Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
1.
ACS Chem Neurosci ; 11(5): 796-805, 2020 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-32056421

RESUMEN

Aggregational states of amyloid ß-protein (Aß) are critical for its neurotoxicity, although they are not well-characterized, particularly after binding to the cell membranes. This is one reason why the mechanisms of Aß neurotoxicity are controversial and elusive. In this study, the effects of toxic Aß-(1-42) fibrils formed in the membrane on cellular processes were investigated using human neuroblastoma SH-SY5Y cells. Consistent with previous observations, fibrillar Aßs formed on the membranes induced activation of caspase-3, the effector caspase for apoptosis. Knockdown analyses of the initiator caspases, caspase-8 and caspase-9, indicated that the apoptosis was induced via activation of caspase-8, followed by activation of caspase-9 and caspase-3. We also found that inflammation signaling pathways including Toll-like receptors and inflammasomes NOD-, LRR-, and pyrin domain-containing protein 3 are involved in the initiation of apoptosis by the Aß fibrils. These inflammation-related molecules are promising targets for the prevention of apoptotic cell death induced by Aß.


Asunto(s)
Péptidos beta-Amiloides , Fragmentos de Péptidos , Apoptosis , Caspasa 3 , Caspasas , Línea Celular Tumoral , Humanos
2.
Cell Death Differ ; 27(5): 1539-1553, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31659279

RESUMEN

Among caspase family members, Caspase-8 is unique, with associated critical activities to induce and suppress death receptor-mediated apoptosis and necroptosis, respectively. Caspase-8 inhibits necroptosis by suppressing the function of receptor-interacting protein kinase 1 (RIPK1 or RIP1) and RIPK3 to activate mixed lineage kinase domain-like (MLKL). Disruption of Caspase-8 expression causes embryonic lethality in mice, which is rescued by depletion of either Ripk3 or Mlkl, indicating that the embryonic lethality is caused by activation of necroptosis. Here, we show that knockdown of Caspase-8 expression in embryoid bodies derived from ES cells markedly enhances retinoic acid (RA)-induced cell differentiation and necroptosis, both of which are dependent on Ripk1 and Ripk3; however, the enhancement of RA-induced cell differentiation is independent of Mlkl and necrosome formation. RA treatment obviously enhanced the expression of RA-specific target genes having the retinoic acid response element (RARE) in their promoter regions to induce cell differentiation, and induced marked expression of RIPK1, RIPK3, and MLKL to stimulate necroptosis. Caspase-8 knockdown induced RIPK1 and RIPK3 to translocate into the nucleus and to form a complex with RA receptor (RAR), and RAR interacting with RIPK1 and RIPK3 showed much stronger binding activity to RARE than RAR without RIPK1 or RIPK3. In Caspase-8-deficient as well as Caspase-8- and Mlkl-deficient mouse embryos, the expression of RA-specific target genes was obviously enhanced. Thus, Caspase-8, RIPK1, and RIPK3 regulate RA-induced cell differentiation and necroptosis both in vitro and in vivo.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Necroptosis/efectos de los fármacos , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Tretinoina/farmacología , Animales , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Embrión de Mamíferos/metabolismo , Cuerpos Embrioides/efectos de los fármacos , Cuerpos Embrioides/metabolismo , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Técnicas de Silenciamiento del Gen , Ratones Endogámicos C57BL , Proteínas Quinasas/metabolismo , Transporte de Proteínas/efectos de los fármacos , Receptores de Ácido Retinoico/metabolismo , Transducción de Señal/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
3.
J Biol Chem ; 294(32): 11994-12006, 2019 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-31217278

RESUMEN

Phosphatidylserine (PS), an anionic phospholipid enriched in the inner leaflet of the plasma membrane, is exposed to the outer leaflet during apoptosis. PS exposure was recently shown to be induced during tumor necrosis factor-induced necroptosis. We herein demonstrated that interferon (IFN)-γ induced necroptosis in Caspase-8-knockout mouse-derived embryonic fibroblasts (C8KO MEFs), as well as in WT MEFs co-treated with the pan-caspase inhibitor, z-VAD-fmk. PS exposure and necroptosis were significant after 6- and 24-h treatments with IFN-γ, respectively. To elucidate the molecular mechanisms underlying IFN-γ-induced PS exposure, we generated C8KO MEF-derived cell lines without the expression of RIPK3 (receptor-interacting protein kinase 3), an essential molecule in tumor necrosis factor-induced necroptosis, and IFN-γ-induced PS exposure and necrotic cell death were shown to be specifically inhibited by the loss of RIPK3 expression. Furthermore, the down-regulated expression of MLKL (mixed lineage kinase domain-like protein), a key molecule for inducing membrane rupture downstream of RIPK3 in necroptosis, abolished IFN-γ-induced PS exposure in C8KO MEFs. In human colorectal adenocarcinoma-derived HT29 cells, PS exposure and necroptosis were similarly induced by treatment with IFN-γ in the presence of Smac mimetics and z-VAD-fmk. The removal of IFN-γ from PS-exposing MEFs after a 6-h treatment completely inhibited necroptotic cell death but not the subsequent increase in the number of PS-exposing cells. Therefore, PS exposure mediated by RIPK3-activated MLKL oligomers was induced by a treatment with IFN-γ for a significant interval of time before the induction of necroptosis by membrane rupture.


