Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters











Publication year range
1.
Inflamm Bowel Dis ; 29(6): 960-972, 2023 06 01.
Article in English | MEDLINE | ID: mdl-36661889

ABSTRACT

BACKGROUND: Early life stress (ELS) is an environmental trigger believed to promote increased risk of IBD. Our goal was to identify mechanisms whereby ELS in mice affects susceptibility to and/or severity of gut inflammation. METHODS: We utilized 2 published animal models of ELS. In the first model, newborn mice were separated from the dam daily for 4 to 8 hours starting on postnatal day 2 and then weaned early on postnatal day 17. Control mice were left undisturbed with the dams until weaning on postnatal day 21. In the second model, dams were fed dexamethasone or vehicle ad libitum in drinking water on postpartum days 1 to 14. Plasma and colonic corticosterone were measured in juvenile and adult mice. Colitis was induced in 4-week-old mice via intraperitoneal injection of interleukin (IL)-10 receptor blocking antibody every 5 days for 15 days. Five or 15 days later, colitis scores and transcripts for Tnf, glucocorticoid receptors, and steroidogenic enzymes were measured. RESULTS: Mice exposed to ELS displayed reduced plasma and colonic corticosterone. Control animals showed improvements in indices of inflammation following cessation of interleukin-10 receptor blockade, whereas ELS-exposed animals maintained high levels of Tnf and histological signs of colitis. In colitic animals, prior exposure to ELS was associated with significantly lower expression of genes associated with corticosterone synthesis and responsiveness. Finally, TNF stimulation of colonic crypt cells from ELS mice led to increased inhibition of corticosterone synthesis. CONCLUSIONS: Our study identifies impaired local glucocorticoid production and responsiveness as a potential mechanism whereby ELS predisposes to chronic colitis in susceptible hosts.


Using 2 distinct animal models, this study shows that in mice, early life stress leads to reduced colonic corticosterone and that induction of colitis after stress removal results in reduced transcription of glucocorticoid synthesis genes, increased Tnf, and enhanced chronicity of intestinal inflammation.


Subject(s)
Colitis , Stress, Psychological , Animals , Female , Mice , Colitis/metabolism , Corticosterone/pharmacology , Disease Models, Animal , Glucocorticoids , Inflammation/etiology , Stress, Psychological/complications
2.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Article in English | MEDLINE | ID: mdl-33753483

ABSTRACT

Genome-wide association studies have identified ICOSLG, which encodes the inducible costimulator ligand (ICOSLG or ICOSL) as a susceptibility locus for inflammatory bowel disease. ICOSL has been implicated in the enhancement of pattern recognition receptor signaling in dendritic cells, induction of IL-10 production by CD4 T cells, and the generation of high-affinity antibodies to specific antigens-all of which can potentially explain its involvement in gastrointestinal inflammation. Here, we show that murine ICOSL deficiency results in significant enrichment of IL-10-producing CD4 T cells particularly in the proximal large intestine. Transient depletion of IL-10-producing cells from adult ICOSL-deficient mice induced severe colonic inflammation that was prevented when mice were first treated with metronidazole. ICOSL-deficient mice displayed reduced IgA and IgG antibodies in the colon mucus and impaired serum antibody recognition of microbial antigens, including flagellins derived from mucus-associated bacteria of the Lachnospiraceae family. Confirming the synergy between ICOSL and IL-10, ICOSL deficiency coupled with CD4-specific deletion of the Il10 gene resulted in juvenile onset colitis that was impeded when pups were fostered by ICOSL-sufficient dams. In this setting, we found that both maternally acquired and host-derived antibodies contribute to the life anti-commensal antibody repertoire that mediates this protection in early life. Collectively, our findings reveal a partnership between ICOSL-dependent anti-commensal antibodies and IL-10 in adaptive immune regulation of the microbiota in the large intestine. Furthermore, we identify ICOSL deficiency as an effective platform for exploring the functions of anti-commensal antibodies in host-microbiota mutualism.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Gastrointestinal Microbiome/immunology , Inducible T-Cell Co-Stimulator Ligand/metabolism , Inflammatory Bowel Diseases/immunology , Interleukin-10/metabolism , Animals , Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , CD4-Positive T-Lymphocytes/metabolism , Colon/immunology , Colon/microbiology , Colon/pathology , Disease Models, Animal , Female , Host Microbial Interactions/immunology , Humans , Inducible T-Cell Co-Stimulator Ligand/genetics , Inducible T-Cell Co-Stimulator Protein/genetics , Inducible T-Cell Co-Stimulator Protein/metabolism , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/pathology , Interleukin-10/genetics , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Male , Mice , Mice, Knockout , Signal Transduction/immunology , Symbiosis/immunology
3.
J Immunol ; 202(4): 1039-1044, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30642977

