Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters











Publication year range
1.
J Biol Chem ; 298(7): 102034, 2022 07.
Article in English | MEDLINE | ID: mdl-35588784

ABSTRACT

Surface determinants newly expressed by apoptotic cells that are involved in triggering potent immunosuppressive responses, referred to as "innate apoptotic immunity (IAI)" have not been characterized fully. It is widely assumed, often implicitly, that phosphatidylserine, a phospholipid normally cloistered in the inner leaflet of cells and externalized specifically during apoptosis, is involved in triggering IAI, just as it plays an essential role in the phagocytic recognition of apoptotic cells. It is notable, however, that the triggering of IAI in responder cells is not dependent on the engulfment of apoptotic cells by those responders. Contact between the responder and the apoptotic target, on the other hand, is necessary to elicit IAI. Previously, we demonstrated that exposure of protease-sensitive determinants on the apoptotic cell surface are essential for initiating IAI responses; exposed glycolytic enzyme molecules were implicated in particular. Here, we report our analysis of the involvement of externalized phosphatidylserine in triggering IAI. To analyze the role of phosphatidylserine, we employed a panel of target cells that either externalized phosphatidylserine constitutively, independently of apoptosis, or did not, as well as their WT parental cells that externalized the phospholipid in an apoptosis-dependent manner. We found that the externalization of phosphatidylserine, which can be fully uncoupled from apoptosis, is neither sufficient nor necessary to trigger the profound immunomodulatory effects of IAI. These results reinforce the view that apoptotic immunomodulation and phagocytosis are dissociable and further underscore the significance of protein determinants localized to the cell surface during apoptosis in triggering innate apoptotic immunity.


Subject(s)
Apoptosis , Immunity, Innate , Phagocytosis , Phosphatidylserines , Animals , Apoptosis/physiology , Cell Line , Humans , Immunomodulation , Mice , Phagocytosis/physiology , Phosphatidylserines/metabolism
2.
J Immunol ; 207(2): 436-448, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34215655

ABSTRACT

Phosphatidylserine (PS)-targeting monoclonal Abs (mAbs) that directly target PS and target PS via ß2-gp1 (ß2GP1) have been in preclinical and clinical development for over 10 y for the treatment of infectious diseases and cancer. Although the intended targets of PS-binding mAbs have traditionally included pathogens as well as stressed tumor cells and its associated vasculature in oncology, the effects of PS-targeting mAbs on activated immune cells, notably T cells, which externalize PS upon Ag stimulation, is not well understood. Using human T cells from healthy donor PBMCs activated with an anti-CD3 + anti-CD28 Ab mixture (anti-CD3/CD28) as a model for TCR-mediated PS externalization and T cell stimulation, we investigated effects of two different PS-targeting mAbs, 11.31 and bavituximab (Bavi), on TCR activation and TCR-mediated cytokine production in an ex vivo paradigm. Although 11.31 and Bavi bind selectivity to anti-CD3/28 activated T cells in a PS-dependent manner, surprisingly, they display distinct functional activities in their effect on IFN-γ and TNF-ɑ production, whereby 11.31, but not Bavi, suppressed cytokine production. This inhibitory effect on anti-CD3/28 activated T cells was observed on both CD4+ and CD8+ cells and independently of monocytes, suggesting the effects of 11.31 were directly mediated by binding to externalized PS on activated T cells. Imaging showed 11.31 and Bavi bind at distinct focal depots on the cell membrane. Collectively, our findings indicate that PS-targeting mAb 11.31 suppresses cytokine production by anti-CD3/28 activated T cells.


Subject(s)
Antibodies, Monoclonal/immunology , CD28 Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Interferon-gamma/immunology , Muromonab-CD3/immunology , Phosphatidylserines/immunology , Tumor Necrosis Factor-alpha/immunology , CD3 Complex/immunology , Cell Line , HEK293 Cells , Humans , Leukocytes, Mononuclear/immunology , Lymphocyte Activation/immunology
3.
Article in English | MEDLINE | ID: mdl-31036289

ABSTRACT

Of the roughly one million cells per second dying throughout the body, the vast majority dies by apoptosis, the predominant form of regulated cell death in higher organisms. Long regarded as mere waste, apoptotic cells are now recognized as playing a prominent and active role in homeostatic maintenance, especially resolution of inflammation, and in the sculpting of tissues during development. The activities associated with apoptotic cells are continually expanding, with more recent studies demonstrating their ability to modulate such vital functions as proliferation, survival, differentiation, metabolism, migration, and angiogenesis. In each case, the role of apoptotic cells is active, exerting their effects via new activities acquired during the apoptotic program. Moreover, the capacity to recognize and respond to apoptotic cells is not limited to professional phagocytes. Most, if not all, cells receive and integrate an array of signals from cells dying in their vicinity. These signals comprise a form of biochemical communication. As reviewed in this chapter, this communication is remarkably sophisticated; each of its three critical steps-encoding, transmission, and decoding of the apoptotic cell's "message"-is endowed with exquisite robustness. Together, the abundance and intricacy of the variables at each step comprise the vocabulary and grammar of the language by which dead cells achieve their post-mortem voice. The combinatorial complexity of the resulting communication network permits dying cells, through the signals they emit and the responses those signals elicit, to partake of an expanded role in homeostasis, acting as both sentinels of environmental change and agents of adaptation.


