117 results
Search Results
2. Nanotubes mediate niche-stem-cell signalling in the Drosophila testis.
- Author
-
Inaba, Mayu, Buszczak, Michael, and Yamashita, Yukiko M.
- Subjects
NANOTUBES ,STEM cells ,DROSOPHILA ,MICROTUBULES ,GERM cells ,RECEPTOR-ligand complexes ,CYTOKINES ,LIGANDS (Biochemistry) - Abstract
Stem cell niches provide resident stem cells with signals that specify their identity. Niche signals act over a short range such that only stem cells but not their differentiating progeny receive the self-renewing signals. However, the cellular mechanisms that limit niche signalling to stem cells remain poorly understood. Here we show that the Drosophila male germline stem cells form previously unrecognized structures, microtubule-based nanotubes, which extend into the hub, a major niche component. Microtubule-based nanotubes are observed specifically within germline stem cell populations, and require intraflagellar transport proteins for their formation. The bone morphogenetic protein (BMP) receptor Tkv localizes to microtubule-based nanotubes. Perturbation of microtubule-based nanotubes compromises activation of Dpp signalling within germline stem cells, leading to germline stem cell loss. Moreover, Dpp ligand and Tkv receptor interaction is necessary and sufficient for microtubule-based nanotube formation. We propose that microtubule-based nanotubes provide a novel mechanism for selective receptor-ligand interaction, contributing to the short-range nature of niche-stem-cell signalling. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
3. Antiviral immunity via RIG-I-mediated recognition of RNA bearing 5′-diphosphates.
- Author
-
Goubau, Delphine, Schlee, Martin, Deddouche, Safia, Rehwinkel, Jan, Pruijssers, Andrea J., Iskarpatyoti, Jason A., Zillinger, Thomas, Goldeck, Marion, Schuberth, Christine, Barchet, Winfried, Ludwig, Janos, Hartmann, Gunther, Van der Veen, Annemarthe G., Reis e Sousa, Caetano, Fujimura, Tsutomu, and Dermody, Terence S.
- Subjects
MAMMALIAN cell cycle ,RNA ,CYTOKINES ,GENOMES ,IMMUNE system ,REOVIRUSES - Abstract
Mammalian cells possess mechanisms to detect and defend themselves from invading viruses. In the cytosol, the RIG-I-like receptors (RLRs), RIG-I (retinoic acid-inducible gene I; encoded by DDX58) and MDA5 (melanoma differentiation-associated gene 5; encoded by IFIH1) sense atypical RNAs associated with virus infection. Detection triggers a signalling cascade via the adaptor MAVS that culminates in the production of type I interferons (IFN-α and β; hereafter IFN), which are key antiviral cytokines. RIG-I and MDA5 are activated by distinct viral RNA structures and much evidence indicates that RIG-I responds to RNAs bearing a triphosphate (ppp) moiety in conjunction with a blunt-ended, base-paired region at the 5′-end (reviewed in refs 1, 2, 3). Here we show that RIG-I also mediates antiviral responses to RNAs bearing 5′-diphosphates (5′pp). Genomes from mammalian reoviruses with 5′pp termini, 5′pp-RNA isolated from yeast L-A virus, and base-paired 5′pp-RNAs made by in vitro transcription or chemical synthesis, all bind to RIG-I and serve as RIG-I agonists. Furthermore, a RIG-I-dependent response to 5′pp-RNA is essential for controlling reovirus infection in cultured cells and in mice. Thus, the minimal determinant for RIG-I recognition is a base-paired RNA with 5′pp. Such RNAs are found in some viruses but not in uninfected cells, indicating that recognition of 5′pp-RNA, like that of 5′ppp-RNA, acts as a powerful means of self/non-self discrimination by the innate immune system. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
4. Endothelial-cell FAK targeting sensitizes tumours to DNA-damaging therapy.
- Author
-
Tavora, Bernardo, Alexopoulou, Annika, Hodivala-Dilke, Kairbaan M., Reynolds, Louise E., Batista, Silvia, Demircioglu, Fevzi, Fernandez, Isabelle, Lechertier, Tanguy, Lees, Delphine M., Wong, Ping-Pui, Elia, George, Clear, Andrew, Gribben, John G., Ledoux, Adeline, Hunter, Jill, and Perkins, Neil
- Subjects
DRUG resistance in cancer cells ,ENDOTHELIAL cells ,FOCAL adhesion kinase ,DNA damage ,APOPTOSIS ,LABORATORY mice ,CYTOKINES - Abstract
Chemoresistance is a serious limitation of cancer treatment. Until recently, almost all the work done to study this limitation has been restricted to tumour cells. Here we identify a novel molecular mechanism by which endothelial cells regulate chemosensitivity. We establish that specific targeting of focal adhesion kinase (FAK; also known as PTK2) in endothelial cells is sufficient to induce tumour-cell sensitization to DNA-damaging therapies and thus inhibit tumour growth in mice. The clinical relevance of this work is supported by our observations that low blood vessel FAK expression is associated with complete remission in human lymphoma. Our study shows that deletion of FAK in endothelial cells has no apparent effect on blood vessel function per se, but induces increased apoptosis and decreased proliferation within perivascular tumour-cell compartments of doxorubicin- and radiotherapy-treated mice. Mechanistically, we demonstrate that endothelial-cell FAK is required for DNA-damage-induced NF-κB activation in vivo and in vitro, and the production of cytokines from endothelial cells. Moreover, loss of endothelial-cell FAK reduces DNA-damage-induced cytokine production, thus enhancing chemosensitization of tumour cells to DNA-damaging therapies in vitro and in vivo. Overall, our data identify endothelial-cell FAK as a regulator of tumour chemosensitivity. Furthermore, we anticipate that this proof-of-principle data will be a starting point for the development of new possible strategies to regulate chemosensitization by targeting endothelial-cell FAK specifically. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
5. The alarmin IL-33 promotes regulatory T-cell function in the intestine.
- Author
-
Schiering, Chris, Krausgruber, Thomas, Chomka, Agnieszka, Adelmann, Krista, Griseri, Thibault, Bollrath, Julia, Hegazy, Ahmed N., Owens, Benjamin M. J., Powrie, Fiona, Fröhlich, Anja, Löhning, Max, Wohlfert, Elizabeth A., Pott, Johanna, Harrison, Oliver J., Belkaid, Yasmine, and Fallon, Padraic G.
- Subjects
INTERLEUKIN-33 ,INTESTINES ,TRANSFORMING growth factors-beta ,INFLAMMATORY bowel diseases ,CYTOKINES - Abstract
FOXP3
+ regulatory T cells (Treg cells) are abundant in the intestine, where they prevent dysregulated inflammatory responses to self and environmental stimuli. It is now appreciated that Treg cells acquire tissue-specific adaptations that facilitate their survival and function; however, key host factors controlling the Treg response in the intestine are poorly understood. The interleukin (IL)-1 family member IL-33 is constitutively expressed in epithelial cells at barrier sites, where it functions as an endogenous danger signal, or alarmin, in response to tissue damage. Recent studies in humans have described high levels of IL-33 in inflamed lesions of inflammatory bowel disease patients, suggesting a role for this cytokine in disease pathogenesis. In the intestine, both protective and pathological roles for IL-33 have been described in murine models of acute colitis, but its contribution to chronic inflammation remains ill defined. Here we show in mice that the IL-33 receptor ST2 is preferentially expressed on colonic Treg cells, where it promotes Treg function and adaptation to the inflammatory environment. IL-33 signalling in T cells stimulates Treg responses in several ways. First, it enhances transforming growth factor (TGF)-β1 -mediated differentiation of Treg cells and, second, it provides a necessary signal for Treg -cell accumulation and maintenance in inflamed tissues. Strikingly, IL-23, a key pro-inflammatory cytokine in the pathogenesis of inflammatory bowel disease, restrained Treg responses through inhibition of IL-33 responsiveness. These results demonstrate a hitherto unrecognized link between an endogenous mediator of tissue damage and a major anti-inflammatory pathway, and suggest that the balance between IL-33 and IL-23 may be a key controller of intestinal immune responses. [ABSTRACT FROM AUTHOR]- Published
- 2014
- Full Text
- View/download PDF
6. Sessile alveolar macrophages communicate with alveolar epithelium to modulate immunity.
- Author
-
Westphalen, Kristin, Gusarova, Galina A., Islam, Mohammad N., Subramanian, Manikandan, Cohen, Taylor S., Prince, Alice S., and Bhattacharya, Jahar
- Subjects
PHAGOCYTES ,ALVEOLAR macrophages ,MACROPHAGES ,CYTOKINES ,PATHOGENIC microorganisms - Abstract
The tissue-resident macrophages of barrier organs constitute the first line of defence against pathogens at the systemic interface with the ambient environment. In the lung, resident alveolar macrophages (AMs) provide a sentinel function against inhaled pathogens. Bacterial constituents ligate Toll-like receptors (TLRs) on AMs, causing AMs to secrete proinflammatory cytokines that activate alveolar epithelial receptors, leading to recruitment of neutrophils that engulf pathogens. Because the AM-induced response could itself cause tissue injury, it is unclear how AMs modulate the response to prevent injury. Here, using real-time alveolar imaging in situ, we show that a subset of AMs attached to the alveolar wall form connexin 43 (Cx43)-containing gap junction channels with the epithelium. During lipopolysaccharide-induced inflammation, the AMs remained sessile and attached to the alveoli, and they established intercommunication through synchronized Ca
2+ waves, using the epithelium as the conducting pathway. The intercommunication was immunosuppressive, involving Ca2+ -dependent activation of Akt, because AM-specific knockout of Cx43 enhanced alveolar neutrophil recruitment and secretion of proinflammatory cytokines in the bronchoalveolar lavage. A picture emerges of a novel immunomodulatory process in which a subset of alveolus-attached AMs intercommunicates immunosuppressive signals to reduce endotoxin-induced lung inflammation. [ABSTRACT FROM AUTHOR]- Published
- 2014
- Full Text
- View/download PDF
7. Cell death by pyroptosis drives CD4 T-cell depletion in HIV-1 infection.
- Author
-
Doitsh, Gilad, Galloway, Nicole L. K., Geng, Xin, Yang, Zhiyuan, Monroe, Kathryn M., Zepeda, Orlando, Hunt, Peter W., Hatano, Hiroyu, Sowinski, Stefanie, Muñoz-Arias, Isa, and Greene, Warner C.
