAm). "DNAm age" is defined as the predicted chronological age by the age estimator based on DNAm. The estimator is called the epigenetic clock. The molecular mechanism underlining the epigenetic clock is still unknown. Here, we evaluated the effects of hypoxia and two immortalization factors, hTERT and SV40-LargeT (LT), on the DNAm age of human fibroblasts in vitro. We detected the cell division-associated progression of DNAm age after >10 population doublings. Moreover, the progression of DNAm age was slower under hypoxia (1% oxygen) compared to normoxia (21% oxygen), suggesting that oxygen levels determine the speed of the epigenetic aging. We show that the speed of cell division-associated DNAm age progression depends on the chronological age of the cell donor. hTERT expression did not arrest cell division-associated progression of DNAm age in most cells. SV40LT expression produced inconsistent effects, including rejuvenation of DNAm age. Our results show that a) oxygen and the targets of SV40LT (e.g. p53) modulate epigenetic aging rates and b) the chronological age of donor cells determines the speed of mitosis-associated DNAm age progression in daughter cells.


Subject(s)
Aging/physiology , Biological Clocks , DNA Methylation , Fibroblasts/physiology , Hypoxia/metabolism , Adult , Antigens, Polyomavirus Transforming/genetics , Antigens, Polyomavirus Transforming/metabolism , Epigenesis, Genetic , Humans , Infant, Newborn , Primary Cell Culture , Telomerase/genetics , Telomerase/metabolism
9.
Exp Biol Med (Maywood) ; 244(8): 621-629, 2019 05.
Article in English | MEDLINE | ID: mdl-30836793

ABSTRACT

IMPACT STATEMENT: Bax induces mitochondria-dependent programed cell death. While cytotoxic drugs activating Bax have been developed for cancer treatment, clinically effective therapeutics suppressing Bax-induced cell death rescuing essential cells have not been developed. This mini-review will summarize previously reported Bax inhibitors including peptides, small compounds, and antibodies. We will discuss potential applications and the future direction of these Bax inhibitors.


Subject(s)
Apoptosis/drug effects , Cell-Penetrating Peptides/pharmacology , bcl-2-Associated X Protein/antagonists & inhibitors , Amino Acid Sequence , Animals , Apoptosis/physiology , Brain Ischemia/drug therapy , Brain Ischemia/pathology , Cell-Penetrating Peptides/pharmacokinetics , Cell-Penetrating Peptides/therapeutic use , Cell-Penetrating Peptides/toxicity , Cells, Cultured , Disease Models, Animal , Drug Carriers , Drug Design , Humans , Immunoglobulin Fab Fragments/pharmacology , Ku Autoantigen/metabolism , Mice , Mice, Knockout , Mitochondria/physiology , Mitochondrial Membrane Transport Proteins/drug effects , Mitochondrial Membranes/drug effects , Mitochondrial Permeability Transition Pore , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/pathology , Organ Preservation/methods , Pinocytosis , Protein Multimerization/drug effects , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/pathology , Rats , Retinal Degeneration/drug therapy , Retinal Degeneration/pathology , bcl-2-Associated X Protein/deficiency , bcl-2-Associated X Protein/immunology , bcl-2-Associated X Protein/metabolism
10.
Aging Cell ; 18(2): e12897, 2019 04.
Article in English | MEDLINE | ID: mdl-30712319

ABSTRACT

The age of tissues and cells can be accurately estimated by DNA methylation analysis. The multitissue DNA methylation (DNAm) age predictor combines the DNAm levels of 353 CpG dinucleotides to arrive at an age estimate referred to as DNAm age. Recent studies based on short-term observations showed that the DNAm age of reconstituted blood following allogeneic hematopoietic stem cell transplantation (HSCT) reflects the age of the donor. However, it is not known whether the DNAm age of donor blood remains independent of the recipient's age over the long term. Importantly, long-term studies including child recipients have the potential to clearly reveal whether DNAm age is cell-intrinsic or whether it is modulated by extracellular cues in vivo. Here, we address this question by analyzing blood methylation data from HSCT donor and recipient pairs who greatly differed in chronological age (age differences between 1 and 49 years). We found that the DNAm age of the reconstituted blood was not influenced by the recipient's age, even 17 years after HSCT, in individuals without relapse of their hematologic disorder. However, the DNAm age of recipients with relapse of leukemia was unstable. These data are consistent with our previous findings concerning the abnormal DNAm age of cancer cells, and it can potentially be exploited to monitor the health of HSCT recipients. Our data demonstrate that transplanted human hematopoietic stem cells have an intrinsic DNAm age that is unaffected by the environment in a recipient of a different age.