Subject(s)
Apoptosis , Phagocytosis , Signal Transduction , Animals , Cell Communication , Homeostasis , Humans , Inflammation/metabolism
4.
Front Immunol ; 9: 241, 2018.
Article in English | MEDLINE | ID: mdl-29535707

ABSTRACT

Within an organism, environmental stresses can trigger cell death, particularly apoptotic cell death. Apoptotic cells, themselves, are potent regulators of their cellular environment, involved primarily in effecting homeostatic control. Tumors, especially, exist in a dynamic balance of cell proliferation and cell death. This special feature of the tumorous microenvironment-namely, the prominence and persistence of cell death-necessarily entails a magnification of the extrinsic, postmortem effects of dead cells. In both normal and malignant tissues, apoptotic regulation is exerted through immune as well as non-immune mechanisms. Apoptotic cells suppress the repertoire of immune reactivities, both by attenuating innate (especially inflammatory) responses and by abrogating adaptive responses. In addition, apoptotic cells modulate multiple vital cell activities, including survival, proliferation (cell number), and growth (cell size). While the microenvironment of the tumor may contribute to apoptosis, the postmortem effects of apoptotic cells feature prominently in the reciprocal acclimatization between the tumor and its environment. In much the same way that pathogens evade the host's defenses through exploitation of key aspects of innate and adaptive immunity, cancer cells subvert several normal homeostatic processes, in particular wound healing and organ regeneration, to transform and overtake their environment. In understanding this subversion, it is crucial to view a tumor not simply as a clone of malignant cells, but rather as a complex and highly organized structure in which there exists a multidirectional flow of information between the cancer cells themselves and the multiple other cell types and extracellular matrix components of which the tumor is comprised. Apoptotic cells, therefore, have the unfortunate consequence of facilitating tumorigenesis and tumor survival.


Subject(s)
Apoptosis/immunology , Cell Transformation, Neoplastic/immunology , Macrophages/immunology , Neoplasms/immunology , Tumor Microenvironment/immunology , Adaptive Immunity , Animals , Cell Proliferation , Cell Survival/immunology , Cell Transformation, Neoplastic/pathology , Disease Models, Animal , Humans , Immunity, Innate , Macrophages/pathology , Mice , Neoplasms/pathology
5.
Aging Cell ; 16(3): 585-593, 2017 06.
Article in English | MEDLINE | ID: mdl-28371013

ABSTRACT

Immunosenescence is a state of unbalanced immune responsiveness, characterized by a diverse repertoire of seemingly discreet and paradoxical alterations in all aspects of immunity arising in an aging-associated manner. We asked whether aging-associated alterations in the ability of apoptotic cells to elicit immunomodulatory responses (innate apoptotic immunity; IAI) or in IAI responses themselves might underlie the confounding aging-associated anomalies of immunosenescence. We explored this question by examining, as a function of animal age, responsiveness of murine macrophages on the single cell level. We monitored the expression of pro- and anti-inflammatory cytokines cytofluorimetrically in response to pro-inflammatory Toll-like receptor (TLR) stimulation and anti-inflammatory treatment with apoptotic cells. While we found no alterations with age in the potency of apoptotic cells or in the initiation and magnitude of IAI responses, we did identify a cell-intrinsic deficiency in anti-inflammatory IAI response termination linked with age and preceding manifestations of immunosenescence. Further, we found that an aging-associated deficiency in response termination also is evident following TLR stimulation. These surprising observations reveal that a loss of homeostatic immune control with animal age results from the dysregulation of response termination (as distinct from response initiation) and is exerted on the level of transcription. We suggest that, with advancing age, cells become locked into relatively longer-lived response states. Aging-associated immune dysfunctions may reflect a diminution in the cellular nimbleness of immune responsiveness.


