INVITED REVIEW
Mechanisms of bacterial lipopolysaccharide-induced endothelial apoptosis

Douglas D. Bannerman1 and Simeon E. Goldblum2

1 Immunology and Disease Resistance Laboratory, United States Department of Agriculture-Agricultural Research Service, Beltsville 20705; and 2 Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201


    ABSTRACT
TOP
ABSTRACT
INTRODUCTION
EVIDENCE FOR LPS-INDUCED EC...
CLINICAL RELEVANCE AND IN...
CELL SURFACE RECOGNITION OF...
TLR-4 SIGNALING LEADING TO...
TLR-4 SIGNALING LEADING TO...
ROLE OF NF-kappa B ACTIVATION...
ROLE OF CASPASES
ANTIAPOPTOTIC SIGNALING AND...
ROLE OF BCL-2 FAMILY...
ROLE OF NITRIC OXIDE
ROLE OF REACTIVE OXYGEN...
ROLE OF APOPTOTIC SIGNALING...
FUTURE DIRECTIONS
REFERENCES

Gram-negative bacterial sepsis remains a common, life-threatening event. The prognosis for patients who develop sepsis-related complications, including the development of acute respiratory distress syndrome (ARDS), remains poor. A common finding among patients and experimental animals with sepsis and ARDS is endothelial injury and/or dysfunction. A component of the outer membrane of gram-negative bacteria, lipopolysaccharide (LPS) or endotoxin, has been implicated in the pathogenesis of much of the endothelial cell injury and/or dysfunction associated with these disease states. LPS is a highly proinflammatory molecule that elicits a wide array of endothelial responses, including the upregulation of cytokines, adhesion molecules, and tissue factor. In addition to activation, LPS induces endothelial cell death that is apoptotic in nature. This review summarizes the evidence for LPS-induced vascular endothelial injury and examines the molecular signaling pathways that activate and inhibit LPS-induced endothelial apoptosis. Furthermore, the role of apoptotic signaling molecules in mediating LPS-induced activation of endothelial cells will be considered.

endotoxin; inflammation; nuclear factor-kappa B; sepsis; vascular injury


    INTRODUCTION
TOP
ABSTRACT
INTRODUCTION
EVIDENCE FOR LPS-INDUCED EC...
CLINICAL RELEVANCE AND IN...
CELL SURFACE RECOGNITION OF...
TLR-4 SIGNALING LEADING TO...
TLR-4 SIGNALING LEADING TO...
ROLE OF NF-kappa B ACTIVATION...
ROLE OF CASPASES
ANTIAPOPTOTIC SIGNALING AND...
ROLE OF BCL-2 FAMILY...
ROLE OF NITRIC OXIDE
ROLE OF REACTIVE OXYGEN...
ROLE OF APOPTOTIC SIGNALING...
FUTURE DIRECTIONS
REFERENCES

DESPITE ADVANCES IN ANTIMICROBIAL therapy and overall medical care, gram-negative bacterial sepsis remains a common, life-threatening event (163). Complications arising from sepsis include disseminated intravascular coagulation (133, 197, 201), systemic vascular collapse (88, 201), multiorgan failure (22, 23, 65), and the development of vascular leak syndromes, including acute respiratory distress syndrome (ARDS) (26, 27, 142, 144, 161). One common denominator to all of these complications is endothelial cell (EC) injury and/or dysfunction (46, 47, 80, 132, 148, 153).

The vascular endothelium serves as the key barrier between the intravascular compartment and extravascular tissues and plays a critical role in a large number of physiological and pathological processes (38). ECs are integrally involved in regulating blood flow, coagulation, leukocyte trafficking, edema formation, wound healing, and angiogenesis. Because of their location at the blood-extravascular tissue interface, ECs are constantly exposed to circulating mediators that may perturb the above-mentioned endothelial barrier functions. One such mediator that targets the endothelium is bacterial lipopolysaccharide (LPS) or endotoxin, a highly proinflammatory molecule that is a component of the outer envelope of all gram-negative bacteria (17, 159, 176). LPS is released from the surface of replicating and dying gram-negative bacteria into the circulation, where it interacts with the endothelial lining of the vessel wall (173, 176). Evidence exists that LPS, alone or in concert with other endogenous factors, is responsible for much of the EC injury and/or dysfunction associated with gram-negative sepsis. First, LPS bioactivity has been detected in the bloodstream of gram-negative septicemic patients, and in selected studies, levels of circulating LPS predict the development of multiorgan failure, including ARDS (26). Second, administration of LPS alone to experimental animals reconstitutes the EC injury seen after gram-negative bacterial challenge (27, 201). Third, immunological and pharmacological interventions that specifically target the LPS molecule protect against these same vascular complications (6, 210, 211, 225). Finally, LPS directly elicits several of the EC responses in vitro that are similarly evoked during sepsis, including: 1) the production of the proinflammatory cytokines IL-6 (77, 109), IL-8 (7, 77, 223), and IL-1beta (57, 138); 2) the increased surface expression of the adhesion molecules E-selectin, intercellular adhesion molecule (ICAM)-1, and vascular cell adhesion molecule (VCAM)-1 (34, 59, 77, 108); and 3) increased expression of tissue factor (42, 75). In addition to activation, LPS induces EC-programmed cell death or apoptosis (18, 21, 64, 67, 82, 95, 116, 143, 221), an event that is believed to contribute to the pathogenesis of sepsis and its attendant complications (82, 189). Several of the signaling molecules involved in EC activation leading to increased cytokine and adhesion molecule expression are similarly involved in LPS proapoptotic signaling. Furthermore, there is evidence that LPS proapoptotic signaling molecules have an additional role in regulating LPS-induced NF-kappa B activation and nonapoptotic EC responses to LPS. This review focuses on recent advances in the elucidation of the mechanisms by which LPS elicits EC apoptosis and the cross talk between signaling pathways leading to EC activation and death.


    EVIDENCE FOR LPS-INDUCED EC APOPTOSIS
TOP
ABSTRACT
INTRODUCTION
EVIDENCE FOR LPS-INDUCED EC...
CLINICAL RELEVANCE AND IN...
CELL SURFACE RECOGNITION OF...
TLR-4 SIGNALING LEADING TO...
TLR-4 SIGNALING LEADING TO...
ROLE OF NF-kappa B ACTIVATION...
ROLE OF CASPASES
ANTIAPOPTOTIC SIGNALING AND...
ROLE OF BCL-2 FAMILY...
ROLE OF NITRIC OXIDE
ROLE OF REACTIVE OXYGEN...
ROLE OF APOPTOTIC SIGNALING...
FUTURE DIRECTIONS
REFERENCES

Apoptosis is an ATP-dependent form of cell death, morphologically characterized by chromatin condensation, nuclear fragmentation, cell shrinkage, and blebbing of the plasma membrane (76, 171). The end result of apoptosis is fragmentation of the cell into small membrane-bound bodies that are quickly cleared by phagocytotic cells (45, 181, 187). Biochemically, apoptosis is characterized by the activation of caspases, highly specific proteases that cleave a wide array of intracellular substrates (198). Activation of upstream caspases initiates a proteolytic cascade leading to DNA fragmentation and the cleavage of key regulatory proteins resulting in cell death. In contrast to apoptosis, necrotic cell death is an energy-independent process characterized by cell swelling and lysis (76, 171). Unlike apoptotic cells, necrotic cells release cellular constituents that elicit an inflammatory reaction in surrounding viable tissue.

LPS induces apoptosis in bovine and ovine ECs in vitro (18, 64, 94, 95) and elicits human EC apoptosis in the absence of new gene expression (164). That LPS-induced EC death is apoptotic in nature has been confirmed by several criteria, including morphological changes (64), DNA laddering (18, 99), TdT-mediated dUTP nick end labeling (18, 94, 95), nuclear histone release (15, 21), caspase activation (15, 19), and poly(ADP-ribose)polymerase cleavage (21, 36). Several studies have demonstrated that purified LPS itself, in the absence of host-derived mediators, evokes EC injury and/or apoptosis (18, 36, 64, 67, 82, 95, 116, 221). The ability of LPS to induce EC apoptosis in the absence of non-EC-derived host mediators is compatible with a direct effect. Furthermore, neutralizing antibodies to other known death receptors expressed on EC, including TNF type 1 receptor (TNFR1), Fas, and death receptor 3 (DR3), fails to inhibit LPS-induced apoptosis (36). Finally, LPS-induced EC apoptosis occurs independently of new protein synthesis, thereby precluding the involvement of upregulated gene products (84).


    CLINICAL RELEVANCE AND IN VIVO EVIDENCE
TOP
ABSTRACT
INTRODUCTION
EVIDENCE FOR LPS-INDUCED EC...
CLINICAL RELEVANCE AND IN...
CELL SURFACE RECOGNITION OF...
TLR-4 SIGNALING LEADING TO...
TLR-4 SIGNALING LEADING TO...
ROLE OF NF-kappa B ACTIVATION...
ROLE OF CASPASES
ANTIAPOPTOTIC SIGNALING AND...
ROLE OF BCL-2 FAMILY...
ROLE OF NITRIC OXIDE
ROLE OF REACTIVE OXYGEN...
ROLE OF APOPTOTIC SIGNALING...
FUTURE DIRECTIONS
REFERENCES

Endothelial apoptosis has been implicated in the pathogenesis of several disease states, including atherosclerosis (30), hypertension (73), congestive heart failure (204), and systemic capillary leak syndrome (12). Several studies have reported that EC injury and/or death is a key pathological finding during bacterial sepsis. First, injection of Escherichia coli into rabbits (47) or baboons (40, 41) induces severe microvascular injury and EC detachment. Second, increased numbers of apoptotic ECs are detected in the pulmonary capillaries of a murine model of sepsis (93, 217). Third, evidence of EC injury is observed in postmortem biopsies obtained from patients who have died of sepsis-related ARDS (148). Fourth, an increase in circulating EC is observed in septic patients, and the magnitude of this increase correlates negatively with survival (153).

There are several reports that purified LPS elicits EC injury and apoptosis in vivo. EC injury and/or detachment from the vascular wall has been reported after LPS injection into mice (124), rats (172, 192), rabbits (47, 69, 145), dogs (72, 127), sheep (27, 149), and primates (14). Liver sinusoidal ECs obtained from LPS-treated rats display enhanced activation of caspase-3, a central apoptotic effector protease (49). Disseminated EC apoptosis has been reported in the lung, liver, thymus, and intestine of mice challenged with LPS (67, 82, 116, 122). Finally, injection of a broad spectrum caspase inhibitor decreases EC apoptosis in the lung after LPS administration and improves survival in a murine model of acute lung injury (116). The combined in vitro and in vivo data implicate apoptosis as a key component of the EC response to LPS. However, the extent to which LPS directly induces EC apoptosis in human systems either alone or in concert with other known apoptosis-inducing proinflammatory cytokines, such as TNF-alpha and/or IL-1beta , remains unclear.


    CELL SURFACE RECOGNITION OF LPS
TOP
ABSTRACT
INTRODUCTION
EVIDENCE FOR LPS-INDUCED EC...
CLINICAL RELEVANCE AND IN...
CELL SURFACE RECOGNITION OF...
TLR-4 SIGNALING LEADING TO...
TLR-4 SIGNALING LEADING TO...
ROLE OF NF-kappa B ACTIVATION...
ROLE OF CASPASES
ANTIAPOPTOTIC SIGNALING AND...
ROLE OF BCL-2 FAMILY...
ROLE OF NITRIC OXIDE
ROLE OF REACTIVE OXYGEN...
ROLE OF APOPTOTIC SIGNALING...
FUTURE DIRECTIONS
REFERENCES

As the name implies, LPS is composed of both lipid and polysaccharide components. The lipid portion is composed of a unique lipid, lipid A, which is the most widely conserved region of the LPS molecule (174). It is well established that the lipid A moiety of LPS is responsible for its proinflammatory properties (174). Lipid A alone is capable of eliciting EC responses identical to those induced by LPS (16, 50, 193). Agents that specifically target the lipid A moiety of LPS inhibit EC activation (16, 52) and protect against the development of vascular complications in endotoxin shock models (6, 210). Similar to activation, the lipid A moiety is also responsible for the proapoptotic properties of the LPS molecule (84). Neutralization of lipid A with polymyxin B completely abrogates LPS-induced EC apoptosis (18, 19, 64).

An early identified receptor implicated in cellular recognition of LPS was membrane-associated CD14 (mCD14) (202). mCD14 is a glycoprotein found on cells of monocytic origin and to a lesser extent on neutrophils. Although LPS can directly bind mCD14, its affinity for the receptor is greatly increased when LPS is complexed with the acute phase protein LPS-binding protein (LBP) (159). ECs, which lack mCD14, are activated by LPS in the presence of LBP and soluble CD14 (sCD14), the latter of which is released from mCD14-bearing cells (9, 74). It has been proposed that LBP facilitates the transfer of LPS to sCD14 and that this LPS-sCD14 complex is recognized by ECs (202, 205). Similar to EC activation, a requisite role for sCD14 in mediating LPS-induced apoptosis has been established. In the absence of sCD14, ECs are resistant to LPS-induced apoptosis (9, 64, 219). In the presence of CD14-containing serum, anti-CD14 antibodies inhibit LPS-induced EC cytotoxicity (9, 64, 219). Because CD14 is a glycosyl phosphatidylinositol-anchored protein and lacks an intracellular cytoplasmic domain, it was postulated that another transmembrane receptor must exist that can transduce LPS signaling across the plasma membrane. This receptor, which has since been identified as Toll-like receptor (Tlr)-4, is expressed in cells of monocytic lineage as well as in non-mCD14-bearing ECs (37, 61). There has been some controversy in the past regarding whether another member of the Toll-like receptor family, Tlr-2, mediates LPS-induced signaling. Two groups have reported that LPS-induced activation is mediated by Tlr-2 (121, 218). However, a subsequent study reported that certain commercial preparations of LPS are contaminated with bacterial lipoproteins and that these lipoproteins, not LPS, are responsible for Tlr-2 activation (90). The genetic evidence clearly establishes Tlr-4 as a true LPS receptor (166). C3H/HeJ and C57BL/10ScCr mice, which contain a missense mutation in or a null mutation for the Tlr-4 gene, respectively, are resistant to LPS (166). Furthermore, transfection of Tlr-4 into LPS-insensitive HEK-293 cells confers sensitivity to LPS-induced NF-kappa B activation (37). Finally, anti-Tlr-4 antibodies abrogate LPS-induced EC NF-kappa B activation, whereas anti-Tlr-2 antibodies have no inhibitory effect (61).