- Subjects
CELL death ,CD4 antigen ,T cells ,HIV-positive persons ,CASPASES ,CYTOPLASM ,INFLAMMATION ,CYTOKINES - Abstract
The pathway causing CD4 T-cell death in HIV-infected hosts remains poorly understood although apoptosis has been proposed as a key mechanism. We now show that caspase-3-mediated apoptosis accounts for the death of only a small fraction of CD4 T cells corresponding to those that are both activated and productively infected. The remaining over 95% of quiescent lymphoid CD4 T cells die by caspase-1-mediated pyroptosis triggered by abortive viral infection. Pyroptosis corresponds to an intensely inflammatory form of programmed cell death in which cytoplasmic contents and pro-inflammatory cytokines, including IL-1β, are released. This death pathway thus links the two signature events in HIV infection-CD4 T-cell depletion and chronic inflammation-and creates a pathogenic vicious cycle in which dying CD4 T cells release inflammatory signals that attract more cells to die. This cycle can be broken by caspase 1 inhibitors shown to be safe in humans, raising the possibility of a new class of 'anti-AIDS' therapeutics targeting the host rather than the virus. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
8. Immunosuppressive CD71+ erythroid cells compromise neonatal host defence against infection.
- Author
-
Elahi, Shokrollah, Ertelt, James M., Kinder, Jeremy M., Jiang, Tony T., Zhang, Xuzhe, Xin, Lijun, Chaturvedi, Vandana, Strong, Beverly S., Qualls, Joseph E., Steinbrecher, Kris A., Kalfa, Theodosia A., Shaaban, Aimen F., and Way, Sing Sing
- Subjects
ERYTHROCYTE membranes ,IMMUNOSUPPRESSIVE agents ,FETAL immunology ,NEONATOLOGY ,CYTOKINES ,LABORATORY mice - Abstract
Newborn infants are highly susceptible to infection. This defect in host defence has generally been ascribed to the immaturity of neonatal immune cells; however, the degree of hyporesponsiveness is highly variable and depends on the stimulation conditions. These discordant responses illustrate the need for a more unified explanation for why immunity is compromised in neonates. Here we show that physiologically enriched CD71
+ erythroid cells in neonatal mice and human cord blood have distinctive immunosuppressive properties. The production of innate immune protective cytokines by adult cells is diminished after transfer to neonatal mice or after co-culture with neonatal splenocytes. Neonatal CD71+ cells express the enzyme arginase-2, and arginase activity is essential for the immunosuppressive properties of these cells because molecular inhibition of this enzyme or supplementation with l-arginine overrides immunosuppression. In addition, the ablation of CD71+ cells in neonatal mice, or the decline in number of these cells as postnatal development progresses parallels the loss of suppression, and restored resistance to the perinatal pathogens Listeria monocytogenes and Escherichia coli. However, CD71+ cell-mediated susceptibility to infection is counterbalanced by CD71+ cell-mediated protection against aberrant immune cell activation in the intestine, where colonization with commensal microorganisms occurs swiftly after parturition. Conversely, circumventing such colonization by using antimicrobials or gnotobiotic germ-free mice overrides these protective benefits. Thus, CD71+ cells quench the excessive inflammation induced by abrupt colonization with commensal microorganisms after parturition. This finding challenges the idea that the susceptibility of neonates to infection reflects immune-cell-intrinsic defects and instead highlights processes that are developmentally more essential and inadvertently mitigate innate immune protection. We anticipate that these results will spark renewed investigation into the need for immunosuppression in neonates, as well as improved strategies for augmenting host defence in this vulnerable population. [ABSTRACT FROM AUTHOR]- Published
- 2013
- Full Text
- View/download PDF
9. Induction of mouse germ-cell fate by transcription factors in vitro.
- Author
-
Nakaki, Fumio, Hayashi, Katsuhiko, Ohta, Hiroshi, Kurimoto, Kazuki, Yabuta, Yukihiro, and Saitou, Mitinori
- Subjects
GERM cells ,GAMETES ,ZYGOTES ,CYTOKINES ,EMBRYONIC stem cell research ,TRANSCRIPTION factors ,SPERMATOGENESIS ,OOGENESIS - Abstract
The germ-cell lineage ensures the continuity of life through the generation of male and female gametes, which unite to form a totipotent zygote. We have previously demonstrated that, by using cytokines, embryonic stem cells and induced pluripotent stem cells can be induced into epiblast-like cells (EpiLCs) and then into primordial germ cell (PGC)-like cells with the capacity for both spermatogenesis and oogenesis, creating an opportunity for understanding and regulating mammalian germ-cell development in both sexes in vitro. Here we show that, without cytokines, simultaneous overexpression of three transcription factors, Blimp1 (also known as Prdm1), Prdm14 and Tfap2c (also known as AP2γ), directs EpiLCs, but not embryonic stem cells, swiftly and efficiently into a PGC state. Notably, Prdm14 alone, but not Blimp1 or Tfap2c, suffices for the induction of the PGC state in EpiLCs. The transcription-factor-induced PGC state, irrespective of the transcription factors used, reconstitutes key transcriptome and epigenetic reprogramming in PGCs, but bypasses a mesodermal program that accompanies PGC or PGC-like-cell specification by cytokines including bone morphogenetic protein 4. Notably, the transcription-factor-induced PGC-like cells contribute to spermatogenesis and fertile offspring. Our findings provide a new insight into the transcriptional logic for PGC specification, and create a foundation for the transcription-factor-based reconstitution and regulation of mammalian gametogenesis. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
10. Pathogen blocks host death receptor signalling by arginine GlcNAcylation of death domains.
- Author
-
Li, Shan, Zhang, Li, Yao, Qing, Li, Lin, Dong, Na, Rong, Jie, Gao, Wenqing, Ding, Xiaojun, Sun, Liming, Chen, Xing, Chen, She, and Shao, Feng
- Subjects
TUMOR necrosis factor receptors ,CYTOKINES ,DEATH receptors ,ARGININE ,HOMEOSTASIS ,CELL death ,INFLAMMATION - Abstract
The tumour necrosis factor (TNF) family is crucial for immune homeostasis, cell death and inflammation. These cytokines are recognized by members of the TNF receptor (TNFR) family of death receptors, including TNFR1 and TNFR2, and FAS and TNF-related apoptosis-inducing ligand (TRAIL) receptors. Death receptor signalling requires death-domain-mediated homotypic/heterotypic interactions between the receptor and its downstream adaptors, including TNFR1-associated death domain protein (TRADD) and FAS-associated death domain protein (FADD). Here we discover that death domains in several proteins, including TRADD, FADD, RIPK1 and TNFR1, were directly inactivated by NleB, an enteropathogenic Escherichia coli (EPEC) type III secretion system effector known to inhibit host nuclear factor-κB (NF-κB) signalling. NleB contained an unprecedented N-acetylglucosamine (GlcNAc) transferase activity that specifically modified a conserved arginine in these death domains (Arg 235 in the TRADD death domain). NleB GlcNAcylation (the addition of GlcNAc onto a protein side chain) of death domains blocked homotypic/heterotypic death domain interactions and assembly of the oligomeric TNFR1 complex, thereby disrupting TNF signalling in EPEC-infected cells, including NF-κB signalling, apoptosis and necroptosis. Type-III-delivered NleB also blocked FAS ligand and TRAIL-induced cell death by preventing formation of a FADD-mediated death-inducing signalling complex (DISC). The arginine GlcNAc transferase activity of NleB was required for bacterial colonization in the mouse model of EPEC infection. The mechanism of action of NleB represents a new model by which bacteria counteract host defences, and also a previously unappreciated post-translational modification. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
11. Structural mechanism of cytosolic DNA sensing by cGAS.
- Author
-
Civril, Filiz, Deimling, Tobias, de Oliveira Mann, Carina C., Ablasser, Andrea, Moldt, Manuela, Witte, Gregor, Hornung, Veit, and Hopfner, Karl-Peter
- Subjects
BIOSENSORS ,CYTOSOL ,DNA analysis ,MOLECULAR structure ,CYTOKINES ,INFLAMMATION - Abstract
Cytosolic DNA arising from intracellular bacterial or viral infections is a powerful pathogen-associated molecular pattern (PAMP) that leads to innate immune host defence by the production of type I interferon and inflammatory cytokines. Recognition of cytosolic DNA by the recently discovered cyclic-GMP-AMP (cGAMP) synthase (cGAS) induces the production of cGAMP to activate the stimulator of interferon genes (STING). Here we report the crystal structure of cGAS alone and in complex with DNA, ATP and GTP along with functional studies. Our results explain the broad DNA sensing specificity of cGAS, show how cGAS catalyses dinucleotide formation and indicate activation by a DNA-induced structural switch. cGAS possesses a remarkable structural similarity to the antiviral cytosolic double-stranded RNA sensor 2′-5′oligoadenylate synthase (OAS1), but contains a unique zinc thumb that recognizes B-form double-stranded DNA. Our results mechanistically unify dsRNA and dsDNA innate immune sensing by OAS1 and cGAS nucleotidyl transferases. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
12. Innate lymphoid cells regulate CD4+ T-cell responses to intestinal commensal bacteria.
- Author
-
Hepworth, Matthew R., Monticelli, Laurel A., Fung, Thomas C., Ziegler, Carly G. K., Grunberg, Stephanie, Sinha, Rohini, Mantegazza, Adriana R., Ma, Hak-Ling, Crawford, Alison, Angelosanto, Jill M., Wherry, E. John, Koni, Pandelakis A., Bushman, Frederic D., Elson, Charles O., Eberl, Gérard, Artis, David, and Sonnenberg, Gregory F.
- Subjects
LYMPHOID tissue ,CD4 antigen ,CELLULAR control mechanisms ,T cells ,GUT microbiome ,NATURAL immunity ,CYTOKINES - Abstract
Innate lymphoid cells (ILCs) are a recently characterized family of immune cells that have critical roles in cytokine-mediated regulation of intestinal epithelial cell barrier integrity. Alterations in ILC responses are associated with multiple chronic human diseases, including inflammatory bowel disease, implicating a role for ILCs in disease pathogenesis. Owing to an inability to target ILCs selectively, experimental studies assessing ILC function have predominantly used mice lacking adaptive immune cells. However, in lymphocyte-sufficient hosts ILCs are vastly outnumbered by CD4
+ T cells, which express similar profiles of effector cytokines. Therefore, the function of ILCs in the presence of adaptive immunity and their potential to influence adaptive immune cell responses remain unknown. To test this, we used genetic or antibody-mediated depletion strategies to target murine ILCs in the presence of an adaptive immune system. We show that loss of retinoic-acid-receptor-related orphan receptor-γt-positive (RORγt+ ) ILCs was associated with dysregulated adaptive immune cell responses against commensal bacteria and low-grade systemic inflammation. Remarkably, ILC-mediated regulation of adaptive immune cells occurred independently of interleukin (IL)-17A, IL-22 or IL-23. Genome-wide transcriptional profiling and functional analyses revealed that RORγt+ ILCs express major histocompatibility complex class II (MHCII) and can process and present antigen. However, rather than inducing T-cell proliferation, ILCs acted to limit commensal bacteria-specific CD4+ T-cell responses. Consistent with this, selective deletion of MHCII in murine RORγt+ ILCs resulted in dysregulated commensal bacteria-dependent CD4+ T-cell responses that promoted spontaneous intestinal inflammation. These data identify that ILCs maintain intestinal homeostasis through MHCII-dependent interactions with CD4+ T cells that limit pathological adaptive immune cell responses to commensal bacteria. [ABSTRACT FROM AUTHOR]- Published
- 2013
- Full Text
- View/download PDF
13. M-CSF instructs myeloid lineage fate in single haematopoietic stem cells.
- Author
-
Mossadegh-Keller, Noushine, Sarrazin, Sandrine, Kandalla, Prashanth K., Espinosa, Leon, Stanley, E. Richard, Nutt, Stephen L., Moore, Jordan, and Sieweke, Michael H.
- Subjects
STEM cells ,INFLAMMATION ,PROGENITOR cells ,TRANSCRIPTION factors ,CYTOKINES ,GENE expression - Abstract
Under stress conditions such as infection or inflammation the body rapidly needs to generate new blood cells that are adapted to the challenge. Haematopoietic cytokines are known to increase output of specific mature cells by affecting survival, expansion and differentiation of lineage-committed progenitors, but it has been debated whether long-term haematopoietic stem cells (HSCs) are susceptible to direct lineage-specifying effects of cytokines. Although genetic changes in transcription factor balance can sensitize HSCs to cytokine instruction, the initiation of HSC commitment is generally thought to be triggered by stochastic fluctuation in cell-intrinsic regulators such as lineage-specific transcription factors, leaving cytokines to ensure survival and proliferation of the progeny cells. Here we show that macrophage colony-stimulating factor (M-CSF, also called CSF1), a myeloid cytokine released during infection and inflammation, can directly induce the myeloid master regulator PU.1 and instruct myeloid cell-fate change in mouse HSCs, independently of selective survival or proliferation. Video imaging and single-cell gene expression analysis revealed that stimulation of highly purified HSCs with M-CSF in culture resulted in activation of the PU.1 promoter and an increased number of PU.1
+ cells with myeloid gene signature and differentiation potential. In vivo, high systemic levels of M-CSF directly stimulated M-CSF-receptor-dependent activation of endogenous PU.1 protein in single HSCs and induced a PU.1-dependent myeloid differentiation preference. Our data demonstrate that lineage-specific cytokines can act directly on HSCs in vitro and in vivo to instruct a change of cell identity. This fundamentally changes the current view of how HSCs respond to environmental challenge and implicates stress-induced cytokines as direct instructors of HSC fate. [ABSTRACT FROM AUTHOR]- Published
- 2013
- Full Text
- View/download PDF
14. Sodium chloride drives autoimmune disease by the induction of pathogenic TH17 cells.
- Author
-
Kleinewietfeld, Markus, Manzel, Arndt, Titze, Jens, Kvakan, Heda, Yosef, Nir, Linker, Ralf A., Muller, Dominik N., and Hafler, David A.
- Subjects
GENETICS of autoimmune diseases ,SALT ,PATHOGENIC microorganisms ,T cells ,IR genes ,CYTOKINES - Abstract
There has been a marked increase in the incidence of autoimmune diseases in the past half-century. Although the underlying genetic basis of this class of diseases has recently been elucidated, implicating predominantly immune-response genes, changes in environmental factors must ultimately be driving this increase. The newly identified population of interleukin (IL)-17-producing CD4
+ helper T cells (TH 17 cells) has a pivotal role in autoimmune diseases. Pathogenic IL-23-dependent TH 17 cells have been shown to be critical for the development of experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis, and genetic risk factors associated with multiple sclerosis are related to the IL-23-TH 17 pathway. However, little is known about the environmental factors that directly influence TH 17 cells. Here we show that increased salt (sodium chloride, NaCl) concentrations found locally under physiological conditions in vivo markedly boost the induction of murine and human TH 17 cells. High-salt conditions activate the p38/MAPK pathway involving nuclear factor of activated T cells 5 (NFAT5; also called TONEBP) and serum/glucocorticoid-regulated kinase 1 (SGK1) during cytokine-induced TH 17 polarization. Gene silencing or chemical inhibition of p38/MAPK, NFAT5 or SGK1 abrogates the high-salt-induced TH 17 cell development. The TH 17 cells generated under high-salt conditions display a highly pathogenic and stable phenotype characterized by the upregulation of the pro-inflammatory cytokines GM-CSF, TNF-α and IL-2. Moreover, mice fed with a high-salt diet develop a more severe form of EAE, in line with augmented central nervous system infiltrating and peripherally induced antigen-specific TH 17 cells. Thus, increased dietary salt intake might represent an environmental risk factor for the development of autoimmune diseases through the induction of pathogenic TH 17 cells. [ABSTRACT FROM AUTHOR]- Published
- 2013
- Full Text
- View/download PDF
15. T-helper-1-cell cytokines drive cancer into senescence.
- Author
-
Braumüller, Heidi, Wieder, Thomas, Brenner, Ellen, Aßmann, Sonja, Hahn, Matthias, Alkhaled, Mohammed, Schilbach, Karin, Essmann, Frank, Kneilling, Manfred, Griessinger, Christoph, Ranta, Felicia, Ullrich, Susanne, Mocikat, Ralph, Braungart, Kilian, Mehra, Tarun, Fehrenbacher, Birgit, Berdel, Julia, Niessner, Heike, Meier, Friedegund, and van den Broek, Maries
- Subjects
T helper cells ,CYTOKINES ,TUMOR growth ,CANCER cell proliferation ,TUMOR necrosis factors ,CELL cycle ,CANCER invasiveness - Abstract
Cancer control by adaptive immunity involves a number of defined death and clearance mechanisms. However, efficient inhibition of exponential cancer growth by T cells and interferon-γ (IFN-γ) requires additional undefined mechanisms that arrest cancer cell proliferation. Here we show that the combined action of the T-helper-1-cell cytokines IFN-γ and tumour necrosis factor (TNF) directly induces permanent growth arrest in cancers. To safely separate senescence induced by tumour immunity from oncogene-induced senescence, we used a mouse model in which the Simian virus 40 large T antigen (Tag) expressed under the control of the rat insulin promoter creates tumours by attenuating p53- and Rb-mediated cell cycle control. When combined, IFN-γ and TNF drive Tag-expressing cancers into senescence by inducing permanent growth arrest in G1/G0, activation of p16INK4a (also known as CDKN2A), and downstream Rb hypophosphorylation at serine 795. This cytokine-induced senescence strictly requires STAT1 and TNFR1 (also known as TNFRSF1A) signalling in addition to p16INK4a. In vivo, Tag-specific T-helper 1 cells permanently arrest Tag-expressing cancers by inducing IFN-γ- and TNFR1-dependent senescence. Conversely, Tnfr1
−/− Tag-expressing cancers resist cytokine-induced senescence and grow aggressively, even in TNFR1-expressing hosts. Finally, as IFN-γ and TNF induce senescence in numerous murine and human cancers, this may be a general mechanism for arresting cancer progression. [ABSTRACT FROM AUTHOR]- Published
- 2013
- Full Text
- View/download PDF
16. Mitotic cell rounding accelerates epithelial invagination.
- Author
-
Kondo, Takefumi and Hayashi, Shigeo
- Subjects
EPITHELIAL cells ,CYTOPLASM ,CELL division ,GROWTH factors ,CYTOKINES - Abstract
Mitotic cells assume a spherical shape by increasing their surface tension and osmotic pressure by extensively reorganizing their interphase actin cytoskeleton into a cortical meshwork and their microtubules into the mitotic spindle. Mitotic entry is known to interfere with tissue morphogenetic events that require cell-shape changes controlled by the interphase cytoskeleton, such as apical constriction. However, here we show that mitosis plays an active role in the epithelial invagination of the Drosophila melanogaster tracheal placode. Invagination begins with a slow phase under the control of epidermal growth factor receptor (EGFR) signalling; in this process, the central apically constricted cells, which are surrounded by intercalating cells, form a shallow pit. This slow phase is followed by a fast phase, in which the pit is rapidly depressed, accompanied by mitotic entry, which leads to the internalization of all the cells in the placode. We found that mitotic cell rounding, but not cell division, of the central cells in the placode is required to accelerate invagination, in conjunction with EGFR-induced myosin II contractility in the surrounding cells. We propose that mitotic cell rounding causes the epithelium to buckle under pressure and acts as a switch for morphogenetic transition at the appropriate time. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
17. How insulin engages its primary binding site on the insulin receptor.
- Author
-
Menting, John G., Whittaker, Jonathan, Margetts, Mai B., Whittaker, Linda J., Kong, Geoffrey K.-W., Smith, Brian J., Watson, Christopher J., Žáková, Lenka, Kletvíková, Emília, Jirá?ek, Ji?í, Chan, Shu Jin, Steiner, Donald F., Dodson, Guy G., Brzozowski, Andrzej M., Weiss, Michael A., Ward, Colin W., and Lawrence, Michael C.
- Subjects
INSULIN ,TYPE 2 diabetes ,GROWTH factors ,CYTOKINES ,ALZHEIMER'S disease - Abstract
Insulin receptor signalling has a central role in mammalian biology, regulating cellular metabolism, growth, division, differentiation and survival. Insulin resistance contributes to the pathogenesis of type 2 diabetes mellitus and the onset of Alzheimer's disease; aberrant signalling occurs in diverse cancers, exacerbated by cross-talk with the homologous type 1 insulin-like growth factor receptor (IGF1R). Despite more than three decades of investigation, the three-dimensional structure of the insulin-insulin receptor complex has proved elusive, confounded by the complexity of producing the receptor protein. Here we present the first view, to our knowledge, of the interaction of insulin with its primary binding site on the insulin receptor, on the basis of four crystal structures of insulin bound to truncated insulin receptor constructs. The direct interaction of insulin with the first leucine-rich-repeat domain (L1) of insulin receptor is seen to be sparse, the hormone instead engaging the insulin receptor carboxy-terminal ?-chain (?CT) segment, which is itself remodelled on the face of L1 upon insulin binding. Contact between insulin and L1 is restricted to insulin B-chain residues. The ?CT segment displaces the B-chain C-terminal ?-strand away from the hormone core, revealing the mechanism of a long-proposed conformational switch in insulin upon receptor engagement. This mode of hormone-receptor recognition is novel within the broader family of receptor tyrosine kinases. We support these findings by photo-crosslinking data that place the suggested interactions into the context of the holoreceptor and by isothermal titration calorimetry data that dissect the hormone-insulin receptor interface. Together, our findings provide an explanation for a wealth of biochemical data from the insulin receptor and IGF1R systems relevant to the design of therapeutic insulin analogues. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
18. The NAD-dependent deacetylase SIRT2 is required for programmed necrosis.
- Author
-
Narayan, Nisha, Lee, In Hye, Borenstein, Ronen, Sun, Junhui, Wong, Renee, Tong, Guang, Fergusson, Maria M., Liu, Jie, Rovira, Ilsa I., Cheng, Hwei-Ling, Wang, Guanghui, Gucek, Marjan, Lombard, David, Alt, Fredrick W., Sack, Michael N., Murphy, Elizabeth, Cao, Liu, and Finkel, Toren
- Subjects
TUMOR necrosis factors ,CYTOKINES ,GLYCOPROTEINS ,GROWTH factors ,DEACETYLATION - Abstract
Although initially viewed as unregulated, increasing evidence suggests that cellular necrosis often proceeds through a specific molecular program. In particular, death ligands such as tumour necrosis factor (TNF)-? activate necrosis by stimulating the formation of a complex containing receptor-interacting protein 1 (RIP1) and receptor-interacting protein 3 (RIP3). Relatively little is known regarding how this complex formation is regulated. Here, we show that the NAD-dependent deacetylase SIRT2 binds constitutively to RIP3 and that deletion or knockdown of SIRT2 prevents formation of the RIP1-RIP3 complex in mice. Furthermore, genetic or pharmacological inhibition of SIRT2 blocks cellular necrosis induced by TNF-?. We further demonstrate that RIP1 is a critical target of SIRT2-dependent deacetylation. Using gain- and loss-of-function mutants, we demonstrate that acetylation of RIP1 lysine 530 modulates RIP1-RIP3 complex formation and TNF-?-stimulated necrosis. In the setting of ischaemia-reperfusion injury, RIP1 is deacetylated in a SIRT2-dependent fashion. Furthermore, the hearts of Sirt2
?/? mice, or wild-type mice treated with a specific pharmacological inhibitor of SIRT2, show marked protection from ischaemic injury. Taken together, these results implicate SIRT2 as an important regulator of programmed necrosis and indicate that inhibitors of this deacetylase may constitute a novel approach to protect against necrotic injuries, including ischaemic stroke and myocardial infarction. [ABSTRACT FROM AUTHOR]- Published
- 2012
- Full Text
- View/download PDF
19. Compensatory dendritic cell development mediated by BATF-IRF interactions.
- Author
-
Tussiwand, Roxane, Lee, Wan-Ling, Murphy, Theresa L., Mashayekhi, Mona, KC, Wumesh, Albring, Jörn C., Satpathy, Ansuman T., Rotondo, Jeffrey A., Edelson, Brian T., Kretzer, Nicole M., Wu, Xiaodi, Weiss, Leslie A., Glasmacher, Elke, Li, Peng, Liao, Wei, Behnke, Michael, Lam, Samuel S. K., Aurthur, Cora T., Leonard, Warren J., and Singh, Harinder
- Subjects
DENDRITIC cells ,INTRACELLULAR pathogens ,T cells ,CYTOKINES ,LEUCINE zippers ,VACCINES - Abstract
The AP1 transcription factor Batf3 is required for homeostatic development of CD8?