Subject(s)
Immunity, Innate , Immunosenescence/genetics , Interleukin-10/immunology , Macrophages/drug effects , Transcription, Genetic/immunology , Tumor Necrosis Factor-alpha/immunology , Age Factors , Animals , Apoptosis/immunology , Gene Expression Regulation, Developmental , Homeostasis/genetics , Homeostasis/immunology , Humans , Interleukin-10/genetics , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Macrophages/cytology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Primary Cell Culture , Signal Transduction , Single-Cell Analysis , Tumor Necrosis Factor-alpha/genetics
6.
Microb Pathog ; 105: 218-225, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28242425

ABSTRACT

Infections by pathogenic microorganisms elicit host immune responses, which crucially limit those infections. Pathogens employ various strategies to evade host immunity. We have identified the exploitation of the repertoire of potent immunosuppressive responses elicited normally by apoptotic cells ("Innate Apoptotic Immunity"; IAI) as one of these strategies. In the case of Listeria monocytogenes, an environmentally ubiquitous, foodborne bacterial pathogen capable of causing life-threatening invasive disease in immunocompromised and elderly individuals, the induction of host cell apoptosis appears to play an important role in pathogenesis. Previous studies have documented extensive lymphocyte apoptosis resulting from L. monocytogenes infection and demonstrated paradoxically that lymphocyte-deficient animals exhibit diminished susceptibility to listerial pathogenicity. We speculated that the triggering of IAI following the induction of host cell apoptosis was responsible for enhanced pathogenesis, and that the administration of exogenous apoptotic cells would serve to exert this effect. Importantly, apoptotic cells, which are not susceptible to L. monocytogenes infection, do not provide a niche for bacterial replication. Our experiments confirm that apoptotic cells, including exogenous apoptotic cells induced to die independently of the pathogen, specifically enhance pathogenesis. The recognition of a role of apoptotic cells and Innate Apoptotic Immunity in microbial pathogenesis provides an intriguing and novel insight for therapeutic approaches for the control of pathogenic infections.


Subject(s)
Apoptosis/immunology , Listeria monocytogenes/pathogenicity , Listeriosis/immunology , Listeriosis/pathology , Animals , Cell Line , Cells, Cultured , Cytokines/immunology , Disease Models, Animal , Immunity, Innate , Inflammation/immunology , Inflammation/pathology , Listeria monocytogenes/immunology , Listeriosis/microbiology , Lymphocytes/immunology , Mice, Inbred C57BL , Signal Transduction
7.
Mech Ageing Dev ; 157: 44-59, 2016 07.
Article in English | MEDLINE | ID: mdl-27453067

ABSTRACT

Aging is associated with a waning of normal immune function. This "immunosenescence" is characterized by a diverse repertoire of seemingly discreet and unbalanced immune alterations. A number of studies have suggested that aging-associated alterations in innate immune responsiveness, especially responsiveness dependent on Toll-like Receptor (TLR) engagement, are causally involved. We find, however, that the magnitude and dose-dependency of responsiveness to TLR engagement (assessed with respect to cytokine production) in distinct populations of murine macrophages are not altered generally with animal age or as a consequence of immunosenescence. Responses elicited with a wide array of TLR agonists were examined by extensive functional analyses, principally on the level of the individual cell. These studies reveal an intriguing "all-or-nothing" response behavior of macrophages, independent of animal age. Although reports to the contrary have been cited widely, aging-associated immune decline cannot be attributed to widespread alterations in the extents of TLR-dependent innate immune macrophage responses.


Subject(s)
Aging/immunology , Cytokines/immunology , Immunity, Innate/physiology , Macrophages/immunology , Toll-Like Receptors/immunology , Animals , Mice , Mice, Inbred BALB C , Toll-Like Receptors/agonists
8.
Mol Biol Cell ; 25(11): 1704-14, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24694594

ABSTRACT

Apoptotic recognition is innate and linked to a profound immune regulation (innate apoptotic immunity [IAI]) involving anti-inflammatory and immunosuppressive responses. Many of the molecular and mechanistic details of this response remain elusive. Although immune outcomes can be quantified readily, the initial specific recognition events have been difficult to assess. We developed a sensitive, real-time method to detect the recognition of apoptotic cells by viable adherent responder cells, using a photonic crystal biosensor approach. The method relies on characteristic spectral shifts resulting from the specific recognition and dose-dependent interaction of adherent responder cells with nonadherent apoptotic targets. Of note, the biosensor provides a readout of early recognition-specific events in responder cells that occur distal to the biosensor surface. We find that innate apoptotic cell recognition occurs in a strikingly species-independent manner, consistent with our previous work and inferences drawn from indirect assays. Our studies indicate obligate cytoskeletal involvement, although apoptotic cell phagocytosis is not involved. Because it is a direct, objective, and quantitative readout of recognition exclusively, this biosensor approach affords a methodology with which to dissect the early recognition events associated with IAI and immunosuppression.