    TLR-4 SIGNALING LEADING TO NF-kappa B ACTIVATION
TOP
ABSTRACT
INTRODUCTION
EVIDENCE FOR LPS-INDUCED EC...
CLINICAL RELEVANCE AND IN...
CELL SURFACE RECOGNITION OF...
TLR-4 SIGNALING LEADING TO...
TLR-4 SIGNALING LEADING TO...
ROLE OF NF-kappa B ACTIVATION...
ROLE OF CASPASES
ANTIAPOPTOTIC SIGNALING AND...
ROLE OF BCL-2 FAMILY...
ROLE OF NITRIC OXIDE
ROLE OF REACTIVE OXYGEN...
ROLE OF APOPTOTIC SIGNALING...
FUTURE DIRECTIONS
REFERENCES

Tlr-4 is integrally involved in LPS signaling and has a requisite role in the activation of the transcription factor NF-kappa B. The extracellular domain of Tlr-4 contains repeating leucine-rich motifs characteristic of innate immune response pattern recognition receptors (Fig. 1) (48). The cytoplasmic domain contains regions that are homologous to the intracellular signaling domain of the type 1 IL-1 receptor. Although the exact mechanism by which LPS is recognized by Tlr-4 remains unclear, cell activation is dependent on the cell surface assembly of a multiprotein recognition complex consisting of CD14, MD-2, and Tlr-4 (3) (Fig. 2). After activation of the Tlr-4 receptor complex, the adapter protein myeloid differentiation factor 88 (MyD88) is recruited to the cytoplasmic domain of Tlr-4 through homotypic binding of respective Toll receptor-IL-1 receptor (TIR) domains (146, 154). MyD88 contains an additional protein-binding domain, the death domain (DD) (Fig. 1), which facilitates its association with another DD-containing signaling molecule, IL-1 receptor-associated kinase-1 (IRAK-1) (48). After autophosphorylation, IRAK-1 dissociates from MyD88 and interacts with TNF receptor-associated factor-6 (TRAF-6) (137, 194), resulting in the activation of a downstream kinase cascade involving NF-kappa B-inducing kinase (NIK) and Ikappa B kinase (IKK) (Fig. 2). The IKK-mediated phosphorylation of Ikappa B, an inhibitor of NF-kappa B, leads to Ikappa B degradation through the proteasome pathway and enables NF-kappa B to translocate to the nucleus where it promotes new gene expression (48).


View larger version (25K):
[in this window]
[in a new window]
 
Fig. 1.   Schematic diagram of the binding motifs of signaling molecules involved in LPS apoptotic and NF-kappa B signaling. Toll-like receptor (Tlr)-4, myeloid differentiation factor 88 (MyD88), MyD88 adapter-like protein/Toll receptor-IL-1 receptor domain-containing adpater protein (MAL/TIRAP), IL-1 receptor-associated kinase-1 (IRAK-1), and TNF receptor-associated factor-6 (TRAF-6) have all been shown to promote LPS-induced NF-kappa B and proapoptotic signaling. Fas-associated death domain (FADD) and FADD-like interleukin converting enzyme-like inhibitor protein (FLIP) have been implicated in mediating LPS-induced pro- and antiapoptotic signaling, respectively. LRR, leucine-rich repeats; DD, death domain; TIR, Toll receptor-IL-1 receptor domain; PKD, protein kinase domain; ZFs, zinf finger domains; DED, death effector domains; ICE, interleukin-converting enzyme-like protease domain.



View larger version (30K):
[in this window]
[in a new window]
 
Fig. 2.   Schematic diagram of LPS proapoptotic signaling in endothelial cells (ECs). After LPS binding to Tlr-4, EC signaling pathways leading to nuclear translocation of NF-kappa B and apoptosis are activated. These pathways share some of the same signaling molecules, including MyD88, IRAK-1, and TRAF-6. A redundant signaling pathway involving MAL/TIRAP similarly mediates both NF-kappa B activation and apoptosis. Downstream of TRAF-6, the signaling pathways that activate these two processes diverge. The molecule(s) that links the upstream NF-kappa B signaling molecules to the recruitment and activation of caspases remains unknown. LBP, LPS-binding protein; NIK, NF-kappa B-inducing kinase; IKK, Ikappa B kinase; sCD14, soluble CD14.


    TLR-4 SIGNALING LEADING TO APOPTOSIS
TOP
ABSTRACT
INTRODUCTION
EVIDENCE FOR LPS-INDUCED EC...
CLINICAL RELEVANCE AND IN...
CELL SURFACE RECOGNITION OF...
TLR-4 SIGNALING LEADING TO...
TLR-4 SIGNALING LEADING TO...
ROLE OF NF-kappa B ACTIVATION...
ROLE OF CASPASES
ANTIAPOPTOTIC SIGNALING AND...
ROLE OF BCL-2 FAMILY...
ROLE OF NITRIC OXIDE
ROLE OF REACTIVE OXYGEN...
ROLE OF APOPTOTIC SIGNALING...
FUTURE DIRECTIONS
REFERENCES

Several of the upstream signaling molecules involved in LPS activation of NF-kappa B are similarly involved in promoting LPS-induced apoptosis. Macrophages derived from C3H/HeJ mice, which have a missense mutation in the third exon of Tlr-4 (166), are resistant to LPS-induced apoptosis (110). Furthermore, neutralization of the lipid A moiety of LPS, which is the domain of LPS recognized by Tlr-4 (139), protects against LPS-induced EC apoptosis (18, 19). Downstream of Tlr-4, MyD88 and IRAK-1 have been implicated in mediating LPS-elicited cell death signaling. Expression of either MyD88 or IRAK-1 dominant-negative (D/N) constructs, which inhibit LPS-induced NF-kappa B activation (19, 222), protects against LPS-induced EC apoptosis (19). MyD88 has similarly been shown to mediate Tlr-2 activation of NF-kappa B and proapoptotic signaling (5). Downstream of MyD88 and IRAK-1, TRAF-6 has been implicated in promoting LPS-induced EC apoptosis (100). TRAF-6 participation in LPS-induced apoptosis involves c-jun NH2-terminal kinase, the activation of which lies upstream of caspase activation. Together, these data indicate that Tlr-4 can serve as a death receptor for LPS and that the signaling molecules involved in LPS-induced NF-kappa B activation serve a dual role in promoting LPS-induced apoptosis (Fig. 2).

There is evidence suggesting that Tlr-4 activation of NF-kappa B can occur through a MyD88- and IRAK-1-independent pathway. The LPS-induced DNA-binding activity of NF-kappa B in macrophages derived from either MyD88 or IRAK-1 knockout mice is delayed, but not inhibited, indicating that cellular activation by LPS can occur in the absence of these signaling molecules (115, 194). Recently, an MyD88-like protein has been identified by two independent groups, MyD88 adapter-like protein (MAL) or TIR domain-containing adapter protein (TIRAP), which promote LPS-induced NF-kappa B signaling through IRAK-2 (62, 91). MAL/TIRAP contains a COOH terminus TIR domain but lacks the NH2 terminus DD present in MyD88 (Fig. 1). Similar to MyD88, MAL/TIRAP has an additional role in promoting LPS-induced EC apoptosis (15). These studies demonstrate the presence of an MyD88-independent pathway that serves a redundant signaling role in promoting both LPS-induced NF-kappa B activation and apoptosis in ECs (Fig. 2).

Although Tlr-4 and its respective intracellular binding partners MyD88 and MAL/TIRAP have been shown to mediate LPS-induced apoptosis, questions remain as to how this signaling pathway activates the effector proteases of apoptosis, the caspases. The Fas-associated death domain (FADD) is a proapoptotic adapter protein that couples death receptors to initiator caspases (24, 35, 96, 126). Its role in mediating apoptosis has been well elucidated in death signaling initiated by the Fas and TNF-alpha receptors. FADD recruitment, either directly to Fas or through the intermediary TNF receptor-associated DD protein in the case of the TNF-alpha receptor, is mediated through binding between the two highly conserved DDs found on these proteins. Similarly, FADD and procaspase-8 interact through the death effector domains (DED) contained by each protein (Fig. 1). Procaspase-8 contains partial proteolytic activity that enables it to cleave other procaspase-8 molecules brought into close proximity after recruitment to FADD (155). Activation of caspase-8 initiates a proteolytic cascade resulting in the activation of downstream effector caspases, including caspase-3 (89). Although a role for FADD in mediating TNF-alpha - and Fas-induced apoptosis has been well established, there are conflicting reports as to whether this molecule is involved in LPS proapoptotic signaling. Choi et al. (36) reported that expression of a FADD D/N protected against LPS-induced EC apoptosis, whereas another group showed that the same FADD D/N had no effect (19). It is difficult to reconcile these differences because both studies utilized the same EC type and a comparable retrovirus-based system to stably express identical FADD D/N constructs. Furthermore, both studies were able to demonstrate the efficacy of the FADD D/N construct in protecting against TNF-alpha -elicited apoptosis. One major difference between the studies was in the assay used to measure cell death. In the study by Choi et al. (36), mitochondrial activity was used as a measure of cell viability. This method of viability, however, does not discriminate between necrotic and apoptotic cell death. In the contrasting report, a more selective assay for determining apoptotic cell death, which involved measuring caspase activity, was used (19). One explanation for these differences may be that LPS induces both apoptotic (caspase-dependent) and nonapoptotic (caspase-independent) cell death and that FADD plays a role in mediating the latter.

Tlr-4 is one member of a larger family of pattern recognition receptors involved in the innate immune response. Another member of the Tlr family, Tlr-2, recognizes the cell wall components peptidoglycan and bacterial lipoproteins found in both gram-positive and -negative bacteria as well as gram-positive-restricted lipoteichoic acid (4, 186). Several of the intracellular signal transduction molecules involved in Tlr-4-mediated activation of NF-kappa B, including MyD88, IRAK, TRAF-6, NIK, and IKK, are similarly involved in Tlr-2 signaling. Interestingly, bacterial lipoprotein-elicited Tlr-2 proapoptotic signaling is reportedly mediated by MyD88 and FADD, two molecules similarly involved in LPS/Tlr-4-induced apoptosis (5). In contrast, Tlr-2-mediated apoptosis occurs independently of TRAF-6 involvement (5), a signaling molecule recently established to participate in LPS/Tlr-4 proapoptotic signaling (100). Thus divergence in the apoptotic signaling pathways mediated by Tlr-2 and Tlr-4 occurs downstream of MyD88.


    ROLE OF NF-kappa B ACTIVATION IN MEDIATING LPS-INDUCED APOPTOSIS
TOP
ABSTRACT
INTRODUCTION
EVIDENCE FOR LPS-INDUCED EC...
CLINICAL RELEVANCE AND IN...
CELL SURFACE RECOGNITION OF...
TLR-4 SIGNALING LEADING TO...
TLR-4 SIGNALING LEADING TO...
ROLE OF NF-kappa B ACTIVATION...
ROLE OF CASPASES
ANTIAPOPTOTIC SIGNALING AND...
ROLE OF BCL-2 FAMILY...
ROLE OF NITRIC OXIDE
ROLE OF REACTIVE OXYGEN...
ROLE OF APOPTOTIC SIGNALING...
FUTURE DIRECTIONS
REFERENCES

LPS-induced activation of NF-kappa B is a key signaling event that mediates an array of EC responses, including increased 1) IL-6 (109), IL-8 (7, 223), and IL-1beta (138) production, 2) E-selectin, ICAM-1, and VCAM-1 surface expression (34, 108), and 3) tissue factor activity (42, 75). In addition to its role in promoting the expression of proinflammatory gene products, NF-kappa B has been implicated in both pro- (86, 178) and antiapoptotic signaling (128, 203). In ECs, inhibition of NF-kappa B activation sensitizes human ECs to direct TNF-alpha -induced apoptosis in the absence of cycloheximide, suggesting an antiapoptotic role for NF-kappa B (221). Evidence exists that this sensitization is conferred by inhibition of NF-kappa B-dependent expression of members of the inhibitors of apoptosis (IAP) gene family (190). In contrast to TNF-alpha , inhibition of NF-kappa B does not sensitize human ECs to direct LPS-induced apoptosis (221). These data, therefore, preclude an antiapoptotic role for NF-kappa B in conferring protection against LPS-elicited apoptosis.

There are reports that NF-kappa B signaling has a role in proapoptotic signaling (86, 178). On the basis of the predominant role of NF-kappa B in mediating LPS-elicited EC responses and the finding that MyD88, MAL/TIRAP, and IRAK-1 promote both LPS-induced NF-kappa B activation and apoptosis (Fig. 2), there has been speculation that these events are coupled. Several lines of evidence, however, suggest that these events occur independently of one another. First, the presumed mechanism of the proapoptotic properties of NF-kappa B reported by Ryan et al. (178) is through the promotion of new gene expression, similar to its antiapoptotic role and the induction of antiapoptotic proteins. The finding that LPS elicits EC apoptosis in the absence of new protein synthesis argues against a proapoptotic role for NF-kappa B that involves new gene expression (21). Second, inhibition of NF-kappa B activation by overexpression of an Ikappa Balpha superrepressor fails to block LPS-induced EC apoptosis (19, 21). This latter finding also demonstrates a bifurcation in the NF-kappa B and apoptotic pathways that mediate LPS signaling upstream of Ikappa B degradation. Finally, complete inhibition of apoptosis with the caspase inhibitor zVAD-FMK does not inhibit the ability of LPS to activate NF-kappa B (19). Thus despite a commonality among upstream signaling molecules, LPS/Tlr-4-induced NF-kappa B activation and apoptosis are mutually independent events (Fig. 2).


    ROLE OF CASPASES
TOP
ABSTRACT
INTRODUCTION
EVIDENCE FOR LPS-INDUCED EC...
CLINICAL RELEVANCE AND IN...
CELL SURFACE RECOGNITION OF...
TLR-4 SIGNALING LEADING TO...
TLR-4 SIGNALING LEADING TO...
ROLE OF NF-kappa B ACTIVATION...
ROLE OF CASPASES
ANTIAPOPTOTIC SIGNALING AND...
ROLE OF BCL-2 FAMILY...
ROLE OF NITRIC OXIDE
ROLE OF REACTIVE OXYGEN...
ROLE OF APOPTOTIC SIGNALING...
FUTURE DIRECTIONS
REFERENCES

A hallmark of apoptosis is the activation of highly specific effector proteases of the caspase family. Caspases exist as inactive zymogens (proenzymes) that are activated by proteolytic processing of the procaspase molecule in one of three ways: 1) autoactivation due to low levels of intrinsic catalytic activity, 2) transactivation by other caspases within close proximity, or 3) activation by noncaspase proteases (198). Once activated, caspases cleave and activate other members of the caspase family leading to amplification of a proteolytic cascade. The end result is a series of proteolytic events that lead to the cleavage of intracellular substrates, chromatin condensation, DNA fragmentation, and eventual cell death.