Subject(s)
Cellular Senescence/genetics , DNA, Neoplasm/genetics , Epigenesis, Genetic/genetics , Hematopoietic Stem Cell Transplantation , Leukemia/pathology , Adolescent , Adult , Child , Child, Preschool , DNA Methylation , Humans , Infant , Leukemia/blood , Leukemia/genetics , Middle Aged , Transplantation, Homologous , Young Adult
11.
Aging (Albany NY) ; 10(7): 1758-1775, 2018 Jul 26.
Article in English | MEDLINE | ID: mdl-30048243

ABSTRACT

DNA methylation (DNAm)-based biomarkers of aging have been developed for many tissues and organs. However, these biomarkers have sub-optimal accuracy in fibroblasts and other cell types used in ex vivo studies. To address this challenge, we developed a novel and highly robust DNAm age estimator (based on 391 CpGs) for human fibroblasts, keratinocytes, buccal cells, endothelial cells, lymphoblastoid cells, skin, blood, and saliva samples. High age correlations can also be observed in sorted neurons, glia, brain, liver, and even bone samples. Gestational age correlates with DNAm age in cord blood. When used on fibroblasts from Hutchinson Gilford Progeria Syndrome patients, this age estimator (referred to as the skin & blood clock) uncovered an epigenetic age acceleration with a magnitude that is below the sensitivity levels of other DNAm-based biomarkers. Furthermore, this highly sensitive age estimator accurately tracked the dynamic aging of cells cultured ex vivo and revealed that their proliferation is accompanied by a steady increase in epigenetic age. The skin & blood clock predicts lifespan and it relates to many age-related conditions. Overall, this biomarker is expected to become useful for forensic applications (e.g. blood or buccal swabs) and for a quantitative ex vivo human cell aging assay.


Subject(s)
Biological Clocks/physiology , Blood Cells/physiology , Epigenesis, Genetic/physiology , Progeria/metabolism , Skin Physiological Phenomena , Aging/physiology , Cellular Senescence/physiology , DNA Methylation , Fetal Blood/cytology , Fibroblasts/physiology , Gene Expression Regulation/physiology , Humans
12.
Invest Ophthalmol Vis Sci ; 57(14): 6278-6286, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27893093

ABSTRACT

Purpose: Cluster of differentiation 40 (CD40) is required for retinal capillary degeneration in diabetic mice, a process mediated by the retinal endothelial cells (REC) death. However, CD40 activates prosurvival signals in endothelial cells. The purpose of this study was to identify a mechanism by which CD40 triggers programmed cell death (PCD) of RECs and address this paradox. Methods: Human RECs and Müller cells were incubated with CD154 and L-N6-(1-Iminoethyl)lysine (L-Nil, nitric oxide synthase 2 inhibitor), α-lipoic acid (inhibitor of oxidative stress), anti-Fas ligand antibody, or A-438079 (P2X7 adenosine triphosphate [ATP] receptor inhibitor). Programmed cell death was analyzed by fluorescence-activated cell sorting (FACS) or Hoechst/propidium iodide staining. Release of ATP was measured using a luciferase-based assay. Mice were made diabetic with streptozotocin. Expression of P2X7 was assessed by FACS, quantitative PCR, or immunohistochemistry. Results: Ligation of CD40 in primary RECs did not induce PCD. In contrast, in the presence of primary CD40+ Müller cells, CD40 stimulation caused PCD of RECs that was not impaired by L-Nil, α-lipoic acid, or anti-Fas ligand antibody. We found CD40 did not trigger TNF-α or IL-1ß secretion. Primary Müller cells released extracellular ATP in response to CD40 ligation. Inhibition of P2X7 (A-438079) impaired PCD of RECs; CD40 upregulated P2X7 in RECs, making them susceptible to ATP/P2X7-mediated PCD. Diabetic mice upregulated P2X7 in the retina and RECs in a CD40-dependent manner. Conclusions: Cluster of differentiation 40 induces PCD of RECs through a dual mechanism: ATP release by Müller cells and P2X7 upregulation in RECs. These findings are likely of in vivo relevance since CD40 upregulates P2X7 in RECs in diabetic mice and CD40 is known to be required for retinal capillary degeneration.