Asunto(s)
Caspasa 8/genética , Interferón gamma/farmacología , Necroptosis/efectos de los fármacos , Fosfatidilserinas/metabolismo , Proteínas Quinasas/metabolismo , Clorometilcetonas de Aminoácidos/farmacología , Animales , Caspasa 8/metabolismo , Línea Celular , Células HT29 , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Quinasas/química , Proteínas Quinasas/genética , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/antagonistas & inhibidores , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo
4.
Nat Commun ; 8(1): 540, 2017 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-28912471

RESUMEN

Nucleic acid nanotechnology has great potential for future therapeutic applications. However, the construction of nanostructured devices that control cell fate by detecting and amplifying protein signals has remained a challenge. Here we design and build protein-driven RNA-nanostructured devices that actuate in vitro by RNA-binding-protein-inducible conformational change and regulate mammalian cell fate by RNA-protein interaction-mediated protein assembly. The conformation and function of the RNA nanostructures are dynamically controlled by RNA-binding protein signals. The protein-responsive RNA nanodevices are constructed inside cells using RNA-only delivery, which may provide a safe tool for building functional RNA-protein nanostructures. Moreover, the designed RNA scaffolds that control the assembly and oligomerization of apoptosis-regulatory proteins on a nanometre scale selectively kill target cells via specific RNA-protein interactions. These findings suggest that synthetic RNA nanodevices could function as molecular robots that detect signals and localize target proteins, induce RNA conformational changes, and programme mammalian cellular behaviour.Nucleic acid nanotechnology has great potential for future therapeutic applications. Here the authors build protein-driven RNA nanostructures that can function within mammalian cells and regulate the cell fate.


Asunto(s)
Nanoestructuras/química , Nanotecnología/métodos , Proteínas de Unión al ARN/química , ARN/química , Supervivencia Celular , Células HeLa , Humanos , Nanotecnología/instrumentación , ARN/genética , ARN/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
5.
J Immunol ; 199(3): 1122-1130, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28674179

RESUMEN

CD4+ Th cells play crucial roles in orchestrating immune responses against pathogenic microbes, after differentiating into effector subsets. Recent research has revealed the importance of IFN-γ and IL-17 double-producing CD4+ Th cells, termed Th17/Th1 cells, in the induction of autoimmune and inflammatory diseases. In addition, Th17/Th1 cells are involved in the regulation of infection caused by the intracellular bacterium Mycobacterium tuberculosis in humans. However, the precise mechanism of Th17/Th1 induction during pathogen infection is unclear. In this study, we showed that the inflammasome and Fas-dependent IL-1ß induces Th17/Th1 cells in mice, in response to infection with the pathogenic intracellular bacterium Listeria monocytogenes In the spleens of infected wild-type mice, Th17/Th1 cells were induced, and expressed T-bet and Rorγt. In Pycard-/- mice, which lack the adaptor molecule of the inflammasome (apoptosis-associated speck-like protein containing a caspase recruitment domain), Th17/Th1 induction was abolished. In addition, the Fas-mediated IL-1ß production was required for Th17/Th1 induction during bacterial infection: Th17/Th1 induction was abolished in Fas-/- mice, whereas supplementation with recombinant IL-1ß restored Th17/Th1 induction via IL-1 receptor 1 (IL-1R1), and rescued the mortality of Fas-/- mice infected with Listeria IL-1R1, but not apoptosis-associated speck-like protein containing a caspase recruitment domain or Fas on T cells, was required for Th17/Th1 induction, indicating that IL-1ß stimulates IL-1R1 on T cells for Th17/Th1 induction. These results indicate that IL-1ß, produced by the inflammasome and Fas-dependent mechanisms, contributes cooperatively to the Th17/Th1 induction during bacterial infection. This study provides a deeper understanding of the molecular mechanisms underlying Th17/Th1 induction during pathogenic microbial infections in vivo.