+ classical dendritic cells that prime CD8 T-cell responses against intracellular pathogens. Here we identify an alternative, Batf3-independent pathway in mice for CD8?+ dendritic cell development operating during infection with intracellular pathogens and mediated by the cytokines interleukin (IL)-12 and interferon-?. This alternative pathway results from molecular compensation for Batf3 provided by the related AP1 factors Batf, which also functions in T and B cells, and Batf2 induced by cytokines in response to infection. Reciprocally, physiological compensation between Batf and Batf3 also occurs in T cells for expression of IL-10 and CTLA4. Compensation among BATF factors is based on the shared capacity of their leucine zipper domains to interact with non-AP1 factors such as IRF4 and IRF8 to mediate cooperative gene activation. Conceivably, manipulating this alternative pathway of dendritic cell development could be of value in augmenting immune responses to vaccines. [ABSTRACT FROM AUTHOR]- Published
- 2012
- Full Text
- View/download PDF
20. Melanomas resist T-cell therapy through inflammation-induced reversible dedifferentiation.
- Author
-
Landsberg, Jennifer, Kohlmeyer, Judith, Renn, Marcel, Bald, Tobias, Rogava, Meri, Cron, Mira, Fatho, Martina, Lennerz, Volker, Wölfel, Thomas, Hölzel, Michael, and Tüting, Thomas
- Subjects
MELANOMA treatment ,T cells ,CYTOKINES ,TUMOR necrosis factors ,ANTIGENS ,CANCER relapse ,DISEASE remission ,SPONTANEOUS cancer regression ,THERAPEUTICS - Abstract
Adoptive cell transfer therapies (ACTs) with cytotoxic T cells that target melanocytic antigens can achieve remissions in patients with metastatic melanomas, but tumours frequently relapse. Hypotheses explaining the acquired resistance to ACTs include the selection of antigen-deficient tumour cell variants and the induction of T-cell tolerance. However, the lack of appropriate experimental melanoma models has so far impeded clear insights into the underlying mechanisms. Here we establish an effective ACT protocol in a genetically engineered mouse melanoma model that recapitulates tumour regression, remission and relapse as seen in patients. We report the unexpected observation that melanomas acquire ACT resistance through an inflammation-induced reversible loss of melanocytic antigens. In serial transplantation experiments, melanoma cells switch between a differentiated and a dedifferentiated phenotype in response to T-cell-driven inflammatory stimuli. We identified the proinflammatory cytokine tumour necrosis factor (TNF)-? as a crucial factor that directly caused reversible dedifferentiation of mouse and human melanoma cells. Tumour cells exposed to TNF-? were poorly recognized by T cells specific for melanocytic antigens, whereas recognition by T cells specific for non-melanocytic antigens was unaffected or even increased. Our results demonstrate that the phenotypic plasticity of melanoma cells in an inflammatory microenvironment contributes to tumour relapse after initially successful T-cell immunotherapy. On the basis of our work, we propose that future ACT protocols should simultaneously target melanocytic and non-melanocytic antigens to ensure broad recognition of both differentiated and dedifferentiated melanoma cells, and include strategies to sustain T-cell effector functions by blocking immune-inhibitory mechanisms in the tumour microenvironment. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
21. Novel role of PKR in inflammasome activation and HMGB1 release.
- Author
-
Lu, Ben, Nakamura, Takahisa, Inouye, Karen, Li, Jianhua, Tang, Yiting, Lundbäck, Peter, Valdes-Ferrer, Sergio I., Olofsson, Peder S., Kalb, Thomas, Roth, Jesse, Zou, Yongrui, Erlandsson-Harris, Helena, Yang, Huan, Ting, Jenny P.-Y., Wang, Haichao, Andersson, Ulf, Antoine, Daniel J., Chavan, Sangeeta S., Hotamisligil, Gökhan S., and Tracey, Kevin J.
- Subjects
PROTEIN kinases ,CYTOKINES ,RNA ,MACROPHAGE activation ,AUTOPHOSPHORYLATION ,ESCHERICHIA coli ,GENE transfection - Abstract
The inflammasome regulates the release of caspase activation-dependent cytokines, including interleukin (IL)-1?, IL-18 and high-mobility group box 1 (HMGB1). By studying HMGB1 release mechanisms, here we identify a role for double-stranded RNA-dependent protein kinase (PKR, also known as EIF2AK2) in inflammasome activation. Exposure of macrophages to inflammasome agonists induced PKR autophosphorylation. PKR inactivation by genetic deletion or pharmacological inhibition severely impaired inflammasome activation in response to double-stranded RNA, ATP, monosodium urate, adjuvant aluminium, rotenone, live Escherichia coli, anthrax lethal toxin, DNA transfection and Salmonella typhimurium infection. PKR deficiency significantly inhibited the secretion of IL-1?, IL-18 and HMGB1 in E. coli-induced peritonitis. PKR physically interacts with several inflammasome components, including NOD-like receptor (NLR) family pyrin domain-containing 3 (NLRP3), NLRP1, NLR family CARD domain-containing protein 4 (NLRC4), absent in melanoma 2 (AIM2), and broadly regulates inflammasome activation. PKR autophosphorylation in a cell-free system with recombinant NLRP3, apoptosis-associated speck-like protein containing a CARD (ASC, also known as PYCARD) and pro-caspase-1 reconstitutes inflammasome activity. These results show a crucial role for PKR in inflammasome activation, and indicate that it should be possible to pharmacologically target this molecule to treat inflammation. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
22. ZNRF3 promotes Wnt receptor turnover in an R-spondin-sensitive manner.
- Author
-
Hao, Huai-Xiang, Xie, Yang, Zhang, Yue, Charlat, Olga, Oster, Emma, Avello, Monika, Lei, Hong, Mickanin, Craig, Liu, Dong, Ruffner, Heinz, Mao, Xiaohong, Ma, Qicheng, Zamponi, Raffaella, Bouwmeester, Tewis, Finan, Peter M., Kirschner, Marc W., Porter, Jeffery A., Serluca, Fabrizio C., and Cong, Feng
- Subjects
PROTEINS ,GROWTH factors ,CELL polarity ,CYTOKINES ,STEM cells ,CELL receptors - Abstract
R-spondin proteins strongly potentiate Wnt signalling and function as stem-cell growth factors. Despite the biological and therapeutic significance, the molecular mechanism of R-spondin action remains unclear. Here we show that the cell-surface transmembrane E3 ubiquitin ligase zinc and ring finger 3 (ZNRF3) and its homologue ring finger 43 (RNF43) are negative feedback regulators of Wnt signalling. ZNRF3 is associated with the Wnt receptor complex, and inhibits Wnt signalling by promoting the turnover of frizzled and LRP6. Inhibition of ZNRF3 enhances Wnt/?-catenin signalling and disrupts Wnt/planar cell polarity signalling in vivo. Notably, R-spondin mimics ZNRF3 inhibition by increasing the membrane level of Wnt receptors. Mechanistically, R-spondin interacts with the extracellular domain of ZNRF3 and induces the association between ZNRF3 and LGR4, which results in membrane clearance of ZNRF3. These data suggest that R-spondin enhances Wnt signalling by inhibiting ZNRF3. Our study provides new mechanistic insights into the regulation of Wnt receptor turnover, and reveals ZNRF3 as a tractable target for therapeutic exploration. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
23. Acquisition of a multifunctional IgA+ plasma cell phenotype in the gut.
- Author
-
Fritz, Jörg H., Rojas, Olga Lucia, Simard, Nathalie, McCarthy, Douglas D., Hapfelmeier, Siegfried, Rubino, Stephen, Robertson, Susan J., Larijani, Mani, Gosselin, Jean, Ivanov, Ivaylo I., Martin, Alberto, Casellas, Rafael, Philpott, Dana J., Girardin, Stephen E., McCoy, Kathy D., Macpherson, Andrew J., Paige, Christopher J., and Gommerman, Jennifer L.
- Subjects
PLASMA cells ,GASTROINTESTINAL system ,PHENOTYPES ,IMMUNOGLOBULIN receptors ,BACTERIA ,CYTOKINES - Abstract
The largest mucosal surface in the body is in the gastrointestinal tract, a location that is heavily colonized by microbes that are normally harmless. A key mechanism required for maintaining a homeostatic balance between this microbial burden and the lymphocytes that densely populate the gastrointestinal tract is the production and transepithelial transport of poly-reactive IgA (ref. 1). Within the mucosal tissues, B cells respond to cytokines, sometimes in the absence of T-cell help, undergo class switch recombination of their immunoglobulin receptor to IgA, and differentiate to become plasma cells. However, IgA-secreting plasma cells probably have additional attributes that are needed for coping with the tremendous bacterial load in the gastrointestinal tract. Here we report that mouse IgA
+ plasma cells also produce the antimicrobial mediators tumour-necrosis factor-? (TNF-?) and inducible nitric oxide synthase (iNOS), and express many molecules that are commonly associated with monocyte/granulocytic cell types. The development of iNOS-producing IgA+ plasma cells can be recapitulated in vitro in the presence of gut stroma, and the acquisition of this multifunctional phenotype in vivo and in vitro relies on microbial co-stimulation. Deletion of TNF-? and iNOS in B-lineage cells resulted in a reduction in IgA production, altered diversification of the gut microbiota and poor clearance of a gut-tropic pathogen. These findings reveal a novel adaptation to maintaining homeostasis in the gut, and extend the repertoire of protective responses exhibited by some B-lineage cells. [ABSTRACT FROM AUTHOR]- Published
- 2012
- Full Text
- View/download PDF
24. Sustained axon regeneration induced by co-deletion of PTEN and SOCS3.
- Author
-
Sun, Fang, Park, Kevin K., Belin, Stephane, Wang, Dongqing, Lu, Tao, Chen, Gang, Zhang, Kang, Yeung, Cecil, Feng, Guoping, Yankner, Bruce A., and He, Zhigang
- Subjects
AXONS ,REGENERATION (Biology) ,CENTRAL nervous system ,PHOSPHATASES ,RAPAMYCIN ,CYTOKINES ,GENE expression - Abstract
A formidable challenge in neural repair in the adult central nervous system (CNS) is the long distances that regenerating axons often need to travel in order to reconnect with their targets. Thus, a sustained capacity for axon regeneration is critical for achieving functional restoration. Although deletion of either phosphatase and tensin homologue (PTEN), a negative regulator of mammalian target of rapamycin (mTOR), or suppressor of cytokine signalling 3 (SOCS3), a negative regulator of Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathway, in adult retinal ganglion cells (RGCs) individually promoted significant optic nerve regeneration, such regrowth tapered off around 2?weeks after the crush injury. Here we show that, remarkably, simultaneous deletion of both PTEN and SOCS3 enables robust and sustained axon regeneration. We further show that PTEN and SOCS3 regulate two independent pathways that act synergistically to promote enhanced axon regeneration. Gene expression analyses suggest that double deletion not only results in the induction of many growth-related genes, but also allows RGCs to maintain the expression of a repertoire of genes at the physiological level after injury. Our results reveal concurrent activation of mTOR and STAT3 pathways as key for sustaining long-distance axon regeneration in adult CNS, a crucial step towards functional recovery. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
25. STING is a direct innate immune sensor of cyclic di-GMP.
- Author
-
Burdette, Dara L., Monroe, Kathryn M., Sotelo-Troha, Katia, Iwig, Jeff S., Eckert, Barbara, Hyodo, Mamoru, Hayakawa, Yoshihiro, and Vance, Russell E.
- Subjects
NATURAL immunity ,CYTOKINES ,INTERFERONS ,PROTEIN crosslinking ,TRANSCRIPTION factors ,IMMUNOLOGICAL adjuvants ,MICROBIAL genetics ,MOLECULAR immunology - Abstract
The innate immune system detects infection by using germline-encoded receptors that are specific for conserved microbial molecules. The recognition of microbial ligands leads to the production of cytokines, such as type I interferons (IFNs), that are essential for successful pathogen elimination. Cytosolic detection of pathogen-derived DNA is one major mechanism of inducing IFN production, and this process requires signalling through TANK binding kinase 1 (TBK1) and its downstream transcription factor, IFN-regulatory factor 3 (IRF3). In addition, a transmembrane protein called STING (stimulator of IFN genes; also known as MITA, ERIS, MPYS and TMEM173) functions as an essential signalling adaptor, linking the cytosolic detection of DNA to the TBK1-IRF3 signalling axis. Recently, unique nucleic acids called cyclic dinucleotides, which function as conserved signalling molecules in bacteria, have also been shown to induce a STING-dependent type I IFN response. However, a mammalian sensor of cyclic dinucleotides has not been identified. Here we report evidence that STING itself is an innate immune sensor of cyclic dinucleotides. We demonstrate that STING binds directly to radiolabelled cyclic diguanylate monophosphate (c-di-GMP), and we show that unlabelled cyclic dinucleotides, but not other nucleotides or nucleic acids, compete with c-di-GMP for binding to STING. Furthermore, we identify mutations in STING that selectively affect the response to cyclic dinucleotides without affecting the response to DNA. Thus, STING seems to function as a direct sensor of cyclic dinucleotides, in addition to its established role as a signalling adaptor in the IFN response to cytosolic DNA. Cyclic dinucleotides have shown promise as novel vaccine adjuvants and immunotherapeutics, and our results provide insight into the mechanism by which cyclic dinucleotides are sensed by the innate immune system. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
26. Small molecule inhibitors reveal Niemann-Pick C1 is essential for Ebola virus infection.
- Author
-
Côté, Marceline, Misasi, John, Ren, Tao, Bruchez, Anna, Lee, Kyungae, Filone, Claire Marie, Hensley, Lisa, Li, Qi, Ory, Daniel, Chandran, Kartik, and Cunningham, James
- Subjects
NIEMANN-Pick diseases ,EBOLA virus disease ,CYTOKINES ,PIPERAZINE ,GLYCOPROTEINS ,PROTEOLYTIC enzymes ,ANTIVIRAL agents - Abstract
Ebola virus (EboV) is a highly pathogenic enveloped virus that causes outbreaks of zoonotic infection in Africa. The clinical symptoms are manifestations of the massive production of pro-inflammatory cytokines in response to infection and in many outbreaks, mortality exceeds 75%. The unpredictable onset, ease of transmission, rapid progression of disease, high mortality and lack of effective vaccine or therapy have created a high level of public concern about EboV. Here we report the identification of a novel benzylpiperazine adamantane diamide-derived compound that inhibits EboV infection. Using mutant cell lines and informative derivatives of the lead compound, we show that the target of the inhibitor is the endosomal membrane protein Niemann-Pick C1 (NPC1). We find that NPC1 is essential for infection, that it binds to the virus glycoprotein (GP), and that antiviral compounds interfere with GP binding to NPC1. Combined with the results of previous studies of GP structure and function, our findings support a model of EboV infection in which cleavage of the GP1 subunit by endosomal cathepsin proteases removes heavily glycosylated domains to expose the amino-terminal domain, which is a ligand for NPC1 and regulates membrane fusion by the GP2 subunit. Thus, NPC1 is essential for EboV entry and a target for antiviral therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
27. Non-apoptotic role of BID in inflammation and innate immunity.
- Author
-
Yeretssian, Garabet, Correa, Ricardo G., Doiron, Karine, Fitzgerald, Patrick, Dillon, Christopher P., Green, Douglas R., Reed, John C., and Saleh, Maya
- Subjects
NATURAL immunity ,NUCLEOTIDES ,ANTI-infective agents ,APOPTOSIS ,CYTOKINES ,INFLAMMATION ,COLITIS - Abstract
Innate immunity is a fundamental defence response that depends on evolutionarily conserved pattern recognition receptors for sensing infections or danger signals. Nucleotide-binding and oligomerization domain (NOD) proteins are cytosolic pattern-recognition receptors of paramount importance in the intestine, and their dysregulation is associated with inflammatory bowel disease. They sense peptidoglycans from commensal microorganisms and pathogens and coordinate signalling events that culminate in the induction of inflammation and anti-microbial responses. However, the signalling mechanisms involved in this process are not fully understood. Here, using genome-wide RNA interference, we identify candidate genes that modulate the NOD1 inflammatory response in intestinal epithelial cells. Our results reveal a significant crosstalk between innate immunity and apoptosis and identify BID, a BCL2 family protein, as a critical component of the inflammatory response. Colonocytes depleted of BID or macrophages from Bid
−/− mice are markedly defective in cytokine production in response to NOD activation. Furthermore, Bid−/− mice are unresponsive to local or systemic exposure to NOD agonists or their protective effect in experimental colitis. Mechanistically, BID interacts with NOD1, NOD2 and the IκB kinase (IKK) complex, impacting NF-κB and extracellular signal-regulated kinase (ERK) signalling. Our results define a novel role of BID in inflammation and immunity independent of its apoptotic function, furthering the mounting evidence of evolutionary conservation between the mechanisms of apoptosis and immunity. [ABSTRACT FROM AUTHOR]- Published
- 2011
- Full Text
- View/download PDF
28. Lessons on the pathogenesis of aneurysm from heritable conditions.
- Author
-
Lindsay, Mark E. and Dietz, Harry C.
- Subjects
AORTIC aneurysms ,AORTA ,EXTRACELLULAR matrix proteins ,CYTOKINES ,GENETICS - Abstract
Aortic aneurysm is common, accounting for 1-2% of all deaths in industrialized countries. Early theories of the causes of human aneurysm mostly focused on inherited or acquired defects in components of the extracellular matrix in the aorta. Although several mutations in the genes encoding extracellular matrix proteins have been recognized, more recent discoveries have shown important perturbations in cytokine signalling cascades and intracellular components of the smooth muscle contractile apparatus. The modelling of single-gene heritable aneurysm disorders in mice has shown unexpected involvement of the transforming growth factor-β cytokine pathway in aortic aneurysm, highlighting the potential for new therapeutic strategies. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
29. Suppression of TH17 differentiation and autoimmunity by a synthetic ROR ligand.
- Author
-
Solt, Laura A., Kumar, Naresh, Nuhant, Philippe, Yongjun Wang, Lauer, Janelle L., Jin Liu, Istrate, Monica A., Kamenecka, Theodore M., Roush, William R., Vidović, Dušica, Schürer, Stephan C., Jihong Xu, Wagoner, Gail, Drew, Paul D., Griffin, Patrick R., and Burris, Thomas P.
- Subjects
INTERLEUKINS ,NUCLEAR receptors (Biochemistry) ,CYTOKINES ,AUTOIMMUNE disease treatment ,ACETAMIDE ,CELL differentiation ,T-cell receptor genes ,THERAPEUTICS - Abstract
T-helper cells that produce interleukin-17 (T
H 17 cells) are a recently identified CD4+ T-cell subset with characterized pathological roles in autoimmune diseases. The nuclear receptors retinoic-acid-receptor-related orphan receptors α and γt (RORα and RORγt, respectively) have indispensible roles in the development of this cell type. Here we present SR1001, a high-affinity synthetic ligand-the first in a new class of compound-that is specific to both RORα and RORγt and which inhibits TH 17 cell differentiation and function. SR1001 binds specifically to the ligand-binding domains of RORα and RORγt, inducing a conformational change within the ligand-binding domain that encompasses the repositioning of helix 12 and leads to diminished affinity for co-activators and increased affinity for co-repressors, resulting in suppression of the receptors' transcriptional activity. SR1001 inhibited the development of murine TH 17 cells, as demonstrated by inhibition of interleukin-17A gene expression and protein production. Furthermore, SR1001 inhibited the expression of cytokines when added to differentiated murine or human TH 17 cells. Finally, SR1001 effectively suppressed the clinical severity of autoimmune disease in mice. Our data demonstrate the feasibility of targeting the orphan receptors RORα and RORγt to inhibit specifically TH 17 cell differentiation and function, and indicate that this novel class of compound has potential utility in the treatment of autoimmune diseases. [ABSTRACT FROM AUTHOR]- Published
- 2011
- Full Text
- View/download PDF
30. Linear ubiquitination prevents inflammation and regulates immune signalling.
- Author
-
Gerlach, Björn, Cordier, Stefanie M., Schmukle, Anna C., Emmerich, Christoph H., Rieser, Eva, Haas, Tobias L., Webb, Andrew I., Rickard, James A., Anderton, Holly, Wong, Wendy W.-L., Nachbur, Ueli, Gangoda, Lahiru, Warnken, Uwe, Purcell, Anthony W., Silke, John, and Walczak, Henning
- Subjects
TUMOR necrosis factors ,SKIN inflammation ,MASS spectrometry ,CYTOKINES ,GLYCOPROTEINS - Abstract
Members of the tumour necrosis factor (TNF) receptor superfamily have important functions in immunity and inflammation. Recently linear ubiquitin chains assembled by a complex containing HOIL-1 and HOIP (also known as RBCK1 and RNF31, respectively) were implicated in TNF signalling, yet their relevance in vivo remained uncertain. Here we identify SHARPIN as a third component of the linear ubiquitin chain assembly complex, recruited to the CD40 and TNF receptor signalling complexes together with its other constituents, HOIL-1 and HOIP. Mass spectrometry of TNF signalling complexes revealed RIP1 (also known as RIPK1) and NEMO (also known as IKKγ or IKBKG) to be linearly ubiquitinated. Mutation of the Sharpin gene (Sharpin
cpdm/cpdm ) causes chronic proliferative dermatitis (cpdm) characterized by inflammatory skin lesions and defective lymphoid organogenesis. Gene induction by TNF, CD40 ligand and interleukin-1β was attenuated in cpdm-derived cells which were rendered sensitive to TNF-induced death. Importantly, Tnf gene deficiency prevented skin lesions in cpdm mice. We conclude that by enabling linear ubiquitination in the TNF receptor signalling complex, SHARPIN interferes with TNF-induced cell death and, thereby, prevents inflammation. Our results provide evidence for the relevance of linear ubiquitination in vivo in preventing inflammation and regulating immune signalling. [ABSTRACT FROM AUTHOR]- Published
- 2011
- Full Text
- View/download PDF
31. A random cell motility gradient downstream of FGF controls elongation of an amniote embryo.
- Author
-
Bénazéraf, Bertrand, Francois, Paul, Baker, Ruth E., Denans, Nicolas, Little, Charles D., and Pourquié, Olivier
- Subjects
CELL motility ,AMNIOTES ,EMBRYOS ,MESODERM ,MICROSCOPY ,EXTRACELLULAR matrix ,FIBROBLAST growth factors ,MITOGEN-activated protein kinases ,CYTOKINES - Abstract
Vertebrate embryos are characterized by an elongated antero-posterior (AP) body axis, which forms by progressive cell deposition from a posterior growth zone in the embryo. Here, we used tissue ablation in the chicken embryo to demonstrate that the caudal presomitic mesoderm (PSM) has a key role in axis elongation. Using time-lapse microscopy, we analysed the movements of fluorescently labelled cells in the PSM during embryo elongation, which revealed a clear posterior-to-anterior gradient of cell motility and directionality in the PSM. We tracked the movement of the PSM extracellular matrix in parallel with the labelled cells and subtracted the extracellular matrix movement from the global motion of cells. After subtraction, cell motility remained graded but lacked directionality, indicating that the posterior cell movements associated with axis elongation in the PSM are not intrinsic but reflect tissue deformation. The gradient of cell motion along the PSM parallels the fibroblast growth factor (FGF)/mitogen-activated protein kinase (MAPK) gradient, which has been implicated in the control of cell motility in this tissue. Both FGF signalling gain- and loss-of-function experiments lead to disruption of the motility gradient and a slowing down of axis elongation. Furthermore, embryos treated with cell movement inhibitors (blebbistatin or RhoK inhibitor), but not cell cycle inhibitors, show a slower axis elongation rate. We propose that the gradient of random cell motility downstream of FGF signalling in the PSM controls posterior elongation in the amniote embryo. Our data indicate that tissue elongation is an emergent property that arises from the collective regulation of graded, random cell motion rather than by the regulation of directionality of individual cellular movements. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
32. Helical assembly in the MyD88–IRAK4–IRAK2 complex in TLR/IL-1R signalling.
- Author
-
Lin, Su-Chang, Lo, Yu-Chih, and Wu, Hao
- Subjects
OLIGOMERS ,PHOSPHORYLATION ,CELLULAR signal transduction ,MUTAGENESIS ,DROSOPHILA ,CYTOKINES ,ELECTRONS ,MOLECULES ,ELECTROSTATICS - Abstract
MyD88, IRAK4 and IRAK2 are critical signalling mediators of the TLR/IL1-R superfamily. Here we report the crystal structure of the MyD88–IRAK4–IRAK2 death domain (DD) complex, which surprisingly reveals a left-handed helical oligomer that consists of 6 MyD88, 4 IRAK4 and 4 IRAK2 DDs. Assembly of this helical signalling tower is hierarchical, in which MyD88 recruits IRAK4 and the MyD88–IRAK4 complex recruits the IRAK4 substrates IRAK2 or the related IRAK1. Formation of these Myddosome complexes brings the kinase domains of IRAKs into proximity for phosphorylation and activation. Composite binding sites are required for recruitment of the individual DDs in the complex, which are confirmed by mutagenesis and previously identified signalling mutations. Specificities in Myddosome formation are dictated by both molecular complementarity and correspondence of surface electrostatics. The MyD88–IRAK4–IRAK2 complex provides a template for Toll signalling in Drosophila and an elegant mechanism for versatile assembly and regulation of DD complexes in signal transduction. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
33. IL25 elicits a multipotent progenitor cell population that promotes TH2 cytokine responses.
- Author
-
Saenz, Steven A., Siracusa, Mark C., Perrigoue, Jacqueline G., Spencer, Sean P., Urban Jr, Joseph F., Tocker, Joel E., Budelsky, Alison L., Kleinschek, Melanie A., Kastelein, Robert A., Kambayashi, Taku, Bhandoola, Avinash, and Artis, David
- Subjects
HELMINTHIASIS ,CELL proliferation ,LYMPHOID tissue ,GRANULOCYTES ,CYTOKINES ,IMMUNE response ,CELL populations - Abstract
CD4
+ T helper 2 (TH 2) cells secrete interleukin (IL)4, IL5 and IL13, and are required for immunity to gastrointestinal helminth infections. However, TH 2 cells also promote chronic inflammation associated with asthma and allergic disorders. The non-haematopoietic-cell-derived cytokines thymic stromal lymphopoietin, IL33 and IL25 (also known as IL17E) have been implicated in inducing TH 2 cell-dependent inflammation at mucosal sites, but how these cytokines influence innate immune responses remains poorly defined. Here we show that IL25, a member of the IL17 cytokine family, promotes the accumulation of a lineage-negative (Lin- ) multipotent progenitor (MPP) cell population in the gut-associated lymphoid tissue that promotes TH 2 cytokine responses. The IL25-elicited cell population, termed MPPtype2 cells, was defined by the expression of Sca-1 (also known as Ly6a) and intermediate expression of c-Kit (c-Kitint ), and exhibited multipotent capacity, giving rise to cells of monocyte/macrophage and granulocyte lineages both in vitro and in vivo. Progeny of MPPtype2 cells were competent antigen presenting cells, and adoptive transfer of MPPtype2 cells could promote TH 2 cytokine responses and confer protective immunity to helminth infection in normally susceptible Il25-/- mice. The ability of IL25 to induce the emergence of an MPPtype2 cell population identifies a link between the IL17 cytokine family and extramedullary haematopoiesis, and suggests a previously unrecognized innate immune pathway that promotes TH 2 cytokine responses at mucosal sites. [ABSTRACT FROM AUTHOR]- Published
- 2010
- Full Text
- View/download PDF
34. Nuocytes represent a new innate effector leukocyte that mediates type-2 immunity.
- Author
-
Neill, Daniel R., See Heng Wong, Bellosi, Agustin, Flynn, Robin J., Daly, Maria, Langford, Theresa K. A., Bucks, Christine, Kane, Colleen M., Fallon, Padraic G., Pannell, Richard, Jolin, Helen E., and McKenzie, Andrew N. J.
- Subjects
NIPPOSTRONGYLUS brasiliensis ,LEUCOCYTES ,CYTOKINES ,INTERLEUKINS ,CELLULAR immunity ,HELMINTHIASIS - Abstract
Innate immunity provides the first line of defence against invading pathogens and provides important cues for the development of adaptive immunity. Type-2 immunity—responsible for protective immune responses to helminth parasites and the underlying cause of the pathogenesis of allergic asthma—consists of responses dominated by the cardinal type-2 cytokines interleukin (IL)4, IL5 and IL13 (ref. 5). T cells are an important source of these cytokines in adaptive immune responses, but the innate cell sources remain to be comprehensively determined. Here, through the use of novel Il13-eGFP reporter mice, we present the identification and functional characterization of a new innate type-2 immune effector leukocyte that we have named the nuocyte. Nuocytes expand in vivo in response to the type-2-inducing cytokines IL25 and IL33, and represent the predominant early source of IL13 during helminth infection with Nippostrongylus brasiliensis. In the combined absence of IL25 and IL33 signalling, nuocytes fail to expand, resulting in a severe defect in worm expulsion that is rescued by the adoptive transfer of in vitro cultured wild-type, but not IL13-deficient, nuocytes. Thus, nuocytes represent a critically important innate effector cell in type-2 immunity. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
35. Vascular endothelial growth factor B controls endothelial fatty acid uptake.
- Author
-
Hagberg, Carolina E., Falkevall, Annelie, Xun Wang, Larsson, Erik, Huusko, Jenni, Nilsson, Ingrid, van Meeteren, Laurens A., Samen, Erik, Li Lu, Vanwildemeersch, Maarten, Klar, Joakim, Genove, Guillem, Pietras, Kristian, Stone-Elander, Sharon, Claesson-Welsh, Lena, Ylä-Herttuala, Seppo, Lindahl, Per, and Eriksson, Ulf
- Subjects
BIOMOLECULES ,LIPIDS ,CELLULAR control mechanisms ,STEROIDS ,FATTY acids ,CYTOKINES ,MICE ,CONNECTIVE tissues ,ENDOCRINE diseases ,ENDOTHELINS - Abstract
The vascular endothelial growth factors (VEGFs) are major angiogenic regulators and are involved in several aspects of endothelial cell physiology. However, the detailed role of VEGF-B in blood vessel function has remained unclear. Here we show that VEGF-B has an unexpected role in endothelial targeting of lipids to peripheral tissues. Dietary lipids present in circulation have to be transported through the vascular endothelium to be metabolized by tissue cells, a mechanism that is poorly understood. Bioinformatic analysis showed that Vegfb was tightly co-expressed with nuclear-encoded mitochondrial genes across a large variety of physiological conditions in mice, pointing to a role for VEGF-B in metabolism. VEGF-B specifically controlled endothelial uptake of fatty acids via transcriptional regulation of vascular fatty acid transport proteins. As a consequence, Vegfb
-/- mice showed less uptake and accumulation of lipids in muscle, heart and brown adipose tissue, and instead shunted lipids to white adipose tissue. This regulation was mediated by VEGF receptor 1 and neuropilin 1 expressed by the endothelium. The co-expression of VEGF-B and mitochondrial proteins introduces a novel regulatory mechanism, whereby endothelial lipid uptake and mitochondrial lipid use are tightly coordinated. The involvement of VEGF-B in lipid uptake may open up the possibility for novel strategies to modulate pathological lipid accumulation in diabetes, obesity and cardiovascular diseases. [ABSTRACT FROM AUTHOR]- Published
- 2010
- Full Text
- View/download PDF
36. B-cell-derived lymphotoxin promotes castration-resistant prostate cancer.
- Author
-
Ammirante, Massimo, Jun-Li Luo, Grivennikov, Sergei, Nedospasov, Sergei, and Karin, Michael
- Subjects
TUMOR necrosis factors ,PROSTATE cancer ,CANCER patients ,PROSTATECTOMY ,PROSTATE surgery ,CANCER cells ,CARCINOGENESIS ,LEUCOCYTES ,CYTOKINES - Abstract
Prostate cancer (CaP) progresses from prostatic intraepithelial neoplasia through locally invasive adenocarcinoma to castration-resistant metastatic carcinoma. Although radical prostatectomy, radiation and androgen ablation are effective therapies for androgen-dependent CaP, metastatic castration-resistant CaP is a major complication with high mortality. Androgens stimulate growth and survival of prostate epithelium and early CaP. Although most patients initially respond to androgen ablation, many develop castration-resistant CaP within 12–18 months. Despite extensive studies, the mechanisms underlying the emergence of castration-resistant CaP remain poorly understood and their elucidation is critical for developing improved therapies. Curiously, castration-resistant CaP remains androgen-receptor dependent, and potent androgen-receptor antagonists induce tumour regression in castrated mice. The role of inflammation in castration-resistant CaP has not been addressed, although it was reported that intrinsic NF-κB activation supports its growth. Inflammation is a localized protective reaction to injury or infection, but it also has a pathogenic role in many diseases, including cancer. Whereas acute inflammation is critical for host defence, chronic inflammation contributes to tumorigenesis and metastatic progression. The inflammation-responsive IκB kinase (IKK)-β and its target NF-κB have important tumour-promoting functions within malignant cells and inflammatory cells. The latter, including macrophages and lymphocytes, are important elements of the tumour microenvironment, but the mechanisms underlying their recruitment remain obscure, although they are thought to depend on chemokine and cytokine production. We found that CaP progression is associated with inflammatory infiltration and activation of IKK-α, which stimulates metastasis by an NF-κB-independent, cell autonomous mechanism. Here we show that androgen ablation causes infiltration of regressing androgen-dependent tumours with leukocytes, including B cells, in which IKK-β activation results in production of cytokines that activate IKK-α and STAT3 in CaP cells to enhance hormone-free survival. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
37. Systemic signals regulate ageing and rejuvenation of blood stem cell niches.
- Author
-
Mayack, Shane R., Shadrach, Jennifer L., Kim, Francis S., and Wagers, Amy J.
- Subjects
AGING ,STEM cells ,HEMATOPOIESIS ,SOMATOMEDIN ,TISSUES ,PHENOTYPES ,HEMATOPOIETIC stem cells ,CYTOKINES ,DEVELOPMENTAL biology - Abstract
Ageing in multicellular organisms typically involves a progressive decline in cell replacement and repair processes, resulting in several physiological deficiencies, including inefficient muscle repair, reduced bone mass, and dysregulation of blood formation (haematopoiesis). Although defects in tissue-resident stem cells clearly contribute to these phenotypes, it is unclear to what extent they reflect stem cell intrinsic alterations or age-related changes in the stem cell supportive microenvironment, or niche. Here, using complementary in vivo and in vitro heterochronic models, we show that age-associated changes in stem cell supportive niche cells deregulate normal haematopoiesis by causing haematopoietic stem cell dysfunction. Furthermore, we find that age-dependent defects in niche cells are systemically regulated and can be reversed by exposure to a young circulation or by neutralization of the conserved longevity regulator, insulin-like growth factor-1, in the marrow microenvironment. Together, these results show a new and critical role for local and systemic factors in signalling age-related haematopoietic decline, and highlight a new model in which blood-borne factors in aged animals act through local niche cells to induce age-dependent disruption of stem cell function. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
38. Innate production of TH2 cytokines by adipose tissue-associated c-Kit+Sca-1+ lymphoid cells.
- Author
-
Moro, Kazuyo, Yamada, Taketo, Tanabe, Masanobu, Takeuchi, Tsutomu, Ikawa, Tomokatsu, Kawamoto, Hiroshi, Furusawa, Jun-ichi, Ohtani, Masashi, Fujii, Hideki, and Koyasu, Shigeo
- Subjects
BACTERIA ,KILLER cells ,LYMPHOCYTES ,NATURAL immunity ,CELLULAR immunity ,B cells ,ADIPOSE tissues ,LYMPHOID tissue ,CYTOKINES ,HELMINTHIASIS - Abstract
Innate immune responses are important in combating various microbes during the early phases of infection. Natural killer (NK) cells are innate lymphocytes that, unlike T and B lymphocytes, do not express antigen receptors but rapidly exhibit cytotoxic activities against virus-infected cells and produce various cytokines. Here we report a new type of innate lymphocyte present in a novel lymphoid structure associated with adipose tissues in the peritoneal cavity. These cells do not express lineage (Lin) markers but do express c-Kit, Sca-1 (also known as Ly6a), IL7R and IL33R. Similar lymphoid clusters were found in both human and mouse mesentery and we term this tissue ‘FALC’ (fat-associated lymphoid cluster). FALC Lin
- c-Kit+ Sca-1+ cells are distinct from lymphoid progenitors and lymphoid tissue inducer cells. These cells proliferate in response to IL2 and produce large amounts of TH 2 cytokines such as IL5, IL6 and IL13. IL5 and IL6 regulate B-cell antibody production and self-renewal of B1 cells. Indeed, FALC Lin- c-Kit+ Sca-1+ cells support the self-renewal of B1 cells and enhance IgA production. IL5 and IL13 mediate allergic inflammation and protection against helminth infection. After helminth infection and in response to IL33, FALC Lin- c-Kit+ Sca-1+ cells produce large amounts of IL13, which leads to goblet cell hyperplasia—a critical step for helminth expulsion. In mice devoid of FALC Lin- c-Kit+ Sca-1+ cells, such goblet cell hyperplasia was not induced. Thus, FALC Lin- c-Kit+ Sca-1+ cells are TH 2-type innate lymphocytes, and we propose that these cells be called ‘natural helper cells’. [ABSTRACT FROM AUTHOR]- Published
- 2010
- Full Text
- View/download PDF
39. CD8+ T lymphocyte mobilization to virus-infected tissue requires CD4+ T-cell help.
- Author
-
Nakanishi, Yusuke, Lu, Bao, Gerard, Craig, and Iwasaki, Akiko
- Subjects
T cells ,LYMPHOCYTES ,CELL-mediated cytotoxicity ,CHEMOKINES ,CELL death ,CYTOKINES ,INFLAMMATORY mediators ,PEPTIDES ,CELL-mediated lympholysis ,INTERLEUKIN-2 ,DISEASES - Abstract
CD4
+ T helper cells are well known for their role in providing critical signals during priming of cytotoxic CD8+ T lymphocyte (CTL) responses in vivo. T-cell help is required for the generation of primary CTL responses as well as in promoting protective CD8+ memory T-cell development. However, the role of CD4 help in the control of CTL responses at the effector stage is unknown. Here we show that fully helped effector CTLs are themselves not self-sufficient for entry into the infected tissue, but rely on the CD4+ T cells to provide the necessary cue. CD4+ T helper cells control the migration of CTL indirectly through the secretion of IFN-γ and induction of local chemokine secretion in the infected tissue. Our results reveal a previously unappreciated role of CD4 help in mobilizing effector CTL to the peripheral sites of infection where they help to eliminate infected cells. [ABSTRACT FROM AUTHOR]- Published
- 2009
- Full Text
- View/download PDF
40. Central control of fever and female body temperature by RANKL/RANK.
- Author
-
Hanada, Reiko, Leibbrandt, Andreas, Hanada, Toshikatsu, Kitaoka, Shiho, Furuyashiki, Tomoyuki, Fujihara, Hiroaki, Trichereau, Jean, Paolino, Magdalena, Qadri, Fatimunnisa, Plehm, Ralph, Klaere, Steffen, Komnenovic, Vukoslav, Mimata, Hiromitsu, Yoshimatsu, Hironobu, Takahashi, Naoyuki, von Haeseler, Arndt, Bader, Michael, Kilic, Sara Sebnem, Ueta, Yoichi, and Pifl, Christian
- Subjects
BODY temperature regulation ,EXANTHEMA ,NERVOUS system ,CYTOKINES ,DRUG administration ,MAMMARY glands ,PHYSICAL diagnosis ,VITAL signs ,LABORATORY rodents - Abstract
Receptor-activator of NF-κB ligand (TNFSF11, also known as RANKL, OPGL, TRANCE and ODF) and its tumour necrosis factor (TNF)-family receptor RANK are essential regulators of bone remodelling, lymph node organogenesis and formation of a lactating mammary gland. RANKL and RANK are also expressed in the central nervous system. However, the functional relevance of RANKL/RANK in the brain was entirely unknown. Here we report that RANKL and RANK have an essential role in the brain. In both mice and rats, central RANKL injections trigger severe fever. Using tissue-specific Nestin-Cre and GFAP-Cre rank
floxed deleter mice, the function of RANK in the fever response was genetically mapped to astrocytes. Importantly, Nestin-Cre and GFAP-Cre rankfloxed deleter mice are resistant to lipopolysaccharide-induced fever as well as fever in response to the key inflammatory cytokines IL-1β and TNFα. Mechanistically, RANKL activates brain regions involved in thermoregulation and induces fever via the COX2-PGE2 /EP3R pathway. Moreover, female Nestin-Cre and GFAP-Cre rankfloxed mice exhibit increased basal body temperatures, suggesting that RANKL and RANK control thermoregulation during normal female physiology. We also show that two children with RANK mutations exhibit impaired fever during pneumonia. These data identify an entirely novel and unexpected function for the key osteoclast differentiation factors RANKL/RANK in female thermoregulation and the central fever response in inflammation. [ABSTRACT FROM AUTHOR]- Published
- 2009
- Full Text
- View/download PDF
41. Materials engineering for immunomodulation.
- Author
-
Hubbell, Jeffrey A., Thomas, Susan N., and Swartz, Melody A.
- Subjects
IMMUNOGLOBULINS ,IMMUNOREGULATION ,T cells ,LYMPHOCYTES ,IMMUNOLOGICAL adjuvants ,CELLULAR immunity ,IMMUNITY ,IMMUNOMODULATORS ,CYTOKINES - Abstract
The engineering of materials that can modulate the immune system is an emerging field that is developing alongside immunology. For therapeutic ends such as vaccine development, materials are now being engineered to deliver antigens through specific intracellular pathways, allowing better control of the way in which antigens are presented to one of the key types of immune cell, T cells. Materials are also being designed as adjuvants, to mimic specific 'danger' signals in order to manipulate the resultant cytokine environment, which influences how antigens are interpreted by T cells. In addition to offering the potential for medical advances, immunomodulatory materials can form well-defined model systems, helping to provide new insight into basic immunobiology. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
42. Resolvin D2 is a potent regulator of leukocytes and controls microbial sepsis.
- Author
-
Spite, Matthew, Norling, Lucy V., Summers, Lisa, Yang, Rong, Cooper, Dianne, Petasis, Nicos A., Flower, Roderick J., Perretti, Mauro, and Serhan, Charles N.
- Subjects
INFLAMMATORY mediators ,LEUCOCYTES ,SEPSIS ,EXUDATES & transudates ,NEUTROPHILS ,CYTOKINES ,NITRIC oxide ,TISSUE adhesions ,MACROPHAGES ,PHAGOCYTOSIS - Abstract
A growing body of evidence indicates that resolution of acute inflammation is an active process. Resolvins are a new family of lipid mediators enzymatically generated within resolution networks that possess unique and specific functions to orchestrate catabasis, the phase in which disease declines. Resolvin D2 (RvD2) was originally identified in resolving exudates, yet its individual contribution in resolution remained to be elucidated. Here, we establish RvD2’s potent stereoselective actions in reducing excessive neutrophil trafficking to inflammatory loci. RvD2 decreased leukocyte–endothelial interactions in vivo by endothelial-dependent nitric oxide production, and by direct modulation of leukocyte adhesion receptor expression. In mice with microbial sepsis initiated by caecal ligation and puncture, RvD2 sharply decreased both local and systemic bacterial burden, excessive cytokine production and neutrophil recruitment, while increasing peritoneal mononuclear cells and macrophage phagocytosis. These multi-level pro-resolving actions of RvD2 translate to increased survival from sepsis induced by caecal ligation and puncture and surgery. Together, these results identify RvD2 as a potent endogenous regulator of excessive inflammatory responses that acts via multiple cellular targets to stimulate resolution and preserve immune vigilance. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
43. Membrane-bound Fas ligand only is essential for Fas-induced apoptosis.
- Author
-
O'Reilly, Lorraine A., Lin Tai, Lee, Lily, Kruse, Elizabeth A., Grabow, Stephanie, Fairlie, W. Douglas, Haynes, Nicole M., Tarlinton, David M., Jian-Guo Zhang, Belz, Gabrielle T., Smyth, Mark J., Bouillet, Philippe, Robb, Lorraine, and Strasser, Andreas
- Subjects
APOPTOSIS ,CELL death ,LIGANDS (Biochemistry) ,TUMOR necrosis factors ,CYTOKINES ,AUTOIMMUNITY ,GENETIC mutation ,METALLOPROTEINASES ,CELL membranes ,GAMMA globulins - Abstract
Fas ligand (FasL), an apoptosis-inducing member of the TNF cytokine family, and its receptor Fas are critical for the shutdown of chronic immune responses and prevention of autoimmunity. Accordingly, mutations in their genes cause severe lymphadenopathy and autoimmune disease in mice and humans. FasL function is regulated by deposition in the plasma membrane and metalloprotease-mediated shedding. Here we generated gene-targeted mice that selectively lack either secreted FasL (sFasL) or membrane-bound FasL (mFasL) to resolve which of these forms is required for cell killing and to explore their hypothesized non-apoptotic activities. Mice lacking sFasL (FasL
Δs/Δs ) appeared normal and their T cells readily killed target cells, whereas T cells lacking mFasL (FasLΔm/Δm ) could not kill cells through Fas activation. FasLΔm/Δm mice developed lymphadenopathy and hyper-gammaglobulinaemia, similar to FasLgld/gld mice, which express a mutant form of FasL that cannot bind Fas, but surprisingly, FasLΔm/Δm mice (on a C57BL/6 background) succumbed to systemic lupus erythematosus (SLE)-like autoimmune kidney destruction and histiocytic sarcoma, diseases that occur only rarely and much later in FasLgld/gld mice. These results demonstrate that mFasL is essential for cytotoxic activity and constitutes the guardian against lymphadenopathy, autoimmunity and cancer, whereas excess sFasL appears to promote autoimmunity and tumorigenesis through non-apoptotic activities. [ABSTRACT FROM AUTHOR]- Published
- 2009
- Full Text
- View/download PDF
44. A dimerization-dependent mechanism drives RAF catalytic activation.
- Author
-
Rajakulendran, Thanashan, Sahmi, Malha, Lefrançois, Martin, Sicheri, Frank, and Therrien, Marc
- Subjects
CELLULAR control mechanisms ,PROTEIN-tyrosine kinases ,MICROBIAL genetics ,PHOSPHORYLATION ,CARCINOGENESIS ,ONCOGENES ,CYTOKINES ,GROWTH factors ,GENETIC mutation - Abstract
The ERK (extracellular signal-regulated kinase) pathway is an evolutionarily conserved signal transduction module that controls cellular growth, differentiation and survival. Activation of receptor tyrosine kinases (RTKs) by the binding of growth factors initiates GTP loading of RAS, which triggers the initial steps in the activation of the ERK pathway by modulating RAF family kinase function. Once activated, RAF participates in a sequential cascade of phosphorylation events that activate MEK, and in turn ERK. Unbridled signalling through the ERK pathway caused by activating mutations in RTKs, RAS or RAF has been linked to several human cancers. Of note, one member of the RAF family, BRAF, is the most frequently mutated oncogene in the kinase superfamily. Not surprisingly, there has been a colossal effort to understand the underlying regulation of this family of kinases. In particular, the process by which the RAF kinase domain becomes activated towards its substrate MEK remains of topical interest. Here, using Drosophila Schneider S2 cells, we demonstrate that RAF catalytic function is regulated in response to a specific mode of dimerization of its kinase domain, which we term the side-to-side dimer. Moreover, we find that the RAF-related pseudo-kinase KSR (kinase suppressor of Ras) also participates in forming side-to-side heterodimers with RAF and can thereby trigger RAF activation. This mechanism provides an elegant explanation for the longstanding conundrum about RAF catalytic activation, and also provides an explanation for the capacity of KSR, despite lacking catalytic function, to directly mediate RAF activation. We also show that RAF side-to-side dimer formation is essential for aberrant signalling by oncogenic BRAF mutants, and identify an oncogenic mutation that acts specifically by promoting side-to-side dimerization. Together, our data identify the side-to-side dimer interface of RAF as a potential therapeutic target for intervention in BRAF-dependent tumorigenesis. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
45. Riboflavin kinase couples TNF receptor 1 to NADPH oxidase.
- Author
-
Yazdanpanah, Benjamin, Wiegmann, Katja, Tchikov, Vladimir, Krut, Oleg, Pongratz, Carola, Schramm, Michael, Kleinridders, Andre, Wunderlich, Thomas, Kashkar, Hamid, Utermöhlen, Olaf, Brüning, Jens C., Schütze, Stefan, and Krönke, Martin
- Subjects
VITAMIN B2 ,CYTOKINES ,CARRIER proteins ,OXIDASES ,AMINE oxidase ,CHEMICAL reactions ,TUMOR necrosis factors - Abstract
Reactive oxygen species (ROS) produced by NADPH oxidase function as defence and signalling molecules related to innate immunity and various cellular responses. The activation of NADPH oxidase in response to plasma membrane receptor activation depends on the phosphorylation of cytoplasmic oxidase subunits, their translocation to membranes and the assembly of all NADPH oxidase components. Tumour necrosis factor (TNF) is a prominent stimulus of ROS production, but the molecular mechanisms by which TNF activates NADPH oxidase are poorly understood. Here we identify riboflavin kinase (RFK, formerly known as flavokinase) as a previously unrecognized TNF-receptor-1 (TNFR1)-binding protein that physically and functionally couples TNFR1 to NADPH oxidase. In mouse and human cells, RFK binds to both the TNFR1-death domain and to p22
phox , the common subunit of NADPH oxidase isoforms. RFK-mediated bridging of TNFR1 and p22phox is a prerequisite for TNF-induced but not for Toll-like-receptor-induced ROS production. Exogenous flavin mononucleotide or FAD was able to substitute fully for TNF stimulation of NADPH oxidase in RFK-deficient cells. RFK is rate-limiting in the synthesis of FAD, an essential prosthetic group of NADPH oxidase. The results suggest that TNF, through the activation of RFK, enhances the incorporation of FAD in NADPH oxidase enzymes, a critical step for the assembly and activation of NADPH oxidase. [ABSTRACT FROM AUTHOR]- Published
- 2009
- Full Text
- View/download PDF
46. The AP-1 transcription factor Batf controls TH17 differentiation.
- Author
-
Schraml, Barbara U., Hildner, Kai, Ise, Wataru, Wan-Ling Lee, Smith, Whitney A.-E., Solomon, Ben, Sahota, Gurmukh, Sim, Julia, Mukasa, Ryuta, Cemerski, Saso, Hatton, Robin D., Stormo, Gary D., Weaver, Casey T., Russell, John H., Murphy, Theresa L., and Murphy, Kenneth M.
- Subjects
TRANSCRIPTION factors ,NF-kappa B ,NUCLEAR receptors (Biochemistry) ,LEUCINE zippers ,HELIX-loop-helix motifs ,DNA-binding proteins ,CELLULAR immunity ,ENCEPHALOMYELITIS ,CYTOKINES - Abstract
Activator protein 1 (AP-1, also known as JUN) transcription factors are dimers of JUN, FOS, MAF and activating transcription factor (ATF) family proteins characterized by basic region and leucine zipper domains. Many AP-1 proteins contain defined transcriptional activation domains, but BATF and the closely related BATF3 (refs 2, 3) contain only a basic region and leucine zipper, and are considered to be inhibitors of AP-1 activity. Here we show that Batf is required for the differentiation of IL17-producing T helper (T
H 17) cells. TH 17 cells comprise a CD4+ T-cell subset that coordinates inflammatory responses in host defence but is pathogenic in autoimmunity. Batf-/- mice have normal TH 1 and TH 2 differentiation, but show a defect in TH 17 differentiation, and are resistant to experimental autoimmune encephalomyelitis. Batf-/- T cells fail to induce known factors required for TH 17 differentiation, such as RORγt (encoded by Rorc) and the cytokine IL21 (refs 14–17). Neither the addition of IL21 nor the overexpression of RORγt fully restores IL17 production in Batf-/- T cells. The Il17 promoter is BATF-responsive, and after TH 17 differentiation, BATF binds conserved intergenic elements in the Il17a–Il17f locus and to the Il17, Il21 and Il22 (ref. 18) promoters. These results demonstrate that the AP-1 protein BATF has a critical role in TH 17 differentiation. [ABSTRACT FROM AUTHOR]- Published
- 2009
- Full Text
- View/download PDF
47. A parallel circuit of LIF signalling pathways maintains pluripotency of mouse ES cells.
- Author
-
Niwa, Hitoshi, Ogawa, Kazuya, Shimosato, Daisuke, and Adachi, Kenjiro
- Subjects
CYTOKINES ,LEUKEMIA ,LEUCOCYTOSIS ,EMBRYONIC stem cells ,STEM cells ,PROTEIN kinases ,PHOSPHOTRANSFERASES - Abstract
The cytokine leukaemia inhibitory factor (LIF) integrates signals into mouse embryonic stem (ES) cells to maintain pluripotency. Although the Jak–Stat3 pathway is essential and sufficient to mediate LIF signals, it is still unclear how these signals are linked to the core circuitry of pluripotency-associated transcription factors, consisting of Oct3/4 (also called Pou5f1), Sox2 and Nanog. Here we show that two LIF signalling pathways are each connected to the core circuitry via different transcription factors. In mouse ES cells, Klf4 is mainly activated by the Jak–Stat3 pathway and preferentially activates Sox2, whereas Tbx3 is preferentially regulated by the phosphatidylinositol-3-OH kinase–Akt and mitogen-activated protein kinase pathways and predominantly stimulates Nanog. In the absence of LIF, artificial expression of Klf4 or Tbx3 is sufficient to maintain pluripotency while maintaining the expression of Oct3/4. Notably, overexpression of Nanog supports LIF-independent self-renewal of mouse ES cells in the absence of Klf4 and Tbx3 activity. Therefore, Klf4 and Tbx3 are involved in mediating LIF signalling to the core circuitry but are not directly associated with the maintenance of pluripotency, because ES cells keep pluripotency without their expression in the particular context. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
48. Down's syndrome suppression of tumour growth and the role of the calcineurin inhibitor DSCR1.
- Author
-
Kwan-Hyuck Baek, Zaslavsky, Alexander, Lynch, Ryan C., Britt, Carmella, Okada, Yoshiaki, Siarey, Richard J., Lensch, M. William, Park, In-Hyun, Yoon, Sam S., Minami, Takashi, Korenberg, Julie R., Folkman, Judah, Daley, George Q., Aird, William C., Galdzicki, Zygmunt, and Ryeom, Sandra
- Subjects
DOWN syndrome ,HUMAN chromosome abnormalities ,INTELLECTUAL disabilities ,CANCER prevention ,TUMOR prevention ,VASCULAR endothelial growth factors ,NEOVASCULARIZATION ,GENETICS ,CYTOKINES - Abstract
The incidence of many cancer types is significantly reduced in individuals with Down’s syndrome, and it is thought that this broad cancer protection is conferred by the increased expression of one or more of the 231 supernumerary genes on the extra copy of chromosome 21. One such gene is Down’s syndrome candidate region-1 (DSCR1, also known as RCAN1), which encodes a protein that suppresses vascular endothelial growth factor (VEGF)-mediated angiogenic signalling by the calcineurin pathway. Here we show that DSCR1 is increased in Down’s syndrome tissues and in a mouse model of Down’s syndrome. Furthermore, we show that the modest increase in expression afforded by a single extra transgenic copy of Dscr1 is sufficient to confer significant suppression of tumour growth in mice, and that such resistance is a consequence of a deficit in tumour angiogenesis arising from suppression of the calcineurin pathway. We also provide evidence that attenuation of calcineurin activity by DSCR1, together with another chromosome 21 gene Dyrk1a, may be sufficient to markedly diminish angiogenesis. These data provide a mechanism for the reduced cancer incidence in Down’s syndrome and identify the calcineurin signalling pathway, and its regulators DSCR1 and DYRK1A, as potential therapeutic targets in cancers arising in all individuals. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
49. Genes that mediate breast cancer metastasis to the brain.
- Author
-
Bos, Paula D., Zhang, Xiang H.-F., Nadal, Cristina, Weiping Shu, Gomis, Roger R., Nguyen, Don X., Minn, Andy J., van de Vijver, Marc J., Gerald, William L., Foekens, John A., and Massagué, Joan
- Subjects
BREAST cancer ,RISK of metastasis ,CANCER invasiveness ,CANCER of unknown primary origin ,EPIDERMAL growth factor ,CARDIOPULMONARY system ,GENETIC regulation ,CYCLOOXYGENASES ,CYTOKINES ,THERAPEUTICS - Abstract
The molecular basis for breast cancer metastasis to the brain is largely unknown. Brain relapse typically occurs years after the removal of a breast tumour, suggesting that disseminated cancer cells must acquire specialized functions to take over this organ. Here we show that breast cancer metastasis to the brain involves mediators of extravasation through non-fenestrated capillaries, complemented by specific enhancers of blood–brain barrier crossing and brain colonization. We isolated cells that preferentially infiltrate the brain from patients with advanced disease. Gene expression analysis of these cells and of clinical samples, coupled with functional analysis, identified the cyclooxygenase COX2 (also known as PTGS2), the epidermal growth factor receptor (EGFR) ligand HBEGF, and the α2,6-sialyltransferase ST6GALNAC5 as mediators of cancer cell passage through the blood–brain barrier. EGFR ligands and COX2 were previously linked to breast cancer infiltration of the lungs, but not the bones or liver, suggesting a sharing of these mediators in cerebral and pulmonary metastases. In contrast, ST6GALNAC5 specifically mediates brain metastasis. Normally restricted to the brain, the expression of ST6GALNAC5 in breast cancer cells enhances their adhesion to brain endothelial cells and their passage through the blood–brain barrier. This co-option of a brain sialyltransferase highlights the role of cell-surface glycosylation in organ-specific metastatic interactions. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
50. Haematopoietic stem cells depend on Gαs-mediated signalling to engraft bone marrow.
- Author
-
Adams, Gregor B., Alley, Ian R., Chung, Ung-il, Chabner, Karissa T., Jeanson, Nathaniel T., Lo Celso, Cristina, Marsters, Emily S., Chen, Min, Weinstein, Lee S., Lin, Charles P., Kronenberg, Henry M., and Scadden, David T.
- Subjects
STEM cell research ,BONE grafting ,BONE marrow ,FETAL development ,G proteins ,LABORATORY mice ,CHEMOTAXIS ,CYTOKINES - Abstract
Haematopoietic stem and progenitor cells (HSPCs) change location during development and circulate in mammals throughout life, moving into and out of the bloodstream to engage bone marrow niches in sequential steps of homing, engraftment and retention. Here we show that HSPC engraftment of bone marrow in fetal development is dependent on the guanine-nucleotide-binding protein stimulatory α subunit (Gα
s ). HSPCs from adult mice deficient in Gαs (Gαs -/- ) differentiate and undergo chemotaxis, but also do not home to or engraft in the bone marrow in adult mice and demonstrate a marked inability to engage the marrow microvasculature. If deleted after engraftment, Gαs deficiency did not lead to lack of retention in the marrow, rather cytokine-induced mobilization into the blood was impaired. Testing whether activation of Gαs affects HSPCs, pharmacological activators enhanced homing and engraftment in vivo. Gαs governs specific aspects of HSPC localization under physiological conditions in vivo and may be pharmacologically targeted to improve transplantation efficiency. [ABSTRACT FROM AUTHOR]- Published
- 2009
- Full Text
- View/download PDF
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.