Subject(s)
Apoptosis , CD40 Ligand/metabolism , Diabetic Retinopathy/metabolism , Endothelial Cells/metabolism , Ependymoglial Cells/metabolism , Gene Expression Regulation , Receptors, Purinergic P2X7/genetics , Adenosine Triphosphate/metabolism , Animals , Cell Differentiation , Cells, Cultured , Diabetes Mellitus, Experimental , Diabetic Retinopathy/genetics , Diabetic Retinopathy/pathology , Endothelial Cells/pathology , Enzyme-Linked Immunosorbent Assay , Ependymoglial Cells/pathology , Flow Cytometry , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Purinergic P2X Receptor Antagonists/pharmacology , Pyridines/pharmacology , Real-Time Polymerase Chain Reaction , Receptors, Purinergic P2X7/metabolism , Retina/metabolism , Retina/pathology , Tetrazoles/pharmacology
13.
Exp Biol Med (Maywood) ; 241(12): 1265-71, 2016 06.
Article in English | MEDLINE | ID: mdl-27302174

ABSTRACT

Cells with DNA damage undergo apoptosis or cellular senescence if the damage cannot be repaired. Recent studies highlight that cellular senescence plays a major role in aging. However, age-associated diseases, including emphysema and neurodegenerative disorders, are caused by apoptosis of lung alveolar epithelial cells and neurons, respectively. Therefore, enhanced apoptosis also promotes aging and shortens the life span depending on the cell type. Recently, we reported that ku70(-) (/) (-)bax(-) (/) (-) and ku70(-) (/) (-)bax(+/) (-) mice showed significantly extended life span in comparison with ku70(-) (/) (-)bax(+/+) mice. Ku70 is essential for non-homologous end joining pathway for DNA double strand break repair, and Bax plays an important role in apoptosis. Our study suggests that Bax-induced apoptosis has a significant impact on shortening the life span of ku70(-) (/) (-) mice, which are defective in one of DNA repair pathways. The lung alveolar space gradually enlarges during aging, both in mouse and human, and this age-dependent change results in the decrease of respiration capacity during aging that can lead to emphysema in more severe cases. We found that emphysema occurred in ku70(-) (/) (-) mice at the age of three-months old, and that Bax deficiency was able to suppress it. These results suggest that Bax-mediated apoptosis induces emphysema in ku70(-) (/) (-) mice. We also found that the number of cells, including bronchiolar epithelial cells and type 2 alveolar epithelial cells, shows a higher DNA double strand break damage response in ku70 KO mouse lung than in wild type. Recent studies suggest that non-homologous end joining activity decreases with increased age in mouse and rat model. Together, we hypothesize that the decline of Ku70-dependent DNA repair activity in lung alveolar epithelial cells is one of the causes of age-dependent decline of lung function resulting from excess Bax-mediated apoptosis of lung alveolar epithelial cells (and their progenitor cells).


Subject(s)
Apoptosis , DNA Repair , Emphysema/pathology , Ku Autoantigen/deficiency , Longevity , bcl-2-Associated X Protein/metabolism , Animals , Mice , Mice, Knockout , Survival Analysis
14.
Oncotarget ; 6(29): 27388-402, 2015 Sep 29.
Article in English | MEDLINE | ID: mdl-26317541

ABSTRACT

Bcl-2 inhibits apoptosis by two distinct mechanisms but only one is targeted to treat Bcl-2-positive malignancies. In this mechanism, the BH1-3 domains of Bcl-2 form a hydrophobic pocket, binding and inhibiting pro-apoptotic proteins, including Bim. In the other mechanism, the BH4 domain mediates interaction of Bcl-2 with inositol 1,4, 5-trisphosphate receptors (IP3Rs), inhibiting pro-apoptotic Ca2+ signals. The current anti-Bcl-2 agents, ABT-263 (Navitoclax) and ABT-199 (Venetoclax), induce apoptosis by displacing pro-apoptotic proteins from the hydrophobic pocket, but do not inhibit Bcl-2-IP3R interaction. Therefore, to target this interaction we developed BIRD-2 (Bcl-2 IP3 Receptor Disruptor-2), a decoy peptide that binds to the BH4 domain, blocking Bcl-2-IP3R interaction and thus inducing Ca2+-mediated apoptosis in chronic lymphocytic leukemia, multiple myeloma, and follicular lymphoma cells, including cells resistant to ABT-263, ABT-199, or the Bruton's tyrosine kinase inhibitor Ibrutinib. Moreover, combining BIRD-2 with ABT-263 or ABT-199 enhances apoptosis induction compared to single agent treatment. Overall, these findings provide strong rationale for developing novel therapeutic agents that mimic the action of BIRD-2 in targeting the BH4 domain of Bcl-2 and disrupting Bcl-2-IP3R interaction.