Subject(s)
Apoptosis , Biosensing Techniques/methods , Photons , Animals , Cell Adhesion , Cell Communication , Cell Survival , Crystallization , Cytoskeleton/metabolism , Humans , Jurkat Cells , Mice , Phagocytosis , Species Specificity
9.
J Virol ; 88(5): 2658-69, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24352454

ABSTRACT

UNLABELLED: Cells that undergo apoptosis in response to chemical or physical stimuli repress inflammatory reactions, but cells that undergo nonapoptotic death in response to such stimuli lack this activity. Whether cells dying from viral infection exhibit a cell death-type modulatory effect on inflammatory reactions is unknown. We compared the effects on macrophage inflammatory responses of cells dying an apoptotic or a nonapoptotic death as a result of adenoviral infection. The results were exactly opposite to the predictions from the conventional paradigm. Cells dying by apoptosis induced by infection with an adenovirus type 5 (Ad5) E1B 19-kilodalton (E1B 19K) gene deletion mutant did not repress macrophage NF-κB activation or cytokine responses to proinflammatory stimuli, whereas cells dying a nonapoptotic death from infection with E1B 19K-competent, wild-type Ad5 repressed these macrophage inflammatory responses as well as cells undergoing classical apoptosis in response to chemical injury. The immunorepressive, E1B 19K-related cell death activity depended upon direct contact of the virally infected corpses with responder macrophages. Replacement of the viral E1B 19K gene with the mammalian Bcl-2 gene in cis restored the nonapoptotic, immunorepressive cell death activity of virally infected cells. These results define a novel function of the antiapoptotic, adenoviral E1B 19K protein that may limit local host innate immune inflammation during accumulation of virally infected cells at sites of infection and suggest that E1B 19K-deleted, replicating adenoviral vectors might induce greater inflammatory responses to virally infected cells than E1B 19K-positive vectors, because of the net effect of their loss-of-function mutation. IMPORTANCE: We observed that cells dying a nonapoptotic cell death induced by adenovirus infection repressed macrophage proinflammatory responses while cells dying by apoptosis induced by infection with an E1B 19K deletion mutant virus did not repress macrophage proinflammatory responses and enhanced some cytokine responses. Our results define a new function of the antiapoptotic, adenoviral protein E1B 19K, which we have termed "apoptotic mimicry." Our studies suggest the possibility that the presence or absence of this E1B 19K function could alter the immunological outcome of both natural and therapeutic adenoviral infections. For example, emerging, highly immunopathogenic adenovirus serotypes might induce increased host inflammatory responses as a result of altered E1B 19K function or expression. It is also possible that engineered variations in E1B 19K expression/function could be created during adenovirus vector design that would increase the therapeutic efficacy of replicating adenovirus vectors for vaccines or oncolytic viral targeting of neoplastic cells.


Subject(s)
Adenoviridae Infections/immunology , Adenoviridae/immunology , Adenovirus E1B Proteins/immunology , Apoptosis/immunology , Immunity, Innate , Adenoviridae/genetics , Adenoviridae Infections/genetics , Adenoviridae Infections/metabolism , Adenovirus E1B Proteins/genetics , Adenovirus E1B Proteins/metabolism , Animals , Cell Communication/immunology , Cell Death/immunology , Cell Line , Cytopathogenic Effect, Viral , Defective Viruses/genetics , Defective Viruses/immunology , Enzyme Activation , Gene Expression , Humans , Inflammation Mediators/metabolism , Lung/immunology , Lung/virology , Macrophages/immunology , Macrophages/metabolism , Mice , NF-kappa B/metabolism , Phenotype , Proto-Oncogene Proteins c-bcl-2/metabolism , Sequence Deletion , Transcriptional Activation
10.
J Biol Chem ; 287(17): 13761-77, 2012 Apr 20.
Article in English | MEDLINE | ID: mdl-22396534

ABSTRACT

Apoptosis allows for the removal of damaged, aged, and/or excess cells without harm to surrounding tissue. To accomplish this, cells undergoing apoptosis acquire new activities that enable them to modulate the fate and function of nearby cells. We have shown that receptor-mediated recognition of apoptotic versus necrotic target cells by viable kidney proximal tubular epithelial cells (PTEC) modulates the activity of several signaling pathways critically involved in regulation of proliferation and survival. Generally, apoptotic and necrotic targets have opposite effects with apoptotic targets inhibiting and necrotic targets stimulating the activity of these pathways. Here we explore the consequences of these signaling differences. We show that recognition of apoptotic targets induces a profound decrease in PTEC viability through increased responder cell death and decreased proliferation. In contrast, necrotic targets promote viability through decreased death and increased proliferation. Both target types mediate their effects through a network of Akt-dependent and -independent events. Apoptotic targets modulate Akt-dependent viability in part through a reduction in cellular ß-catenin and decreased inactivation of Bad. In contrast, Akt-independent modulation of viability occurs through activation of caspase-8, suggesting that death receptor-dependent pathways are involved. Apoptotic targets also activate p38, which partially protects responders from target-induced death. The response of epithelial cells varies depending on their tissue origin. Some cell lines, like PTEC, demonstrate decreased viability, whereas others (e.g. breast-derived) show increased viability. By acting as sentinels of environmental change, apoptotic targets allow neighboring cells, especially non-migratory epithelial cells, to monitor and potentially adapt to local stresses.