Asunto(s)
Inflamasomas/inmunología , Interleucina-1beta/inmunología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Células TH1/inmunología , Células Th17/inmunología , Receptor fas/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/deficiencia , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Adaptadoras de Señalización CARD , Diferenciación Celular , Interleucina-1beta/administración & dosificación , Listeria monocytogenes/patogenicidad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Receptores Tipo I de Interleucina-1/genética , Receptores Tipo I de Interleucina-1/inmunología , Bazo/inmunología , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Receptor fas/deficiencia , Receptor fas/genética
7.
Gastroenterology ; 148(7): 1452-65, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25683115

RESUMEN

BACKGROUND & AIMS: The KRAS gene is mutated in most pancreatic ductal adenocarcinomas (PDAC). Expression of this KRAS oncoprotein in mice is sufficient to initiate carcinogenesis but not progression to cancer. Activation of phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K) is required for KRAS for induction and maintenance of PDAC in mice. The somatostatin receptor subtype 2 (sst2) inhibits PI3K, but sst2 expression is lost during the development of human PDAC. We investigated the effects of sst2 loss during KRAS-induced PDAC development in mice. METHODS: We analyzed tumor growth in mice that expressed the oncogenic form of KRAS (KRAS(G12D)) in pancreatic precursor cells, as well as sst2+/- and sst2-/-, and in crossed KRAS(G12D);sst2+/- and KRAS(G12D);sst2-/- mice. Pancreatic tissues and acini were collected and assessed by histologic, immunoblot, immunohistochemical, and reverse-transcription polymerase chain reaction analyses. We also compared protein levels in paraffin-embedded PDAC samples from patients vs heathy pancreatic tissues from individuals without pancreatic cancer. RESULTS: In sst2+/- mice, PI3K was activated and signaled via AKT (PKB; protein kinase B); when these mice were crossed with KRAS(G12D) mice, premalignant lesions, tumors, and lymph node metastases developed more rapidly than in KRAS(G12D) mice. In crossed KRAS(G12D);sst2+/- mice, activation of PI3K signaling via AKT resulted in activation of nuclear factor-κB (NF-κB), which increased KRAS activity and its downstream pathways, promoting initiation and progression of neoplastic lesions. We found this activation loop to be mediated by PI3K-induced production of the chemokine CXCL16. Administration of a CXCL16-neutralizing antibody to KRAS(G12D) mice reduced activation of PI3K signaling to AKT and NF-κB, blocking carcinogenesis. Levels of CXCL16 and its receptor CXCR6 were significantly higher in PDAC tissues and surrounding acini than in healthy pancreatic tissues from mice or human beings. In addition, expression of sst2 was progressively lost, involving increased PI3K activity, in mouse lesions that expressed KRAS(G12D) and progressed to PDAC. CONCLUSIONS: Based on analyses of mice, loss of sst2 from pancreatic tissues activates PI3K signaling via AKT, leading to activation of NF-κB, amplification of oncogenic KRAS signaling, increased expression of CXCL16, and pancreatic tumor formation. CXCL16 might be a therapeutic target for PDAC.


Asunto(s)
Carcinoma Ductal Pancreático/enzimología , Proliferación Celular , Quimiocina CXCL6/metabolismo , Mutación , Neoplasias Pancreáticas/enzimología , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Receptores de Somatostatina/deficiencia , Transducción de Señal , Animales , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/secundario , Estudios de Casos y Controles , Línea Celular Tumoral , Quimiocina CXCL16 , Quimiocinas CXC/metabolismo , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Humanos , Metástasis Linfática , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Fenotipo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Depuradores/metabolismo , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo , Factores de Tiempo , Transfección , Carga Tumoral , Regulación hacia Arriba
8.
PLoS One ; 9(9): e108032, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25238250

RESUMEN

FLICE/caspase-8-associated huge protein (FLASH)/casp8ap2 is involved in various cellular functions, such as cell cycle progression, transcriptional regulation, the regulation of apoptosis, and the regulation of histone gene expression. The down-regulated expression of FLASH has been shown to inhibit cell cycle progression in the S phase in many kinds of mice and human cell lines and the inhibition of cell cycle progression may be attributed to the suppressed expression of replication-dependent histone genes. We here demonstrated that the induced knockout of FLASH never affected cell cycle progression in ES cells, in which the expression of core histone genes was decreased to levels similar to those in human KB cells sensitive to the knockdown of FLASH. In addition, the FLASH conditional knockout ES cells could differentiate normally into not only mesodermal and endodermal cells, but also trophoblasts. In order to investigate the function of FLASH in early embryogenesis in vivo, we also examined a FLASH mutant mouse, in which FLASH mutant allele did not express FLASH mRNA in embryos and most adult organs, except for the testis. FLASH mutant embryos died between E3.5 and E8.5. Furthermore, the in vitro cultivation of FLASH mutant embryos generated by in vitro fertilization showed embryonic lethality at the pre-implantation stage by inhibiting the hatching of embryos and their adherence to substrates. Taken together, these results indicate that FLASH plays an important role in early embryogenesis, but is not essential for either the proliferation or differentiation of ES cells.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/fisiología , Proteínas de Unión al Calcio/fisiología , Diferenciación Celular/genética , Proliferación Celular/genética , Desarrollo Embrionario/genética , Células Madre Embrionarias/citología , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Células Madre Embrionarias/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Técnicas de Inactivación de Genes , Histonas/genética , Humanos , Ratones , Mutación
9.
Int Immunol ; 26(4): 221-31, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24343821