Subject(s)
Lymphoma, Follicular/pathology , Multiple Myeloma/pathology , Peptides/chemistry , Proto-Oncogene Proteins c-bcl-2/metabolism , Aniline Compounds/therapeutic use , Animals , Apoptosis , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Calcium Signaling , Cell Line, Tumor , Cell Survival , Drug Resistance, Neoplasm , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Immunohistochemistry , Inositol 1,4,5-Trisphosphate Receptors/chemistry , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Lymphoma, Follicular/drug therapy , Mice , Mice, Nude , Multiple Myeloma/drug therapy , NIH 3T3 Cells , Neoplasm Transplantation , Protein Structure, Tertiary , Sulfonamides/therapeutic use , bcl-2-Associated X Protein/metabolism
15.
Exp Eye Res ; 123: 27-36, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24726920

ABSTRACT

The current study investigates the cellular events which trigger activation of proapoptotic Bcl-2-associated × protein (Bax) in retinal cell death induced by all-trans-retinal (atRAL). Cellular events which activate Bax, such as DNA damage by oxidative stress and phosphorylation of p53, were evaluated by immunochemical and biochemical methods using ARPE-19 cells, 661 W cells, cultured neural retinas and a retinal degeneration model, Abca4(-/-)Rdh8(-/-) mice. atRAL-induced Bax activation in cultured neural retinas was examined by pharmacological and genetic methods. Other Bax-related cellular events were also evaluated by pharmacological and biochemical methods. Production of 8-OHdG, a DNA damage indicator, and the phosphorylation of p53 at Ser46 were detected prior to Bax activation in ARPE-19 cells incubated with atRAL. Light exposure to Abca4(-/-)Rdh8(-/-) mice also caused the above mentioned events in conditions of short term intense light exposure and regular room lighting conditions. Incubation with Bax inhibiting peptide and deletion of the Bax gene partially protected retinal cells from atRAL toxicity in cultured neural retina. Necrosis was demonstrated not to be the main pathway in atRAL mediated cell death. Bcl-2-interacting mediator and Bcl-2 expression levels were not altered by atRAL in vitro. atRAL-induced oxidative stress results in DNA damage leading to the activation of Bax by phosphorylated p53. This cascade is closely associated with an apoptotic cell death mechanism rather than necrosis.


Subject(s)
Apoptosis/drug effects , DNA Damage , Retina/pathology , Retinal Pigment Epithelium/pathology , Retinaldehyde/toxicity , bcl-2-Associated X Protein/metabolism , 8-Hydroxy-2'-Deoxyguanosine , ATP-Binding Cassette Transporters/genetics , Alcohol Oxidoreductases/genetics , Animals , Cell Line , Colorimetry , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Disease Models, Animal , Gene Deletion , Humans , Immunoblotting , Immunohistochemistry , L-Lactate Dehydrogenase/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Culture Techniques , Phosphorylation , Retina/metabolism , Retinal Degeneration/metabolism , Retinal Degeneration/prevention & control , Retinal Pigment Epithelium/metabolism , Tomography, Optical Coherence , Tumor Suppressor Protein p53/metabolism
16.
Autophagy ; 8(2): 236-51, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22240589

ABSTRACT

Autophagy regulates cell survival and cell death upon various cellular stresses, yet the molecular signaling events involved are not well defined. Here, we established the function of a proteolytic Cyclin E fragment (p18-CycE) in DNA damage-induced autophagy, apoptosis, and senescence. p18-CycE was identified in hematopoietic cells undergoing DNA damage-induced apoptosis. In epithelial cells exposed to DNA damage, chronic but not transient expression of p18-CycE leads to higher turnover of LC3 I/II and increased emergence of autophagosomes and autolysosomes. Levels of p18-CycE, which was generated by proteolytic cleavage of endogenous Cyclin E, were greatly increased by chloroquine and correlated with LC 3II conversion. Preventing p18-CycE genesis blocked conversion of LC3 I to LC3 II. Upon DNA damage, cytoplasmic ataxia-telangiectasia-mutated (ATM) was phosphorylated in p18-CycE-expressing cells resulting in sustained activation of the adenosine-mono-phosphate-dependent kinase (AMPK). These lead to sustained activation of mammalian autophagy-initiating kinase ULK1, which was abrogated upon inhibiting ATM and AMPK phosphorylation. Moreover, p18-CycE was degraded via autophagy followed by induction of senescence. Both autophagy and senescence were prevented by inhibiting autophagy, which leads to increased apoptosis in p18-CycE-expressing cells by stabilizing p18-CycE expression. Senescence was further associated with cytoplasmic co-localization and degradation of p18-CycE and Ku70. In brief, chronic p18-CycE expression-induced autophagy leads to clearance of p18-CycE following DNA damage and induction of senescence. Autophagy inhibition stabilized the cytoplasmic p18-CycE-Ku70 complex leading to apoptosis. Thus, our findings define how chronic apoptotic stress and DNA damage initiate autophagy and regulate cell survival through senescence and/or apoptosis.