ABSTRACT

The ICOS pathway has been implicated in the development and functions of regulatory T (Treg) cells, including those producing IL-10. Treg cell-derived IL-10 is indispensable for the establishment and maintenance of intestinal immune homeostasis. We examined the possible involvement of the ICOS pathway in the accumulation of murine colonic Foxp3- and/or IL-10-expressing cells. We show that ICOS deficiency does not impair induction of IL-10 by intestinal CD4 T cells but, instead, triggers substantial reductions in gut-resident and peripherally derived Foxp3+ Treg cells. ICOS deficiency is associated with reduced demethylation of Foxp3 CNS2 and enhanced loss of Foxp3. This instability significantly limits the ability of ICOS-deficient Treg cells to reverse ongoing inflammation. Collectively, our results identify a novel role for ICOS costimulation in imprinting the functional stability of Foxp3 that is required for the retention of full Treg cell function in the periphery.


Subject(s)
Down-Regulation , Forkhead Transcription Factors/metabolism , Inducible T-Cell Co-Stimulator Protein/metabolism , Interleukin-10/metabolism , T-Lymphocytes, Regulatory/metabolism , Animals , Down-Regulation/immunology , Forkhead Transcription Factors/immunology , Inducible T-Cell Co-Stimulator Protein/deficiency , Inducible T-Cell Co-Stimulator Protein/immunology , Inflammation/immunology , Inflammation/metabolism , Interleukin-10/immunology , Mice , Mice, Knockout , Mice, Transgenic , T-Lymphocytes, Regulatory/immunology
4.
Oncotarget ; 8(5): 8670-8678, 2017 Jan 31.
Article in English | MEDLINE | ID: mdl-28055968

ABSTRACT

Malignant peripheral nerve sheath tumors (MPNSTs) are aggressive, Schwann cell-derived neoplasms of the peripheral nervous system that have recently been shown to possess an autocrine CXCL12/CXCR4 signaling loop that promotes tumor cell proliferation and survival. Importantly, the CXCL12/CXCR4 signaling axis is driven by availability of the CXCL12 ligand rather than CXCR4 receptor levels alone. Therefore, pharmacological reduction of CXCL12 expression could be a potential chemotherapeutic target for patients with MPNSTs or other pathologies wherein the CXCL12/CXCR4 signaling axis is active. AT101 is a well-established BCL-2 homology domain 3 (BH3) mimetic that we recently demonstrated functions as an iron chelator and thus acts as a hypoxia mimetic. In this study, we found that AT101 significantly reduces CXCL12 mRNA and secreted protein in established human MPNST cell lines in vitro. This effect was recapitulated by other BH3 mimetics [ABT-737 (ABT), obatoclax (OBX) and sabutoclax (SBX)] but not by desferrioxamine (DFO), an iron chelator and known hypoxia mimetic. These data suggest that CXCL12 reduction is a function of AT101's BH3 mimetic property rather than its iron chelation ability. Additionally, this study investigates a potential mechanism of BH3 mimetic-mediated CXCL12 suppression: liberation of a negative CXCL12 transcriptional regulator, poly (ADP-Ribose) polymerase I (PARP1) from its physical interaction with BCL-2. These data suggest that clinically available BH3 mimetics might prove therapeutically useful at least in part by virtue of their ability to suppress CXCL12 expression.


Subject(s)
Antineoplastic Agents/pharmacology , Biphenyl Compounds/pharmacology , Gossypol/analogs & derivatives , Molecular Mimicry , Neurilemmoma/drug therapy , Nitrophenols/pharmacology , Pyrroles/pharmacology , Sulfonamides/pharmacology , Cell Line, Tumor , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Down-Regulation , Gene Expression Regulation, Neoplastic , Gossypol/pharmacology , Humans , Indoles , Neurilemmoma/genetics , Neurilemmoma/metabolism , Neurilemmoma/pathology , Piperazines/pharmacology , Poly (ADP-Ribose) Polymerase-1/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Proto-Oncogene Proteins c-bcl-2/metabolism
5.
J Neuropathol Exp Neurol ; 75(10): 946-954, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27516117

ABSTRACT

Glioblastomas (GBMs) are the most common and aggressive primary human malignant brain tumors. 4-Hydroxy tamoxifen (OHT) is an active metabolite of the tamoxifen (TMX) prodrug and a well-established estrogen receptor (ER) and estrogen-related receptor antagonist. A recent study from our laboratory demonstrated that OHT induced ER-independent malignant peripheral nerve sheath tumor (MPNST) cell death by autophagic degradation of the prosurvival protein Kirsten rat sarcoma viral oncogene homolog. Because both MPNST and GBM are glial in cell origin, we hypothesized that OHT could mediate similar effects in GBM. OHT induced a concentration-dependent reduction in cell viability that was largely independent of caspase activation in a human GBM cell line and 2 patient-derived xenolines. Further, OHT induced both cytotoxic autophagy and a concentration-dependent decrease in epidermal growth factor receptor (EGFR) protein levels. A GBM cell line expressing EGFR variant III (EGFRvIII) was relatively resistant to OHT-induced death and EGFRvIII was refractory to OHT-induced degradation. Thus, OHT induces GBM cell death through a caspase-independent, autophagy-related mechanism and should be considered as a potential therapeutic agent in patients with GBM whose tumors express wild-type EGFR.