RESUMEN

Fas mutant mice are well recognized as autoimmune mouse models, which develop symptoms similar to human systemic lupus erythematosus. Although disease severity in Fas mutant mice is greatly affected by the genetic background, the mechanisms affecting pathological heterogeneity among different strains of Fas mutant mice are poorly understood. In this study, we examined the phenotypic differences between Fas-deficient (Fas (-/-)) mice on the BALB/c and C57BL/6 backgrounds to gain insight into the etiological and pathological heterogeneity of monogenic autoimmune diseases. Fas (-/-) mice on the BALB/c background (BALB/c-Fas (-/-)) developed more severe autoimmune disease with high serum auto-antibodies and renal disease compared with those on the C57BL/6 background (C57BL/6-Fas (-/-)). Splenic B cells were highly activated, and germinal center formation was enhanced in BALB/c-Fas (-/-) but not in C57BL/6-Fas (-/-) mice. Follicular helper T (Tfh) cells were equally abundant in the spleens from both strains of Fas (-/-) mice. However, Tfh cells from BALB/c-Fas (-/-) mice produced much higher amounts of B-cell-activating cytokines, including IL-4 and IL-10, a phenotype reminiscent of Th2-type Tfh cells described in human studies. Our results revealed a qualitative difference in Tfh cells between the two strains of Fas (-/-) mice. We propose that the pathogenic Th2-type Tfh cells in BALB/c-Fas (-/-) mice contribute to the excessive activation of B cells, resulting in high serum immunoglobulin levels and the severe lupus phenotype, which may account for the differential outcomes of human monogenic autoimmune diseases.


Asunto(s)
Linfocitos B/inmunología , Lupus Eritematoso Sistémico/inmunología , Células Th2/inmunología , Animales , Autoanticuerpos/sangre , Células Cultivadas , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Interleucina-10/metabolismo , Interleucina-4/metabolismo , Lupus Eritematoso Sistémico/genética , Activación de Linfocitos/genética , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Receptor fas/genética
10.
J Immunol ; 190(8): 4245-54, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23509366

RESUMEN

The molecular mechanisms of Fas (CD95/Apo-1)-mediated apoptosis are increasingly understood. However, the role of Fas-mediated production of proinflammatory cytokines such as IL-18 and IL-1ß in bacterial infection is unclear. We demonstrate the importance of Fas-mediated signaling in IL-18/IL-1ß production postinfection with Listeria monocytogenes without the contribution of caspase-1 inflammasome. IL-18/IL-1ß production in L. monocytogenes-infected peritoneal exudate cells from Fas-deficient mice was lower than those from wild type mice, indicating that Fas signaling contributes to cytokine production. L. monocytogenes infection induced Fas ligand expression on NK cells, which stimulates Fas expressed on the infected macrophages, leading to the production of IL-18/IL-1ß. This was independent of caspase-1, caspase-11, and nucleotide-binding domain and leucine-rich repeat-containing receptors (NLRs) such as Nlrp3 and Nlrc4, but dependent on apoptosis-associated speck-like protein containing a caspase recruitment domain. Wild type cells exhibited caspase-8 activation, whereas Fas-deficient cells did not. L. monocytogenes-induced caspase-8 activation was abrogated by inhibitor for intracellular reactive oxygen species, N-acetyl-L-cysteine. L. monocytogenes-infected macrophages produced type-I IFNs such as IFN-ß1, which was required for Il18 gene expression. Thus, Fas signaling regulates innate inflammatory cytokine production in L. monocytogenes infection.


Asunto(s)
Citocinas/biosíntesis , Mediadores de Inflamación/fisiología , Listeriosis/inmunología , Listeriosis/patología , Receptor fas/fisiología , Animales , Proteína Ligando Fas/biosíntesis , Femenino , Inmunidad Innata , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/microbiología , Mediadores de Inflamación/metabolismo , Interleucina-18/biosíntesis , Interleucina-1beta/biosíntesis , Listeriosis/metabolismo , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Transducción de Señal/inmunología , Receptor fas/biosíntesis , Receptor fas/metabolismo
11.
Int Immunol ; 25(6): 373-82, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23411727

RESUMEN

Fas (CD95), a member of the tumor necrosis factor receptor superfamily, mediates apoptosis-inducing signals in its expressing cells, especially in self-reactive cells. We recently reported that Fas(-/-) mice with a BALB/c background (BALB/c Fas(-/-) mice) developed blepharitis with allergic inflammation that was accompanied by hyper-IgE production. Here, we found a novel type of immunocyte in the spleen of BALB/c Fas(-/-) mice, which enhanced the production of IgE by B cells in the presence of IL-4 and CD40 signaling in vitro. The immunocyte did not express lineage markers but expressed Thy-1 and Sca-1 just like recently identified type 2 innate lymphoid cells, such as natural helper (NH) cells and nuocytes. However, they did not express c-Kit, IL-7R and IL-33R (T1/ST2), important markers of type 2 innate lymphoid cells. Instead, our identified Lin(-)Thy-1(+)Sca-1(+) cells expressed IL-18R and secreted Th2 cytokines when co-cultured with B cells or when stimulated with IL-18 and IL-2. Moreover, we found essentially the same type of cells in BALB/c wild-type mice as in BALB/c Fas(-/-) mice, which enhanced IgE production in contact with B cells in vitro. These cells from BALB/c wild-type mice expressed Fas and were sensitive to Fas-mediated apoptosis. Collectively, the newly identified Lin(-)Thy-1(+)Sca-1(+) cell, which we designated a F-NH cell (Fas-expressing natural helper cell), is a novel type 2 innate immunocyte with activity to enhance IgE production from B cells with the help of IL-4 and CD40 signaling. F-NH cells may play an important role in the development of chronic allergic inflammation.


Asunto(s)
Inmunidad Innata/inmunología , Inmunoglobulina E/biosíntesis , Linfocitos/inmunología , Animales , Células Cultivadas , Inmunoglobulina E/inmunología , Linfocitos/citología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor fas/deficiencia , Receptor fas/metabolismo
12.
Nat Genet ; 45(1): 98-103, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23202126

RESUMEN

There is great interest in tumor stem cells (TSCs) as potential therapeutic targets; however, cancer therapies targeting TSCs are limited. A drawback is that TSC markers are often shared by normal stem cells (NSCs); thus, therapies that target these markers may cause severe injury to normal tissues. To identify a potential TSC-specific marker, we focused on doublecortin-like kinase 1 (Dclk1). Dclk1 was reported as a candidate NSC marker in the gut, but recent reports have implicated it as a marker of differentiated cells (for example, Tuft cells). Using lineage-tracing experiments, we show here that Dclk1 does not mark NSCs in the intestine but instead marks TSCs that continuously produce tumor progeny in the polyps of Apc(Min/+) mice. Specific ablation of Dclk1-positive TSCs resulted in a marked regression of polyps without apparent damage to the normal intestine. Our data suggest the potential for developing a therapy for colorectal cancer based on targeting Dclk1-positive TSCs.


Asunto(s)
Mucosa Intestinal/metabolismo , Neoplasias Intestinales/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Células Madre Neoplásicas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Células Madre/metabolismo , Animales , Quinasas Similares a Doblecortina , Femenino , Orden Génico , Neoplasias Intestinales/patología , Pólipos Intestinales/genética , Pólipos Intestinales/metabolismo , Pólipos Intestinales/patología , Intestinos/patología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Ratones , Ratones Transgénicos , Proteínas Serina-Treonina Quinasas/metabolismo
13.
Int Immunol ; 25(5): 287-93, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23220580

RESUMEN

Fas (CD95) is a cell surface death receptor belonging to the tumor necrosis factor receptor superfamily, which mediates apoptosis-inducing signaling when activated by Fas ligand or its agonistic antibody. lpr mice with a loss of apoptosis-inducing function mutation in the Fas gene develop systemic autoimmune disease and lymphadenopathy but not allergic inflammation. In the case of Fas mutations including lpr and knockout (KO), background genes determine the incidence and severity of lymphadenopathy and histopathological manifestation of systemic autoimmunity: MRL-lpr/lpr mice and C57BL/6-lpr/lpr or C57BL/6 Fas KO mice develop severe and minimum disease, respectively. We generated Fas KO mice with the Balb/c background that show severer autoimmune phenotypes than MRL-lpr/lpr mice, such as critical infiltration of mononuclear cells into lung, liver and spleen, elevated serum levels of auto-antibodies and a decreased life span. To our astonishment, Balb/c Fas KO mice spontaneously develop blepharitis with not only autoimmune inflammation with deposition of auto-antibody but also allergic inflammation with infiltration by eosinophils and mast cells and show the capacity to strongly increase serum level of IgE and IgG1 along with their aging. Thus, Fas expression regulates development of not only autoimmune disease but also allergic inflammation.


Asunto(s)
Enfermedades Autoinmunes/metabolismo , Blefaritis/metabolismo , Hipersensibilidad/metabolismo , Inmunoglobulina E/biosíntesis , Inflamación/metabolismo , Receptor fas/deficiencia , Animales , Enfermedades Autoinmunes/patología , Blefaritis/patología , Hipersensibilidad/patología , Inmunoglobulina E/sangre , Inmunoglobulina E/metabolismo , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/sangre , Inmunoglobulina G/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados
14.
J Biol Chem ; 287(49): 41165-73, 2012 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-23071110

RESUMEN

Caspase-8 has an important role as an initiator caspase during death receptor-mediated apoptosis. Moreover, it has been reported to contribute to the regulation of cell fate in various types of cells including T-cells. In this report, we show that caspase-8 has an essential role in cell survival in mouse T-lymphoma-derived L5178Y cells. The knockdown of caspase-8 expression decreased the growth rate and increased cell death, both of which were induced by the absence of protease activity of procaspase-8. The cell death was associated with reactive oxygen species (ROS) accumulation, caspase activation, and autophagosome formation. The cell death was inhibited completely by treatment with ROS scavengers, but only partly by treatment with caspase inhibitors, expression of Bcl-xL, and knockdown of caspase-3 or Atg-7 which completely inhibits apoptosis or autophagosome formation, respectively, indicating that apoptosis and autophagy-associated cell death are induced simultaneously by the knockdown of caspase-8 expression. Further analysis indicated that RIP1 and RIP3 regulate this multiple cell death, because the cell death as well as ROS production was completely inhibited by not only treatment with the RIP1 inhibitor necrostatin-1, but also by knockdown of RIP3. Thus, in the absence of protease activity of procaspase-8, RIP1 and RIP3 simultaneously induce not only nonapoptotic cell death conceivably including autophagic cell death and necroptosis but also apoptosis through ROS production in mouse T-lymphoma cells.


Asunto(s)
Apoptosis , Caspasa 8/química , Proteínas de Complejo Poro Nuclear/química , Péptido Hidrolasas/química , Proteínas de Unión al ARN/química , Proteína Serina-Treonina Quinasas de Interacción con Receptores/química , Animales , Autofagia , Caspasa 8/metabolismo , Muerte Celular , Línea Celular Tumoral , Regulación Enzimológica de la Expresión Génica , Humanos , Linfoma de Células T/metabolismo , Ratones , Ratones Endogámicos BALB C , Necrosis , Especies Reactivas de Oxígeno , Linfocitos T/metabolismo
15.
J Biol Chem ; 287(12): 9568-78, 2012 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-22308035

RESUMEN

The transcriptional coactivator Yes-associated protein (YAP) has been implicated in tumorigenesis by regulating cell proliferation and apoptosis. YAP interacts with the transcription factor TEAD and is essential in mediating TEAD-dependent gene expression. Here we show that YAP is hyperphosphorylated and activated in response to genotoxic stress such as UV irradiation and cisplatin treatment. Using high resolution mobility shift assay for phosphorylated proteins, we identified multiple sites of phosphorylation induced by UV irradiation. Pretreatment with p38 and JNK inhibitors completely suppressed the mobility retardation of phosphorylated YAP in UV-irradiated cells. Co-immunoprecipitation experiments showed that the physical interaction of YAP with TEAD was markedly enhanced by UV irradiation or CDDP treatment but suppressed by pretreatment with p38 and JNK inhibitors. Similarly, pretreatment with p38 and JNK inhibitors suppressed the expression of YAP/TEAD target genes, which were elevated on exposure to genotoxic stress. Using phosphomimetic and phosphorylation-deficient YAP mutants, we showed that the coactivator activity of YAP correlated with its state of phosphorylation and sensitivity to cisplatin-induced apoptosis. Our results demonstrate that multisite phosphorylation of YAP induces YAP/TEAD-dependent gene expression and provides a mechanism by which YAP regulates apoptosis differently depending on cellular context.


Asunto(s)
Apoptosis , Células/citología , Proteínas Nucleares/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Activación Transcripcional , Proteínas de Ciclo Celular , Células/metabolismo , Daño del ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células HEK293 , Humanos , Proteínas Nucleares/genética , Fosforilación , Unión Proteica , Factores de Transcripción de Dominio TEA , Transactivadores/genética , Factores de Transcripción/genética
16.
Int Immunol ; 24(5): 283-92, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22301689

RESUMEN

B-cell receptor (BCR)-mediated apoptosis is critical for B-cell development and homeostasis. CD40 signaling has been shown to protect immature or mature B cells from BCR-mediated apoptosis. In this study, to understand the fate of CD40-pre-activated splenic B cells stimulated by BCR engagement in the presence of CD40 signaling, murine splenic B cells were cultured with anti-Igκ and anti-CD40 antibodies after pre-activation with anti-CD40 antibody. We found that apoptosis was induced in the cultured B cells even in the presence of CD40 signaling during the 3-4 days cultivation. We detected up-regulation of Bim expression followed by Bax activation in this apoptotic process and cessation of the apoptosis in Bim-deficient B cells, indicating that Bim is a key regulator of the BCR-mediated apoptosis in the presence of CD40 signaling in CD40-pre-activated B cells. Importantly, this BCR-mediated apoptosis in CD40-pre-activated B cells was shown to be induced at the initiation of plasma cell differentiation at around the preplasmablast stage, and Bim-deficient B cells cultured under these conditions differentiated into plasma cells. Additionally, transforming growth factor-ß was found to protect CD40-pre-activated B cells from BCR-mediated apoptosis in the presence of CD40 signaling. Our identified BCR-mediated apoptosis, which is unpreventable by CD40 signaling, suggests a potential mechanism that regulates the elimination of peripheral B cells, which should be derived from nonspecific T-dependent activation of bystander B cells and continuous stimulation with antigens including self-antigens in the presence of T cell help through CD40.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis/inmunología , Antígenos CD40/metabolismo , Diferenciación Celular , Proteínas de la Membrana/metabolismo , Células Plasmáticas/citología , Células Plasmáticas/inmunología , Proteínas Proto-Oncogénicas/metabolismo , Receptores de Antígenos de Linfocitos B/inmunología , Transducción de Señal/inmunología , Animales , Proteínas Reguladoras de la Apoptosis/deficiencia , Proteína 11 Similar a Bcl2 , Antígenos CD40/inmunología , Diferenciación Celular/inmunología , Activación de Linfocitos , Proteínas de la Membrana/deficiencia , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas/deficiencia , Bazo/citología , Bazo/inmunología
17.
Gut ; 60(11): 1494-505, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21471570

RESUMEN

BACKGROUND AND AIMS: Inflammatory bowel disease (IBD) is initiated and perpetuated by a dysregulated immune response to unknown environmental antigens such as luminal bacteria in genetically susceptible hosts. SR-PSOX/CXCL16, a scavenger receptor that binds phosphatidylserine and oxidised lipoprotein, has both phagocytic activity and chemotactic properties. The aim of this study was to investigate the role of SR-PSOX/CXCL16 in patients with IBD and experimental murine colitis. METHODS: The serum levels of SR-PSOX/CXCL16 were measured in patients with IBD. The roles of SR-PSOX/CXCL16 in phagocytosis of bacterial components and cytokine production by macrophages from wild-type (WT) and SR-PSOX/CXCL16 knockout (KO) mice were assessed. Colitis was induced by administering dextran sulfate sodium (DSS) to WT and SR-PSOX/CXCL16 KO mice. Colonic inflammation was analysed by clinical, histological and immunological parameters. Finally, the effect of a monoclonal antibody (mAb) to SR-PSOX/CXCL16 on DSS-induced colitis and trinitrobenzene sulfonic acid-induced colitis models was evaluated. RESULTS: Serum levels of SR-PSOX/CXCL16 correlated significantly with the disease activity of patients with IBD. Ex vivo experiments showed that SR-PSOX/CXCL16 was involved in both phagocytosis of bacterial antigens and the T helper 1 immune response through the production of interleukin 12 and interferon γ. In vivo murine experiments demonstrated the upregulated gene expression of SR-PSOX/CXCL16 in inflamed colonic tissues and the predominant expression of SR-PSOX/CXCL16 on macrophages. SR-PSOX/CXCL16 KO mice were less susceptible to colonic inflammation than were their WT littermates. Administration of SR-PSOX/CXCL16 mAb ameliorated the condition in the two different experimental colitis models. CONCLUSIONS: SR-PSOX/CXCL16 plays a critical role in colonic inflammation and could be a potential therapeutic target for patients with IBD.


Asunto(s)
Quimiocina CXCL6/fisiología , Quimiocinas CXC/fisiología , Enfermedades Inflamatorias del Intestino/inmunología , Receptores Depuradores/fisiología , Adulto , Animales , Quimiocina CXCL16 , Colon/patología , Citocinas/metabolismo , Progresión de la Enfermedad , Femenino , Expresión Génica/inmunología , Humanos , Enfermedades Inflamatorias del Intestino/fisiopatología , Interleucina-12/biosíntesis , Interleucina-12/inmunología , Macrófagos Peritoneales/inmunología , Masculino , Ratones , Ratones Noqueados , Membrana Mucosa/citología , Fagocitosis/fisiología , Adulto Joven
18.
J Biol Chem ; 285(33): 25418-25, 2010 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-20566630

RESUMEN

Caspases are central to apoptosis, and the principal executioner caspases, caspase-3 and -7, were reported to be similar in activity, primary structure, and three-dimensional structure. Here, we identified different activity in caspase-3 and -7 within cells and examined the relationship between their structure and function using human cells expressing almost equal amounts of exogenous caspase-3, caspase-7, and/or chimeric constructs after down-regulation of endogenous caspase-3 and -7 expression. Caspase-3 (produced in human cells) showed much stronger cleaving activity than caspase-7 against a low molecular weight substrate in vitro dependent on four specific amino acid regions. Within cells, however, an additional three regions were required for caspase-3 to exert much stronger protease activity than caspase-7 against cellular substrates. Three of the former four regions and the latter three regions were shown to form two different three-dimensional structures that were located at the interface of the homodimer of procaspase-7 on opposite sides. In addition, procaspase-3 and -7 revealed specific homodimer-forming activity within cells dependent on five amino acid regions, which were included in the regions critical to the cleaving activity within cells. Thus, human caspase-3 and -7 exhibit differences in protease activity, specific homodimer-forming activity, and three-dimensional structural features, all of which are closely interrelated.


Asunto(s)
Caspasa 3/metabolismo , Caspasa 7/metabolismo , Apoptosis/genética , Apoptosis/fisiología , Western Blotting , Caspasa 3/genética , Caspasa 7/genética , Células Cultivadas , Colorimetría , Células HeLa , Humanos , Células Jurkat
19.
Mol Cell ; 36(2): 193-206, 2009 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-19854130

RESUMEN

Budding yeast Cdc13, Stn1, and Ten1 form the CST complex to protect telomeres from lethal DNA degradation. It remains unknown whether similar complexes are conserved in higher eukaryotes or not. Here we isolated mammalian STN1 and TEN1 homologs and CTC1 (conserved telomere maintenance component 1). The three proteins contain putative OB-fold domains and form a complex called CST, which binds to single-stranded DNA with high affinity in a sequence-independent manner. CST associates with a fraction of telomeres consistently during the cell cycle, in quiescent cells and Pot1-knockdown cells. It does not colocalize with replication foci in S phase. Significant increases in the abundance of single-stranded G-strand telomeric DNA were observed in Stn1-knockdown cells. We propose that CST is a replication protein A (RPA)-like complex that is not directly involved in conventional DNA replication at forks but plays a role in DNA metabolism frequently required by telomeres.


Asunto(s)
ADN de Cadena Simple/metabolismo , Proteína de Replicación A/metabolismo , Proteínas de Unión a Telómeros/metabolismo , Telómero/metabolismo , Animales , Secuencia de Bases , Células HeLa , Humanos , Ratones , Datos de Secuencia Molecular , Complejos Multiproteicos/metabolismo , Proteínas Mutantes/metabolismo , Unión Proteica , Multimerización de Proteína , Estructura Terciaria de Proteína , Transporte de Proteínas , Proteínas Recombinantes , Homología de Secuencia de Aminoácido , Complejo Shelterina , Proteínas de Unión a Telómeros/química
20.
Mol Cell Biol ; 29(17): 4729-41, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19546234

RESUMEN

FLASH has been shown to be required for S phase progression and to interact with a nuclear protein, ataxia-telangiectasia locus (NPAT), a component of Cajal bodies in the nucleus and an activator of histone transcription. We investigated the role of human FLASH by using an inducible FLASH knockdown system in the presence or absence of various mutant forms of mouse FLASH. While carboxyl-terminal deletion mutants of FLASH, which do not interact with NPAT, can support S phase progression, its amino-terminal deletion mutants, which are unable to self associate, cannot support S phase progression, replication-dependent histone transcription, or the formation of Cajal bodies. Furthermore, FLASH was shown to be associated with arsenite resistance protein 2 (ARS2) through its central region, which is composed of only 13 amino acids. The expression of ARS2 and the interaction between FLASH and ARS2 are required for S phase progression. Taking these results together, FLASH functions in S phase progression through interaction with ARS2.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Proteínas de Ciclo Celular/metabolismo , Ciclo Celular/fisiología , Proteínas Nucleares/metabolismo , Fase S/fisiología , Secuencia de Aminoácidos , Animales , Proteínas de Unión al Calcio/genética , Proteínas de Ciclo Celular/genética , Línea Celular , Histonas/genética , Histonas/metabolismo , Humanos , Ratones , Datos de Secuencia Molecular , Proteínas Nucleares/genética , ARN/genética , ARN/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...