Subject(s)
Apoptosis , Epithelial Cells/immunology , Gene Expression Regulation , Animals , CHO Cells , Cell Proliferation , Cell Survival , Cricetinae , Epithelial Cells/cytology , Epithelial Cells/metabolism , HeLa Cells , Homeostasis , Humans , Immune System , Kidney/metabolism , Necrosis , Phagocytes/cytology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , beta Catenin/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
11.
J Biol Chem ; 287(13): 10325-10343, 2012 Mar 23.
Article in English | MEDLINE | ID: mdl-22262862

ABSTRACT

The intriguing cell biology of apoptotic cell death results in the externalization of numerous autoantigens on the apoptotic cell surface, including protein determinants for specific recognition, linked to immune responses. Apoptotic cells are recognized by phagocytes and trigger an active immunosuppressive response ("innate apoptotic immunity" (IAI)) even in the absence of engulfment. IAI is responsible for the lack of inflammation associated normally with the clearance of apoptotic cells; its failure also has been linked to inflammatory and autoimmune pathology, including systemic lupus erythematosus and rheumatic diseases. Apoptotic recognition determinants underlying IAI have yet to be identified definitively; we argue that these molecules are surface-exposed (during apoptotic cell death), ubiquitously expressed, protease-sensitive, evolutionarily conserved, and resident normally in viable cells (SUPER). Using independent and unbiased quantitative proteomic approaches to characterize apoptotic cell surface proteins and identify candidate SUPER determinants, we made the surprising discovery that components of the glycolytic pathway are enriched on the apoptotic cell surface. Our data demonstrate that glycolytic enzyme externalization is a common and early aspect of cell death in different cell types triggered to die with distinct suicidal stimuli. Exposed glycolytic enzyme molecules meet the criteria for IAI-associated SUPER determinants. In addition, our characterization of the apoptosis-specific externalization of glycolytic enzyme molecules may provide insight into the significance of previously reported cases of plasminogen binding to α-enolase on mammalian cells, as well as mechanisms by which commensal bacteria and pathogens maintain immune privilege.


Subject(s)
Apoptosis , Glycolysis , Immunity, Innate , Animals , Biomarkers/metabolism , Humans , Jurkat Cells , Lupus Erythematosus, Systemic/enzymology , Lupus Erythematosus, Systemic/pathology , Mice , Proteomics , U937 Cells
12.
J Immunol ; 186(12): 7144-55, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21593381

ABSTRACT

Efficient execution of apoptotic cell death followed by efficient clearance mediated by professional macrophages is a key mechanism in maintaining tissue homeostasis. Removal of apoptotic cells usually involves three central elements: 1) attraction of phagocytes via soluble "find me" signals, 2) recognition and phagocytosis via cell surface-presenting "eat me" signals, and 3) suppression or initiation of inflammatory responses depending on additional innate immune stimuli. Suppression of inflammation involves both direct inhibition of proinflammatory cytokine production and release of anti-inflammatory factors, which all contribute to the resolution of inflammation. In the current study, using wild-type and adenosine A(2A) receptor (A2AR) null mice, we investigated whether A2ARs, known to mediate anti-inflammatory signals in macrophages, participate in the apoptotic cell-mediated immunosuppression. We found that macrophages engulfing apoptotic cells release adenosine in sufficient amount to trigger A2ARs, and simultaneously increase the expression of A2ARs, as a result of possible activation of liver X receptor and peroxisome proliferators activated receptor δ. In macrophages engulfing apoptotic cells, stimulation of A2ARs suppresses the NO-dependent formation of neutrophil migration factors, such as macrophage inflammatory protein-2, using the adenylate cyclase/protein kinase A pathway. As a result, loss of A2ARs results in elevated chemoattractant secretion. This was evident as pronounced neutrophil migration upon exposure of macrophages to apoptotic cells in an in vivo peritonitis model. Altogether, our data indicate that adenosine is one of the soluble mediators released by macrophages that mediate engulfment-dependent apoptotic cell suppression of inflammation.