7.
PLoS One ; 9(5): e96733, 2014.
Article in English | MEDLINE | ID: mdl-24824755

ABSTRACT

Malignant peripheral nerve sheath tumors (MPNSTs) are aggressive Schwann cell-derived sarcomas and are the leading cause of mortality in patients with neurofibromatosis type 1 (NF1). Current treatment modalities have been largely ineffective, resulting in a high rate of MPNST recurrence and poor five-year patient survival. This necessitates the exploration of alternative chemotherapeutic options for MPNST patients. This study sought to assess the cytotoxic effect of the BH3-mimetic AT101 [(-)-gossypol] on MPNST cells in vitro and to identify key regulators of AT101-induced MPNST cell death. We found that AT101 caused caspase-independent, non-apoptotic MPNST cell death, which was accompanied by autophagy and was mediated through HIF-1α induced expression of the atypical BH3-only protein BNIP3. These effects were mediated by intracellular iron chelation, a previously unreported mechanism of AT101 cytotoxicity.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cell Death/drug effects , Gossypol/analogs & derivatives , Membrane Proteins/metabolism , Neurilemmoma/drug therapy , Proto-Oncogene Proteins/metabolism , Antineoplastic Agents, Phytogenic/therapeutic use , Autophagy/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Gossypol/pharmacology , Gossypol/therapeutic use , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Iron/metabolism , Neurilemmoma/metabolism , Neurilemmoma/pathology , Schwann Cells/drug effects , Schwann Cells/metabolism , Schwann Cells/pathology
8.
Cancer Res ; 73(14): 4395-405, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23722551

ABSTRACT

Tamoxifen is widely used to treat estrogen receptor-positive breast cancer. Recent findings that tamoxifen and its derivative 4-hydroxytamoxifen (OHT) can exert estrogen receptor-independent cytotoxic effects have prompted the initiation of clinical trials to evaluate its use in estrogen receptor-negative malignancies. For example, tamoxifen and OHT exert cytotoxic effects in malignant peripheral nerve sheath tumors (MPNST) where estrogen is not involved. In this study, we gained insights into the estrogen receptor-independent cytotoxic effects of OHT by studying how it kills MPNST cells. Although caspases were activated following OHT treatment, caspase inhibition provided no protection from OHT-induced death. Rather, OHT-induced death in MPNST cells was associated with autophagic induction and attenuated by genetic inhibition of autophagic vacuole formation. Mechanistic investigations revealed that OHT stimulated autophagic degradation of K-Ras, which is critical for survival of MPNST cells. Similarly, we found that OHT induced K-Ras degradation in breast, colon, glioma, and pancreatic cancer cells. Our findings describe a novel mechanism of autophagic death triggered by OHT in tumor cells that may be more broadly useful clinically in cancer treatment.


Subject(s)
Autophagy/drug effects , Cell Death/drug effects , Nerve Sheath Neoplasms/drug therapy , Proto-Oncogene Proteins/metabolism , Tamoxifen/analogs & derivatives , ras Proteins/metabolism , Autophagy/genetics , Caspases/genetics , Caspases/metabolism , Cell Death/genetics , Cell Line, Tumor , Down-Regulation/drug effects , ErbB Receptors/genetics , ErbB Receptors/metabolism , HCT116 Cells , Humans , MCF-7 Cells , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Nerve Sheath Neoplasms/enzymology , Nerve Sheath Neoplasms/metabolism , Nerve Sheath Neoplasms/pathology , Protein Kinase C/genetics , Protein Kinase C/metabolism , Proteolysis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins p21(ras) , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Tamoxifen/pharmacology , ras Proteins/genetics
9.
ACS Chem Neurosci ; 3(12): 1063-72, 2012 Dec 19.
Article in English | MEDLINE | ID: mdl-23259041

ABSTRACT

Rotenone, which selectively inhibits mitochondrial complex I, induces oxidative stress, α-synuclein accumulation, and dopaminergic neuron death, principal pathological features of Parkinson's disease. The autophagy-lysosome pathway degrades damaged proteins and organelles for the intracellular maintenance of nutrient and energy balance. While it is known that rotenone causes autophagic vacuole accumulation, the mechanism by which this effect occurs has not been thoroughly investigated. Treatment of differentiated SH-SY5Y cells with rotenone (10 µM) induced the accumulation of autophagic vacuoles at 6 h and 24 h as indicated by Western blot analysis for microtubule associated protein-light chain 3-II (MAP-LC3-II). Assessment of autophagic flux at these time points indicated that autophagic vacuole accumulation resulted from a decrease in their effective lysosomal degradation, which was substantiated by increased levels of autophagy substrates p62 and α-synuclein. Inhibition of lysosomal degradation may be explained by the observed decrease in cellular ATP levels, which in turn may have caused the observed concomitant increase in acidic vesicle pH. The early (6 h) effects of rotenone on cellular energetics and autophagy-lysosome pathway function preceded the induction of cell death and apoptosis. These findings indicate that the classical mitochondrial toxin rotenone has a pronounced effect on macroautophagy completion that may contribute to its neurotoxic potential.


Subject(s)
Autophagy/drug effects , Cell Death/drug effects , Neurons/drug effects , Rotenone/pharmacology , Uncoupling Agents/pharmacology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Line, Tumor , Humans , Lysosomal-Associated Membrane Protein 1/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Lysosomes/pathology , Neurons/metabolism , Neurons/pathology , Vacuoles/drug effects , Vacuoles/metabolism , Vacuoles/pathology
10.
PLoS One ; 7(6): e39586, 2012.
Article in English | MEDLINE | ID: mdl-22761832

ABSTRACT

Endoplasmic reticulum (ER) stress-induced apoptosis has been implicated in various neurodegenerative diseases including Parkinson Disease, Alzheimer Disease and Huntington Disease. PUMA (p53 upregulated modulator of apoptosis) and BIM (BCL2 interacting mediator of cell death), pro-apoptotic BH3 domain-only, BCL2 family members, have previously been shown to regulate ER stress-induced cell death, but the upstream signaling pathways that regulate this response in neuronal cells are incompletely defined. Consistent with previous studies, we show that both PUMA and BIM are induced in response to ER stress in neuronal cells and that transcriptional induction of PUMA regulates ER stress-induced cell death, independent of p53. CHOP (C/EBP homologous protein also known as GADD153; gene name Ddit3), a critical initiator of ER stress-induced apoptosis, was found to regulate both PUMA and BIM expression in response to ER stress. We further show that CHOP knockdown prevents perturbations in the AKT (protein kinase B)/FOXO3a (forkhead box, class O, 3a) pathway in response to ER stress. CHOP co-immunoprecipitated with FOXO3a in tunicamycin treated cells, suggesting that CHOP may also regulate other pro-apoptotic signaling cascades culminating in PUMA and BIM activation and cell death. In summary, CHOP regulates the expression of multiple pro-apoptotic BH3-only molecules through multiple mechanisms, making CHOP an important therapeutic target relevant to a number of neurodegenerative conditions.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Endoplasmic Reticulum Stress/physiology , Forkhead Transcription Factors/metabolism , Membrane Proteins/metabolism , Neurons/metabolism , Proto-Oncogene Proteins/metabolism , Transcription Factor CHOP/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Apoptosis/drug effects , Apoptosis/physiology , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Cell Survival/drug effects , Cell Survival/physiology , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Membrane Proteins/genetics , Mice , Neurons/cytology , Neurons/drug effects , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Telencephalon/cytology , Telencephalon/drug effects , Telencephalon/metabolism , Transcription Factor CHOP/genetics , Tumor Suppressor Proteins/genetics , Tunicamycin/pharmacology
11.
J Histochem Cytochem ; 59(11): 976-83, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22043021

ABSTRACT

The BCL-2 family includes both pro- and anti-apoptotic proteins, which regulate programmed cell death during development and in response to various apoptotic stimuli. The BH3-only subgroup of pro-apoptotic BCL-2 family members is critical for the induction of apoptotic signaling, by binding to and neutralizing anti-apoptotic BCL-2 family members. During embryonic development, the anti-apoptotic protein BCL-X(L) plays a critical role in the survival of neuronal populations by regulating the multi-BH domain protein BAX. In this study, the authors investigated the role of Harakiri (HRK), a relatively recently characterized BH3-only molecule in disrupting the BAX-BCL-X(L) interaction during nervous system development. Results indicate that HRK deficiency significantly reduces programmed cell death in the nervous system. However, HRK deficiency does not significantly attenuate the widespread apoptosis seen in the Bcl-x (-/-) embryonic nervous system, indicating that other BH3-only molecules, alone or in combination, may regulate BAX activation in immature neurons.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Nervous System/embryology , Neurons/cytology , Neuropeptides/metabolism , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Cell Death , Gene Deletion , Mice , Mice, Inbred C57BL , Neuropeptides/genetics , bcl-X Protein/genetics
12.
Neuro Oncol ; 12(5): 473-81, 2010 May.
Article in English | MEDLINE | ID: mdl-20406898

ABSTRACT

Glioblastoma (GBM) is a high-grade central nervous system malignancy and despite aggressive treatment strategies, GBM patients have a median survival time of just 1 year. Chloroquine (CQ), an antimalarial lysosomotropic agent, has been identified as a potential adjuvant in the treatment regimen of GBMs. However, the mechanism of CQ-induced tumor cell death is poorly defined. We and others have shown that CQ-mediated cell death may be p53-dependent and at least in part due to the intrinsic apoptotic death pathway. Here, we investigated the effects of CQ on 5 established human GBM lines, differing in their p53 gene status. CQ was found to induce a concentration-dependent death in each of these cell lines. Although CQ treatment increased caspase-3-like enzymatic activity in all 5 cell lines, a broad-spectrum caspase inhibitor did not significantly attenuate death. Moreover, CQ caused an accumulation of autophagic vacuoles in all cell lines and was found to affect the levels and subcellular distribution of cathepsin D, suggesting that altered lysosomal function may also play a role in CQ-induced cell death. Thus, CQ can induce p53-independent death in gliomas that do not require caspase-mediated apoptosis. To potentially identify more potent chemotherapeutics, various CQ derivatives and lysosomotropic compounds were tested on the GBM cells. Quinacrine and mefloquine were found to be more potent than CQ in killing GBM cells in vitro and given their superior blood-brain barrier penetration compared with CQ may prove more efficacious as chemotherapeutic agents for GBM patients.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/genetics , Brain Neoplasms/genetics , Chloroquine/pharmacology , Glioma/genetics , Tumor Suppressor Protein p53/genetics , Autophagy/drug effects , Blotting, Western , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Glioma/metabolism , Glioma/pathology , Humans , Immunohistochemistry , Mefloquine/pharmacology , Quinacrine/pharmacology , Tumor Suppressor Protein p53/metabolism , Vacuoles/drug effects , Vacuoles/genetics , Vacuoles/pathology
13.
J Biol Chem ; 285(14): 10497-507, 2010 Apr 02.
Article in English | MEDLINE | ID: mdl-20123985

ABSTRACT

Macroautophagy (autophagy) is a process wherein bulk cytosolic proteins and damaged organelles are sequestered and degraded via the lysosome. Alterations in autophagy-associated proteins have been shown to cause neural tube closure defects, neurodegeneration, and tumor formation. Normal lysosome function is critical for autophagy completion and when altered may lead to an accumulation of autophagic vacuoles (AVs) and caspase activation. The tumor suppressor p53 is highly expressed in neural precursor cells (NPCs) and has an important role in the regulation of both autophagy and apoptosis. We hypothesized that altered lysosome function would lead to NPC death via an interaction between autophagy- and apoptosis-associated proteins. To test our hypothesis, we utilized FGF2-expanded NPCs and the neural stem cell line, C17.2, in combination with the lysosomotropic agent chloroquine (CQ) and the vacuolar ATPase inhibitor bafilomycin A1 (Baf A1). Both CQ and Baf A1 caused concentration- and time-dependent AV accumulation, p53 phosphorylation, increased damage regulator autophagy modulator levels, caspase-3 activation, and cell death. Short hairpin RNA knockdown of Atg7, but not Beclin1, expression significantly inhibited CQ- and Baf A1-induced cell death, indicating that Atg7 is an upstream mediator of lysosome dysfunction-induced cell death. Cell death and/or caspase-3 activation was also attenuated by protein synthesis inhibition, p53 deficiency, or Bax deficiency, indicating involvement of the intrinsic apoptotic death pathway. In contrast to lysosome dysfunction, starvation-induced AV accumulation was inhibited by either Atg7 or Beclin1 knockdown, and Atg7 knockdown had no effect on starvation-induced death. These findings indicate that Atg7- and Beclin1-induced autophagy plays a cytoprotective role during starvation but that Atg7 has a unique pro-apoptotic function in response to lysosome dysfunction.


Subject(s)
Apoptosis , Cerebellum/metabolism , Lysosomes/metabolism , Microtubule-Associated Proteins/metabolism , Neurons/metabolism , Stem Cells/metabolism , Animals , Antifungal Agents/pharmacology , Antimalarials/pharmacology , Autophagy , Autophagy-Related Protein 7 , Blotting, Western , Caspases/metabolism , Cerebellum/cytology , Chloroquine/pharmacology , Fibroblast Growth Factor 2 , Fluorescent Antibody Technique , Lysosomes/pathology , Macrolides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/genetics , Neurons/cytology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/cytology , Tumor Suppressor Protein p53/physiology , bcl-2-Associated X Protein/physiology
14.
J Histochem Cytochem ; 58(3): 265-75, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19901272

ABSTRACT

The prodeath effects of p53 are typically mediated via its transcriptional upregulation of proapoptotic Bcl-2 family members, including PUMA, Noxa, and/or Bax. We previously reported that staurosporine (STS), a broad-spectrum kinase inhibitor and prototypical apoptosis-inducing agent, produced p53-dependent, Bax-dependent, neural precursor cell (NPC) apoptosis, but that this effect occurred independently of new gene transcription and PUMA expression. To further characterize the mechanism by which p53 regulates NPC death, we used primary cerebellar NPCs derived from wild-type, p53-deficient, and Bax-deficient neonatal mice and the mouse cerebellar neural stem cell line, C17.2. We found that STS rapidly increased p53 cytoplasmic immunoreactivity in neuritic-like processes in C17.2 cells, which preceded Bax activation and caspase-3 cleavage. Confocal microscopy analysis of STS-treated cells revealed partial colocalization of p53 with the mitochondrial marker pyruvate dehydrogenase as well as with conformationally altered "activated" Bax, suggesting an interaction between these proapoptotic molecules in triggering apoptotic death. Nucleophosmin (NPM), a CRM1-dependent nuclear chaperone, also exhibited partial colocalization with both activated Bax and p53 following STS treatment. These observations suggest that cytoplasmic p53 can trigger transcription-independent NPC apoptosis through its potential interaction with NPM and activated Bax.


Subject(s)
Apoptosis , Cytoplasm/metabolism , Neurons/physiology , Stem Cells/physiology , Tumor Suppressor Protein p53/physiology , bcl-2-Associated X Protein/physiology , Animals , Animals, Newborn , Caspase 3/metabolism , Cell Line , Cerebellum/cytology , Enzyme Activation , Mice , Mice, Knockout , Mitochondria/metabolism , Nuclear Proteins/metabolism , Nucleophosmin , Transcription, Genetic , Tumor Suppressor Protein p53/genetics , bcl-2-Associated X Protein/genetics
15.
J Neuropathol Exp Neurol ; 68(12): 1326-38, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19915483

ABSTRACT

Perinatal hypoxia-ischemia may result in long-term neurological deficits. In addition to producing neuron death, HI causes death of neural precursor cells (NPCs) in the developing brain. To characterize the molecular pathways that regulate hypoxia-induced death of NPCs, we treated a mouse neural stem cell line (C17.2 cells) and fibroblastic growth factor II-expanded primary NPCs derived from wild-type or gene-disrupted mice, with oxygen glucose deprivation or the hypoxia mimetics desferrioxamine or cobalt chloride. Neural precursor cells undergoing hypoxia exhibited time- and concentration-dependent caspase-3 activation and cell death, which was significantly reduced by treatment with a broad caspase inhibitor or protein synthesis inhibition. Bax/Bak-deficient NPCs were protected from desferrioxamine-induced death and exhibited minimal caspase-3 activation. Oxygen glucose deprivation or hypoxia-mimetic exposure also resulted in increased hypoxia-inducible factor alpha and bcl-2/adenovirus E1B 19-kd interacting protein 3 (BNIP3) expression. BNIP3 shRNA treatment failed to affect hypoxia-induced caspase-3 activation but inhibited cell death and nuclear translocation of apoptosis-inducing factor, indicating that BNIP3 is an important regulator of caspase-independent NPC death after hypoxia. These studies demonstrate that hypoxia activates both caspase-dependent and -independent NPC death pathways that are critically regulated by multiple Bcl-2 family members.


Subject(s)
Apoptosis/physiology , Membrane Proteins/metabolism , Mitochondrial Proteins/metabolism , Neurons/metabolism , Signal Transduction/physiology , Stem Cells/metabolism , Animals , Apoptosis/drug effects , Blotting, Western , Caspase 3/metabolism , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Neurons/drug effects , Neurons/pathology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/drug effects , Stem Cells/pathology
16.
J Neuropathol Exp Neurol ; 68(7): 747-56, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19535997

ABSTRACT

Synaptogenesis in humans occurs in the last trimester of gestation and in the first few years of life, whereas it occurs in the postnatal period in rodents. A single exposure of neonatal rodents to ethanol during this period evokes extensive neuronal apoptosis. Previous studies indicate that ethanol triggers the intrinsic apoptotic pathway in neurons, and that this requires the multi-BH domain, proapoptotic Bcl-2 family member Bax. To define the upstream regulators of this apoptotic pathway, we examined the possible roles of p53 and a subclass of proapoptotic Bcl-2 family members (i.e. the BH3 domain-only proteins) in neonatal wild-type and gene-targeted mice that lack these cell death inducers. Acute ethanol exposure produced greater caspase-3 activation and neuronal apoptosis in wild-type mice than in saline-treated littermate controls. Loss of p53-upregulated mediator of apoptosis (Puma) resulted in marked protection from ethanol-induced caspase-3 activation and apoptosis. Although Puma expression has been reported to be regulated by p53, p53-deficient mice exhibited a similar extent of ethanol-induced caspase-3 activation and neuronal apoptosis as wild-type mice. Mice deficient in other proapoptotic BH3-only proteins, including Noxa, Bim, or Hrk, showed no significant protection from ethanol-induced neuronal apoptosis. Collectively, these studies indicate a p53-independent, Bax- and Puma-dependent mechanism of neuronal apoptosis and identify Puma as a possible molecular target for inhibiting the effects of intrauterine ethanol exposure in humans.


Subject(s)
Apoptosis/physiology , Ethanol/toxicity , Neurons/physiology , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , bcl-2-Associated X Protein/metabolism , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Blotting, Western , Caspase 3/metabolism , Immunohistochemistry , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Neuropeptides/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Time Factors , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics , bcl-2-Associated X Protein/genetics
17.
J Histochem Cytochem ; 56(10): 921-7, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18606610

ABSTRACT

Members of the Bcl-2 family include pro- and antiapoptotic proteins that regulate programmed cell death of developing tissues and death in response to cellular damage. In developing mice, the antiapoptotic Bcl-x(L) is necessary for survival of neural and hematopoietic cells, and consequently, bcl-x-deficient mice die around Day 13.5 of embryogenesis. Furthermore, adult bcl-x(+/-) heterozygous male mice have reduced fertility because of testicular degeneration. Bax, a multi-BH (Bcl-2 homology) domain proapoptotic member of the Bcl-2 family, is regulated by Bcl-x(L) and is required for the neuropathological abnormalities seen in bcl-x-deficient embryos. The BH3 domain only subgroup of the Bcl-2 family includes proapoptotic members that are essential for the initiation of apoptotic signaling. In this study, we investigated the role for Bim, a BH3 domain only protein, in the embryonic lethality and increased developmental cell death in bcl-x-deficient animals and the perturbed testicular function in bcl-x(+/-) adults. Our studies show that bim deficiency attenuates hematopoietic cell death in the fetal liver of bcl-x-deficient animals, indicating that Bim contributes to programmed cell death in this cell population. In addition, we found that testicular degeneration of adult bcl-x(+/-) males was rescued by concomitant Bim deficiency. However, concomitant Bim deficiency had no effect on the embryonic lethality and widespread nervous system abnormalities caused by bcl-x deficiency. Our work identifies Bim as an important regulator of bcl-x deficiency-induced cell death during hematopoiesis and testicular development.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Apoptosis , Germ Cells/cytology , Hematopoietic Stem Cells/cytology , Liver/cytology , Membrane Proteins/physiology , Neurons/cytology , Proto-Oncogene Proteins/physiology , bcl-X Protein/genetics , Animals , Bcl-2-Like Protein 11 , Germ Cells/metabolism , Hematopoietic Stem Cells/metabolism , Liver/embryology , Liver/metabolism , Male , Mice , Mice, Knockout , Neurons/metabolism , Protein Structure, Tertiary , Testis/abnormalities , Testis/cytology , Testis/embryology
18.
Autophagy ; 3(3): 222-9, 2007.
Article in English | MEDLINE | ID: mdl-17297299

ABSTRACT

Cathepsin D (CD) is an essential lysosomal protease and mice lacking this enzyme exhibit neuropathology similar to that observed in brains of patients with neuronal ceroid lipofuscinosces (NCL/Batten disease), a group of autosomal recessive pediatric neurodegenerative diseases. CD-deficient (CD-/-) brains exhibit a dramatic induction of autophagic stress as defined by the aberrant accumulation of autophagosomes, which is concomitant with markers of apoptosis. However, the signaling abnormalities which lead to CD deficiency-induced neurodegeneration are poorly defined. Since phosphatidylinositol-3 kinase (PI3-K) is known to regulate both apoptosis and autophagy, PI3-K-mediated signaling events were assessed in CD-/- brain at P14 and P25-26. Compared to WT littermate controls, CD-/- cortical neurons exhibited a widespread decrease in phosphorylation of Akt (inactivation) and GSK3beta (disinhibition) at P25-26, while levels of total Akt and GSK3beta remained unchanged. This P25-26-specific decrease in phosphorylation of Akt and GSK-3beta in CD-/- brain coincided temporally with markers of apoptosis but followed the induction of autophagic stress observed at both P14 and P25-26. In addition, levels and/or activation of mTOR and Beclin were not affected by CD deficiency, suggesting that the accumulation of autophagosomes is not due to an increased synthesis of autophagosomes but rather from an inhibition of autophagosome recycling, due most likely to a compromise in lysosome function. Together these observations indicate a pronounced decrease in pro-survival PI3-K signaling in CD-/- brain that may contribute to autophagic stress-induced and apoptotic neuropathology.


Subject(s)
Brain/metabolism , Cathepsin D/deficiency , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Animals , Apoptosis , Apoptosis Regulatory Proteins , Autophagy , Beclin-1 , Brain/cytology , Brain/enzymology , Cathepsin D/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Mice , Mice, Inbred C57BL , Neurons/metabolism , Phagosomes/metabolism , Phosphorylation , Protein Kinases/metabolism , Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases
19.
J Neurosci ; 27(8): 2081-90, 2007 Feb 21.
Article in English | MEDLINE | ID: mdl-17314303

ABSTRACT

Neuronal ceroid lipofuscinosces/Batten disease (NCL) is a devastating group of neurodegenerative diseases caused by genetic disruptions in lysosomal function. Cathepsin D (CD) is a major lysosomal protease, and mutations in CD that render it enzymatically defective have been reported recently in subsets of NCL patients. The targeted deletion of CD in mice results in extensive neuropathology, including biochemical and morphological evidence of apoptosis and autophagic stress (aberrant autophagosome accumulation), effects that are similar to those observed in NCL. To determine the contribution of Bax-dependent apoptosis in this mouse model of NCL, combined Bax- and CD-deficient mice were generated. Morphological analysis of CD-deficient mouse brains indicated large numbers of pyknotic neurons and neurons with marked cytoplasmic swellings containing undigested lipofuscin. Cell death and apoptosis were evidenced by increases in terminal deoxynucleotidyl transferase-mediated biotinylated UTP nick end labeling (TUNEL) reactivity and activation of caspase-3, respectively. DeOlmos silver-positive neurons were abundant in CD-deficient brain and correlated with neuron loss, as indicated by significant decreases in NeuN (neuronal nuclear antigen)-positive neurons. Lysosome dysfunction and autophagic stress were apparent in CD-deficient brain as indicated by the accumulation of autofluorescent storage material and by increased levels of LC3-II (light chain 3-II, a selective autophagosome marker), respectively. Bax deletion significantly inhibited caspase-3 activation and hippocampal TUNEL reactivity but did not prevent the majority of CD deficiency-induced neuropathology, including the persistence of pyknotic neurons, elevated cortical TUNEL reactivity, lysosome dysfunction and autophagic stress, neurodegeneration, and neuron loss. Together, these results suggest that CD deficiency-induced neuropathology does not require Bax-dependent apoptosis and highlights the importance of caspase-independent neuron death and neurodegeneration resulting from the genetic disruption of lysosome function.


Subject(s)
Apoptosis , Cathepsin D/deficiency , Nerve Degeneration/etiology , Nerve Degeneration/physiopathology , bcl-2-Associated X Protein/deficiency , Animals , Autophagy , Brain/metabolism , Brain/pathology , Caspase 3/metabolism , Cell Death , DNA-Binding Proteins , Disease Models, Animal , Enzyme Activation , Hippocampus/physiopathology , In Situ Nick-End Labeling , Lipofuscin/metabolism , Lysosomes , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Nerve Tissue Proteins/metabolism , Neuronal Ceroid-Lipofuscinoses , Neurons/metabolism , Neurons/pathology , Nuclear Proteins/metabolism
20.
J Neuropathol Exp Neurol ; 66(1): 66-74, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17204938

ABSTRACT

Regulation of cerebellar neural precursor cell (NPC) death is important for both normal brain development and prevention of brain tumor formation. The tumor suppressor p53 is an important regulator of NPC apoptosis, but the precise mechanism of p53-regulated cerebellar NPC death remains largely unknown. Here, by using primary cerebellar NPCs and a mouse cerebellar NPC line, we compared the molecular regulation of cerebellar NPC death produced by staurosporine (STS), a broad-spectrum kinase inhibitor, with that caused by genotoxic agents. We found that both STS- and genotoxin-induced cerebellar NPC death were markedly inhibited by p53 or Bax deficiency. Genotoxin-induced cerebellar NPC death required new protein synthesis and PUMA, a p53 transcriptionally regulated BH3-only molecule. In contrast, STS caused cerebellar NPC death without requiring new protein synthesis or PUMA expression. In addition, genotoxic agents increased nuclear p53 immunoreactivity, whereas STS produced rapid cytoplasmic p53 accumulation. Interestingly, STS-induced death of cerebellar granule neurons was p53-independent, indicating a differentiation-dependent feature of neuronal apoptotic regulation. These results suggest that STS-induced cerebellar NPC death requires a direct effect of p53 on cytoplasmic apoptotic mediators, whereas genotoxin-induced death requires p53-dependent gene transcription of PUMA. Thus, p53 has multiple death promoting mechanisms in cerebellar NPCs.


Subject(s)
Apoptosis/physiology , Cerebellum/cytology , Neurons/cytology , Stem Cells/cytology , Transcriptional Activation/physiology , Tumor Suppressor Protein p53/metabolism , Analysis of Variance , Animals , Animals, Newborn , Apoptosis Regulatory Proteins , Blotting, Western/methods , Caspases/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Immunohistochemistry/methods , Mice , Mice, Knockout , Neurons/drug effects , Neurons/physiology , Protein Synthesis Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-2/deficiency , Staurosporine/pharmacology , Stem Cells/drug effects , Stem Cells/physiology , Time Factors , Tumor Suppressor Proteins/deficiency , bcl-2 Homologous Antagonist-Killer Protein/deficiency , bcl-2-Associated X Protein/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL