Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters










Publication year range
1.
Apoptosis ; 9(4): 423-7, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15192324

ABSTRACT

The complex sequence of events leading to apoptotic cell death is governed by an elaborate regulatory scheme involving the actions of both initiator and executioner proteases. Among the most intensively studied of the initiator caspases is caspase-9, an essential throughput element in the so-called intrinsic or mitochondrially gated pathway of apoptosis. Previous reviews have described the proteolytic processing and activation of this protease in much detail; here we provide an update on caspase-9 regulation. A comprehensive description of the intra- and intermolecular events involved in modulating protein expression and activity are presented. Particular emphasis is placed on the role alternative splicing plays in the expression of functionally divergent protein isoforms, as well as, the participation of specific post-translational events in regulating caspase-9 activity. Such discrete modulation in reported activity characterizes, not only the pivotal role of this protease in the final commitment process itself, but also emphasizes the more general interplay that exists between mutually opposing cytotoxic and cytoprotective influences in maintaining cellular homeostasis.


Subject(s)
Caspases/metabolism , Alternative Splicing , Animals , Caspase 9 , Caspases/genetics , Cell Nucleus/enzymology , Cytosol/enzymology , Gene Expression Regulation , Humans , Mitochondria/enzymology , Nitric Oxide/metabolism , Phosphorylation , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Processing, Post-Translational , Protein Transport
2.
Leukemia ; 17(11): 2140-8, 2003 Nov.
Article in English | MEDLINE | ID: mdl-12970763

ABSTRACT

The present studies examined performance of SAPK cascades and apoptotic commitment following ribosomal trauma in REH lymphoid leukemia cells. Ribostatic insults included disruption of ribosomal activity by mechanistically dissimilar agents such as blasticidin-S (BCS) (which binds 28S-rRNA to block peptidyl bond formation), kasugamycin (KSM) (which binds 18S-rRNA to prevent translational initiation), and cycloheximide (CHX) (which blocks A-site to P-site translocation of peptidyl-tRNA). Exposure of REH cells to BCS elicited DNA degradation and apoptotic cytolysis. BCS stimulated JNK1/JNK2 and p38, and their shared targets c-Jun and ATF2. Inhibition of JNK1/JNK2 (but not of p38) antagonized blasticidin-induced apoptosis, whereas targeting alternative ribosomal sites with KSM or CHX limited translation, but failed to activate the SAPK cascade or initiate apoptosis. Our findings indicate that interference with 28S-rRNA by BCS initiates apoptosis in REH cells through recruitment of SAPK-JNK signaling. Disparities between the lethal actions of BCS, KSM, and CHX appear to reflect established differences in the subribosomal targets of these agents. We propose that the SAPK cascade comprises an essential mechanism for the transduction of specific lethal stress signals emanating from active ribosomes, and that interference with the 28S-rRNA, rather than the peptidyl transfer center of the large subunit, is critical to apoptotic commitment.


Subject(s)
Apoptosis/physiology , MAP Kinase Signaling System/physiology , Caspases/metabolism , Cycloheximide/pharmacology , DNA, Neoplasm/drug effects , Enzyme Inhibitors/pharmacology , Humans , JNK Mitogen-Activated Protein Kinases , Leukemia, Lymphoid , Mitogen-Activated Protein Kinases/metabolism , Nucleosides/toxicity , Peptidyl Transferases/antagonists & inhibitors , Tumor Cells, Cultured
3.
Oncogene ; 18(50): 7016-25, 1999 Nov 25.
Article in English | MEDLINE | ID: mdl-10597302

ABSTRACT

Determinants of differentiation and apoptosis in myelomonocytic leukemia cells (U937) exposed to the novel hybrid polar compound SAHA (suberoylanilide hydroxamic acid) have been examined. In contrast to hexamethylenbisacetamide (HMBA), SAHA-related maturation was limited and accompanied by marked cytoxicity. SAHA-mediated apoptosis occurred within the G0G1 and S phase populations, and was associated with decreased mitochondrial membrane potential, caspase-3 activation, PARP degradation, hypophosphorylation/cleavage of pRB, and down-regulation of c-Myc, c-Myb, and B-Myb. Enforced expression of Bcl-2 or Bcl-XL inhibited SAHA-induced apoptosis, but only modestly potentiated differentiation. While SAHA induced the cyclin-dependent kinase inhibitor p21CIP1, antisense ablation of this CDKI increased, rather than decreased, SAHA-related lethality. In contrast, conditional expression of wild-type p53 failed to modify SAHA actions, but markedly potentiated HMBA-induced apoptosis. Finally, SAHA modestly increased expression/activation of the stress-activated protein kinase (SAPK/JNK); moreover, SAHA-related lethality was partially attenuated by a dominant-negative c-Jun mutant protein (TAM67). SAHA did not stimulate mitogen-activated protein kinase (MAPK), nor was lethality diminished by the specific MEK/MAPK inhibitor PD98059. These findings indicate that SAHA potently induces apoptosis in human leukemia cells via a pathway that is p53-independent but at least partially regulated by Bcl-2/Bcl-XL, p21CIP1, and the c-Jun/AP-1 signaling cascade.


Subject(s)
Apoptosis/drug effects , Cyclins/metabolism , Hydroxamic Acids/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Cyclin-Dependent Kinase Inhibitor p21 , Down-Regulation , Humans , Leukemia/metabolism , Leukemia/pathology , Proto-Oncogene Proteins c-myb/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Tumor Suppressor Protein p53/metabolism , U937 Cells , Vorinostat , bcl-X Protein
4.
J Immunol ; 163(12): 6442-7, 1999 Dec 15.
Article in English | MEDLINE | ID: mdl-10586034

ABSTRACT

The observation that follicular dendritic cells (FDC) reduce apoptosis in B cells prompted the hypothesis that FDC might enhance tumor cell survival by protecting malignant B cells from apoptotic death. To test this notion, apoptosis was induced in B cell lymphomas by anti-Fas or various antineoplastic agents in the presence and absence of FDC. Apoptosis was detected and quantified by TUNEL analysis. Induction of apoptosis with anti-Fas, etoposide, cyclophosphamide, and busulfan was markedly antagonized by FDC at FDC to B cell ratios of >/=1:16. For example, treatment with 10 ng/ml anti-Fas caused 60-90% of A20 cells to undergo apoptosis in 6 h, whereas addition of FDC reduced apoptosis to background levels (3-15%). Similarly, treatment with busulfan induced apoptosis in 55-80% of A20 cells, whereas addition of FDC reduced B cell death to

Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/immunology , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Dendritic Cells, Follicular/immunology , fas Receptor/immunology , Animals , Apoptosis/drug effects , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Cell Separation , Female , Mice , Mice, Inbred BALB C , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/metabolism , Stem Cells/immunology , Stem Cells/pathology , Tumor Cells, Cultured , bcl-2-Associated X Protein , bcl-X Protein
5.
Biochim Biophys Acta ; 1452(1): 1-11, 1999 Oct 13.
Article in English | MEDLINE | ID: mdl-10525155

ABSTRACT

Over two dozen alternative splice variants of CaMK-II, the type II Ca(2+)/CaM-dependent protein kinase, are encoded from four genes (alpha, beta, gamma and delta) in mammalian cells. Isozymes of alpha and beta CaMK-II are well characterized in brain; however, an understanding of the relative endogenous levels of CaMK-II isozymes in a wide variety of non-neuronal cells has not yet been described. In this study, we have demonstrated that CaMK-II consists primarily of the 54 kDa delta CaMK-II (delta(2) or delta(C)) isozyme in rodent fibroblasts. beta and gamma CaMK-II isozymes are minor and alpha CaMK-II was not expressed. The primary delta CaMK-II in human fibroblasts and the MCF10A mammary epithelial cell line was the 52 kDa delta(4) CaMK-II, an isozyme identical to delta(2) except for a missing 21-amino-acid C-terminal tail. delta CaMK-II levels were diminished in both human and rodent fibroblasts after SV40 transformation and in the mammary adenocarcinoma MCF7 cell line when compared to MCF10A cells. In fact, most tumor cells exhibited CaMK-II specific activities which were two- to tenfold lower than in untransformed fibroblasts. We conducted complementary CaMK-II studies on the NGF-induced differentiation of rat PC-12 cells. Although no new synthesis of CaMK-II occurs, neurite outgrowth in these cells is accompanied by a preferential activation of delta CaMK-II. Endogenous delta CaMK-II has a perinuclear distribution in fibroblasts and extends along neurites in PC-12 cells. These findings point to a role for delta CaMK-II isozymes in cellular differentiation.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , 3T3 Cells , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/biosynthesis , Cell Differentiation , Cell Line, Transformed , Down-Regulation , Humans , Immunoblotting , Isoenzymes/metabolism , Mice , PC12 Cells , Rats , Tumor Cells, Cultured
6.
J Pharmacol Exp Ther ; 290(3): 1384-92, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10454518

ABSTRACT

We recently demonstrated that physiological induction of apoptosis by cytotoxic sphingolipid messengers proceeds via activating protein-1 (AP1)-dependent and AP1-independent mechanisms in U937 human monoblastic leukemia cells. Here we examine involvement of the stress-activated protein kinase (SAPK) cascade and AP1 in the initiation of apoptosis in U937 cells by podophyllotoxin-derived inhibitors of topoisomerase II. Induction of apoptotic cell death and DNA damage by treatment of U937 cells with etoposide (100 microM) was associated with phosphorylation and activation of the c-Jun NH(2)-terminal kinase (JNK1) SAPK enzymes p46 and p54-JNK2 and transient increases in expression of the transcription factor c-Jun, a primary JNK substrate. These responses were accompanied by a modest, but sustained, recruitment of the mitogen-activated protein kinases p42-extracellular signal receptor-activated kinase (ERK)1 and p44-extracellular signal receptor-activated kinase 2. The capacity of etoposide to promote double-stranded DNA degradation and cell death was unaffected by manipulations that interfere with SAPK signaling outflow through c-Jun/AP1, including: 1) pharmacological inhibition of AP1 activity by diferuloylmethane and 2) molecular ablation of normal c-Jun function by the Jun dominant-negative mutant TAM-67. Cytotoxicity of the structurally related compound teniposide was similarly unaffected. In parallel trials, the lethal actions of ceramide (but not of sphingosine) were markedly diminished by pretreatment with diferuloylmethane or expression of TAM-67, confirming the effectiveness of these interventions in suppression of SAPK/AP1-dependent apoptosis. The involvement of AP1 in the proapoptotic actions of other inhibitors of topoisomerase II activity was also evaluated. Induction of cell death by the anthracyclines daunorubicin, daunorubicin, and idarubicin was found to be insensitive to pretreatment with diferuloylmethane or expression of TAM-67. Collectively, the present data indicate that induction of apoptosis by etoposide and related inhibitors of topoisomerase II is mediated through a cell death pathway that does not require SAPK-dependent recruitment of AP1. These findings additionally suggest that activation of the SAPK represents a consequence, rather than an underlying cause, of etoposide-induced apoptosis in myeloid leukemia cells.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Etoposide/pharmacology , Protein Synthesis Inhibitors/pharmacology , Proto-Oncogene Proteins c-jun/physiology , Transcription Factor AP-1/physiology , Antibiotics, Antineoplastic/pharmacology , Curcumin/pharmacology , Humans , Leukemia, Monocytic, Acute/drug therapy , Leukemia, Monocytic, Acute/pathology , Proto-Oncogene Proteins c-jun/metabolism , Transcription Factor AP-1/metabolism , Tumor Cells, Cultured
7.
Invest New Drugs ; 17(3): 227-40, 1999.
Article in English | MEDLINE | ID: mdl-10665476

ABSTRACT

Neoplastic cell survival is governed by a balance between pro-apoptotic and anti-apoptotic signals. Noteworthy among several anti-apoptotic signaling elements is the protein kinase C (PKC) isoenzyme family, which mediates a central cytoprotective effect in the regulation of cell survival. Activation of PKC, and subsequent recruitment of numerous downstream elements such as the mitogen-activated protein kinase (MAPK) cascade, opposes initiation of the apoptotic cell death program by diverse cytotoxic stimuli. The understanding that the lethal actions of numerous antineoplastic agents are, in many instances, antagonized by cytoprotective signaling systems has been an important stimulus for the development of novel antineoplastic strategies. In this regard, inhibition of PKC, which has been shown to initiate apoptosis in a variety of malignant cell types, has recently been the focus of intense interest. Furthermore, there is accumulating evidence that selective targeting of PKC may prove useful in improving the therapeutic efficacy of established antineoplastic agents. Such chemosensitizing strategies can involve either (a) direct inhibition of PKC (e.g., following acute treatment with relatively specific inhibitors such as the synthetic sphingoid base analog safingol, or the novel staurosporine derivatives UCN-01 and CGP-41251) or (b) down-regulation (e.g., following chronic treatment with the non-tumor-promoting PKC activator bryostatin 1). In preclinical model systems, suppression of the cytoprotective function(s) of PKC potentiates the activity of cytotoxic agents (e.g., cytarabine) as well as ionizing radiation, and efforts to translate these findings into the clinical arena in humans are currently underway. Although the PKC-driven cytoprotective signaling systems affected by these treatments have not been definitively characterized, interference with PKC activity has been associated with loss of the mitogen-activated protein kinase (MAPK) response. Accordingly, recent pre-clinical studies have demonstrated that pharmacological disruption of the primary MEK-ERK module can mimic the chemopotentiating and radiopotentiating actions of PKC inhibition and/or down-regulation.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Protein Kinase C/antagonists & inhibitors , Animals , Bryostatins , Cell Survival , Cytoprotection , Humans , Lactones/pharmacology , Macrolides , Mitogen-Activated Protein Kinase Kinases/physiology , Naphthalenes/pharmacology , Protein Kinase C/physiology , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Staurosporine/pharmacology
8.
Leukemia ; 12(12): 1843-50, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9844914

ABSTRACT

During the last 10 years, multiple signal transduction pathways within cells have been discovered. These pathways have been linked to the regulation of many diverse cellular events such as proliferation, senescence, differentiation and apoptosis. This review will focus upon the many roles of signaling by the p42/p44 mitogen-activated protein (MAP) kinase pathway. Recent evidence suggests that signaling by the MAP kinase pathway can both enhance proliferation by increased expression of molecules such as cyclin D1, but also cause growth arrest by increased expression of molecules such as the cyclin kinase inhibitor protein p21(Cip-1/MDA6/WAF1). These differential effects on growth have been correlated to the amplitude and duration of the MAP kinase activity signal. Furthermore several laboratories are reporting data suggesting that inhibition of the MAP kinase pathway, as well as a family of upstream MAP kinase activators, the protein kinase C family, represent an important route to both radio- and chemo-sensitization of tumor cells. Herein, we describe the historical discovery and characterization of the MAP kinase pathway. In addition we describe potential mechanisms by which inhibition of protein kinase C, the MAP kinase pathway, and potentially of p21(Cip-1/MDA6/WAF1) expression, may alter the sensitivities of leukemic and carcinoma cells to cytotoxic insults, leading to increased apoptosis and loss of clonogenicity.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/physiology , Cyclins/physiology , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinases , Signal Transduction/physiology , Cell Cycle/physiology , Cell Division/physiology , Cell Survival/physiology , Cell Survival/radiation effects , Cyclin-Dependent Kinase Inhibitor p21 , Enzyme Activation , Leukemia/drug therapy , Leukemia/enzymology , Leukemia/pathology , Leukemia/radiotherapy , Mitogen-Activated Protein Kinase 3 , Protein Kinase C/physiology , Transcription Factors/metabolism , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/radiation effects
9.
Curr Opin Oncol ; 10(6): 552-9, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9818235

ABSTRACT

The sphingolipid messenger ceramide has been implicated in the initiation of apoptotic cell death in a variety of physiologic settings. Recent investigation has shown that ceramide-dependent stress signaling is associated with chemotherapy-related apoptosis. It is not entirely clear, however, whether drug-mediated generation of ceramide is essential for execution of the cell death program, or simply represents a component of the genotoxic stress response. For example, there is evidence that ceramide subserves an important role in certain stresses (e.g., ionizing radiation, daunorubicin) but represents a secondary process in others (e.g., cytarabine). The review presents evidence for and against a cytotoxic effector function for ceramide in the lethal actions of conventional antineoplastic agents.


Subject(s)
Apoptosis/physiology , Ceramides/physiology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Ceramides/pharmacology , Humans , Receptors, Tumor Necrosis Factor/drug effects , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction , fas Receptor/drug effects , fas Receptor/metabolism
10.
Mol Pharmacol ; 54(5): 844-56, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9804619

ABSTRACT

The stress-activated protein kinase (SAPK) and mitogen-activated protein kinase (MAPK) cascades mediate cytotoxic and cytoprotective functions, respectively, in the regulation of leukemic cell survival. Involvement of these signaling systems in the cytotoxicity of 1-beta-D-arabinofuranosylcytosine (ara-C) and modulation of ara-C lethality by protein kinase C PKC inhibition/down-regulation was examined in HL-60 promyelocytic leukemia cells. Exposure to ara-C (10 microM) for 6 hr promoted extensive apoptotic DNA damage and cell death, as well as activation of PKC. This response was accompanied by downstream activation of the SAPK and MAPK cascades. PKC-dependent MAPK activity seemed to limit ara-C action in that the toxicity of ara-C was enhanced by pharmacological reductions of PKC, MAPK, or both. Thus, ara-C action was (1) partially attenuated by diradylglycerols, which stimulated PKC and MAPK, but (2) dramatically amplified by sphingoid bases, which inhibited PKC and MAPK. The cytotoxicity of ara-C also was substantially increased by pharmacological reductions of PKC, including down-regulation of PKC by chronic preexposure to the macrocyclic lactone bryostatin 1 or inhibition of PKC by acute coexposure to the dihydrosphingosine analog safingol. Significantly, both of these manipulations prevented activation of MAPK by ara-C. Moreover, acute disruption of the MAPK module by AMF, a selective inhibitor of MEK1, suppressed both basal and drug-stimulated MAPK activity and sharply increased the cytotoxicity of ara-C, suggesting the direct involvement of MAPK as a downstream antiapoptotic effector for PKC. None of these chemopotentiating agents enhanced ara-CTP formation. Ceramide-driven SAPK activity did not seem to mediate drug-induced apoptosis, given that (1) neutralization of endogenous tumor necrosis factor-alpha with monoclonal antibodies or soluble tumor necrosis factor receptor substantially reduced ceramide generation and SAPK activation by ara-C, whereas the induction of apoptosis was unaffected; (2) pharmacological inhibition of sphingomyelinase by 3-O-methoxysphingomyelin reduced ceramide generation and SAPK activation without limiting the drug's cytotoxicity; and (3) potentiation of ara-C action by bryostatin 1 or safingol was not associated with further stimulation of SAPK. These observations collectively suggest a primary role for decreased MAPK, rather than increased SAPK, in the potentiation of ara-C cytotoxicity by interference with PKC-dependent signaling.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Cytarabine/pharmacology , Protein Kinase Inhibitors , Protein Kinases/physiology , Signal Transduction/drug effects , Antimetabolites, Antineoplastic/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/physiology , Bryostatins , Calcium-Calmodulin-Dependent Protein Kinases/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cytarabine/metabolism , Diglycerides/pharmacology , Down-Regulation/drug effects , Enzyme Activation , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , HL-60 Cells/drug effects , HL-60 Cells/enzymology , HL-60 Cells/pathology , Humans , Lactones/pharmacology , Macrolides , Protein Kinases/metabolism , Signal Transduction/physiology , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Stereoisomerism
11.
Int J Oncol ; 12(4): 927-34, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9499457

ABSTRACT

The goal of the present study was to determine whether partial restoration of the differentiation-inducing capacity of the PKC activator bryostatin 1 by the calcium ionophore A23187 is accompanied by enhancement of apoptosis in ara-C-pretreated human leukemia cells. When HL-60 cells were exposed to ara-C (10 or 100 microM;6 h) followed by bryostatin 1 alone (10 nM; 24 h), no increase in apoptosis was noted. In contrast, subsequent exposure of ara-C-pretreated cells to A23187 (250 nM; 24 h) increased apoptosis by approximately 100%. When ara-C-pretreated cells were incubated with A23187 and bryostatin 1, no further potentiation of cell death (compared to cells exposed to A23187 alone) was observed. Nevertheless, the combination of bryostatin 1 and A23187 substantially increased inhibition of clonogenicity in cells preincubated with ara-C (e.g., by > or = 2 logs). This effect was associated with morphological and functional evidence (i.e., plastic adherence) of enhanced leukemic cell maturation. The differentiating capacity of the combination of bryostatin 1 and A23187 was significantly weaker than that of the phorbol diester, PMA (10 nM), and unaccompanied (at 24 h) by induction of the cyclin-dependent kinase inhibitors (CDKIs) p21WAF1/CIP1 and p27KIP1. However, the extent of apoptosis was comparable in cells exposed to ara-C followed by PMA or bryostatin 1 + A23187, suggesting that differentiation per se is not solely responsible for enhancement of cell death in ara-C-pretreated cells. Coadministration of bryostatin 1 and the organotellurium compound AS101, which mimics the actions of A23187 in some systems, after ara-C also led to enhanced antiproliferative effects which were unaccompanied by an increase in apoptosis. Finally, exposure of cells to ara-C followed by other differentiation-inducing agents, including dimethylsulfoxide and sodium butyrate also resulted in increases in cell death in this cell line. These findings indicate that the inability of bryostatin 1 to potentiate apoptosis in ara-C-pretreated HL-60 cells may involve factors other than an inadequate differentiation stimulus. They also suggest that loss of leukemic self-renewal capacity following exposure to cytotoxic and differentiation-inducing agents may involve mechanisms other than, or in addition to, potentiation of apoptosis, particularly cellular maturation.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Calcimycin/pharmacology , Cytarabine/pharmacology , Lactones/pharmacology , Acetamides/pharmacology , Bryostatins , Calcium/metabolism , Cell Differentiation/drug effects , DNA Fragmentation , HL-60 Cells , Humans , Macrolides , Tetradecanoylphorbol Acetate/pharmacology
12.
J Lipid Res ; 39(1): 1-16, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9469581

ABSTRACT

The metazoan nervous system gives rise intradevelopmentally to many more neurons than ultimately survive in the adult. Such excess cells are eliminated through programmed cell death or apoptosis. As is true for cells of other lineages, neuronal survival is sustained by an array of growth factors, such that withdrawal of neurotrophic support results in apoptotic cell death. Apoptosis is therefore believed to represent a beneficial process essential to normal development of central and peripheral nervous system (CNS and PNS) structures. Although the initiation of neuronal apoptosis in response to numerous extracellular agents has been widely reported, the regulatory mechanisms underlying this mode of cell death remain incompletely understood. In recent years, the contribution of lipid-dependent signaling systems, such as the sphingomyelin pathway, to regulation of cell survival has received considerable attention, leading to the identification of lethal functions for the lipid effectors ceramide and sphingosine in both normal and pathophysiological conditions. Moreover, the apoptotic capacities of several cytotoxic receptor systems (e.g., CD120a, CD95) and many environmental stresses (e.g., ionizing radiation, heat-shock, oxidative stress) are now known to derive from the activation of multiple signaling cascades by ceramide or, under some circumstances, by sphingosine. Inappropriate initiation of apoptosis has been proposed to underlie the progressive neuronal attrition associated with various neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and other neurological disorders that are characterized by the gradual loss of specific populations of neurons. In such pathophysiological states, neuronal cell death can result in specific disorders of movement and diverse impairments of CNS and PNS function. In some autoimmune neurological diseases such as Guillain-Barré syndrome, demyelinating polyneuropathy, and motoneuron disease, persistent immunological attack of microvascular endothelial cells by glycolipid-directed autoantibodies may lead to extensive cellular damages, resulting in increased permeability across brain-nerve barrier (BNB) and/or blood-brain barrier (BBB).


Subject(s)
Apoptosis , Neurodegenerative Diseases/pathology , Sphingolipids/physiology , Animals , Gangliosides/physiology , Glycolipids/physiology , Humans , Lipids/physiology , Second Messenger Systems
13.
Mol Pharmacol ; 52(6): 1000-9, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9396780

ABSTRACT

The effects of the non-tumor-promoting protein kinase C (PKC) activator bryostatin 1 and the PKC inhibitors staurosporine and UCN-01 were examined with respect to modulation of 1-[beta-D-arabinofuranosyl]cytosine (ara-C)-induced apoptosis in human myeloid leukemia cells (HL-60) overexpressing the antiapoptotic protein Bcl-2. HL-60/Bcl-2 cells displayed a 5-fold increase in Bcl-2 protein compared with empty-vector counter-parts (HL-60/pCEP4) but comparable levels of Bax, Mcl-1, and Bcl-xL. After exposure to an equimolar concentration of ara-C (10 microM for 6 hr), HL-60/Bcl-2 cells were significantly less susceptible to apoptosis, DNA fragmentation, and loss of clonogenicity than HL-60/pCEP4 cells. The protective effect of increased Bcl-2 expression was manifested by a failure of ara-C to induce activation/cleavage of the Yama protease (CPP32; caspase-3) and degradation of one of its substrates, poly(ADP-ribose)polymerase to an 85-kDa cleavage product. When HL-60/Bcl-2 cells were preincubated with bryostatin 1 (10 nM; 24 hr) or coincubated with either staurosporine (50 nM; 6 hr) or UCN-01 (300 nM; 6 hr) after a 1-hr preincubation, exposures that exerted minimal effects alone, ara-C-induced apoptosis and DNA fragmentation were restored to levels equivalent to, or greater than, those observed in empty-vector controls. These events were accompanied by restoration of the ability of ara-C to induce CPP32 cleavage and activation, poly(ADP-ribose) polymerase degradation, and inhibition of colony formation. Western analysis of Bcl-2 protein obtained from overexpressing cells treated with bryostatin 1, staurosporine, or UCN-01 revealed the appearance of a slowly migrating species and a general broadening of the protein band, effects that were insensitive to the protein synthesis inhibitor cycloheximide. Alterations in Bcl-2 protein mobility on sodium dodecyl sulfate-polyacrylamide gel electrophoresis were reversed by treatment of lysates with alkaline phosphatase or protein phosphatase 2A; actions of the latter were blocked by the specific phosphatase inhibitor okadaic acid. In vivo labeling studies of Bcl-2 protein demonstrated increased incorporation of [32PO4]orthophosphate in drug-treated cells. Last, phosphorylated Bcl-2 failed to display decreased binding to the proapoptotic protein Bax. Collectively, these findings indicate that bryostatin 1, which down-regulates PKC, and staurosporine and UCN-01, which directly inhibit the enzyme, circumvent resistance of Bcl-2-overexpressing leukemic cells to ara-C-induced apoptosis and activation of the protease cascade. They also raise the possibility that modulation of Bcl-2 phosphorylation status contributes to this effect.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Cytarabine/pharmacology , HL-60 Cells/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Alkaloids/pharmacology , Antineoplastic Agents/pharmacology , Blotting, Western , Bryostatins , DNA, Neoplasm/drug effects , DNA, Neoplasm/metabolism , Down-Regulation/drug effects , Drug Resistance, Neoplasm , Drug Synergism , Endopeptidases/metabolism , Enzyme Inhibitors/pharmacology , HL-60 Cells/drug effects , Humans , Lactones/pharmacology , Macrolides , Phosphorylation , Protein Kinase C/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Staurosporine/pharmacology
14.
Biochem Pharmacol ; 54(5): 563-73, 1997 Sep 01.
Article in English | MEDLINE | ID: mdl-9337072

ABSTRACT

The role of the nuclear phosphoprotein c-Myc has been examined with respect to the regulation of 1-beta-D-arabinofuranosylcytosine (ara-C)-induced apoptosis in human leukemia cells exposed to bryostatin 1 and other pharmacologic protein kinase C (PKC) activators. Pretreatment of HL-60 cells for 24 hr with 10 nM bryostatin 1 significantly potentiated the ability of ara-C (10 microM; 6 hr) to induce apoptosis without reducing the expression of c-Myc protein. In contrast, equivalent exposure to the stage 2 tumor-promoting PKC activator mezerein (10 nM) in conjunction with ara-C reduced c-Myc levels by 87% and failed to potentiate apoptosis. Co-administration of bryostatin 1 with mezerein before ara-C prevented down-regulation of c-Myc and augmented cell death, whereas co-treatment with the calcium ionophore A23187 (250 nM) and bryostatin 1 reduced c-Myc levels by 80% and abrogated the increase in ara-C-induced apoptosis. When cells were exposed for 24 hr to a c-myc antisense oligonucleotide (AS-ODN;10 microM) but not to a scrambled sequence ODN (SS-ODN) prior to ara-C, c-Myc expression was reduced by 81%, and apoptosis and cell viability were unperturbed. However, AS-ODN (but not SS-ODN) reduced c-Myc protein in cells pre-exposed to bryostatin 1 by 74% and abrogated potentiation of ara-C-induced apoptosis. The actions of c-myc AS-ODN did not stem from proximal G1 arrest/differentiation or biochemical events, since they were not associated with a reduction in the S-phase cell fraction, p21(WAF1/CIP1) induction, pRb hypophosphorylation, or alterations in ara-C metabolism. Together, these findings indicate that HL-60 cell apoptosis proceeds by both c-Myc-dependent and -independent pathways, and that only the former are involved in the potentiation of ara-C-mediated cell death by bryostatin 1.


Subject(s)
Apoptosis/drug effects , Cell Cycle/drug effects , Cytarabine/pharmacology , Diterpenes , HL-60 Cells/drug effects , Lactones/pharmacology , Oligonucleotides, Antisense/pharmacology , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Antineoplastic Agents/pharmacology , Bryostatins , DNA Fragmentation , Drug Synergism , Enzyme Activation , Humans , Kinetics , Macrolides , Phorbol 12,13-Dibutyrate/pharmacology , Proto-Oncogene Proteins c-myc/genetics , Terpenes/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Thionucleotides
15.
Mol Cell Biol ; 17(7): 3556-65, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9199291

ABSTRACT

To study the mechanisms by which catecholamines regulate hepatocyte proliferation after partial hepatectomy (PHX), hepatocytes were isolated from adult male rats 24 h after sham operation or two-thirds PHX and treated with catecholamines and other agonists. In freshly isolated sham cells, p42 mitogen-activated protein (MAP) kinase activity was stimulated by the alpha1-adrenergic agonist phenylephrine (PHE). Activation of p42 MAP kinase by growth factors was blunted by pretreatment of sham hepatocytes with glucagon but not by that with the beta2-adrenergic agonist isoproterenol (ISO). In PHX cells, the ability of PHE to activate p42 MAP kinase was dramatically reduced, whereas ISO became competent to inhibit p42 MAP kinase activation. PHE treatment of sham but not PHX and ISO treatment of PHX but not sham hepatocytes also activated the stress-activated protein (SAP) kinases p46/54 SAP kinase and p38 SAP kinase. These data demonstrate that an alpha1- to beta2-adrenergic receptor switch occurs upon PHX and results in an increase in SAP kinase versus MAP kinase signaling by catecholamines. In primary cultures of hepatocytes, ISO treatment of PHX but not sham cells inhibited [3H]thymidine incorporation. In contrast, PHE treatment of sham but not PHX cells stimulated [3H]thymidine incorporation, which was reduced by approximately 25 and approximately 95% with specific inhibitors of p42 MAP kinase and p38 SAP kinase function, respectively. Inhibition of the p38 SAP kinase also dramatically reduced basal [3H]thymidine incorporation. These data suggest that p38 SAP kinase plays a permissive role in liver regeneration. Alterations in the abilities of catecholamines to modulate the activities of protein kinase A and the MAP and SAP kinase pathways may represent one physiological mechanism by which these agonists can regulate hepatocyte proliferation after PHX.


Subject(s)
Adrenergic Agonists/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Liver Regeneration , Liver/physiology , Mitogen-Activated Protein Kinases , Receptors, Adrenergic, alpha-1/physiology , Receptors, Adrenergic, beta-2/physiology , Stress, Physiological/physiopathology , Animals , Cell Division , Cyclic AMP-Dependent Protein Kinases/physiology , JNK Mitogen-Activated Protein Kinases , Liver/cytology , Male , Phosphorylases/metabolism , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-raf , Rats , Rats, Sprague-Dawley , Time Factors , p38 Mitogen-Activated Protein Kinases
16.
FEBS Lett ; 412(1): 9-14, 1997 Jul 21.
Article in English | MEDLINE | ID: mdl-9257680

ABSTRACT

The role of protein kinase C (PKC) and p42(MAP kinase) signaling in the regulation of proliferation and apoptosis was investigated in freshly isolated and primary cultured rat hepatocytes. Acute treatment of freshly isolated hepatocytes with phenylephrine and EGF caused rapid phasic activations of p42(MAP kinase) and JNK1. Acute pre-treatment of hepatocytes with the PKC inhibitors sphingosine, chelerythrine and bis-indolylmaleimide abolished the ability of phenylephrine, but not EGF, to activate p42(MAP kinase) and JNK1. Acute pretreatments with all of the PKC inhibitors alone increased JNK1 basal activity approximately 2-fold. Acute treatments of primary cultures of hepatocytes with an inhibitor of MEK1 activation (PD98059) also caused inhibition of p42(MAP kinase) and a approximately 2-fold activation of JNK1. These data demonstrate that PKC can function as both a proximal activator and a distal inhibitor of signaling through the JNK1/SAP kinase pathway. Treatments (4 h) of primary cultured hepatocytes with sphingosine, chelerythrine, bis-indolylmaleimide and PD98059 did not induce apoptosis as judged by propidium iodide staining. Similar acute treatments of HepG2 cells rapidly induced cell death. These data demonstrate that acute inhibition of either PKC or p42(MAP kinase) function is sufficient to rapidly induce apoptosis in transformed, but not in non-transformed hepatocytes.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Liver/enzymology , Mitogen-Activated Protein Kinase Kinases , Mitogen-Activated Protein Kinases , Protein Kinase C/metabolism , Protein-Tyrosine Kinases/metabolism , Animals , Cells, Cultured , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Epidermal Growth Factor/pharmacology , JNK Mitogen-Activated Protein Kinases , Kinetics , MAP Kinase Kinase 1 , Male , Mitogen-Activated Protein Kinase 1 , Phenylephrine/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Rats , Rats, Sprague-Dawley
17.
Cell Death Differ ; 4(4): 294-303, 1997 May.
Article in English | MEDLINE | ID: mdl-16465244

ABSTRACT

We have previously reported that pretreatment of HL-60 human promyelocytic leukemia cells with the non-tumor-promoting protein kinase C (PKC) activator bryostatin 1 potentiates induction of apoptosis by the antimetabolite 1-[beta-D-arabinofuranosyl]cytosine (ara-C) (Biochem Pharmacol 47:839,1994). To determine whether this phenomenon results from altered expression of Bcl-2 or related proteins, Northern and Western analysis was employed to assess the effects of bryostatin 1 and other PKC activators on steady-state levels of Bcl-2, Bax, Bcl-x, and Mcl-1 mRNA and protein. Pretreatment of cells for 24 h with 10 nM bryostatin 1, or, to a lesser extent, the stage-1 tumor-promoter phorbol dibutyrate (PDB) significantly potentiated apoptosis induced by ara-C (100 microM; 6 h); in contrast, equivalent exposure to the stage-2 tumor promoter, mezerein (MZN), which, unlike bryostatin 1, is a potent inducer of differentiation in this cell line, failed to modify ara-C-related cell death. Neither bryostatin 1 nor PDB altered expression of bcl-2/Bcl-2 over this time frame. In contrast, MZN down-regulated bcl-2 mRNA levels, but this effect was not accompanied by altered expression of Bcl-2 protein. None of the PKC activators modified expression of Bax or Bcl-x(L) mRNA or protein; levels of Bcl-x(S) were undetectable in both treated and untreated cells. However, expression of Mcl-1 mRNA and protein increased modestly after treatment with either bryostatin 1 or PDB, and to a greater extent following exposure to MZN. Combined treatment of cells with bryostatin 1 and MZN resulted in undiminished potentiation of ara-C-mediated apoptosis and by antagonism of cellular maturation. These effects were accompanied by unaltered expression of Bcl-2, Bax, and Bcl-x(L), and by a further increase in Mcl-1 protein levels. When cells were co-incubated with bryostatin 1 and calcium ionophore (A23187), an identical pattern of expression of Bcl-2 family members was observed, despite the loss of bryostatin 1's capacity to potentiate apoptosis, and the restoration of its ability to induce differentiation. Finally, treatment of cells with bryostatin 1+/-ara-C (but not ara-C alone) resulted in a diffuse broadening of the Bcl-2 protein band, whereas exposure of cells to taxol (250 nM, 6 h) led to the appearance of a distinct Bcl-2 species with reduced mobility, phenomena compatible with protein phosphorylation. Together, these findings indicate that the ability of bryostatin 1 to facilitate drug-induced apoptosis in human myeloid leukemia cells involves factors other than quantitative changes in the expression of Bcl-2 family members, and raise the possibility that qualitative alterations in the Bcl-2 protein, such as phosphorylation status, may contribute to this capacity. They also suggest that increased expression of Mcl-1 occurs early in the pre-commitment stage of myeloid cell differentiation, and that this event does not protect cells from drug-induced apoptosis.

18.
Mol Pharmacol ; 52(6): 935-47, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9415703

ABSTRACT

We characterized participation of the stress-activated protein kinase (SAPK) cascade in the lethal actions of the cytotoxic lipid messengers ceramide and sphingosine in U937 human monoblastic leukemia cells. Acute exposure of U937 cells to either lipid resulted in loss of proliferative capacity, degradation of genomic DNA, and manifestation of apoptotic cytoarchitecture. Ceramide robustly stimulated p46-JNK1/p54-JNK2 activity and increased expression of c-jun mRNA and c-Jun protein; in contrast, sphingosine moderately stimulated p46-JNK1/p54-JNK2 and failed to modify c-jun/c-Jun expression. Dominant-negative blockade of normal c-Jun activity by transfection with the TAM-67 c-Jun NH2-terminal deletion mutant abolished the lethal actions of ceramide but was without effect on those of sphingosine, indicating that ceramide-related apoptosis is directly dependent on activation of c-Jun, whereas sphingosine-induced cell death proceeds via an unrelated downstream mechanism. Characterization of the mitogen-activated protein kinase (MAPK) cascade in these responses revealed a further functional disparity between the two lipids: basal p42-ERK1/ p44-ERK2 activity was gradually reduced by ceramide but immediately and completely suppressed by sphingosine. Moreover, blockade of the MAPK cascade by the aminomethoxyflavone MEK1 inhibitor PD-98059 unexpectedly activated p46-JNK1/p54-JNK2 and induced apoptosis in a manner qualitatively resembling that of sphingosine. Both lipids sharply increased p38-RK activity; selective pharmacological inhibition of p38-RK by the pyridinyl imidazole SB-203580 failed to mitigate the cytotoxicity associated with either ceramide or sphingosine, suggesting that p38-RK is not essential for lipid-induced apoptosis. These findings demonstrate that reciprocal alterations in the SAPK and MAPK cascades are associated with the apoptotic influence of either lipid inasmuch as (i) ceramide-mediated lethality is primarily associated with strong stimulation of SAPK and weak inhibition of MAPK, whereas (ii) sphingosine-mediated lethality is primarily associated with weak stimulation of SAPK and strong inhibition of MAPK. We therefore propose that leukemic cell survival depends on the maintenance of an imbalance of the outputs from the MAPK and SAPK systems such that the dominant basal influence of the MAPK cascade allows sustained proliferation, whereas acute redirection of this balance toward the SAPK cascade initiates apoptotic cell death.


Subject(s)
Apoptosis/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Ceramides/pharmacology , Mitogen-Activated Protein Kinases , Sphingosine/pharmacology , Apoptosis/physiology , Calcium-Calmodulin-Dependent Protein Kinases/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , DNA Damage , Enzyme Activation , Humans , JNK Mitogen-Activated Protein Kinases , Leukemia, Myeloid/enzymology , Leukemia, Myeloid/pathology , Signal Transduction/drug effects , Signal Transduction/physiology
20.
Endocrinology ; 137(10): 4419-26, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8828503

ABSTRACT

Interleukin-6 (IL-6) is a B-cell differentiation-inducing cytokine that affects the secretion of several neuroendocrine hormones. Normal rat anterior pituitary (AP) cells synthesize and release IL-6, suggesting a paracrine role for the stimulation of AP hormone release by this cytokine. We have previously reported that IL-1 beta enhances IL-6 release and phospholipase A2 (PLA2)-mediated hydrolysis of phosphatidylcholine (PC) in AP cells. Because lysophosphatidylcholine (LPC) may function as a second messenger for IL-1 beta, we have investigated the effects of exogenous LPC on IL-6 release from AP cells in vitro. AP cells from male Long-Evans rats were dispersed and cultured for 5-6 days in 96-well (100,000 cells/well) culture plates. Cells were rinsed and incubated in the absence or presence of 1.25-40 microM LPC 18:0 (stearoyl) for 6 h, and IL-6 concentrations determined using the 7-TD1 cell bioassay. LPC 18:0 significantly (P < 0.01) stimulated IL-6 release up to 10-fold in a concentration-related manner. In contrast, LPC 18:0 did not affect PRL release. LPC species substituted with progressively shorter saturated 1-acyl chains (16:0-10:0) were less effective for IL-6 induction. Examination of structurally related glycerophospholipid species revealed the specificity of the LPC stimulation of IL-6 release. Thus, 1.25-40 microM lysophosphatidylethanolamine (LPE; 18:0) and lysophosphatidic acid (LPA; 18:0) were without significant effect on AP IL-6 release, demonstrating the specific functional requirement for the phosphorylcholine headgroup. Hydrolysis of the structurally related choline-linked phospholipid sphingomyelin (SM) has been implicated in IL-1 beta action in certain cell types. Similarly, 1.25-20 microM lysosphingomyelin (sphingosylphosphorylcholine; SPC) also significantly (P < or = 0.01) stimulated IL-6 release from AP cells, although SPC exhibited discernibly lower potency and efficacy than LPC. An acyl analog of platelet-activation factor (PAF), i.e. 18:0-2:0 PC (1-stearoyl-2-acetoyl-sn-glycero-3-phosphorylcholine), differs from LPC by an acetyl group in the sn-2 position; PAF was at least as effective as LPC for the stimulation of IL-6 release from AP cells in vitro. Stimulation of IL-6 release by LPC 18:0 was completely suppressed by pharmacological inhibitors of protein kinase C such as H7 (20 microM) and chelerythrine (5 microM). In addition, H7 (20 microM) abolished the stimulation of IL-6 release by IL-1 beta (0.16-100 ng/mL). These findings demonstrate that LPC, acyl PAF, or SPC (but not other lysophospholipids) stimulate IL-6 release from AP cells in vitro. We conclude that LPC-mediated activation of protein kinase C is involved in the stimulatory actions of IL-1 beta in AP cells.


Subject(s)
Interleukin-6/metabolism , Lysophosphatidylcholines/pharmacology , Pituitary Gland, Anterior/metabolism , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Alkaloids , Animals , Benzophenanthridines , Cells, Cultured , Enzyme Inhibitors/pharmacology , Interleukin-1/pharmacology , Male , Phenanthridines/pharmacology , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/pharmacology , Pituitary Gland, Anterior/cytology , Pituitary Gland, Anterior/drug effects , Platelet Activating Factor/pharmacology , Prolactin/metabolism , Protein Kinase C/antagonists & inhibitors , Rats , Rats, Inbred Strains , Sphingosine/analogs & derivatives , Sphingosine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...