Subject(s)
Adenosine/immunology , Apoptosis/immunology , Inflammation/immunology , Phagocytosis/immunology , Receptor, Adenosine A2A/immunology , Animals , Inflammation/pathology , Macrophages/immunology , Mice , Mice, Knockout , Peritonitis/immunology
13.
J Biol Chem ; 285(3): 1829-40, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19910463

ABSTRACT

During apoptosis, cells acquire new activities that enable them to modulate the fate and function of interacting phagocytes, particularly macrophages (m). Although the best known of these activities is anti-inflammatory, apoptotic targets also influence m survival and proliferation by modulating proximal signaling events, such as MAPK modules and Akt. We asked whether modulation of these same signaling events extends to epithelial cells, a minimally phagocytic cell type. We used BU.MPT cells, a mouse kidney epithelial cell line, as our primary model, but we also evaluated several epithelial cell lines of distinct tissue origins. Like m, mouse kidney epithelial cells recognized apoptotic and necrotic targets through distinct non-competing receptors, albeit with lower binding capacity and markedly reduced phagocytosis. Also, modulation of inflammatory activity and MAPK-dependent signaling by apoptotic and necrotic targets was indistinguishable in kidney epithelial cells and m. In contrast, modulation of Akt-dependent signaling differed dramatically between kidney epithelial cells and m. In kidney epithelial cells, modulation of Akt was linked to target cell recognition, independently of phagocytosis, whereas in m, modulation was linked to phagocytosis. Moreover, recognition of apoptotic and necrotic targets by kidney epithelial cells elicited opposite responses; apoptotic targets inhibited whereas necrotic targets stimulated Akt activity. These data confirm that nonprofessional phagocytes recognize and respond to dying cells, albeit in a manner partially distinct from m. By acting as sentinels of environmental change, apoptotic and necrotic targets may permit neighboring viable cells, especially non-migratory epithelial cells, to monitor and adapt to local stresses.


Subject(s)
Apoptosis , Epithelial Cells/cytology , Signal Transduction , Adaptation, Physiological , Animals , Cell Line , Cell Survival , Cricetinae , Homeostasis , Humans , Inflammation/metabolism , MAP Kinase Signaling System , Macrophages/cytology , Macrophages/metabolism , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Necrosis/metabolism , Organ Specificity , Proto-Oncogene Proteins c-akt/metabolism , Stress, Physiological
14.
Autoimmunity ; 42(4): 317-21, 2009 May.
Article in English | MEDLINE | ID: mdl-19811288

ABSTRACT

Virtually all cells in the body have the capacity to recognize and respond to dead cells. Viable cells discriminate apo from nec targets via distinct cell surface receptors. Engagement of these receptors induces "recognition-dependent" signaling events in viable responding cells that differ for apo vs. nec targets. Although "engulfment-dependent" signaling events also contribute to the response by viable cells, these events do not differ for apo vs. nec targets. While many signaling events are conserved across diverse cell lineages, other signaling events, especially those involving Akt, demonstrate lineage-specific variation. Whereas apo targets activate Akt in MPhi, they inhibit Akt in kidney epithelial cells. Differences in the responses to dead targets by viable migratory cells, such as MPhi, and viable fixed cells, such as kidney epithelial cells, permit cell-specific adaptations to local environmental change or stress. We propose that dead cells (apo and nec) act as sentinels to alert nearby viable cells to local environmental change or stress.


Subject(s)
Apoptosis/immunology , Inflammation/immunology , Necrosis/immunology , Signal Transduction/immunology , Stress, Physiological/immunology , Animals , Humans
15.
Eur Cytokine Netw ; 18(4): 172-80, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17993451

ABSTRACT

Tumor necrosis factor (TNF) is known to be a mediator of a variety of cellular responses including apoptotic death or proliferation depending on the target cell and the environmental conditions. We show here that TNF triggers both growth and death signals in NIH 3T3 murine fibroblasts. In cells arrested in G(0) by serum deprivation, TNF drives approximately 50% of them to enter the cell cycle, but kills the cells that remain quiescent. The presence of serum prevents toxic effects of TNF, suggesting that TNF can cooperate to drive cells through the cell cycle, but is unable to do so by itself and alternatively it triggers death signals in cells unable to proliferate. Interestingly, TNF induces a similar toxic effect in cells forced to stay at the G(1)/S border, S or M phases. We have explored the TNF apoptotic pathway in arrested cells. This mechanism is not due to the loss of the anti-apoptotic capacity of NFkappaB and is mediated by mitochondria since Bcl-2 overexpression partially inhibits cell death. There are, however, interesting differences in the kinetics of mitochondrial events which indicate that this form of sensitization to TNF leads to an apoptotic mechanism different from that observed after sensitization by RNA synthesis inhibition.


Subject(s)
Apoptosis , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Cycle , Cell Proliferation , Cytokines/metabolism , DNA/metabolism , Fibroblasts/metabolism , Intracellular Membranes/metabolism , Mice , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , NIH 3T3 Cells , Proto-Oncogene Proteins c-bcl-2/metabolism , Rhodamines/pharmacology
16.
Autoimmunity ; 40(4): 274-80, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17516209

ABSTRACT

Growing evidence exists for a new role for apoptotic cell recognition and clearance in immune homeostasis. Apoptotic cells at all stages, irrespective of membrane integrity, elicit a signature set of signaling events in responding phagocytes, both professional and non-professional. These signaling events are initiated by receptor-mediated recognition of apoptotic determinants, independently of species, cell type, or apoptotic stimulus. We propose that the ability of phagocytes to respond to apoptotic targets with a characteristic set of signaling events comprises a second distinct dimension of innate immunity, as opposed to the traditional innate discrimination of self vs. non-self. We further propose that a loss or abnormality of the signaling events elicited by apoptotic cells, as distinct from the actual clearance of those cells, may predispose to autoimmunity.


Subject(s)
Apoptosis/immunology , Autoimmunity , Phagocytes/immunology , Phagocytosis/immunology , Signal Transduction/immunology , Animals , Humans
17.
J Biol Chem ; 281(43): 32741-54, 2006 Oct 27.
Article in English | MEDLINE | ID: mdl-16931517

ABSTRACT

The mechanisms by which secretory phospholipases A(2) (PLA(2)s) exert cellular effects are not fully understood. Group IIF PLA(2) (gIIFPLA(2)) is a structurally unique secretory PLA(2) with a long C-terminal extension. Homology modeling suggests that the membrane-binding surface of this acidic PLA(2) contains hydrophobic residues clustered near the C-terminal extension. Vesicle leakage and monolayer penetration measurements showed that gIIFPLA(2) had a unique ability to penetrate and disrupt compactly packed monolayers and bilayers whose lipid composition recapitulates that of the outer plasma membrane of mammalian cells. Fluorescence imaging showed that gIIFPLA(2) could also readily enter and deform plasma membrane-mimicking giant unilamellar vesicles. Mutation analysis indicates that hydrophobic residues (Tyr(115), Phe(116), Val(118), and Tyr(119)) near the C-terminal extension are responsible for these activities. When gIIFPLA(2) was exogenously added to HEK293 cells, it initially bound to the plasma membrane and then rapidly entered the cells in an endocytosis-independent manner, but the cell entry did not lead to a significant degree of phospholipid hydrolysis. GIIFPLA(2) mRNA was detected endogenously in human CD4(+) helper T cells after in vitro stimulation and exogenously added gIIFPLA(2) inhibited the proliferation of a T cell line, which was not seen with group IIA PLA(2). Collectively, these data suggest that unique membrane-binding properties of gIIFPLA(2) may confer special functionality on this secretory PLA(2) under certain physiological conditions.


Subject(s)
Cell Membrane/metabolism , Phospholipases A/metabolism , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Cell Division/drug effects , Cell Line , Cell Membrane/chemistry , Escherichia coli/genetics , Group II Phospholipases A2 , Humans , Hybridomas/drug effects , Hydrolysis , Hydrophobic and Hydrophilic Interactions , In Vitro Techniques , Male , Membrane Lipids/chemistry , Membrane Lipids/metabolism , Mice , Models, Molecular , Molecular Sequence Data , Mutation , Phospholipases A/chemistry , Phospholipases A/genetics , Phospholipases A/pharmacology , Phospholipases A2 , Phospholipids/chemistry , Phospholipids/metabolism , RNA, Messenger/metabolism , Sequence Homology, Amino Acid , Static Electricity , Substrate Specificity , T-Lymphocytes/drug effects
18.
Exp Cell Res ; 312(13): 2465-75, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16730350

ABSTRACT

It has been proposed that C. elegans LIN-9 functions downstream of CDK4 in a pathway that regulates cell proliferation. Here, we report that mammalian BARA/LIN-9 is a predominantly nuclear protein that inhibits cell proliferation. More importantly, we demonstrate that BARA/LIN-9 also acts downstream of cyclin D/CDK4 in mammalian cells since (i) its antiproliferative effect is partially blocked by coexpression of cyclin D1, and (ii) a mutant form that lacks the first 84 amino acids rescues several phenotypic alterations observed in mice null for cdk4. Interestingly, mutation of BARA/LIN-9 restores the expression of E2F target genes in CDK4 null MEFs, indicating that the wild-type protein plays a role in the expression of genes required for the G1/S transition.


Subject(s)
Alleles , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cyclin-Dependent Kinase 4/deficiency , E2F Transcription Factors/antagonists & inhibitors , Mutation/genetics , Repressor Proteins/antagonists & inhibitors , Animals , Cell Cycle , Cyclin-Dependent Kinase 4/genetics , DNA/biosynthesis , E2F Transcription Factors/metabolism , Embryo, Mammalian/embryology , Female , Fertility/genetics , Fibroblasts/cytology , Gene Deletion , Gene Expression Regulation , Humans , Male , Mice , NIH 3T3 Cells , Nuclear Proteins/metabolism , Ovary/cytology , Phenotype , Pituitary Gland/cytology , Repressor Proteins/metabolism , Testis/cytology , Tumor Suppressor Proteins/metabolism
19.
J Biol Chem ; 281(29): 20055-67, 2006 Jul 21.
Article in English | MEDLINE | ID: mdl-16707494

ABSTRACT

The purpose of physiological cell death is the noninflammatory clearance of cells that have become inappropriate or nonfunctional. Consistent with this function, the recognition of apoptotic cells by professional phagocytes, including macrophages and dendritic cells, triggers a set of potent anti-inflammatory responses manifest on multiple levels. The immediate-early inhibition of proinflammatory cytokine gene transcription in the phagocyte is a proximate consequence of recognition of the apoptotic corpse, independent of subsequent engulfment and soluble factor involvement. Here, we show that recognition is linked to a characteristic signature of responses, including MAPK signaling events and the ablation of proinflammatory transcription and cytokine secretion. Specific recognition and response occurs without regard to the origin (species, tissue type, or suicidal stimulus) of the apoptotic cell and does not involve Toll-like receptor signaling. These features mark this as an innate immunity fundamentally distinct from the discrimination of "self" versus "other" considered to be the hallmark of conventional immunity. This profound unconventional innate immune discrimination of effete from live cells is as ubiquitous as apoptotic cell death itself, manifest by professional and nonprofessional phagocytes and nonphagocytic cell types alike. Innate apoptotic immunity provides an intrinsic anti-inflammatory circuit that attenuates proinflammatory responses dynamically and may act systemically as a powerful physiological regulator of immunity.


Subject(s)
Apoptosis/physiology , Immunity, Innate , Macrophages/immunology , Animals , Cell Death , Cell Line , Dendritic Cells/immunology , HeLa Cells , Humans , Jurkat Cells , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , Phagocytes/immunology , Transcription, Genetic
20.
J Biol Chem ; 281(8): 4663-70, 2006 Feb 24.
Article in English | MEDLINE | ID: mdl-16377620

ABSTRACT

Current models of autoimmunity suggest that delayed clearance of apoptotic cells leads to the presentation of apoptotic antigens in the context of inflammatory signals, with resultant autoimmunity. These models implicitly assume that, in contrast to early apoptotic cells (that retain membrane integrity), late apoptotic cells (with compromised membranes) act like necrotic cells (which also lack intact membranes), possibly because of the release of proinflammatory intracellular contents. We showed previously that early apoptotic and necrotic cells induce distinct mitogen-activated protein kinase modules in macrophages with which they interact. Exposure to apoptotic cells led to nearly complete inhibition of both basal and macrophage colony-stimulating factor-induced ERK1/2 by macrophages. In contrast, necrotic cells induced ERK1/2. We show here that apoptotic cells also strongly induced both c-Jun N-terminal kinase and p38, whereas necrotic cells had no detectable effect on c-Jun N-terminal kinase and p38. We also compared the signaling events induced in macrophages by exposure to early apoptotic cells, late apoptotic cells, and necrotic cells. The signaling events induced by late apoptotic cells were identical to and just as potent as those induced by early apoptotic cells. Thus, apoptotic cells are functionally equivalent throughout the cell death process, irrespective of membrane integrity. Moreover, the effects of both early and late apoptotic cells on signaling were dominant over those of necrotic cells. These data show that apoptotic cells do not become proinflammatory upon the loss of membrane integrity and are inconsistent with the notion that delayed clearance alone can lead to autoimmunity.


Subject(s)
Apoptosis , Autoimmunity , Necrosis , Signal Transduction , Animals , Blotting, Western , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Membrane/metabolism , Cell Physiological Phenomena , Cell Proliferation , Humans , Inflammation , Jurkat Cells , MAP Kinase Kinase 4/metabolism , MAP Kinase Signaling System , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Models, Biological , Phosphorylation , Protein Structure, Tertiary , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL