scispace - formally typeset
Search or ask a question

Showing papers by "Richard A. Flavell published in 2000"


Journal ArticleDOI
TL;DR: It is demonstrated here that carbon monoxide, a by-product of heme catabolism by heme oxygenase, mediates potent anti-inflammatory effects and may have an important protective function in inflammatory disease states and thus has potential therapeutic uses.
Abstract: The stress-inducible protein heme oxygenase-1 provides protection against oxidative stress. The anti-inflammatory properties of heme oxygenase-1 may serve as a basis for this cytoprotection. We demonstrate here that carbon monoxide, a by-product of heme catabolism by heme oxygenase, mediates potent anti-inflammatory effects. Both in vivo and in vitro, carbon monoxide at low concentrations differentially and selectively inhibited the expression of lipopolysaccharide-induced pro-inflammatory cytokines tumor necrosis factor-α, interleukin-1β, and macrophage inflammatory protein-1β and increased the lipopolysaccharide-induced expression of the anti-inflammatory cytokine interleukin-10. Carbon monoxide mediated these anti-inflammatory effects not through a guanylyl cyclase–cGMP or nitric oxide pathway, but instead through a pathway involving the mitogen-activated protein kinases. These data indicate the possibility that carbon monoxide may have an important protective function in inflammatory disease states and thus has potential therapeutic uses.

2,035 citations


Journal ArticleDOI
05 May 2000-Science
TL;DR: It is shown here that JNK is required for UV-induced apoptosis in primary murine embryonic fibroblasts, and data indicate that mitochondria are influenced by proapoptotic signal transduction through the JNK pathway.
Abstract: The c-Jun NH2-terminal kinase (JNK) is activated when cells are exposed to ultraviolet (UV) radiation. However, the functional consequence of JNK activation in UV-irradiated cells has not been established. It is shown here that JNK is required for UV-induced apoptosis in primary murine embryonic fibroblasts. Fibroblasts with simultaneous targeted disruptions of all the functional Jnk genes were protected against UV-stimulated apoptosis. The absence of JNK caused a defect in the mitochondrial death signaling pathway, including the failure to release cytochrome c. These data indicate that mitochondria are influenced by proapoptotic signal transduction through the JNK pathway.

1,738 citations


Journal ArticleDOI
01 Feb 2000-Immunity
TL;DR: It is shown that T cell homeostasis requires T GFβ signaling in T cells, and a transgenic approach to abrogate the TGFβ response in key immune cells is developed.

943 citations


Journal ArticleDOI
TL;DR: Insight is provided on the significance and mechanism of neural cell death in mammalian brain development and the apoptosis of progenitor cells during early embryonic development and postmitotic neurons at later stage of brain development have distinct roles and mechanisms.

436 citations


Journal ArticleDOI
TL;DR: It is demonstrated that TGF-β inhibits GATA-3 expression in developing Th cells, which is a major mechanism of inhibition of Th2 differentiation by T GF-β as ectopic expression of Gata-3 in developing T cells overcomes the ability of TGF -β to inhibit Th 2 differentiation.
Abstract: TGF-beta is an important immunomodulatory cytokine that can inhibit differentiation of effector T cells. In this report, we address the molecular mechanisms through which TGF-beta inhibits differentiation of CD4(+) cells into Th type 2 cells. We demonstrate that TGF-beta inhibits GATA-3 expression in developing Th cells. We also show that inhibition of GATA-3 expression by TGF-beta is a major mechanism of inhibition of Th2 differentiation by TGF-beta as ectopic expression of GATA-3 in developing T cells overcomes the ability of TGF-beta to inhibit Th2 differentiation. TGF-beta likely inhibits GATA-3 expression at the transcriptional level and does so without interfering with IL-4 signaling.

377 citations


Journal ArticleDOI
04 May 2000-Nature
TL;DR: Three new mouse models in which peripheral T cells completely lack JNK proteins or signalling are used, to test whether the JNK signalling pathway is crucial for IL-2 expression and T-cell activation, and it is found that these T cells made moreIL-2 and proliferated better than wild-type cells.
Abstract: The hallmark of T-cell activation is the production of interleukin 2 (IL-2). c-Jun amino-terminal kinase (JNK), a MAP kinase that phosphorylates c-Jun and other components of the AP-1 group of transcription factors, has been implicated in the activation of IL-2 expression. Previously, we found that T cells from mice deficient in the Jnk1 or Jnk2 gene can be activated and produce IL-2 normally, but are deficient in functional differentiation into Th1 or Th2 subsets. However, studies of mice with compound mutations indicate that JNK1 and JNK2 are redundant during mouse development. Here we use three new mouse models in which peripheral T cells completely lack JNK proteins or signalling, to test whether the JNK signalling pathway is crucial for IL-2 expression and T-cell activation. Unexpectedly, these T cells made more IL-2 and proliferated better than wild-type cells. However, production of effector T-cell cytokines did require JNK. Thus, JNK is necessary for T-cell differentiation but not for naive T-cell activation.

328 citations


Journal ArticleDOI
TL;DR: It is demonstrated here that elimination of certain caspases was compensated in vivo by the activation of othercaspases, providing direct experimental evidence for compensatory pathways of caspase activation.
Abstract: Dysregulation of apoptosis contributes to the pathogenesis of many human diseases. As effectors of the apoptotic machinery, caspases are considered potential therapeutic targets. Using an established in vivo model of Fas-mediated apoptosis, we demonstrate here that elimination of certain caspases was compensated in vivo by the activation of other caspases. Hepatocyte apoptosis and mouse death induced by the Fas agonistic antibody Jo2 required proapoptotic Bcl-2 family member Bid and used a Bid-mediated mitochondrial pathway of caspase activation; deficiency in caspases essential for this pathway, caspase-9 or caspase-3, unexpectedly resulted in rapid activation of alternate caspases after injection of Jo2, and therefore failed to protect mice against Jo2 toxicity. Moreover, both ultraviolet and gamma irradiation, two established inducers of the mitochondrial caspase-activation pathway, also elicited compensatory activation of caspases in cultured caspase-3(-/-) hepatocytes, indicating that the compensatory caspase activation was mediated through the mitochondria. Our findings provide direct experimental evidence for compensatory pathways of caspase activation. This issue should therefore be considered in developing caspase inhibitors for therapeutic applications.

319 citations


Journal ArticleDOI
01 Nov 2000-Immunity
TL;DR: Signals emanating from the pre-TCR are mediated at least in part by NF-kappa B, which provides a selective survival signal for developing thymocytes with productive beta chain rearrangements.

286 citations


Journal ArticleDOI
23 Jun 2000-Science
TL;DR: A complementary DNA subtraction method, representational display analysis, is used to show that the small guanosine triphosphatase Rac2 is expressed selectively in murine TH1 cells, which activates TH1-specific signaling and IFN-gamma gene expression.
Abstract: T helper 1 (TH1) cells mediate cellular immunity, whereas TH2 cells potentiate antiparasite and humoral immunity We used a complementary DNA subtraction method, representational display analysis, to show that the small guanosine triphosphatase Rac2 is expressed selectively in murine TH1 cells Rac induces the interferon-γ (IFN-γ) promoter through cooperative activation of the nuclear factor kappa B and p38 mitogen-activated protein kinase pathways Tetracycline-regulated transgenic mice expressing constitutively active Rac2 in T cells exhibited enhanced IFN-γ production Dominant-negative Rac inhibited IFN-γ production in murine T cells Moreover, T cells from Rac2−/− mice showed decreased IFN-γ production under TH1 conditions in vitro Thus, Rac2 activates TH1-specific signaling and IFN-γ gene expression

172 citations


Journal ArticleDOI
TL;DR: In this paper, the role of the mitogen-activated protein (MAP) kinase family members, which include the extracellular response kinases (ERK), p38, and c-Jun amino terminal kinases(JNK), play a role in mediating signals triggered by cytokines, growth factors, and environmental stress.

166 citations


Journal ArticleDOI
TL;DR: It is demonstrated that the calcineurin targeting domain of NFATc1 is phosphorylated and inactivated by the c-Jun NH2-terminal kinase (JNK), which inhibits the nuclear accumulation and transcription activity of NFatc1 and accounts for the observation that Jnk1 −/− T cells exhibit greatly increased NFAT c1-dependent nuclear responses.
Abstract: The c-Jun NH2-terminal kinase (JNK) group of mitogen-activated protein kinases is activated by treatment of cells with cytokines or exposure to environmental stress (19). Gene disruption studies indicate that JNK protein kinases are required for multiple biological processes (15, 23, 32, 41, 42). However, the molecular mechanisms that account for the phenotypes displayed by JNK knockout mice remain unclear. For example, the disruption of the Jnk1 gene causes a severe defect in the response of CD4+ helper T (TH) cells to antigen-presenting cells (15). Wild-type naive TH cells can differentiate to TH1 or TH2 effector cells, which mediate inflammatory and humoral responses, respectively. In contrast, Jnk1−/− CD4 TH cells preferentially differentiate to TH2 effector cells and secrete large amounts of the TH2 cytokines, including interleukin-4 (IL-4). As a consequence, these mice are highly susceptible to infection with Leishmania. These effects of Jnk1 gene disruption were associated with increased nuclear accumulation of the NFATc1 transcription factor (15), which is known to act at the IL-4 promoter and is essential for TH2 responses (30, 44). The phenotype of Jnk1−/− mice suggests that NFATc1 is negatively regulated by JNK1 (15). However, this phenotype could be a direct or an indirect consequence of Jnk1 gene disruption. Transcription factor NFAT was first identified as an important regulator of IL-2 gene expression (16, 20). More recently, it has been established that NFAT contributes to the expression of several cytokines and participates in multiple physiological processes (12, 31). In resting T cells, NFAT is restricted to the cytoplasm (12, 31). Following T-cell activation, a sustained increase in intracellular calcium (37) activates the phosphatase calcineurin (11, 21). The activated calcineurin dephosphorylates NFAT and leads to increased nuclear accumulation (12, 31). The nuclear NFAT transcription factor then increases the expression of target genes, including IL-2, IL-4, and Fas ligand. The interaction of calcineurin with NFAT is a critical element in the signal transduction pathway that leads to increased NFAT-dependent gene expression. Interestingly, this interaction is mediated by a targeting domain (PxIxIT motif) that is present in the NH2-terminal region of the NFAT transcription factor (1, 2, 8). This targeting domain is required for efficient NFAT activation in vivo. Furthermore, ectopic expression of the targeting domain causes profound and specific inhibition of NFAT-mediated gene expression in cultured cells (2, 8). Studies of transgenic mice also demonstrate inhibition of NFAT-mediated gene expression caused by expression of the calcineurin targeting domain (8). Together, these data indicate that the calcineurin targeting domain is a critical component of the regulatory mechanism that controls NFAT activity. The purpose of this study was to examine the role of JNK1 in NFATc1 regulation. Since the phenotype of Jnk1−/− mice suggests a correlation between the JNK1 signaling pathway and NFATc1 (15), we tested whether NFATc1 is directly regulated by JNK1. We report that JNK1 binds and phosphorylates NFATc1 on sites located near the PxIxIT calcineurin targeting domain. This phosphorylation inhibits the interaction of calcineurin with the targeting domain and blocks nuclear accumulation. This observation provides a molecular mechanism for the observation of increased NFATc1 nuclear accumulation in Jnk1−/− mice.

Journal ArticleDOI
TL;DR: Recent advancement in understanding of transcriptional and signaling regulation of the differentiation process is summarized, which will become important in the future to develop means in treating immune disorders.
Abstract: After activation CD4+ helper T cells differentiate into T-helper (Th) 1 or Th2 effector cells. These two subsets are characterized by their distinct cytokine expression pattern and the immune function they mediate. Over the past years, a number of factors have been identified to affect helper T cell lineage determination, including antigen receptor, coreceptors and, most importantly, cytokine environment. In this review, we also summarize recent advancement in understanding of transcriptional and signaling regulation of the differentiation process. This knowledge will become important in the future to develop means in treating immune disorders.

Journal ArticleDOI
TL;DR: A role of programmed cell death is established in regulating the size of progenitor population in the central nervous system, a function that is distinct from the classic role of cell death in matching postmitotic neuronal population with postsynaptic targets.
Abstract: The number of neurons in the mammalian brain is determined by a balance between cell proliferation and programmed cell death. Recent studies indicated that Bcl-XL prevents, whereas Caspase-3 mediates, cell death in the developing nervous system, but whether Bcl-XL directly blocks the apoptotic function of Caspase-3 in vivo is not known. To examine this question, we generated bcl-x/caspase-3 double mutants and found that caspase-3 deficiency abrogated the increased apoptosis of postmitotic neurons but not the increased hematopoietic cell death and embryonic lethality caused by the bcl-x mutation. In contrast, caspase-3, but not bcl-x, deficiency changed the normal incidence of neuronal progenitor cell apoptosis, consistent with the lack of expression of Bcl-XL in the proliferative population of the embryonic cortex. Thus, although Caspase-3 is epistatically downstream to Bcl-XL in postmitotic neurons, it independently regulates apoptosis of neuronal founder cells. Taken together, these results establish a role of programmed cell death in regulating the size of progenitor population in the central nervous system, a function that is distinct from the classic role of cell death in matching postmitotic neuronal population with postsynaptic targets.

Journal ArticleDOI
TL;DR: Neither calpain inhibition nor microtubule stabilization with Taxol protected telencephalic neurons from Aβ-induced caspase activation or apoptosis, which have potential implications regarding the underlying pathophysiology of AD and towards AD treatment strategies.
Abstract: Fibrillar amyloid beta (Abeta) peptides are major constituents of senile plaques in Alzheimer disease (AD) brain and cause neuronal apoptosis in vitro. Bax and caspase-3 have been implicated in the pathogenesis of AD and are components of a well-defined molecular pathway of neuronal apoptosis. To determine whether Abeta-induced neuronal apoptosis involves bax and/or caspase-3 activation, we examined the effect of Abeta on wild-type, bax-deficient, and caspase-3-deficient telencephalic neurons in vitro. In wild-type cultures, Abeta produced time- and concentration-dependent caspase-3 activation, apoptotic nuclear changes, and neuronal death. These neurotoxic effects of Abeta were not observed in bax-deficient cultures. Caspase-3 deficiency, or pharmacological inhibition of caspase activity, prevented caspase-3 activation and blocked the appearance of apoptotic nuclear features but not Abeta-induced neuronal death. Neither calpain inhibition nor microtubule stabilization with Taxol protected telencephalic neurons from Abeta-induced caspase activation or apoptosis. These results have potential implications regarding the underlying pathophysiology of AD and towards AD treatment strategies.

Journal ArticleDOI
TL;DR: It is demonstrated that B. burgdorferi bbK32 and bbk50 are expressed during tick engorgement and that BBK32 antisera can interfere with spirochete transmission at various stages of the vector-host life cycle.
Abstract: Antisera to BBK32 (a Borrelia burgdorferi fibronectin-binding protein) and BBK50, two Ags synthesized during infection, protect mice from experimental syringe-borne Lyme borreliosis. Therefore, B. burgdorferi bbk32 and bbk50 expression within Ixodes scapularis ticks and the murine host, and the effect of BBK32 and BBK50 antisera on spirochetes throughout the vector-host life cycle were investigated. bbk32 and bbk50 mRNA and protein were first detected within engorged ticks, demonstrating regulated expression within the vector. Then bbk32 expression increased in mice at the cutaneous site of inoculation. During disseminated murine infection, bbk32 and bbk50 were expressed in several murine tissues, and mRNA levels were greatest in the heart and spleen at 30 days. BBK32 antisera protected mice from tick-borne B. burgdorferi infection and spirochete numbers were reduced by 90% within nymphs that engorged on immunized mice. Moreover, 75% of these ticks did not retain spirochetes upon molting, and subsequent B. burgdorferi transmission by adult ticks was impaired. Larval acquisition of B. burgdorferi by I. scapularis was also inhibited by BBK32 antisera. These data demonstrate that bbk32 and bbk50 are expressed during tick engorgement and that BBK32 antisera can interfere with spirochete transmission at various stages of the vector-host life cycle. These studies provide insight into mechanisms of immunity to Lyme borreliosis and other vector-borne diseases.

Journal ArticleDOI
TL;DR: It is shown that the p38 mitogen-activated protein (MAP) kinase signaling pathway is strictly regulated during the differentiation of CD4−CD8− thymocytes, a critical regulatory element of differentiation and proliferation during the early stages of in vivo thymocyte development.
Abstract: The development of T cells in the thymus is coordinated by cell-specific gene expression programs that involve multiple transcription factors and signaling pathways. Here, we show that the p38 mitogen-activated protein (MAP) kinase signaling pathway is strictly regulated during the differentiation of CD4−CD8− thymocytes. Persistent activation of p38 MAP kinase blocks fetal thymocyte development at the CD25+CD44− stage in vivo, and results in the lack of T cells in the peripheral immune system of adult mice. Inactivation of p38 MAP kinase is required for further differentiation of these cells into CD4+CD8+ thymocytes. The arrest of cell cycle in mitosis is partially responsible for the blockade of differentiation. Therefore, the p38 MAP kinase pathway is a critical regulatory element of differentiation and proliferation during the early stages of in vivo thymocyte development.

Journal ArticleDOI
TL;DR: An understanding of the pathophysiology of common autoimmune diseases will require elucidation of many different systems that interact in complex ways, of which the process of apoptosis is just one.

Journal ArticleDOI
TL;DR: What these studies have revealed about the role of individual caspase in development, apoptosis, and inflammation are discussed, with a particular focus on the predictable phenotypes versus the surprises based on in vitro results, as well as the implications of these findings.

Journal ArticleDOI
TL;DR: Direct in vivo evidence for the diabetogenic effect of this human MHC class II molecule is provided and a unique “humanized” animal model of spontaneous diabetes is provided.
Abstract: Although DQA1*0301/DQB1*0302 is the human histocompatibility leukocyte antigen (HLA) class II gene most commonly associated with human type 1 diabetes, direct in vivo experimental evidence for its diabetogenic role is lacking. Therefore, we generated C57BL/6 transgenic mice that bear this molecule and do not express mouse major histocompatibility complex (MHC) class II molecules (DQ8+/mII−). They did not develop insulitis or spontaneous diabetes. However, when DQ8+/mII− mice were bred with C57BL/6 mice expressing costimulatory molecule B7-1 on β cells (which normally do not develop diabetes), 81% of the DQ8+/mII−/B7-1+ mice developed spontaneous diabetes. The diabetes was accompanied by severe insulitis composed of both T cells (CD4+ and CD8+) and B cells. T cells from the diabetic mice secreted large amounts of interferon γ, but not interleukin 4, in response to DQ8+ islets and the putative islet autoantigens, insulin and glutamic acid decarboxylase (GAD). Diabetes could also be adoptively transferred to irradiated nondiabetic DQ8+/mII−/B7-1+ mice. In striking contrast, none of the transgenic mice in which the diabetes protective allele (DQA1*0103/DQB1*0601, DQ6 for short) was substituted for mouse MHC class II molecules but remained for the expression of B7-1 on pancreatic β cells (DQ6+/mII−/B7-1+) developed diabetes. Only 7% of DQ−/mII−/B7-1+ mice developed diabetes at an older age, and none of the DQ−/mII+/B7-1+ mice or DQ8+/mII+/B7-1+ mice developed diabetes. In conclusion, substitution of HLA-DQA1*0301/DQB1*0302, but not HLA-DQA1*0103/DQB1*0601, for murine MHC class II provokes autoimmune diabetes in non–diabetes-prone rat insulin promoter (RIP).B7-1 C57BL/6 mice. Our data provide direct in vivo evidence for the diabetogenic effect of this human MHC class II molecule and a unique “humanized” animal model of spontaneous diabetes.

Journal ArticleDOI
TL;DR: It is demonstrated that JNK is regulated by a second, novel mechanism that contributes to the regulation of the JNK pathway to ensure proper control during the course of an immune response.
Abstract: The c-Jun NH2-terminal kinases (JNKs) are a group of mitogen-activated protein (MAP) kinases that participate in signal transduction events mediating specific cellular functions. Activation of JNK is regulated by phosphorylation in response to cellular stress and inflammatory cytokines. Here, we demonstrate that JNK is regulated by a second, novel mechanism. Induction of Jnk gene expression is required in specific tissues before activation of this signaling pathway. The in vivo and in vitro ligation of the T cell receptor (TCR) leads to induction of JNK gene and protein expression. TCR signals are sufficient to induce JNK expression, whereas JNK phosphorylation also requires CD28-mediated costimulatory signals. Therefore, both expression and activation contribute to the regulation of the JNK pathway to ensure proper control during the course of an immune response.

Journal ArticleDOI
01 May 2000-Immunity
TL;DR: In this article, the temporal importance of TNFα in diabetes development in mice expressing islet-specific B7-1 and TNF α has been discussed, where the authors showed that the duration of the inflammatory cytokine mediated inflammation, not the putative maturity of the immune system at the time of expression, determines diabetes progression.

Journal ArticleDOI
TL;DR: The CD154/CD40 pathway plays a critical role in Th1-induced chronic experimental colitis and blocking CD154, even after the onset of disease, ameliorates colitis but does not induce a T helper 2 (Th2) phenotype.

Journal ArticleDOI
TL;DR: It is found that, following K+ deprivation, neurons from −/−CPP32 mice die to the same extent as those from normal (+/+) mice, and caspase‐3 is required for DNA fragmentation and chromatin condensation as judged by the absence of these apoptotic features in +/+ neurons.
Abstract: Caspases are crucial effectors of the cell death pathway activated by virtually all apoptosis-inducing stimuli within neurons and nonneuronal cells. Among the caspases, caspase-3 (CPP32) appears to play a pivotal role and has been found to be necessary for developmentally regulated cell death in the brain. We have used mice lacking caspase-3 (−/−CPP32) to examine its involvement in cultured cerebellar granule neurons induced to undergo apoptosis by potassium deprivation (K+). We find that, following K+ deprivation, neurons from −/−CPP32 mice die to the same extent as those from normal (+/+) mice. Although a small delay in the induction of cell death is observed in −/−CPP32 neurons, the rate of cell death is generally comparable to that of +/+ cultures. Though not critical for neuronal death, caspase-3 is required for DNA fragmentation and chromatin condensation as judged by the absence of these apoptotic features in −/−CPP32 neurons. Boc.Asp.fmk, a pan caspase inhibitor, partially protects +/+ neurons from low-K+-mediated cell death and does so to the same extent in −/−CPP32 cultures, suggesting the involvement of a caspase other than caspase-3 in cell death. However, the protective effect of boc.Asp.fmk is not seen beyond 24 hr, suggesting that the effect of caspase inhibition is one of delaying rather than preventing apoptosis. The more selective caspase inhibitors DEVD.fmk, IETD.fmk, and VEID.fmk fail to affect cell death, indicating that members inhibited by these agents (such as caspases - 6 ,7, 8, 9 and 10) are also not involved in low-K+-mediated apoptosis. J. Neurosci. Res. 59:24–31, 2000 © 2000 Wiley-Liss, Inc.

Journal ArticleDOI
TL;DR: This study suggests a potential molecular mechanism by which CGRP can induce cell death in thymocytes, and demonstrates for the first time the selective implication of the NF-κB signaling pathway in the regulatory function of the neuropeptide C GRP.

Journal ArticleDOI
TL;DR: It is shown here that immature B cells fail to home to the lymph nodes, the first example of autocrine regulation of immune cell migration to sites of foreign antigen presentation.
Abstract: The mechanism by which immature B cells are sequestered from encountering foreign antigens present in lymph nodes or sites of inflammation, before their final maturation in the spleen, has not been elucidated. We show here that immature B cells fail to home to the lymph nodes. These cells can actively exclude themselves from antigen-enriched sites by downregulating their integrin-mediated adhesion to the extracellular matrix protein, fibronectin. This inhibition is mediated by interferon γ secretion. Perturbation of interferon γ activity in vivo leads to the homing of immature B cells to the lymph nodes. This is the first example of autocrine regulation of immune cell migration to sites of foreign antigen presentation.

Journal ArticleDOI
TL;DR: It is indicated that caspase 3 deficiency results in a pervasive, albeit partial, decrease in embryonic neuroblast apoptosis that can account for the observed phenotypic hyperplasia in −/− embryos, and support the additional operation of casp enzyme 3‐independent PCD mechanisms during embryonic CNS development.
Abstract: Caspase 3 (CPP32/Yama/apopain), a mammalian homolog of the Caenorhabditis elegans pro-cell death gene ced-3, is required for normal programmed cell death (PCD) in the nematode. Its prior deletion by homologous recombination in mice resulted in embryonic/early postnatal lethality associated with dramatic central nervous system (CNS) hyperplasia, yet a reported subtle decrease in cell death (Kuida et al. [1996] Nature 384:368‐372). By comparison, the magnitude and distribution of dying cells identified using a DNA end-labeling technique, in situ end-labeling plus (ISEL1) (Blaschke et al. [1996] Development 122:1165‐1174; Blaschke et al. [1998] J. Comp. Neurol. 396:39‐50), supported an alternative explanation where the loss of caspase 3 function produces a more pervasive block in cell death, particularly among neuroblasts. To determine the relationship between loss of caspase 3 and dying cells identified by ISEL1 ,w e analyzed caspase 3 1/1, 1/2, and 2/2 embryos for normal caspase 3 expression and ISEL1 labeling. Both caspase 3 mRNA and active caspase 3 protein are present throughout the 1/1 embryonic CNS, and both are absent from 2/2 embryonic cortices. Quantitation of dying cells identified by ISEL1 reveals a 30% reduction of labeled cells throughout the caspase 3 2/2 embryonic cortices relative to 1/1 littermates. Associated with this decrease is marked expansion of the total population of actively proliferating neuroblasts identified by 5-bromo-29deoxyuridine incorporation that nevertheless appears to maintain histological features of normal neurogenesis rather than dysregulated, neoplastic growth. These data indicate that caspase 3 deficiency results in a pervasive, albeit partial, decrease in embryonic neuroblast apoptosis that

Journal ArticleDOI
TL;DR: Although the results of class-specific ELISAs with recombinant antigens were comparable to those recorded for assays with whole-cell antigen and for individuals with confirmed clinical diagnoses of Lyme borreliosis, immunoblotting is still advised as an adjunct procedure, particularly when there are low antibody titers by an ELISA.
Abstract: Class-specific enzyme-linked immunosorbent assays (ELISAs) with purified recombinant antigens of Borrelia burgdorferi sensu stricto and Western blot analyses with whole cells of this spirochete were used to test human sera to determine which antigens were diagnostically important. In analyses for immunoglobulin M (IgM) antibodies, 14 (82%) of 17 serum samples from persons who had erythema migrans reacted positively by an ELISA with one or more recombinant antigens. There was frequent antibody reactivity to protein 41-G (p41-G), outer surface protein C (OspC), and OspF antigens. In an ELISA for IgG antibodies, 13 (87%) of 15 serum samples had antibodies to recombinant antigens; reactivity to p22, p39, p41-G, OspC, and OspF antigens was frequent. By both ELISAs, serum specimens positive for OspB, OspE, and p37 were uncommon. Analyses of sera obtained from persons who were suspected of having human granulocytic ehrlichiosis (HGE) but who lacked antibodies to ehrlichiae revealed IgM antibodies to all recombinant antigens of B. burgdorferi except OspB and IgG antibodies to all antigens except OspE. Immunoblotting of sera from the study group of individuals suspected of having HGE reaffirmed antibody reactivity to multiple antigens of B. burgdorferi. There was minor cross-reactivity when sera from healthy subjects or persons who had syphilis, oral infections, or rheumatoid arthritis were tested by ELISAs with p37, p41-G, OspB, OspC, OspE, and OspF antigens. Although the results of class-specific ELISAs with recombinant antigens were comparable to those recorded for assays with whole-cell antigen and for individuals with confirmed clinical diagnoses of Lyme borreliosis, immunoblotting is still advised as an adjunct procedure, particularly when there are low antibody titers by an ELISA.

Journal ArticleDOI
TL;DR: It is concluded that, while caspase 1 is not indispensable for release of mature IL-1β in candidiasis, the casp enzyme 1-dependent production of IL-18 may represent an important and novel pathway for the expression of sustained Th1 reactivity to the fungus.
Abstract: Caspase 1, formerly designated interleukin 1β (IL-1β)-converting enzyme, processes pro-IL-1β and pro-IL-18 to yield active cytokines that play a pivotal role in inflammation and cell activation. We show here the effect of caspase 1 deficiency on the inflammatory and adaptive immune responses to the fungus Candida albicans. Caspase 1 deficiency did not affect susceptibility to primary systemic infection with the fungus, as revealed by survival and fungal growth. However, Th1-mediated resistance to reinfection was greatly impaired in caspase 1-deficient mice, and this correlated with low-level production of IL-12 and gamma interferon. Early in infection, production of these cytokines and that of tumor necrosis factor alpha, IL-6, and, interestingly, IL-1β occurred normally in caspase 1-deficient mice, while that of IL-18 was severely impaired. Exogenous administration of IL-18, more than IL-12, restored the Th1-mediated resistance to the infection. We conclude that, while caspase 1 is not indispensable for release of mature IL-1β in candidiasis, the caspase 1-dependent production of IL-18 may represent an important and novel pathway for the expression of sustained Th1 reactivity to the fungus.

Journal ArticleDOI
TL;DR: It is demonstrated that the negative regulation of Th2 cytokine production by the JNK1 signaling pathway is essential for generating Th1-polarized immunity against intracellular pathogens, such as Leishmania major.
Abstract: c-Jun N-terminal kinase (JNK) is a mitogen-activated protein kinase that plays important regulatory roles in helper T cell differentiation. In the current study, we used Jnk1-deficient mice to examine the function of JNK during an in vivo pathogenic infection, leishmaniasis, which is strongly influenced by Th1/Th2 effector mechanisms. The data show that Jnk1-deficient mice, despite their usually genetically resistant background, were unable to resolve Leishmania infections. Jnk1-/- mice displayed reduced delayed-type hypersensitivity in response to the pathogen, which was associated with a T cell defect. We found that, although these mice can direct an apparent Th1-response, there is also simultaneous generation of Leishmania-specific Th2 responses, which possibly down-modulate protective Th1-mediated immune function. These findings demonstrate that the negative regulation of Th2 cytokine production by the JNK1 signaling pathway is essential for generating Th1-polarized immunity against intracellular pathogens, such as Leishmania major.

Journal ArticleDOI
TL;DR: It appears that TNF-α can promote cross-presentation of islet antigen to CD8+ T cells using a unique CD40–CD154-independent pathway, providing new insights into the mechanisms by which inflammatory stimuli can bypass CD154–CD40 immune regulatory signals and cause activation of autoreactive T cells.
Abstract: Neonatal islet-specific expression of tumor necrosis factor (TNF)-α in nonobese diabetic mice promotes diabetes by provoking islet-infiltrating antigen-presenting cells to present islet peptides to autoreactive T cells. Here we show that TNF-α promotes autoaggression of both effector CD4+ and CD8+ T cells. Whereas CD8+ T cells are critical for diabetes progression, CD4+ T cells play a lesser role. TNF-α–mediated diabetes development was not dependent on CD154–CD40 signals or activated CD4+ T cells. Instead, it appears that TNF-α can promote cross-presentation of islet antigen to CD8+ T cells using a unique CD40–CD154-independent pathway. These data provide new insights into the mechanisms by which inflammatory stimuli can bypass CD154–CD40 immune regulatory signals and cause activation of autoreactive T cells.