Of the 14 known caspases expressed in mammalian cells, LPS has been reported to activate caspase-1 (152, 185), caspase-3 (15, 49, 100, 111, 152), caspase-6 (111), and caspase-8 (100, 111). Activation of caspase-1 (interleukin-1 converting enzyme) is primarily responsible for IL-1beta processing and has limited involvement in apoptosis (43). In contrast, caspase-3, -6, and -8 all have been established to prominently participate in apoptosis. Caspase-8, also known as FADD-like interleukin converting enzyme protease (FLICE), is classically described as an initiator caspase that is recruited to such death receptors as TNFR and Fas. Procaspase-8 has intrinsically low levels of proteolytic activity that enables it to cleave other procaspase-8 molecules brought into close proximity after recruitment to the receptor (155). Activation of caspase-8 initiates a proteolytic cascade resulting in the activation of downstream effector caspases and the onset of apoptosis (89). A key target of caspase-8 is procaspase-3. Proteolytic processing of procaspase-3 results in an active caspase-3 molecule that serves as a key effector caspase responsible for much of the proteolysis associated with apoptosis (89, 107). The effects of LPS-induced caspase activation on ECs are quite dramatic, resulting in the cleavage of nuclear proteins as well as structural proteins that mediate cell-cell and cell-substrate adhesion (18, 21). In vivo studies support a role for caspases in mediating LPS-induced EC apoptosis (49, 116). Liver ECs derived from mice administered intravenous LPS display enhanced capase-3 activity (49). Furthermore, inhibition of caspase activation with the cell-permeable, pan-caspase inhibitor peptide zVAD protects against EC apoptosis elicited by LPS in a murine model of endotoxin shock (116).

The mechanism by which LPS/Tlr-4 signaling activates caspases remains unknown. A proximal event in LPS signaling is MyD88 recruitment of IRAK through reciprocal binding of the DD regions on each of these molecules (48). One possibility is that FADD or a FADD-like protein may bind either MyD88 and/or IRAK-1 through homophilic DD-DD interactions (Fig. 2). By a mechanism similar to that reported for other death receptor signaling pathways, including those involving Fas and TNFR1 (11), FADD, which contains an additional DED binding domain, may recruit procaspase-8 via DED-DED interactions to the Tlr-4 signaling complex. The induced proximity of procaspase-8 molecules could lead to respective activation and the onset of apoptosis. In support of this hypothesis, overexpressed MyD88 has been reported to bind FADD via DD-mediated interactions (5, 92). Whether this interaction occurs when these proteins are expressed at physiological levels remains unknown.


    ANTIAPOPTOTIC SIGNALING AND ROLE OF FLICE-LIKE INHIBITOR PROTEIN
TOP
ABSTRACT
INTRODUCTION
EVIDENCE FOR LPS-INDUCED EC...
CLINICAL RELEVANCE AND IN...
CELL SURFACE RECOGNITION OF...
TLR-4 SIGNALING LEADING TO...
TLR-4 SIGNALING LEADING TO...
ROLE OF NF-kappa B ACTIVATION...
ROLE OF CASPASES
ANTIAPOPTOTIC SIGNALING AND...
ROLE OF BCL-2 FAMILY...
ROLE OF NITRIC OXIDE
ROLE OF REACTIVE OXYGEN...
ROLE OF APOPTOTIC SIGNALING...
FUTURE DIRECTIONS
REFERENCES

FLICE-like inhibitor protein (FLIP) is an antiapoptotic protein with significant homology to caspase-8 (FLICE) (106). In FLIP, a substitution of two amino acids that corresponds to the caspase-8 catalytic site renders it catalytically inactive. The role of FLIP in the inhibition of Fas death signaling has been well elucidated. On Fas ligand binding, the adapter protein FADD is recruited to the Fas receptor via an interaction between the DD of each protein (199). FLIP and procaspase-8 are, in turn, recruited to FADD via DED protein-protein interactions contained within all three proteins (Fig. 1). It has been proposed that FLIP can inhibit activation of upstream initiator caspases, including caspase-8, by competitively binding to FADD and blocking assembly of a functional death signaling complex (188). Alternatively, FADD recruitment of FLIP, which lacks intrinsic proteolytic activity, may prevent transactivation of procaspase-8 (182). A role for FLIP in inhibiting TNFR1-elicited apoptosis through a similar mechanism has been reported (97).

Recent studies indicate that FLIP protects against LPS-induced apoptosis. First, decreased expression of FLIP parallels in both a dose- and time-dependent manner with EC sensitization to LPS-induced apoptosis (21, 60). Second, inhibition of proteasome-mediated FLIP degradation protects against LPS-induced apoptosis (21). Third, specific inhibition of FLIP expression with antisense oligonucleotides sensitizes ECs to LPS-induced apoptosis (21). Finally, overexpression of FLIP protects against LPS-evoked apoptosis (60). Together, these data indicate that FLIP confers protection against LPS-induced apoptosis. The mechanism of protection remains unknown. A previous report has implicated a role for FADD in mediating LPS-induced apoptosis (36). One may speculate that Tlr-4 recruitment of FADD or a FADD-like molecule leads to the recruitment of upstream initiator caspases and subsequent caspase activation. In a manner analogous to the Fas and TNFR1 pathways, FLIP recruitment to FADD would be expected to prevent caspase recruitment and/or activation and block the onset of apoptosis (Fig. 2).

Both LPS and TNF-alpha directly induce EC apoptosis in bovine ECs. Human EC sensitization to apoptosis elicited by either of these agents is dependent on the absence of de novo gene expression (164). EC sensitization is conferred by either cycloheximide or actinomycin D, protein or mRNA synthesis inhibitors, respectively. The requisite inhibition of gene expression required for sensitization suggests that either a constitutively expressed protein with a relatively short half-life or an inducible protein confers EC resistance to LPS- and TNF-alpha -induced apoptosis. LPS and TNF-alpha each upregulate the expression of several antiapoptotic proteins, including A1 (99, 112, 113), A20 (58, 99, 220), and cellular inhibitor of apoptosis proteins (cIAPs) (190) in an NF-kappa B-dependent manner. Blocking NF-kappa B activation sensitizes ECs to direct TNF-alpha -induced apoptosis and correlates with the inhibition of expression of these NF-kappa B-inducible antiapoptotic proteins (221). In contrast, inhibition of NF-kappa B activation fails to sensitize ECs to direct LPS-induced apoptosis, suggesting a distinct mechanism by which inhibition of new gene expression sensitizes ECs to LPS- and TNF-alpha -induced apoptosis. Recently, inhibition of proteasome-mediated protein degradation was shown to protect against LPS-induced apoptosis in the presence of cycloheximide (21). That LPS-evoked apoptosis was prevented in the absence of new gene expression precludes a role for an inducible gene product. Interestingly, inhibition of de novo protein synthesis leads to a marked decrease in the expression of FLIP, a constitutively expressed protein implicated in conferring resistance to LPS-induced apoptosis (21, 60). This suggests that cycloheximide sensitization of human ECs to LPS-induced apoptosis is mediated by both the inhibition of de novo synthesis of FLIP and the rapid degradation of preexisting FLIP molecules via the proteasome (Fig. 3).


View larger version (31K):
[in this window]
[in a new window]
 
Fig. 3.   Schematic diagram of the mechanism by which cycloheximide (CHX) or Shiga-like toxin sensitizes ECs to LPS-induced apoptosis. Under physiological conditions, constitutive expression of FLIP confers resistance to LPS-induced apoptosis in human ECs. In the presence of CHX or Shiga-like toxin (SLT), de novo synthesis of FLIP is inhibited. Preexisting molecules of FLIP are rapidly degraded via the proteasome. The end result of CHX or SLT exposure is diminished expression of FLIP and sensitization of human ECs to LPS-induced apoptosis.

Although cycloheximide or actinomycin D have been used as tools to investigate sensitization to LPS-induced apoptosis by inhibiting de novo gene expression, it is difficult to imagine a scenario in which these agents would be present in vivo with LPS. However, naturally occurring agents that inhibit protein synthesis may be expected to confer similar sensitivity. Shiga toxin and Shiga-like toxin-1 (SLT-1), produced by Shigella dysenteriae serotype 1 and certain strains of E. coli, respectively, inhibit protein synthesis (141, 162, 213). Both of these toxins have been implicated in the pathogenesis of hemolytic uremic syndrome and its attendant EC injury (158, 167, 170). Shiga toxin and SLT-1 inhibit protein synthesis by cleaving a specific bond in the 28S rRNA component of the 60S ribosomal subunit, resulting in the release of a single adenine base and the inhibition of aminoacyl tRNA binding to the ribosome (79, 158). Recent findings suggest that SLT-1 sensitizes human ECs to LPS-induced apoptosis by virtue of its protein synthesis inhibitory properties (60) (Fig. 3). SLT-1 inhibition of FLIP expression sensitizes ECs to LPS-induced apoptosis in both a dose- and time-dependent manner. Furthermore, sustained expression of FLIP in the presence of SLT-1 abrogates SLT-induced sensitization of human ECs to LPS-induced apoptosis. Thus strains of E. coli that produce SLT-1 have all the necessary components to elicit EC apoptosis, namely a sensitizing agent, SLT-1, and an inducer of apoptosis, LPS. Other naturally occurring agents reported to have protein synthesis inhibitory properties include Pseudomonas aeruginosa toxin (101), interferons (39), and TNF-alpha (129). The role of these agents in sensitizing ECs to LPS-induced apoptosis during mixed infection and inflammation should be considered. Furthermore, patients in severe sepsis often are in a profound catabolic state (85). Whether this influences the ability of cells to maintain adequate levels of antiapoptotic proteins while being exposed to LPS and/or apoptotic-inducing cytokines remains unknown.


    ROLE OF BCL-2 FAMILY MEMBERS
TOP
ABSTRACT
INTRODUCTION
EVIDENCE FOR LPS-INDUCED EC...
CLINICAL RELEVANCE AND IN...
CELL SURFACE RECOGNITION OF...
TLR-4 SIGNALING LEADING TO...
TLR-4 SIGNALING LEADING TO...
ROLE OF NF-kappa B ACTIVATION...
ROLE OF CASPASES
ANTIAPOPTOTIC SIGNALING AND...
ROLE OF BCL-2 FAMILY...
ROLE OF NITRIC OXIDE
ROLE OF REACTIVE OXYGEN...
ROLE OF APOPTOTIC SIGNALING...
FUTURE DIRECTIONS
REFERENCES

Cell commitment to apoptosis is governed by both pro- and antiapoptotic signaling pathways. The Bcl-2 family of proteins plays a central role in mediating these two opposed pathways (2, 8). The Bcl-2 family is composed of both pro- and antiapoptotic members. A key mechanism by which these proteins mediate apoptosis is through the regulation of cytochrome c release from the mitochondrion (2, 8). Cytochrome c is a cofactor in the activation of caspase-9, the latter of which activates downstream effector caspases, including caspase-3. Proapoptotic and antiapoptotic members of the Bcl-2 family facilitate and restrict, respectively, cytochrome c release from the mitochondrion. The various Bcl-2 family members are characterized by the presence of one or more of four distinct Bcl-2 homology (BH) domains that facilitate protein-protein interactions. Antiapoptotic Bcl-2 family members exert their effect by forming heterodimers via these BH domains with the proapoptotic members, thereby impairing the ability of the proapoptotic members to induce cytochrome c release.

Several lines of evidence suggest that members of the Bcl-2 family mediate sepsis- and LPS-induced apoptosis. First, in a murine model of sepsis induced by cecal ligation and puncture, enhanced endothelial apoptosis parallels a decrement in the expression of the antiapoptotic protein Bcl-2 (217). Second, LPS upregulates EC expression of the proapoptotic Bcl-2 family members Bax (81, 118, 152), Bad (147), and Bak (147). LPS has also been reported to upregulate expression of A1 (99) and to downregulate levels of Bcl-2 (81, 118) and Bcl-xL (147), all of which are antiapoptotic members of the Bcl-2 family. Third, vascular endothelial growth factor (VEGF) (152) and other agents (81, 118) that inhibit LPS-induced upregulation of proapoptotic Bcl-2 family members protect against LPS-induced EC apoptosis. Inhibition of LPS-induced downregulation of antiapoptotic Bcl-2 homologs similarly confers protection (81, 118). Finally, overexpression of A1 or Bcl-xL protects against LPS-induced EC apoptosis (99). Together, these data implicate a role for Bcl-2 homologs in determining EC fate after LPS exposure.


    ROLE OF NITRIC OXIDE
TOP
ABSTRACT
INTRODUCTION
EVIDENCE FOR LPS-INDUCED EC...
CLINICAL RELEVANCE AND IN...
CELL SURFACE RECOGNITION OF...
TLR-4 SIGNALING LEADING TO...
TLR-4 SIGNALING LEADING TO...
ROLE OF NF-kappa B ACTIVATION...
ROLE OF CASPASES
ANTIAPOPTOTIC SIGNALING AND...
ROLE OF BCL-2 FAMILY...
ROLE OF NITRIC OXIDE
ROLE OF REACTIVE OXYGEN...
ROLE OF APOPTOTIC SIGNALING...
FUTURE DIRECTIONS
REFERENCES

Nitric oxide (NO) is a biological messenger molecule with profound influence on the vasculature. NO has been implicated in the regulation of vasomotor tone, inhibition of platelet aggregation and leukocyte adhesion to ECs, activation of transcription factors leading to new gene expression, and the regulation of apoptosis (140). In ECs, NO is generated by both a constitutive nitric oxide synthase and an inducible nitric oxide synthase (iNOS) (168). LPS upregulates the expression of iNOS and the production of NO in ECs (130, 156, 175, 179). These EC responses are further potentiated by LPS coadministration with other proinflammatory cytokines, including interferon-gamma (70, 208).

NO has been reported to inhibit LPS-induced EC apoptosis. ECs exposed to the NO donors S-nitroso-N-acetylpenicillamine or (Z)-1-[2-(2-aminoethyl)-N-(2-ammonioethyl)amino]diazen-1-ium-1,2-diolate demonstrate reduced sensitivity to LPS-induced apoptosis (51, 196). Enhanced production of endogenous NO after overexpression of iNOS has been similarly shown to block LPS-elicited EC apoptosis (31, 200). The mechanism by which NO inhibits LPS-induced EC apoptosis remains unclear. Increased levels of NO have been reported to suppress LPS-induced activation of caspase-3 in ECs (31, 200). In vitro studies using purified human recombinant caspases have shown that NO reversibly inhibits seven distinct members of the caspase family, including caspase-3, through direct S-nitrosylation of a cysteine residue required for caspase catalytic activity (55, 119, 136). Thus the protective effect of NO on LPS-exposed ECs may be mediated by NO inactivation of caspases.

In contrast to a cytoprotective role for NO, other studies have reported that suppression of iNOS induction and inhibitors of NOS activity protect against LPS-elicited EC injury (87, 160, 180), suggesting that NO promotes LPS-induced apoptosis. The differential effects of NO on mediating LPS-induced EC apoptosis appear to be dose dependent. DeMeester et al. (51) have reported that moderate levels of NO confer protection, whereas higher concentrations of NO enhance LPS-induced EC apoptosis. Although the mechanism by which NO potentiates LPS-elicited apoptosis remains unknown, high concentrations of NO can inhibit protein synthesis (214). Because protein synthesis inhibition has been established to sensitize human ECs to LPS-induced apoptosis by inhibiting the expression of the antiapoptotic protein FLIP (21), one may speculate that high levels of NO may result in decreased expression of this cytoprotective protein. Alternatively, increased production of NO after LPS induction of iNOS may result in NO reaction with superoxide anion to form peroxynitrite. EC injury resulting from the generation of peroxynitrite, a potent oxidizer, may synergistically enhance the EC apoptosis elicited by LPS. Further studies are needed to clarify the potential proapoptotic role of NO in mediating LPS-induced EC apoptosis.


    ROLE OF REACTIVE OXYGEN SPECIES
TOP
ABSTRACT
INTRODUCTION
EVIDENCE FOR LPS-INDUCED EC...
CLINICAL RELEVANCE AND IN...
CELL SURFACE RECOGNITION OF...
TLR-4 SIGNALING LEADING TO...
TLR-4 SIGNALING LEADING TO...
ROLE OF NF-kappa B ACTIVATION...
ROLE OF CASPASES
ANTIAPOPTOTIC SIGNALING AND...
ROLE OF BCL-2 FAMILY...
ROLE OF NITRIC OXIDE
ROLE OF REACTIVE OXYGEN...
ROLE OF APOPTOTIC SIGNALING...
FUTURE DIRECTIONS
REFERENCES

In response to stress and/or injury, ECs generate reactive oxygen species (ROS) and nitrogen intermediates (165). Generation of these free radicals can lead to an alteration in the balance of the pro- and antioxidant states, resulting in oxidative stress and potential cell injury (32). Increases in ROS or depletion of antioxidants can lead to apoptosis in a variety of cell types, including ECs (54, 68, 125, 134, 135). LPS elicits an increase in the generation of EC ROS (28, 117, 118, 177, 216). Interestingly, LPS-induced generation of ROS correlates with the onset of apoptosis, and agents that inhibit the formation of ROS confer protection against LPS-evoked EC apoptosis (1, 28, 117, 118, 150). Although the mechanism by which ROS contribute to LPS-induced EC apoptosis remains unknown, ROS have been shown to increase mitochondrial membrane permeability, resulting in the release of proapoptotic factors (10, 29, 63, 195, 224). One of these factors is cytochrome c, a cofactor that drives the assembly of the caspase-9-activating apoptosome (169). Furthermore, ROS have been reported to increase both the expression and activation of caspase-8 (151, 224) as well as the activation of caspase-3 (10, 224).

In addition to the generation of ROS, LPS induces the upregulation of manganese superoxide dismutase (MnSOD) (150, 206) and copper, zinc superoxide dismutase (Cu,Zn-SOD) (123). SODs serve as an important component of the EC antioxidant defense system by accelerating the conversion of superoxide to H2O2. Although both superoxide and H2O2 are potent oxidants, the latter reacts much more slowly with cellular substrates (165). LPS-induced upregulation of MnSOD is mediated, in part, by increased generation of ROS, thus SOD may act to counterbalance the proapoptotic effects of ROS (150). Consistent with this hypothesis, upregulation of SOD has been reported to inhibit apoptosis in a variety of cell types, including ECs (56, 120, 215). Furthermore, overexpression of Cu,Zn-SOD protects against reactive oxygen intermediates (ROI)-elicited induction of caspase-8, activation of caspase-3, and mitochondrial release of cytochrome c after ischemia (151, 191). Finally, overexpression of MnSOD decreases the sensitivity of ECs to LPS-induced apoptosis (31). Clearly, one possible mechanism by which SOD confers protection is by restricting the accumulation of potent oxidizing molecules that may promote apoptosis. Another mechanism may be through SOD's role in accelerating the conversion of superoxide to H2O2. Although H2O2 has been reported to induce apoptosis in some systems, others have reported that high levels of H2O2 inhibit caspase activation and apoptosis (25, 83, 131). Whether LPS-induced upregulation of SODs generates H2O2 at a level that will promote or inhibit LPS-evoked EC apoptosis remains unknown.

The role of ROS in mediating apoptosis is further complicated by ROS activation of signal transduction pathways that inhibit EC apoptosis (105, 165). ROS-activated pathways include: 1) the mitogen-activated protein kinases, which have been implicated in VEGF-induced EC survival (78), 2) Akt, a kinase involved in phosphoinositide 3-kinase signaling that is linked to shear stress and VEGF-induced survival (53, 66, 71), and 3) NF-kappa B, a transcription factor that upregulates the antiapoptotic proteins A1, A20, and IAPs (44, 99, 113, 183, 190, 209, 221). Whether LPS-induced generation of ROS activates these pathways resulting in resistance to LPS proapoptotic signaling remains unknown.


    ROLE OF APOPTOTIC SIGNALING MOLECULES IN MEDIATING LPS-INDUCED EC ACTIVATION
TOP
ABSTRACT
INTRODUCTION
EVIDENCE FOR LPS-INDUCED EC...
CLINICAL RELEVANCE AND IN...
CELL SURFACE RECOGNITION OF...
TLR-4 SIGNALING LEADING TO...
TLR-4 SIGNALING LEADING TO...
ROLE OF NF-kappa B ACTIVATION...
ROLE OF CASPASES
ANTIAPOPTOTIC SIGNALING AND...
ROLE OF BCL-2 FAMILY...
ROLE OF NITRIC OXIDE
ROLE OF REACTIVE OXYGEN...
ROLE OF APOPTOTIC SIGNALING...
FUTURE DIRECTIONS
REFERENCES

A key marker of EC activation is NF-kappa B activation and nuclear translocation, a requisite event for many EC responses, including increased expression of cytokines and adhesion molecules. Recent studies demonstrate that several signaling molecules originally described as mediators of apoptosis also contribute to the regulation of NF-kappa B activation (33, 98, 102, 103, 114, 184, 207). Evidence supporting this dual function for apoptotic signaling molecules includes: 1) enhanced caspase activation inhibits TNF-alpha -induced EC NF-kappa B nuclear translocation and VCAM-1 expression (157), 2) transient overexpression of FADD, FLIP, or caspase-8 augments basal levels of NF-kappa B activation (33, 98, 114), 3) overexpression of FADD upregulates monocyte chemotactic protein-1 and IL-8 expression, the transcription of which is mediated by NF-kappa B (184), and 4) FADD-deficient Jurkat cells display impaired activation of NF-kappa B after exposure to TNF-alpha , TNF-related apoptosis-inducing ligand (TRAIL), or Fas (207).

Similarly, evidence exists that apoptotic signaling molecules also influence the ability of LPS to induce NF-kappa B activation. LPS-insensitive human embryonic kidney 293T cells are sensitized to LPS-induced NF-kappa B activation on induced expression of Nod1, a cytoplasmic protein with structural homology to the apoptosis regulator, Apaf-1 (104). Furthermore, overexpression of Bcl-2 and Bcl-xL inhibits LPS-induced NF-kappa B activation and NF-kappa B-dependent gene expression in ECs (13). This inhibition of NF-kappa B activation corresponds with Bcl-2-mediated inhibition of Ikappa Balpha degradation. The mechanism by which Bcl family members inhibit LPS-induced NF-kappa B activation remains unknown. Identification of a caspase consensus site within Ikappa Balpha has led to speculation that caspases may be involved in its proteolysis (13). Thus Bcl-mediated inhibition of caspase activation may be expected to block Ikappa Balpha degradation and subsequent activation of NF-kappa B.

Further evidence for cross talk between apoptotic signaling molecules and LPS-induced NF-kappa B activation was provided in a recent report demonstrating that the proapoptotic adapter protein FADD downregulates LPS-induced NF-kappa B activation (20). In that study, overexpression of either full-length FADD or the DD of FADD in ECs blocked LPS-induced NF-kappa B activation. Furthermore, mouse embryo fibroblasts (MEF) derived from FADD-/- mice embryos displayed enhanced NF-kappa B activity relative to FADD+/+ MEF after LPS treatment. The production of IL-6 and KC, two NF-kappa B-dependent gene products, were similarly enhanced in FADD-/- MEF after LPS exposure relative to FADD+/+ MEF. Reconstitution of FADD in FADD-/- MEF abrogated the enhanced NF-kappa B activation and IL-6 and KC production elicited by LPS. Together, these data suggest that changes in FADD expression can affect LPS-induced activation of NF-kappa B. The role of FADD in inhibiting this event under basal conditions remains unclear as LPS activates EC NF-kappa B in the presence of physiological levels of FADD.

The mechanism by which FADD downregulates NF-kappa B-dependent gene expression remains unknown. However, the enhanced NF-kappa B activity in the FADD-/- MEF after LPS stimulation correlated with enhanced degradation of the inhibitor of NF-kappa B, Ikappa B, suggesting that FADD exerts its effect upstream of Ikappa B degradation. Interestingly, NF-kappa B activation by IL-1beta , a proinflammatory cytokine that shares the same intracellular signaling pathway leading to NF-kappa B activation as that of LPS, is similarly downregulated by FADD (20).

The data that FADD inhibition of LPS and IL-1beta -induced NF-kappa B activation occurs upstream of Ikappa B degradation and that FADD inhibits a signaling pathway shared by these two distinct stimuli suggests that FADD may exert its effect through inhibiting and/or sequestering signaling molecules involved in both LPS and IL-1beta -induced NF-kappa B activation. A proximal event in LPS and IL-1beta signaling is MyD88 recruitment of IRAK through reciprocal binding of the DD regions on each of these molecules (48, 194, 212) (Fig. 4). One possibility is that FADD, another DD-containing protein, may bind and/or sequester MyD88 and/or IRAK-1 through homophilic DD-DD interactions. In fact, overexpressed MyD88 reportedly binds FADD via DD-mediated interactions (5, 92). Whether this interaction occurs when these proteins are expressed at physiological levels is not known. It has been well established that both MyD88 and IRAK are required for optimal LPS-induced NF-kappa B activation and NF-kappa B-dependent gene expression (115, 194, 222). Thus any protein that can interfere with the recruitment of these molecules to the Tlr-4 receptor-signaling complex, perhaps through DD-DD interactions, would be expected to disrupt LPS-induced NF-kappa B signaling. FADD contains an additional protein-binding domain, the DED (Fig. 1), which facilitates recruitment of FLIP and caspase-8 (Fig. 4). FLIP and caspase-8 have been demonstrated to bind NIK and IKK in a DED-dependent manner (33). This raises another possibility that FADD itself may, through its DED, associate with and/or sequester NIK and IKK, two proteins involved in LPS activation of NF-kappa B. Alternatively, both FLIP and caspase-8 have been demonstrated to potentiate NF-kappa B activation, whereas caspase-8 D/N constructs inhibit NF-kappa B activation induced by a variety of death receptors, including TNFR1, Fas, DR3, DR4, and DR5 (33). These data suggest a role for these proteins in promoting NF-kappa B activation. Therefore, another mechanism by which FADD may inhibit LPS-induced NF-kappa B activation is through the sequestering of FLIP and/or caspase-8 (Fig. 4). Further studies are required to define the role(s) of these DED-containing molecules in mediating LPS signaling.


View larger version (29K):
[in this window]
[in a new window]
 
Fig. 4.   Cross talk between apoptotic and NF-kappa B signaling molecules. FADD downregulates LPS-induced NF-kappa B activation, and this inhibitory effect is exerted upstream of Ikappa B degradation. In overexpression studies, FADD has been demonstrated to bind MyD88 via DD-DD interactions. Whether FADD sequesters MyD88 and prevents its participation in LPS-elicited NF-kappa B signaling remains unknown. Another mode of action may be that FADD binds and sequesters the DD-containing protein IRAK. Alternatively, FLIP and caspase-8 have been reported to bind to NIK and IKK and to potentiate NF-kappa B signaling. The binding of the molecules is mediated, in part, by the DED regions of FLIP and caspase-8. Whether FADD can sequester NIK and IKK either through direct binding with these molecules via its own DED or indirectly through binding of FLIP and caspase-8 remains unknown.


    FUTURE DIRECTIONS
TOP
ABSTRACT
INTRODUCTION
EVIDENCE FOR LPS-INDUCED EC...
CLINICAL RELEVANCE AND IN...
CELL SURFACE RECOGNITION OF...
TLR-4 SIGNALING LEADING TO...
TLR-4 SIGNALING LEADING TO...
ROLE OF NF-kappa B ACTIVATION...
ROLE OF CASPASES
ANTIAPOPTOTIC SIGNALING AND...
ROLE OF BCL-2 FAMILY...
ROLE OF NITRIC OXIDE
ROLE OF REACTIVE OXYGEN...
ROLE OF APOPTOTIC SIGNALING...
FUTURE DIRECTIONS
REFERENCES

The unique position of the endothelium at the blood-extravascular tissue interface exposes ECs to an array of circulating mediators that may be injurious. EC injury and/or dysfunction contribute to the development of complications that often accompany gram-negative sepsis, including ARDS. The pathophysiological implications of EC involvement warrant investigation into the mechanisms of EC injury and/or death. LPS is a key proinflammatory mediator that contributes, at least in part, to the deleterious effects of gram-negative bacteremia and its attendant endotoxemia. LPS is a potent activator of the vascular endothelium and elicits an array of EC responses, including apoptosis. Key areas of future study include: 1) identifying the adapter proteins that link the Tlr-4 signaling molecules to the recruitment and activation of caspases (Fig. 2), 2) determining whether FLIP and Bcl-2 homologs participate in redundant cytoprotective pathways and identifying the mechanism by which these molecules confer EC protection against LPS-induced apoptosis, 3) evaluating the cross talk between apoptotic signaling molecules and NF-kappa B activation with the goal of devising a common therapeutic intervention that could dampen an excessive host inflammatory response and simultaneously protect against host tissue injury (Fig. 4), 4) investigating the synergistic effect that endogenous proinflammatory mediators may have with LPS and one another on promoting EC apoptosis, and 5) extending the promising though limited research evaluating the efficacy of caspase inhibitors in sepsis models.


    FOOTNOTES

Address for reprint requests and other correspondence: D. D. Bannerman, Immunology and Disease Resistance Laboratory, United States Dept. of Agriculture-Agricultural Research Service/ANRI, BARC-East, Bldg. 1040, Rm. #2, Beltsville, MD 20705-2350 (E-mail: dbanner{at}anri.barc.usda.gov).

The costs of publication of this article were defrayed in part by the payment of page charges. The article must therefore be hereby marked "advertisement" in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

10.1152/ajplung.00338.2002


    REFERENCES
TOP
ABSTRACT
INTRODUCTION
EVIDENCE FOR LPS-INDUCED EC...
CLINICAL RELEVANCE AND IN...
CELL SURFACE RECOGNITION OF...
TLR-4 SIGNALING LEADING TO...
TLR-4 SIGNALING LEADING TO...
ROLE OF NF-kappa B ACTIVATION...
ROLE OF CASPASES
ANTIAPOPTOTIC SIGNALING AND...
ROLE OF BCL-2 FAMILY...
ROLE OF NITRIC OXIDE
ROLE OF REACTIVE OXYGEN...
ROLE OF APOPTOTIC SIGNALING...
FUTURE DIRECTIONS
REFERENCES

1.   Abello, PA, Fidler SA, Bulkley GB, and Buchman TG. Antioxidants modulate induction of programmed endothelial cell death (apoptosis) by endotoxin. Arch Surg 129: 134-140, 1994[Abstract].

2.   Adams, JM, and Cory S. The Bcl-2 protein family: arbiters of cell survival. Science 281: 1322-1326, 1998[Abstract/Free Full Text].

3.   Akashi, S, Ogata H, Kirikae F, Kirikae T, Kawasaki K, Nishijima M, Shimazu R, Nagai Y, Fukudome K, Kimoto M, and Miyake K. Regulatory roles for CD14 and phosphatidylinositol in the signaling via Toll-like receptor 4-MD-2. Biochem Biophys Res Commun 268: 172-177, 2000[ISI][Medline].

4.   Aliprantis, AO, Yang RB, Mark MR, Suggett S, Devaux B, Radolf JD, Klimpel GR, Godowski P, and Zychlinsky A. Cell activation and apoptosis by bacterial lipoproteins through Toll-like receptor-2. Science 285: 736-739, 1999[Abstract/Free Full Text].

5.   Aliprantis, AO, Yang RB, Weiss DS, Godowski P, and Zychlinsky A. The apoptotic signaling pathway activated by Toll-like receptor-2. EMBO J 19: 3325-3336, 2000[Abstract/Free Full Text].

6.   Alpert, G, Baldwin G, Thompson C, Wainwright N, Novitsky TJ, Gillis Z, Parsonnet J, Fleisher GR, and Siber GR. Limulus antilipopolysaccharide factor protects rabbits from meningococcal endotoxin shock. J Infect Dis 165: 494-500, 1992[ISI][Medline].

7.   Anrather, J, Csizmadia V, Brostjan C, Soares MP, Bach FH, and Winkler H. Inhibition of bovine endothelial cell activation in vitro by regulated expression of a transdominant inhibitor of NF-kappa B. J Clin Invest 99: 763-772, 1997[Abstract/Free Full Text].

8.   Antonsson, B, and Martinou JC. The Bcl-2 protein family. Exp Cell Res 256: 50-57, 2000[ISI][Medline].

9.   Arditi, M, Zhou J, Dorio R, Rong GW, Goyert SM, and Kim KS. Endotoxin-mediated endothelial cell injury and activation: role of soluble CD14. Infect Immun 61: 3149-3156, 1993[Abstract].

10.   Armstrong, JS, Steinauer KK, Hornung B, Irish JM, Lecane P, Birrell GW, Peehl DM, and Knox SJ. Role of glutathione depletion and reactive oxygen species generation in apoptotic signaling in a human B lymphoma cell line. Cell Death Differ 9: 252-263, 2002[ISI][Medline].

11.   Ashkenazi, A, and Dixit VM. Death receptors: signaling and modulation. Science 281: 1305-1308, 1998[Abstract/Free Full Text].

12.   Assaly, R, Olson D, Hammersley J, Fan PS, Liu J, Shapiro JI, and Kahaleh MB. Initial evidence of endothelial cell apoptosis as a mechanism of systemic capillary leak syndrome. Chest 120: 1301-1308, 2001[Abstract/Free Full Text].

13.   Badrichani, AZ, Stroka DM, Bilbao G, Curiel DT, Bach FH, and Ferran C. Bcl-2 and Bcl-XL serve an anti-inflammatory function in endothelial cells through inhibition of NF-kappa B. J Clin Invest 103: 543-553, 1999[Abstract/Free Full Text].

14.   Balis, JU, Gerber LI, Rappaport ES, and Neville WE. Mechanisms of blood-vascular reactions of the primate lung to acute endotoxemia. Exp Mol Pathol 21: 123-137, 1974[ISI][Medline].

15.   Bannerman, DD, Erwert RD, Winn RK, and Harlan JM. TIRAP mediates endotoxin-induced NF-kappa B activation and apoptosis in endothelial cells. Biochem Biophys Res Commun 295: 157-162, 2002[ISI][Medline].

16.   Bannerman, DD, Fitzpatrick MJ, Anderson DY, Bhattacharjee AK, Novitsky TJ, Hasday JD, Cross AS, and Goldblum SE. Endotoxin-neutralizing protein protects against endotoxin-induced endothelial barrier dysfunction. Infect Immun 66: 1400-1407, 1998[Abstract/Free Full Text].

17.   Bannerman, DD, and Goldblum SE. Direct effects of endotoxin on the endothelium: barrier function and injury. Lab Invest 79: 1181-1199, 1999[ISI][Medline].

18.   Bannerman, DD, Sathyamoorthy M, and Goldblum SE. Bacterial lipopolysaccharide disrupts endothelial monolayer integrity and survival signaling events through caspase cleavage of adherens junction proteins. J Biol Chem 273: 35371-35380, 1998[Abstract/Free Full Text].

19.   Bannerman, DD, Tupper JC, Erwert RD, Winn RK, and Harlan JM. Divergence of bacterial lipopolysaccharide pro-apoptotic signaling downstream of IRAK-1. J Biol Chem 277: 8048-8053, 2002[Abstract/Free Full Text].

20.   Bannerman, DD, Tupper JC, Kelly JD, Winn RK, and Harlan JM. The Fas-associated death domain protein suppresses activation of NF-kappa B by LPS and IL-1beta . J Clin Invest 109: 419-425, 2002[Abstract/Free Full Text].

21.   Bannerman, DD, Tupper JC, Ricketts WA, Bennett CF, Winn RK, and Harlan JM. A constitutive cytoprotective pathway protects endothelial cells from lipopolysaccharide-induced apoptosis. J Biol Chem 276: 14924-14932, 2001[Abstract/Free Full Text].

22.   Beal, AL, and Cerra FB. Multiple organ failure syndrome in the 1990s. Systemic inflammatory response and organ dysfunction. JAMA 271: 226-233, 1994[Abstract].

23.   Bell, RC, Coalson JJ, Smith JD, and Johanson WG, Jr. Multiple organ system failure and infection in adult respiratory distress syndrome. Ann Intern Med 99: 293-298, 1983[ISI][Medline].

24.   Bodmer, JL, Holler N, Reynard S, Vinciguerra P, Schneider P, Juo P, Blenis J, and Tschopp J. TRAIL receptor-2 signals apoptosis through FADD and caspase-8. Nat Cell Biol 2: 241-243, 2000[ISI][Medline].

25.   Borutaite, V, and Brown GC. Caspases are reversibly inactivated by hydrogen peroxide. FEBS Lett 500: 114-118, 2001[ISI][Medline].

26.   Brandtzaeg, P, Kierulf P, Gaustad P, Skulberg A, Bruun JN, Halvorsen S, and Sorensen E. Plasma endotoxin as a predictor of multiple organ failure and death in systemic meningococcal disease. J Infect Dis 159: 195-204, 1989[ISI][Medline].

27.   Brigham, KL, and Meyrick B. Endotoxin and lung injury. Am Rev Respir Dis 133: 913-927, 1986[ISI][Medline].

28.   Brigham, KL, Meyrick B, Berry LC, Jr, and Repine JE. Antioxidants protect cultured bovine lung endothelial cells from injury by endotoxin. J Appl Physiol 63: 840-850, 1987[Abstract/Free Full Text].

29.   Brown, GC, and Borutaite V. Nitric oxide, cytochrome c and mitochondria. Biochem Soc Symp 66: 17-25, 1999[Medline].

30.   Cai, W, Devaux B, Schaper W, and Schaper J. The role of Fas/APO 1 and apoptosis in the development of human atherosclerotic lesions. Atherosclerosis 131: 177-186, 1997[ISI][Medline].

31.   Ceneviva, GD, Tzeng E, Hoyt DG, Yee E, Gallagher A, Engelhardt JF, Kim YM, Billiar TR, Watkins SA, and Pitt BR. Nitric oxide inhibits lipopolysaccharide-induced apoptosis in pulmonary artery endothelial cells. Am J Physiol Lung Cell Mol Physiol 275: L717-L728, 1998[Abstract/Free Full Text].

32.   Chandra, J, Samali A, and Orrenius S. Triggering and modulation of apoptosis by oxidative stress. Free Radic Biol Med 29: 323-333, 2000[ISI][Medline].

33.   Chaudhary, PM, Eby MT, Jasmin A, Kumar A, Liu L, and Hood L. Activation of the NF-kappa B pathway by caspase-8 and its homologs. Oncogene 19: 4451-4460, 2000[ISI][Medline].

34.   Chen, CC, Rosenbloom CL, Anderson DC, and Manning AM. Selective inhibition of E-selectin, vascular cell adhesion molecule-1, and intercellular adhesion molecule-1 expression by inhibitors of Ikappa B-alpha phosphorylation. J Immunol 155: 3538-3545, 1995[Abstract].

35.   Chinnaiyan, AM, Tepper CG, Seldin MF, O'Rourke K, Kischkel FC, Hellbardt S, Krammer PH, Peter ME, and Dixit VM. FADD/MORT1 is a common mediator of CD95 (Fas/APO-1) and tumor necrosis factor receptor-induced apoptosis. J Biol Chem 271: 4961-4965, 1996[Abstract/Free Full Text].

36.   Choi, KB, Wong F, Harlan JM, Chaudhary PM, Hood L, and Karsan A. Lipopolysaccharide mediates endothelial apoptosis by a FADD-dependent pathway. J Biol Chem 273: 20185-20188, 1998[Abstract/Free Full Text].

37.   Chow, JC, Young DW, Golenbock DT, Christ WJ, and Gusovsky F. Toll-like receptor-4 mediates lipopolysaccharide-induced signal transduction. J Biol Chem 274: 10689-10692, 1999[Abstract/Free Full Text].

38.   Cines, DB, Pollak ES, Buck CA, Loscalzo J, Zimmerman GA, McEver RP, Pober JS, Wick TM, Konkle BA, Schwartz BS, Barnathan ES, McCrae KR, Hug BA, Schmidt AM, and Stern DM. Endothelial cells in physiology and in the pathophysiology of vascular disorders. Blood 91: 3527-3561, 1998[Free Full Text].

39.   Clemens, MJ, McNurlan MA, Moore G, and Tilleray VJ. Regulation of protein synthesis in lymphoblastoid cells during inhibition of cell proliferation by human interferons. FEBS Lett 171: 111-116, 1984[ISI][Medline].

40.   Coalson, JJ, Benjamin BA, Archer LT, Beller BK, Spaet RH, and Hinshaw LB. A pathologic study of Escherichia coli shock in the baboon and the response to adrenocorticosteroid treatment. Surg Gynecol Obstet 147: 726-736, 1978[ISI][Medline].

41.   Coalson, JJ, Hinshaw LB, Guenter CA, Berrell EL, and Greenfield LJ. Pathophysiologic responses of the subhuman primate in experimental septic shock. Lab Invest 32: 561-569, 1975[Medline].

42.   Colucci, M, Balconi G, Lorenzet R, Pietra A, Locati D, Donati MB, and Semeraro N. Cultured human endothelial cells generate tissue factor in response to endotoxin. J Clin Invest 71: 1893-1896, 1983[ISI][Medline].

43.   Colussi, PA, and Kumar S. Targeted disruption of caspase genes in mice: what they tell us about the functions of individual caspases in apoptosis. Immunol Cell Biol 77: 58-63, 1999[ISI][Medline].

44.   Cooper, JT, Stroka DM, Brostjan C, Palmetshofer A, Bach FH, and Ferran C. A20 blocks endothelial cell activation through a NF-kappa B-dependent mechanism. J Biol Chem 271: 18068-18073, 1996[Abstract/Free Full Text].

45.   Cox, G, Crossley J, and Xing Z. Macrophage engulfment of apoptotic neutrophils contributes to the resolution of acute pulmonary inflammation in vivo. Am J Respir Cell Mol Biol 12: 232-237, 1995[Abstract].

46.   Curzen, NP, Griffiths MJ, and Evans TW. Role of the endothelium in modulating the vascular response to sepsis. Clin Sci (Lond) 86: 359-374, 1994[ISI][Medline].

47.   Cybulsky, MI, Chan MK, and Movat HZ. Acute inflammation and microthrombosis induced by endotoxin, interleukin-1, and tumor necrosis factor and their implication in gram-negative infection. Lab Invest 58: 365-378, 1988[ISI][Medline].

48.   Daun, JM, and Fenton MJ. Interleukin-1/Toll receptor family members: receptor structure and signal transduction pathways. J Interferon Cytokine Res 20: 843-855, 2000[ISI][Medline].

49.   Deaciuc, IV, Fortunato F, D'Souza NB, Hill DB, Schmidt J, Lee EY, and McClain CJ. Modulation of caspase-3 activity and Fas ligand mRNA expression in rat liver cells in vivo by alcohol and lipopolysaccharide. Alcohol Clin Exp Res 23: 349-356, 1999[ISI][Medline].

50.   Delvos, U, Janssen B, and Muller-Berghaus G. Effect of lipopolysaccharides on cultured human endothelial cells. Relationship between tissue factor activity and prostacyclin release. Blut 55: 101-108, 1987[ISI][Medline].

51.   DeMeester, SL, Qiu Y, Buchman TG, Hotchkiss RS, Dunnigan K, Karl IE, and Cobb JP. Nitric oxide inhibits stress-induced endothelial cell apoptosis. Crit Care Med 26: 1500-1509, 1998[ISI][Medline].

52.   Desch, CE, O'Hara P, and Harlan JM. Antilipopolysaccharide factor from horseshoe crab, Tachypleus tridentatus, inhibits lipopolysaccharide activation of cultured human endothelial cells. Infect Immun 57: 1612-1614, 1989[ISI][Medline].

53.   Dimmeler, S, Assmus B, Hermann C, Haendeler J, and Zeiher AM. Fluid shear stress stimulates phosphorylation of Akt in human endothelial cells: involvement in suppression of apoptosis. Circ Res 83: 334-341, 1998[Abstract/Free Full Text].

54.   Dimmeler, S, Haendeler J, Galle J, and Zeiher AM. Oxidized low-density lipoprotein induces apoptosis of human endothelial cells by activation of CPP32-like proteases. A mechanistic clue to the 'response to injury' hypothesis. Circulation 95: 1760-1763, 1997[Abstract/Free Full Text].

55.   Dimmeler, S, Haendeler J, Nehls M, and Zeiher AM. Suppression of apoptosis by nitric oxide via inhibition of interleukin-1beta -converting enzyme (ICE)-like and cysteine protease protein (CPP)-32-like proteases. J Exp Med 185: 601-607, 1997[Abstract/Free Full Text].

56.   Dimmeler, S, Hermann C, Galle J, and Zeiher AM. Upregulation of superoxide dismutase and nitric oxide synthase mediates the apoptosis-suppressive effects of shear stress on endothelial cells. Arterioscler Thromb Vasc Biol 19: 656-664, 1999[Abstract/Free Full Text].

57.   Dinarello, CA. Proinflammatory and anti-inflammatory cytokines as mediators in the pathogenesis of septic shock. Chest 112: 321S-329S, 1997[Free Full Text].

58.   Dixit, VM, Green S, Sarma V, Holzman LB, Wolf FW, O'Rourke K, Ward PA, Prochownik EV, and Marks RM. Tumor necrosis factor-alpha induction of novel gene products in human endothelial cells including a macrophage-specific chemotaxin. J Biol Chem 265: 2973-2978, 1990[Abstract/Free Full Text].

59.   Engelberts, I, van Hoof SC, Samyo SK, Buurman WA, and van der Linden CJ. Generalized inflammation during peritonitis evidenced by intracutaneous E-selectin expression. Clin Immunol Immunopathol 65: 330-334, 1992[ISI][Medline].

60.   Erwert, RD, Winn RK, Harlan JM, and Bannerman DD. Shiga-like toxin inhibition of FLICE-like inhibitory protein expression sensitizes endothelial cells to bacterial lipopolysaccharide-induced apoptosis. J Biol Chem 277: 40567-40574, 2002[Abstract/Free Full Text].

61.   Faure, E, Equils O, Sieling PA, Thomas L, Zhang FX, Kirschning CJ, Polentarutti N, Muzio M, and Arditi M. Bacterial lipopolysaccharide activates NF-kappa B through Toll-like receptor-4 (TLR-4) in cultured human dermal endothelial cells. Differential expression of tlr-4 and tlr-2 in endothelial cells. J Biol Chem 275: 11058-11063, 2000[Abstract/Free Full Text].

62.   Fitzgerald, KA, Palsson-McDermott EM, Bowie AG, Jefferies CA, Mansell AS, Brady G, Brint E, Dunne A, Gray P, Harte MT, McMurray D, Smith DE, Sims JE, Bird TA, and O'Neill LA. Mal (MyD88-adapter-like) is required for Toll-like receptor-4 signal transduction. Nature 413: 78-83, 2001[ISI][Medline].

63.   Fleury, C, Mignotte B, and Vayssiere JL. Mitochondrial reactive oxygen species in cell death signaling. Biochimie 84: 131-141, 2002[ISI][Medline].

64.   Frey, EA, and Finlay BB. Lipopolysaccharide induces apoptosis in a bovine endothelial cell line via a soluble CD14-dependent pathway. Microb Pathog 24: 101-109, 1998[ISI][Medline].

65.   Fry, DE, Pearlstein L, Fulton RL, and Polk HC, Jr. Multiple system organ failure. The role of uncontrolled infection. Arch Surg 115: 136-140, 1980[Abstract].

66.   Fujio, Y, and Walsh K. Akt mediates cytoprotection of endothelial cells by vascular endothelial growth factor in an anchorage-dependent manner. J Biol Chem 274: 16349-16354, 1999[Abstract/Free Full Text].

67.   Fujita, M, Kuwano K, Kunitake R, Hagimoto N, Miyazaki H, Kaneko Y, Kawasaki M, Maeyama T, and Hara N. Endothelial cell apoptosis in lipopolysaccharide-induced lung injury in mice. Int Arch Allergy Immunol 117: 202-208, 1998[ISI][Medline].

68.   Galle, J, Heermeier K, and Wanner C. Atherogenic lipoproteins, oxidative stress, and cell death. Kidney Int Suppl 71: S62-S65, 1999[Medline].

69.   Gaynor, E, Bouvier C, and Spaet TH. Vascular lesions: possible pathogenetic basis of the generalized Shwartzman reaction. Science 170: 986-988, 1970[ISI][Medline].

70.   Geiger, M, Stone A, Mason SN, Oldham KT, and Guice KS. Differential nitric oxide production by microvascular and macrovascular endothelial cells. Am J Physiol Lung Cell Mol Physiol 273: L275-L281, 1997[Abstract/Free Full Text].

71.   Gerber, HP, McMurtrey A, Kowalski J, Yan M, Keyt BA, Dixit V, and Ferrara N. Vascular endothelial growth factor regulates endothelial cell survival through the phosphatidylinositol 3'-kinase/Akt signal transduction pathway. Requirement for Flk-1/KDR activation. J Biol Chem 273: 30336-30343, 1998[Abstract/Free Full Text].

72.   Gerrity, RG, Richardson M, Caplan BA, Cade JF, Hirsh J, and Schwartz CJ. Endotoxin-induced vascular endothelial injury and repair. II. Focal injury, en face morphology, [3H]thymidine uptake and circulating endothelial cells in the dog. Exp Mol Pathol 24: 59-69, 1976[ISI][Medline].

73.   Gobe, G, Browning J, Howard T, Hogg N, Winterford C, and Cross R. Apoptosis occurs in endothelial cells during hypertension-induced microvascular rarefaction. J Struct Biol 118: 63-72, 1997[ISI][Medline].

74.   Goldblum, SE, Brann TW, Ding X, Pugin J, and Tobias PS. Lipopolysaccharide (LPS)-binding protein and soluble CD14 function as accessory molecules for LPS-induced changes in endothelial barrier function, in vitro. J Clin Invest 93: 692-702, 1994[ISI][Medline].

75.   Golenbock, DT, Bach RR, Lichenstein H, Juan TS, Tadavarthy A, and Moldow CF. Soluble CD14 promotes LPS activation of CD14-deficient PNH monocytes and endothelial cells. J Lab Clin Med 125: 662-671, 1995[ISI][Medline].

76.   Granville, DJ, Carthy CM, Hunt DW, and McManus BM. Apoptosis: molecular aspects of cell death and disease. Lab Invest 78: 893-913, 1998[ISI][Medline].

77.   Grau, GE, Mili N, Lou JN, Morel DR, Ricou B, Lucas R, and Suter PM. Phenotypic and functional analysis of pulmonary microvascular endothelial cells from patients with acute respiratory distress syndrome. Lab Invest 74: 761-770, 1996[ISI][Medline].

78.   Gupta, K, Kshirsagar S, Li W, Gui L, Ramakrishnan S, Gupta P, Law PY, and Hebbel RP. VEGF prevents apoptosis of human microvascular endothelial cells via opposing effects on MAPK/ERK and SAPK/JNK signaling. Exp Cell Res 247: 495-504, 1999[ISI][Medline].

79.   Gyles, CL. Escherichia coli cytotoxins and enterotoxins. Can J Microbiol 38: 734-746, 1992[ISI][Medline].

80.   Hack, CE, and Zeerleder S. The endothelium in sepsis: source of and a target for inflammation. Crit Care Med 29: S21-S27, 2001[ISI][Medline].

81.   Haendeler, J, Zeiher AM, and Dimmeler S. Vitamin C and E prevent lipopolysaccharide-induced apoptosis in human endothelial cells by modulation of Bcl-2 and Bax. Eur J Pharmacol 317: 407-411, 1996[ISI][Medline].

82.   Haimovitz-Friedman, A, Cordon-Cardo C, Bayoumy S, Garzotto M, McLoughlin M, Gallily R, Edwards CK, III, Schuchman EH, Fuks Z, and Kolesnick R. Lipopolysaccharide induces disseminated endothelial apoptosis requiring ceramide generation. J Exp Med 186: 1831-1841, 1997[Abstract/Free Full Text].

83.   Hampton, MB, and Orrenius S. Dual regulation of caspase activity by hydrogen peroxide: implications for apoptosis. FEBS Lett 414: 552-556, 1997[ISI][Medline].

84.   Harlan, JM, Harker LA, Reidy MA, Gajdusek CM, Schwartz SM, and Striker GE. Lipopolysaccharide-mediated bovine endothelial cell injury in vitro. Lab Invest 48: 269-274, 1983[ISI][Medline].

85.   Hawker, FH. How to feed patients with sepsis. Curr Opin Crit Care 6: 247-252, 2000[Medline].

86.   Heimberg, H, Heremans Y, Jobin C, Leemans R, Cardozo AK, Darville M, and Eizirik DL. Inhibition of cytokine-induced NF-kappa B activation by adenovirus-mediated expression of a NF-kappa B super-repressor prevents beta -cell apoptosis. Diabetes 50: 2219-2224, 2001[Abstract/Free Full Text].

87.   Higaki, A, Ninomiya H, Saji M, Maki H, Koike T, and Ohno K. Protective effect of neurotropin against lipopolysaccharide-induced hypotension and lethality linked to suppression of inducible nitric oxide synthase induction. Jpn J Pharmacol 86: 329-335, 2001[ISI][Medline].

88.   Hinshaw, LB. Sepsis/septic shock: participation of the microcirculation: an abbreviated review. Crit Care Med 24: 1072-1078, 1996[ISI][Medline].

89.   Hirata, H, Takahashi A, Kobayashi S, Yonehara S, Sawai H, Okazaki T, Yamamoto K, and Sasada M. Caspases are activated in a branched protease cascade and control distinct downstream processes in Fas-induced apoptosis. J Exp Med 187: 587-600, 1998[Abstract/Free Full Text].

90.   Hirschfeld, M, Ma Y, Weis JH, Vogel SN, and Weis JJ. Cutting edge: repurification of lipopolysaccharide eliminates signaling through both human and murine Toll-like receptor 2. J Immunol 165: 618-622, 2000[Abstract/Free Full Text].

91.   Horng, T, Barton GM, and Medzhitov R. TIRAP: an adapter molecule in the Toll signaling pathway. Nat Immun 2: 835-841, 2001[ISI].

92.   Horng, T, and Medzhitov R. Drosophila MyD88 is an adapter in the Toll signaling pathway. Proc Natl Acad Sci USA 98: 12654-12658, 2001[Abstract/Free Full Text].

93.   Hotchkiss, RS, Swanson PE, Cobb JP, Jacobson A, Buchman TG, and Karl IE. Apoptosis in lymphoid and parenchymal cells during sepsis: findings in normal and T- and B-cell-deficient mice. Crit Care Med 25: 1298-1307, 1997[ISI][Medline].

94.   Hoyt, DG, Mannix RJ, Gerritsen ME, Watkins SC, Lazo JS, and Pitt BR. Integrins inhibit LPS-induced DNA strand breakage in cultured lung endothelial cells. Am J Physiol Lung Cell Mol Physiol 270: L689-L694, 1996[Abstract/Free Full Text].

95.   Hoyt, DG, Mannix RJ, Rusnak JM, Pitt BR, and Lazo JS. Collagen is a survival factor against LPS-induced apoptosis in cultured sheep pulmonary artery endothelial cells. Am J Physiol Lung Cell Mol Physiol 269: L171-L177, 1995[Abstract/Free Full Text].

96.   Hsu, H, Shu HB, Pan MG, and Goeddel DV. TRADD-TRAF2 and TRADD-FADD interactions define two distinct TNF receptor 1 signal transduction pathways. Cell 84: 299-308, 1996[ISI][Medline].

97.   Hu, S, Vincenz C, Ni J, Gentz R, and Dixit VM. I-FLICE, a novel inhibitor of tumor necrosis factor receptor-1- and CD95-induced apoptosis. J Biol Chem 272: 17255-17257, 1997[Abstract/Free Full Text].

98.   Hu, WH, Johnson H, and Shu HB. Activation of NF-kappa B by FADD, Casper, and caspase-8. J Biol Chem 275: 10838-10844, 2000[Abstract/Free Full Text].

99.   Hu, X, Yee E, Harlan JM, Wong F, and Karsan A. Lipopolysaccharide induces the antiapoptotic molecules, A1 and A20, in microvascular endothelial cells. Blood 92: 2759-2765, 1998[Abstract/Free Full Text].

100.   Hull, C, McLean G, Wong F, Duriez PJ, and Karsan A. Lipopolysaccharide signals an endothelial apoptosis pathway through TNF receptor-associated factor 6-mediated activation of c-Jun NH2-terminal kinase. J Immunol 169: 2611-2618, 2002[Abstract/Free Full Text].

101.   Iglewski, BH, and Kabat D. NAD-dependent inhibition of protein synthesis by Pseudomonas aeruginosa toxin. Proc Natl Acad Sci USA 72: 2284-2288, 1975[Abstract].

102.   Inohara, N, Koseki T, del Peso L, Hu Y, Yee C, Chen S, Carrio R, Merino J, Liu D, Ni J, and Nunez G. Nod1, an Apaf-1-like activator of caspase-9 and nuclear factor-kappa B. J Biol Chem 274: 14560-14567, 1999[Abstract/Free Full Text].

103.   Inohara, N, Koseki T, Lin J, del Peso L, Lucas PC, Chen FF, Ogura Y, and Nunez G. An induced proximity model for NF-kappa B activation in the Nod1/RICK and RIP signaling pathways. J Biol Chem 275: 27823-27831, 2000[Abstract/Free Full Text].

104.   Inohara, N, Ogura Y, Chen FF, Muto A, and Nunez G. Human Nod1 confers responsiveness to bacterial lipopolysaccharides. J Biol Chem 276: 2551-2554, 2001[Abstract/Free Full Text].

105.   Irani, K. Oxidant signaling in vascular cell growth, death, and survival: a review of the roles of reactive oxygen species in smooth muscle and endothelial cell mitogenic and apoptotic signaling. Circ Res 87: 179-183, 2000[Abstract/Free Full Text].

106.   Irmler, M, Thome M, Hahne M, Schneider P, Hofmann K, Steiner V, Bodmer JL, Schroter M, Burns K, Mattmann C, Rimoldi D, French LE, and Tschopp J. Inhibition of death receptor signals by cellular FLIP. Nature 388: 190-195, 1997[ISI][Medline].

107.   Janicke, RU, Sprengart ML, Wati MR, and Porter AG. Caspase-3 is required for DNA fragmentation and morphological changes associated with apoptosis. J Biol Chem 273: 9357-9360, 1998[Abstract/Free Full Text].

108.   Jersmann, HP, Hii CS, Ferrante JV, and Ferrante A. Bacterial lipopolysaccharide and tumor necrosis factor-alpha synergistically increase expression of human endothelial adhesion molecules through activation of NF-kappa B and p38 mitogen-activated protein kinase signaling pathways. Infect Immun 69: 1273-1279, 2001[Abstract/Free Full Text].

109.   Jirik, FR, Podor TJ, Hirano T, Kishimoto T, Loskutoff DJ, Carson DA, and Lotz M. Bacterial lipopolysaccharide and inflammatory mediators augment IL-6 secretion by human endothelial cells. J Immunol 142: 144-147, 1989[Abstract/Free Full Text].

110.   Karahashi, H, and Amano F. Lipopolysaccharide (LPS)-induced cell death of C3H mouse peritoneal macrophages in the presence of cycloheximide: different susceptibilities of C3H/HeN and C3H/HeJ mice macrophages. J Endotoxin Res 6: 33-39, 2000[ISI][Medline].

111.   Karahashi, H, and Amano F. Changes of caspase activities involved in apoptosis of a macrophage-like cell line J774.1/JA-4 treated with lipopolysaccharide (LPS) and cycloheximide. Biol Pharm Bull 23: 140-144, 2000[ISI][Medline].

112.   Karsan, A, Yee E, and Harlan JM. Endothelial cell death induced by tumor necrosis factor-alpha is inhibited by the Bcl-2 family member, A1. J Biol Chem 271: 27201-27204, 1996[Abstract/Free Full Text].

113.   Karsan, A, Yee E, Kaushansky K, and Harlan JM. Cloning of human Bcl-2 homologue: inflammatory cytokines induce human A1 in cultured endothelial cells. Blood 87: 3089-3096, 1996[Abstract/Free Full Text].

114.   Kataoka, T, Budd RC, Holler N, Thome M, Martinon F, Irmler M, Burns K, Hahne M, Kennedy N, Kovacsovics M, and Tschopp J. The caspase-8 inhibitor FLIP promotes activation of NF-kappa B and Erk signaling pathways. Curr Biol 10: 640-648, 2000[ISI][Medline].

115.   Kawai, T, Adachi O, Ogawa T, Takeda K, and Akira S. Unresponsiveness of MyD88-deficient mice to endotoxin. Immunity 11: 115-122, 1999[ISI][Medline].

116.   Kawasaki, M, Kuwano K, Hagimoto N, Matsuba T, Kunitake R, Tanaka T, Maeyama T, and Hara N. Protection from lethal apoptosis in lipopolysaccharide-induced acute lung injury in mice by a caspase inhibitor. Am J Pathol 157: 597-603, 2000[Abstract/Free Full Text].

117.   Kim, KY, Kim BG, Kim SO, Yoo SE, Kwak YG, Chae SW, and Hong KW. Prevention of lipopolysaccharide-induced apoptosis by (2S,3S,4R)-N"-cyano-N-(6-amino-3,4-dihydro-3-hydroxy-2-methyl-2-dimethoxym ethyl-2H-benzopyran-4-yl)-N'-benzylguanidine, a benzopyran analog, in endothelial cells. J Pharmacol Exp Ther 300: 535-542, 2002[Abstract/Free Full Text].

118.   Kim, KY, Shin HK, Choi JM, and Hong KW. Inhibition of lipopolysaccharide-induced apoptosis by cilostazol in human umbilical vein endothelial cells. J Pharmacol Exp Ther 300: 709-715, 2002[Abstract/Free Full Text].

119.   Kim, YM, Talanian RV, and Billiar TR. Nitric oxide inhibits apoptosis by preventing increases in caspase-3-like activity via two distinct mechanisms. J Biol Chem 272: 31138-31148, 1997[Abstract/Free Full Text].

120.   Kiningham, KK, Oberley TD, Lin S, Mattingly CA, and St Clair DK. Overexpression of manganese superoxide dismutase protects against mitochondrial-initiated poly(ADP-ribose) polymerase-mediated cell death. FASEB J 13: 1601-1610, 1999[Abstract/Free Full Text].

121.   Kirschning, CJ, Wesche H, Merrill Ayres T, and Rothe M. Human Toll-like receptor 2 confers responsiveness to bacterial lipopolysaccharide. J Exp Med 188: 2091-2097, 1998[Abstract/Free Full Text].

122.   Koide, N, Abe K, Narita K, Kato Y, Sugiyama T, Jiang GZ, and Yokochi T. Apoptotic cell death of vascular endothelial cells and renal tubular cells in the generalized Shwartzman reaction. FEMS Immunol Med Microbiol 16: 205-211, 1996[ISI][Medline].

123.   Kong, XJ, and Fanburg BL. Regulation of Cu,Zn-superoxide dismutase in bovine pulmonary artery endothelial cells. J Cell Physiol 153: 491-497, 1992[ISI][Medline].

124.   Koshi, R, Mathan VI, David S, and Mathan MM. Enteric vascular endothelial response to bacterial endotoxin. Int J Exp Pathol 74: 593-601, 1993[ISI][Medline].

125.   Kotamraju, S, Chitambar CR, Kalivendi SV, Joseph J, and Kalyanaraman B. Transferrin receptor-dependent iron uptake is responsible for doxorubicin-mediated apoptosis in endothelial cells: role of oxidant-induced iron signaling in apoptosis. J Biol Chem 277: 17179-17187, 2002[Abstract/Free Full Text].

126.   Kuang, AA, Diehl GE, Zhang J, and Winoto A. FADD is required for DR4- and DR5-mediated apoptosis: lack of trail-induced apoptosis in FADD-deficient mouse embryonic fibroblasts. J Biol Chem 275: 25065-25068, 2000[Abstract/Free Full Text].

127.   Kux, M, Coalson JJ, Massion WH, and Guenter CA. Pulmonary effects of E. coli endotoxin: role of leukocytes and platelets. Ann Surg 175: 26-34, 1972[ISI][Medline].

128.   LaCasse, EC, Baird S, Korneluk RG, and MacKenzie AE. The inhibitors of apoptosis (IAPs) and their emerging role in cancer. Oncogene 17: 3247-3259, 1998[ISI][Medline].

129.   Lang, CH, Frost RA, Nairn AC, MacLean DA, and Vary TC. TNF-alpha impairs heart and skeletal muscle protein synthesis by altering translation initiation. Am J Physiol Endocrinol Metab 282: E336-E347, 2002[Abstract/Free Full Text].

130.   Laskin, DL, Heck DE, Gardner CR, Feder LS, and Laskin JD. Distinct patterns of nitric oxide production in hepatic macrophages and endothelial cells following acute exposure of rats to endotoxin. J Leukoc Biol 56: 751-758, 1994[Abstract].

131.   Lee, YJ, and Shacter E. Hydrogen peroxide inhibits activation, not activity, of cellular caspase-3 in vivo. Free Radic Biol Med 29: 684-692, 2000[ISI][Medline].

132.   Lehr, HA, Bittinger F, and Kirkpatrick CJ. Microcirculatory dysfunction in sepsis: a pathogenetic basis for therapy? J Pathol 190: 373-386, 2000[ISI][Medline].

133.   Levi, M, ten Cate H, van der Poll T, and van Deventer SJ. Pathogenesis of disseminated intravascular coagulation in sepsis. JAMA 270: 975-979, 1993[Abstract].

134.   Li, AE, Ito H, Rovira II, Kim KS, Takeda K, Yu ZY, Ferrans VJ, and Finkel T. A role for reactive oxygen species in endothelial cell anoikis. Circ Res 85: 304-310, 1999[Abstract/Free Full Text].

135.   Li, D, Yang B, and Mehta JL. Ox-LDL induces apoptosis in human coronary artery endothelial cells: role of PKC, PTK, bcl-2, and Fas. Am J Physiol Heart Circ Physiol 275: H568-H576, 1998[Abstract/Free Full Text].

136.   Li, J, Billiar TR, Talanian RV, and Kim YM. Nitric oxide reversibly inhibits seven members of the caspase family via S-nitrosylation. Biochem Biophys Res Commun 240: 419-424, 1997[ISI][Medline].

137.   Li, L, Cousart S, Hu J, and McCall CE. Characterization of interleukin-1 receptor-associated kinase in normal and endotoxin-tolerant cells. J Biol Chem 275: 23340-23345, 2000[Abstract/Free Full Text].

138.   Libby, P, Ordovas JM, Auger KR, Robbins AH, Birinyi LK, and Dinarello CA. Endotoxin and tumor necrosis factor induce interleukin-1 gene expression in adult human vascular endothelial cells. Am J Pathol 124: 179-185, 1986[Abstract].

139.   Lien, E, Means TK, Heine H, Yoshimura A, Kusumoto S, Fukase K, Fenton MJ, Oikawa M, Qureshi N, Monks B, Finberg RW, Ingalls RR, and Golenbock DT. Toll-like receptor 4 imparts ligand-specific recognition of bacterial lipopolysaccharide. J Clin Invest 105: 497-504, 2000[Abstract/Free Full Text].

140.   Lopez-Farre, A, Rodriguez-Feo JA, Sanchez de Miguel L, Rico L, and Casado S. Role of nitric oxide in the control of apoptosis in the microvasculature. Int J Biochem Cell Biol 30: 1095-1106, 1998[ISI][Medline].

141.   Louise, CB, and Obrig TG. Shiga toxin-associated hemolytic uremic syndrome: combined cytotoxic effects of shiga toxin and lipopolysaccharide (endotoxin) on human vascular endothelial cells in vitro. Infect Immun 60: 1536-1543, 1992[Abstract].

142.   Macnaughton, PD, and Evans TW. Management of adult respiratory distress syndrome. Lancet 339: 469-472, 1992[ISI][Medline].

143.   Maeda, K, Abello PA, Abraham MR, Wetzel RC, Robotham JL, and Buchman TG. Endotoxin induces organ-specific endothelial cell injury. Shock 3: 46-50, 1995[ISI][Medline].

144.   Martin, MA, and Silverman HJ. Gram-negative sepsis and the adult respiratory distress syndrome. Clin Infect Dis 14: 1213-1228, 1992[ISI][Medline].

145.   McCuskey, RS, Urbaschek R, and Urbaschek B. The microcirculation during endotoxemia. Cardiovasc Res 32: 752-763, 1996[ISI][Medline].

146.   Medzhitov, R, Preston-Hurlburt P, Kopp E, Stadlen A, Chen C, Ghosh S, and Janeway CA, Jr. MyD88 is an adaptor protein in the hToll/IL-1 receptor family signaling pathways. Mol Cell 2: 253-258, 1998[ISI][Medline].

147.   Messmer, UK, Briner VA, and Pfeilschifter J. Tumor necrosis factor-alpha and lipopolysaccharide induce apoptotic cell death in bovine glomerular endothelial cells. Kidney Int 55: 2322-2337, 1999[ISI][Medline].

148.   Meyrick, B. Pathology of the adult respiratory distress syndrome. Crit Care Clin 2: 405-428, 1986[Medline].

149.   Meyrick, BO. Endotoxin-mediated pulmonary endothelial cell injury. Fed Proc 45: 19-24, 1986[ISI][Medline].

150.   Mitchell, J, Jiang H, Berry L, and Meyrick B. Effect of antioxidants on lipopolysaccharide-stimulated induction of mangano superoxide dismutase mRNA in bovine pulmonary artery endothelial cells. J Cell Physiol 169: 333-340, 1996[ISI][Medline].

151.   Morita-Fujimura, Y, Fujimura M, Yoshimoto T, and Chan PH. Superoxide during reperfusion contributes to caspase-8 expression and apoptosis after transient focal stroke. Stroke 32: 2356-2361, 2001[Abstract/Free Full Text].

152.   Munshi, N, Fernandis AZ, Cherla RP, Park IW, and Ganju RK. Lipopolysaccharide-induced apoptosis of endothelial cells and its inhibition by vascular endothelial growth factor. J Immunol 168: 5860-5866, 2002[Abstract/Free Full Text].

153.   Mutunga, M, Fulton B, Bullock R, Batchelor A, Gascoigne A, Gillespie JI, and Baudouin SV. Circulating endothelial cells in patients with septic shock. Am J Respir Crit Care Med 163: 195-200, 2001[Abstract/Free Full Text].

154.   Muzio, M, Natoli G, Saccani S, Levrero M, and Mantovani A. The human Toll signaling pathway: divergence of nuclear factor-kappa B and JNK/SAPK activation upstream of tumor necrosis factor receptor-associated factor 6 (TRAF6). J Exp Med 187: 2097-2101, 1998[Abstract/Free Full Text].

155.   Muzio, M, Stockwell BR, Stennicke HR, Salvesen GS, and Dixit VM. An induced proximity model for caspase-8 activation. J Biol Chem 273: 2926-2930, 1998[Abstract/Free Full Text].

156.   Naassila, M, Roux F, Beauge F, and Daoust M. Ethanol potentiates lipopolysaccharide- or interleukin-1beta -induced nitric oxide generation in RBE4 cells. Eur J Pharmacol 313: 273-277, 1996[ISI][Medline].

157.   Neuzil, J, Schroder A, von Hundelshausen P, Zernecke A, Weber T, Gellert N, and Weber C. Inhibition of inflammatory endothelial responses by a pathway involving caspase activation and p65 cleavage. Biochemistry 40: 4686-4692, 2001[ISI][Medline].

158.   O'Loughlin, EV, and Robins-Browne RM. Effect of Shiga toxin and Shiga-like toxins on eukaryotic cells. Microbes Infect 3: 493-507, 2001[ISI][Medline].

159.   Olson, NC, Hellyer PW, and Dodam JR. Mediators and vascular effects in response to endotoxin. Br Vet J 151: 489-522, 1995[ISI][Medline].

160.   Palmer, RM, Bridge L, Foxwell NA, and Moncada S. The role of nitric oxide in endothelial cell damage and its inhibition by glucocorticoids. Br J Pharmacol 105: 11-12, 1992[Abstract].

161.   Parsons, PE, Worthen GS, Moore EE, Tate RM, and Henson PM. The association of circulating endotoxin with the development of the adult respiratory distress syndrome. Am Rev Respir Dis 140: 294-301, 1989[ISI][Medline].

162.   Pijpers, AH, van Setten PA, van den Heuvel LP, Assmann KJ, Dijkman HB, Pennings AH, Monnens LA, and van Hinsbergh VW. Verocytotoxin-induced apoptosis of human microvascular endothelial cells. J Am Soc Nephrol 12: 767-778, 2001[Abstract/Free Full Text].

163.   Pinner, RW, Teutsch SM, Simonsen L, Klug LA, Graber JM, Clarke MJ, and Berkelman RL. Trends in infectious diseases mortality in the United States. JAMA 275: 189-193, 1996[Abstract].

164.   Pohlman, TH, and Harlan JM. Human endothelial cell response to lipopolysaccharide, interleukin-1, and tumor necrosis factor is regulated by protein synthesis. Cell Immunol 119: 41-52, 1989[ISI][Medline].

165.   Pohlman, TH, and Harlan JM. Adaptive responses of the endothelium to stress. J Surg Res 89: 85-119, 2000[ISI][Medline].

166.   Poltorak, A, He X, Smirnova I, Liu MY, Huffel CV, Du X, Birdwell D, Alejos E, Silva M, Galanos C, Freudenberg M, Ricciardi-Castagnoli P, Layton B, and Beutler B. Defective LPS signaling in C3H/HeJ and C57BL/10ScCr mice: mutations in Tlr4 gene. Science 282: 2085-2088, 1998[Abstract/Free Full Text].

167.   Proulx, F, Seidman EG, and Karpman D. Pathogenesis of Shiga toxin-associated hemolytic uremic syndrome. Pediatr Res 50: 163-171, 2001[Abstract/Free Full Text].

168.   Radomski, MW, Palmer RM, and Moncada S. Glucocorticoids inhibit the expression of an inducible, but not the constitutive, nitric oxide synthase in vascular endothelial cells. Proc Natl Acad Sci USA 87: 10043-10047, 1990[Abstract].

169.   Ravagnan, L, Roumier T, and Kroemer G. Mitochondria, the killer organelles and their weapons. J Cell Physiol 192: 131-137, 2002[ISI][Medline].

170.   Ray, PE, and Liu XH. Pathogenesis of Shiga toxin-induced hemolytic uremic syndrome. Pediatr Nephrol 16: 823-839, 2001[ISI][Medline].

171.   Reed, JC. Mechanisms of apoptosis. Am J Pathol 157: 1415-1430, 2000[Abstract/Free Full Text].

172.   Reidy, MA, and Schwartz SM. Endothelial injury and regeneration. IV. Endotoxin: a nondenuding injury to aortic endothelium. Lab Invest 48: 25-34, 1983[ISI][Medline].

173.   Rietschel, ET, Brade H, Holst O, Brade L, Muller-Loennies S, Mamat U, Zahringer U, Beckmann F, Seydel U, Brandenburg K, Ulmer AJ, Mattern T, Heine H, Schletter J, Loppnow H, Schonbeck U, Flad HD, Hauschildt S, Schade UF, Di Padova F, Kusumoto S, and Schumann RR. Bacterial endotoxin: chemical constitution, biological recognition, host response, and immunological detoxification. Curr Top Microbiol Immunol 216: 39-81, 1996[ISI][Medline].

174.   Rietschel, ET, Kirikae T, Schade FU, Mamat U, Schmidt G, Loppnow H, Ulmer AJ, Zahringer U, Seydel U, Di Padova F, Schreier M, and Brade H. Bacterial endotoxin: molecular relationships of structure to activity and function. FASEB J 8: 217-225, 1994[Abstract/Free Full Text].

175.   Rockey, DC, and Chung JJ. Regulation of inducible nitric oxide synthase in hepatic sinusoidal endothelial cells. Am J Physiol Gastrointest Liver Physiol 271: G260-G267, 1996[Abstract/Free Full Text].

176.   Rubenstein, HS, Fine J, and Coons AH. Localization of endotoxin in the walls of the peripheral vascular system during lethal endotoxemia. Proc Soc Exp Biol Med 111: 458-467, 1962.

177.   Rubin, DB, Reznik G, Weiss EA, and Young PR. Non-protein thiols flux to S-nitrosothiols in endothelial cells: an LPS redox signal. Shock 14: 200-207, 2000[ISI][Medline].

178.   Ryan, KM, Ernst MK, Rice NR, and Vousden KH. Role of NF-kappa B in p53-mediated programmed cell death. Nature 404: 892-897, 2000[ISI][Medline].

179.   Salvemini, D, Korbut R, Anggard E, and Vane J. Immediate release of a nitric oxide-like factor from bovine aortic endothelial cells by Escherichia coli lipopolysaccharide. Proc Natl Acad Sci USA 87: 2593-2597, 1990[Abstract].

180.   Sato, I, Kaji K, and Murota S. Age-related decline in cytokine-induced nitric oxide synthase activation and apoptosis in cultured endothelial cells: minimal involvement of nitric oxide in the apoptosis. Mech Ageing Dev 81: 27-36, 1995[ISI][Medline].

181.   Savill, JS, Wyllie AH, Henson JE, Walport MJ, Henson PM, and Haslett C. Macrophage phagocytosis of aging neutrophils in inflammation. Programmed cell death in the neutrophil leads to its recognition by macrophages. J Clin Invest 83: 865-875, 1989[ISI][Medline].

182.   Scaffidi, C, Schmitz I, Krammer PH, and Peter ME. The role of c-FLIP in modulation of CD95-induced apoptosis. J Biol Chem 274: 1541-1548, 1999[Abstract/Free Full Text].

183.   Scatena, M, Almeida M, Chaisson ML, Fausto N, Nicosia RF, and Giachelli CM. NF-kappa B mediates alphavbeta3 integrin-induced endothelial cell survival. J Cell Biol 141: 1083-1093, 1998[Abstract/Free Full Text].

184.   Schaub, FJ, Han DK, Liles WC, Adams LD, Coats SA, Ramachandran RK, Seifert RA, Schwartz SM, and Bowen-Pope DF. Fas/FADD-mediated activation of a specific program of inflammatory gene expression in vascular smooth muscle cells. Nat Med 6: 790-796, 2000[ISI][Medline].

185.   Schumann, RR, Belka C, Reuter D, Lamping N, Kirschning CJ, Weber JR, and Pfeil D. Lipopolysaccharide activates caspase-1 (interleukin-1-converting enzyme) in cultured monocytic and endothelial cells. Blood 91: 577-584, 1998[Abstract/Free Full Text].

186.   Schwandner, R, Dziarski R, Wesche H, Rothe M, and Kirschning CJ. Peptidoglycan- and lipoteichoic acid-induced cell activation is mediated by Toll-like receptor 2. J Biol Chem 274: 17406-17409, 1999[Abstract/Free Full Text].

187.   Sladek, Z, and Rysanek D. Apoptosis of polymorphonuclear leukocytes of the juvenile bovine mammary gland during induced influx. Vet Res 31: 553-563, 2000[ISI][Medline].

188.   Srinivasula, SM, Ahmad M, Ottilie S, Bullrich F, Banks S, Wang Y, Fernandes-Alnemri T, Croce CM, Litwack G, Tomaselli KJ, Armstrong RC, and Alnemri ES. FLAME-1, a novel FADD-like anti-apoptotic molecule that regulates Fas/TNFR1-induced apoptosis. J Biol Chem 272: 18542-18545, 1997[Abstract/Free Full Text].

189.   Stefanec, T. Endothelial apoptosis: could it have a role in the pathogenesis and treatment of disease? Chest 117: 841-854, 2000[Abstract/Free Full Text].

190.   Stehlik, C, de Martin R, Kumabashiri I, Schmid JA, Binder BR, and Lipp J. Nuclear factor (NF)-kappa B-regulated X-chromosome-linked iap gene expression protects endothelial cells from tumor necrosis factor-alpha -induced apoptosis. J Exp Med 188: 211-216, 1998[Abstract/Free Full Text].

191.   Sugawara, T, Noshita N, Lewen A, Gasche Y, Ferrand-Drake M, Fujimura M, Morita-Fujimura Y, and Chan PH. Overexpression of copper/zinc superoxide dismutase in transgenic rats protects vulnerable neurons against ischemic damage by blocking the mitochondrial pathway of caspase activation. J Neurosci 22: 209-217, 2002[Abstract/Free Full Text].

192.   Sutton, ET, Zhou Z, Baker CH, Price JM, and Chen Y. Differences in arterial and arteriolar endothelial structure during endotoxin shock. Circ Shock 41: 71-76, 1993[ISI][Medline].

193.   Suttorp, N, Galanos C, and Neuhof H. Endotoxin alters arachidonate metabolism in pulmonary endothelial cells. Am J Physiol Cell Physiol 253: C384-C390, 1987[Abstract/Free Full Text].

194.   Swantek, JL, Tsen MF, Cobb MH, and Thomas JA. IL-1 receptor-associated kinase modulates host responsiveness to endotoxin. J Immunol 164: 4301-4306, 2000[Abstract/Free Full Text].

195.   Takeyama, N, Miki S, Hirakawa A, and Tanaka T. Role of the mitochondrial permeability transition and cytochrome c release in hydrogen peroxide-induced apoptosis. Exp Cell Res 274: 16-24, 2002[ISI][Medline].

196.   Tang, ZL, Wasserloos KJ, Liu X, Stitt MS, Reynolds IJ, Pitt BR, and St. Croix CM. Nitric oxide decreases the sensitivity of pulmonary endothelial cells to LPS-induced apoptosis in a zinc-dependent fashion. Mol Cell Biochem 234-235: 211-217, 2002[ISI].

197.   Thijs, LG, de Boer JP, de Groot MC, and Hack CE. Coagulation disorders in septic shock. Intensive Care Med 19: S8-S15, 1993[Medline].

198.   Thornberry, NA, and Lazebnik Y. Caspases: enemies within. Science 281: 1312-1316, 1998[Abstract/Free Full Text].

199.   Tschopp, J, Irmler M, and Thome M. Inhibition of fas death signals by FLIPs. Curr Opin Immunol 10: 552-558, 1998[ISI][Medline].

200.   Tzeng, E, Kim YM, Pitt BR, Lizonova A, Kovesdi I, and Billiar TR. Adenoviral transfer of the inducible nitric oxide synthase gene blocks endothelial cell apoptosis. Surgery 122: 255-263, 1997[ISI][Medline].

201.   Ulevitch, RJ, Cochrane CG, Henson PM, Morrison DC, and Doe WF. Mediation systems in bacterial lipopolysaccharide-induced hypotension and disseminated intravascular coagulation. I. The role of complement. J Exp Med 142: 1570-1590, 1975[Abstract].

202.   Ulevitch, RJ, and Tobias PS. Receptor-dependent mechanisms of cell stimulation by bacterial endotoxin. Annu Rev Immunol 13: 437-457, 1995[ISI][Medline].

203.   Van Antwerp, DJ, Martin SJ, Kafri T, Green DR, and Verma IM. Suppression of TNF-alpha -induced apoptosis by NF-kappa B. Science 274: 787-789, 1996[Abstract/Free Full Text].

204.   Vescovo, G, Zennaro R, Sandri M, Carraro U, Leprotti C, Ceconi C, Ambrosio GB, and Dalla Libera L. Apoptosis of skeletal muscle myofibers and interstitial cells in experimental heart failure. J Mol Cell Cardiol 30: 2449-2459, 1998[ISI][Medline].

205.   Viriyakosol, S, and Kirkland T. Knowledge of cellular receptors for bacterial endotoxin. Clin Infect Dis 21 Suppl 2: S190-S195, 1995[ISI][Medline].

206.   Visner, GA, Block ER, Burr IM, and Nick HS. Regulation of manganese superoxide dismutase in porcine pulmonary artery endothelial cells. Am J Physiol Lung Cell Mol Physiol 260: L444-L449, 1991[Abstract/Free Full Text].

207.   Wajant, H, Haas E, Schwenzer R, Muhlenbeck F, Kreuz S, Schubert G, Grell M, Smith C, and Scheurich P. Inhibition of death receptor-mediated gene induction by a cycloheximide-sensitive factor occurs at the level of or upstream of Fas-associated death domain protein (FADD). J Biol Chem 275: 24357-24366, 2000[Abstract/Free Full Text].

208.   Walter, R, Schaffner A, and Schoedon G. Differential regulation of constitutive and inducible nitric oxide production by inflammatory stimuli in murine endothelial cells. Biochem Biophys Res Commun 202: 450-455, 1994[ISI][Medline].

209.   Wang, CY, Mayo MW, Korneluk RG, Goeddel DV, and Baldwin AS, Jr. NF-kappa B antiapoptosis: induction of TRAF1 and TRAF2 and c-IAP1 and c-IAP2 to suppress caspase-8 activation. Science 281: 1680-1683, 1998[Abstract/Free Full Text].

210.   Warren, HS, Glennon ML, Wainwright N, Amato SF, Black KM, Kirsch SJ, Riveau GR, Whyte RI, Zapol WM, and Novitsky TJ. Binding and neutralization of endotoxin by Limulus antilipopolysaccharide factor. Infect Immun 60: 2506-2513, 1992[Abstract].

211.   Warren, HS, Novitsky TJ, Bucklin A, Kania SA, and Siber GR. Endotoxin neutralization with rabbit antisera to Escherichia coli J5 and other gram-negative bacteria. Infect Immun 55: 1668-1673, 1987[ISI][Medline].

212.   Wesche, H, Henzel WJ, Shillinglaw W, Li S, and Cao Z. MyD88: an adapter that recruits IRAK to the IL-1 receptor complex. Immunity 7: 837-847, 1997[ISI][Medline].

213.   Williams, JM, Boyd B, Nutikka A, Lingwood CA, Barnett Foster DE, Milford DV, and Taylor CM. A comparison of the effects of verocytotoxin-1 on primary human renal cell cultures. Toxicol Lett 105: 47-57, 1999[ISI][Medline].

214.   Wolkow, PP. Involvement and dual effects of nitric oxide in septic shock. Inflamm Res 47: 152-166, 1998[ISI][Medline].

215.   Wong, GH. Protective roles of cytokines against radiation: induction of mitochondrial MnSOD. Biochim Biophys Acta 1271: 205-209, 1995[ISI][Medline].

216.   Wong, HR, Mannix RJ, Rusnak JM, Boota A, Zar H, Watkins SC, Lazo JS, and Pitt BR. The heat-shock response attenuates lipopolysaccharide-mediated apoptosis in cultured sheep pulmonary artery endothelial cells. Am J Respir Cell Mol Biol 15: 745-751, 1996[Abstract].

217.   Wu, R, Song X, Xu Y, and Meng X. Apoptosis of endothelial cells in alteration of microvascular permeability in lung during sepsis. Zhonghua Wai Ke Za Zhi 38: 385-387, 2000[Medline].

218.   Yang, RB, Mark MR, Gray A, Huang A, Xie MH, Zhang M, Goddard A, Wood WI, Gurney AL, and Godowski PJ. Toll-like receptor-2 mediates lipopolysaccharide-induced cellular signalling. Nature 395: 284-288, 1998[ISI][Medline].

219.   Yang, Z, Breider MA, Carroll RC, Miller MS, and Bochsler PN. Soluble CD14 and lipopolysaccharide-binding protein from bovine serum enable bacterial lipopolysaccharide-mediated cytotoxicity and activation of bovine vascular endothelial cells in vitro. J Leukoc Biol 59: 241-247, 1996[Abstract].

220.   Zen, K, Karsan A, Eunson T, Yee E, and Harlan JM. Lipopolysaccharide-induced NF-kappa B activation in human endothelial cells involves degradation of Ikappa Balpha but not Ikappa Bbeta . Exp Cell Res 243: 425-433, 1998[ISI][Medline].

221.   Zen, K, Karsan A, Stempien-Otero A, Yee E, Tupper J, Li X, Eunson T, Kay MA, Wilson CB, Winn RK, and Harlan JM. NF-kappa B activation is required for human endothelial survival during exposure to tumor necrosis factor-alpha but not to interleukin-1beta or lipopolysaccharide. J Biol Chem 274: 28808-28815, 1999[Abstract/Free Full Text].

222.   Zhang, FX, Kirschning CJ, Mancinelli R, Xu XP, Jin Y, Faure E, Mantovani A, Rothe M, Muzio M, and Arditi M. Bacterial lipopolysaccharide activates nuclear factor-kappa B through interleukin-1 signaling mediators in cultured human dermal endothelial cells and mononuclear phagocytes. J Biol Chem 274: 7611-7614, 1999[Abstract/Free Full Text].

223.   Zhao, B, Bowden RA, Stavchansky SA, and Bowman PD. Human endothelial cell response to gram-negative lipopolysaccharide assessed with cDNA microarrays. Am J Physiol Cell Physiol 281: C1587-C1595, 2001[Abstract/Free Full Text].

224.   Zhuang, S, Lynch MC, and Kochevar IE. Caspase-8 mediates caspase-3 activation and cytochrome c release during singlet oxygen-induced apoptosis of HL-60 cells. Exp Cell Res 250: 203-212, 1999[ISI][Medline].

225.   Ziegler, EJ, McCutchan JA, Fierer J, Glauser MP, Sadoff JC, Douglas H, and Braude AI. Treatment of gram-negative bacteremia and shock with human antiserum to a mutant Escherichia coli. N Engl J Med 307: 1225-1230, 1982[Abstract].


Am J Physiol Lung Cell Mol Physiol 284(6):L899-L914