Subject(s)
Apoptosis , Autophagy , Cellular Senescence , DNA Damage , Stress, Physiological , Adenylate Kinase/metabolism , Animals , Antigens, Nuclear/metabolism , Cell Line , Cyclin E/metabolism , Cytoplasm/metabolism , DNA-Binding Proteins/metabolism , Enzyme Activation , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Ku Autoantigen , Microscopy, Electron, Scanning , Peptide Fragments/metabolism , Proteolysis
17.
J Biol Chem ; 286(46): 40083-90, 2011 Nov 18.
Article in English | MEDLINE | ID: mdl-21953454

ABSTRACT

Expression of the clusterin (CLU) gene results in the synthesis of a conventional secretory isoform set (pre- and mature secretory clusterin proteins, psCLU/sCLU), as well as another set of intracellular isoforms, appearing in the cytoplasm (pre-nuclear CLU, pnCLU) and in the nucleus as an ∼55-kDa mature nuclear clusterin (nCLU) form. These two isoform sets have opposing cell functions: pro-survival and pro-death, respectively. Although much is known about the regulation and function of sCLU as a pro-survival factor, the regulation and function of endogenous nCLU in cell death are relatively unexplored. Here, we show that depletion of endogenous nCLU protein using siRNA specific to its truncated mRNA increased clonogenic survival of ionizing radiation (IR)-exposed cells. nCLU-mediated apoptosis was Bax-dependent, and lethality correlated with accumulation of mature nCLU protein. nCLU accumulation was regulated by CRM1 because binding between CRM1 and nCLU proteins was significantly diminished by leptomycin B (LMB), and nuclear levels of nCLU protein were significantly enhanced by LMB and IR co-treatment. Moreover, LMB treatment significantly enhanced IR-induced nCLU-mediated cell death responses. Importantly, bax(-/-) and bax(-/-)/bak(-/-) double knock-out cells were resistant to nCLU-mediated cell death, whereas bak(-/-) or wild-type bax(+/+)/bak(+/+) cells were hypersensitive. The regulation of nCLU by CRM1 nuclear export/import may explain recent clinical results showing that highly malignant tumors have lost the ability to accumulate nCLU levels, thereby avoiding growth inhibition and cell death.


Subject(s)
Apoptosis/radiation effects , Cell Nucleus/metabolism , Clusterin/metabolism , Gamma Rays , Karyopherins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , bcl-2-Associated X Protein/metabolism , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/genetics , Active Transport, Cell Nucleus/radiation effects , Animals , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Cell Nucleus/genetics , Clusterin/genetics , Fatty Acids, Unsaturated/pharmacology , Humans , Karyopherins/genetics , Mice , Mice, Knockout , Protein Isoforms/genetics , Protein Isoforms/metabolism , Radiation Tolerance/drug effects , Radiation Tolerance/genetics , Receptors, Cytoplasmic and Nuclear/genetics , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/genetics , Exportin 1 Protein
18.
J Biol Chem ; 286(34): 30181-9, 2011 Aug 26.
Article in English | MEDLINE | ID: mdl-21733849

ABSTRACT

Glucocorticoid hormones, including dexamethasone, induce apoptosis in lymphocytes and consequently are used clinically as chemotherapeutic agents in many hematologic malignancies. Dexamethasone also induces autophagy in lymphocytes, although the mechanism is not fully elucidated. Through gene expression analysis, we found that dexamethasone induces the expression of a gene encoding a stress response protein variously referred to as Dig2, RTP801, or REDD1. This protein is reported to inhibit mammalian target of rapamycin (mTOR) signaling. Because autophagy is one outcome of mTOR inhibition, we investigated the hypothesis that Dig2/RTP801/REDD1 elevation contributes to autophagy induction in dexamethasone-treated lymphocytes. In support of this hypothesis, RNAi-mediated suppression of Dig2/RTP801/REDD1 reduces mTOR inhibition and autophagy in glucocorticoid-treated lymphocytes. We observed similar results in Dig2/Rtp801/Redd1 knock-out murine thymocytes treated with dexamethasone. Dig2/RTP801/REDD1 knockdown also leads to increased levels of dexamethasone-induced cell death, suggesting that Dig2/RTP801/REDD1-mediated autophagy promotes cell survival. Collectively, these findings demonstrate for the first time that elevation of Dig2/RTP801/REDD1 contributes to the induction of autophagy.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Autophagy/drug effects , Dexamethasone/pharmacology , Lymphocytes/metabolism , Transcription Factors/biosynthesis , Animals , Autophagy/physiology , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Lymphocytes/cytology , Mice , Mice, Knockout , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/genetics
19.
Blood ; 117(10): 2924-34, 2011 Mar 10.
Article in English | MEDLINE | ID: mdl-21193695

ABSTRACT

Bcl-2 contributes to the pathophysiology and therapeutic resistance of chronic lymphocytic leukemia (CLL). Therefore, developing inhibitors of this protein based on a thorough understanding of its mechanism of action is an active and promising area of inquiry. One approach centers on agents (eg, ABT-737) that compete with proapoptotic members of the Bcl-2 protein family for binding in the hydrophobic groove formed by the BH1-BH3 domains of Bcl-2. Another region of Bcl-2, the BH4 domain, also contributes to the antiapoptotic activity of Bcl-2 by binding to the inositol 1,4,5-trisphosphate receptor (IP3R) Ca²(+) channel, inhibiting IP(3)-dependent Ca²(+) release from the endoplasmic reticulum. We report that a novel synthetic peptide, modeled after the Bcl-2-interacting site on the IP3R, binds to the BH4 domain of Bcl-2 and functions as a competitive inhibitor of the Bcl-2-IP3R interaction. By disrupting the Bcl-2-IP3R interaction, this peptide induces an IP3R-dependent Ca²(+) elevation in lymphoma and leukemia cell lines and in primary CLL cells. The Ca²(+) elevation evoked by this peptide induces apoptosis in CLL cells, but not in normal peripheral blood lymphocytes, suggesting the involvement of the Bcl-2-IP3R interaction in the molecular mechanism of CLL and indicating the potential merit of targeting this interaction therapeutically.


Subject(s)
Apoptosis/physiology , Calcium/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Peptides/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Binding, Competitive , Blotting, Western , Cell Line, Tumor , Humans , Immunoprecipitation
20.
Methods Mol Biol ; 683: 465-71, 2011.
Article in English | MEDLINE | ID: mdl-21053150

ABSTRACT

The first series of cell-penetrating penta-peptides (CPP5s) were discovered as cytoprotective penta-peptides designed from the Bax-inhibiting domain of Ku70. Bax is an inducer of programmed cell death, and Ku70 is a multifunctional protein maintaining genomic stability and protecting cells from death by inhibiting the cytotoxic activity of Bax. Since these peptides bind and inhibit Bax, they are named Bax-inhibiting peptides (BIPs). The second series of CPP5s were developed by mutating BIP's amino acid sequences to abolish the Bax-binding activity. These peptides were used as negative control peptides to evaluate the Bax-inhibiting activity of BIPs. CPP5s are able to enter cells when they are added to the culture medium. The mechanism of cell entry of CPP5s is not yet understood. Numerous studies showed that BIP rescued cells from cytotoxic stresses both in cell culture and animal model, suggesting the therapeutic potential of BIP. Both BIPs and noncytoprotective CPP5s did not show significant toxicity even at 1.6 mM concentration in cell culture. Our recent study suggests that CPP5s has the protein transduction activity, though only green fluorescent protein (GFP) has been tested as a cargo protein. If CPP5s can deliver wide range of cargo molecules into the cell, CPP5s may be utilized as nontoxic drug delivery tool. In this article, we describe our laboratory's protocols of how to synthesize, store, and apply CPP5s for the examination of their activities of cell penetration and cytoprotection.


Subject(s)
Cell-Penetrating Peptides/metabolism , Cell-Penetrating Peptides/pharmacology , Oligopeptides/metabolism , Oligopeptides/pharmacology , Amino Acid Sequence , Animals , Cell Line , Cell-Penetrating Peptides/chemical synthesis , Cell-Penetrating Peptides/chemistry , Cytoprotection/drug effects , Humans , Mice , Oligopeptides/chemistry , Protein Transport
SELECTION OF CITATIONS
SEARCH DETAIL
...