Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Toxicol Lett ; 396: 1-10, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38588756

RESUMEN

The surge in opioid-related deaths, driven predominantly by fentanyl and its synthetic derivatives, has become a critical public health concern, which is particularly evident in the United States. While the situation is less severe in Europe, the European Monitoring Centre for Drugs and Drug Addiction reports a rise in drug overdose deaths, with emerging concerns about the impact of fentanyl-related molecules. Synthetic opioids, initially designed for medical use, have infiltrated illicit markets due to their low production costs and high potency, with carfentanil posing additional threats, including potential chemical weaponization. Existing overdose mitigation heavily relies on naloxone, requiring timely intervention and caregiver presence, while therapeutic prevention strategies face many access challenges. To provide an additional treatment option, we propose the use of a fentanyl-specific monoclonal antibody (mAb), as a non-opioid method of prophylaxis against fentanyl and carfentanil. This mAb shows protection from opioid effects in a pre-clinical murine model. mAbs could emerge as a versatile countermeasure in civilian and biodefense settings, offering a novel approach to combat opioid-associated mortality.


Asunto(s)
Analgésicos Opioides , Anticuerpos Monoclonales , Fentanilo , Fentanilo/análogos & derivados , Fentanilo/inmunología , Animales , Ratones , Humanos
2.
PLoS Negl Trop Dis ; 17(9): e0011621, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37656766

RESUMEN

Long-term immune evasion by the African trypanosome is achieved through repetitive cycles of surface protein replacement with antigenically distinct versions of the dense Variant Surface Glycoprotein (VSG) coat. Thousands of VSG genes and pseudo-genes exist in the parasite genome that, together with genetic recombination mechanisms, allow for essentially unlimited immune escape from the adaptive immune system of the host. The diversity space of the "VSGnome" at the protein level was thought to be limited to a few related folds whose structures were determined more than 30 years ago. However, recent progress has shown that the VSGs possess significantly more architectural variation than had been appreciated. Here we combine experimental X-ray crystallography (presenting structures of N-terminal domains of coat proteins VSG11, VSG21, VSG545, VSG558, and VSG615) with deep-learning prediction using Alphafold to produce models of hundreds of VSG proteins. We classify the VSGnome into groups based on protein architecture and oligomerization state, contextualize recent bioinformatics clustering schemes, and extensively map VSG-diversity space. We demonstrate that in addition to the structural variability and post-translational modifications observed thus far, VSGs are also characterized by variations in oligomerization state and possess inherent flexibility and alternative conformations, lending additional variability to what is exposed to the immune system. Finally, these additional experimental structures and the hundreds of Alphafold predictions confirm that the molecular surfaces of the VSGs remain distinct from variant to variant, supporting the hypothesis that protein surface diversity is central to the process of antigenic variation used by this organism during infection.


Asunto(s)
Variación Antigénica , Glicoproteínas de Membrana , Proteínas Protozoarias , Trypanosoma , Proteínas de la Membrana
3.
Cell Rep ; 42(3): 112262, 2023 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-36943866

RESUMEN

The African trypanosome survives the immune response of its mammalian host by antigenic variation of its major surface antigen (the variant surface glycoprotein or VSG). Here we describe the antibody repertoires elicited by different VSGs. We show that the repertoires are highly restricted and are directed predominantly to distinct epitopes on the surface of the VSGs. They are also highly discriminatory; minor alterations within these exposed epitopes confer antigenically distinct properties to these VSGs and elicit different repertoires. We propose that the patterned and repetitive nature of the VSG coat focuses host immunity to a restricted set of immunodominant epitopes per VSG, eliciting a highly stereotyped response, minimizing cross-reactivity between different VSGs and facilitating prolonged immune evasion through epitope variation.


Asunto(s)
Trypanosoma brucei brucei , Trypanosoma , Animales , Epítopos Inmunodominantes , Evasión Inmune , Glicoproteínas Variantes de Superficie de Trypanosoma , Variación Antigénica , Epítopos , Mamíferos
4.
Cell Rep ; 42(2): 112049, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36719797

RESUMEN

Poorly immunogenic small molecules pose challenges for the production of clinically efficacious vaccines and antibodies. To address this, we generate an immunization platform derived from the immunogenic surface coat of the African trypanosome. Through sortase-based conjugation of the target molecules to the variant surface glycoprotein (VSG) of the trypanosome surface coat, we develop VSG-immunogen array by sortase tagging (VAST). VAST elicits antigen-specific memory B cells and antibodies in a murine model after deploying the poorly immunogenic molecule fentanyl as a proof of concept. We also develop a single-cell RNA sequencing (RNA-seq)-based computational method that synergizes with VAST to specifically identify memory B cell-encoded antibodies. All computationally selected antibodies bind to fentanyl with picomolar affinity. Moreover, these antibodies protect mice from fentanyl effects after passive immunization, demonstrating the ability of these two coupled technologies to elicit therapeutic antibodies to challenging immunogens.


Asunto(s)
Trypanosoma brucei brucei , Trypanosoma , Tripanosomiasis Africana , Animales , Ratones , Trypanosoma brucei brucei/genética , Tripanosomiasis Africana/tratamiento farmacológico , Analgésicos Opioides , Fentanilo/farmacología , Fentanilo/uso terapéutico , Glicoproteínas Variantes de Superficie de Trypanosoma , Inmunoterapia
5.
Nat Commun ; 13(1): 3308, 2022 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-35676259

RESUMEN

During the ongoing COVID-19 pandemic, PCR testing and antigen tests have proven critical for helping to stem the spread of its causative agent, SARS-CoV-2. However, these methods suffer from either general applicability and/or sensitivity. Moreover, the emergence of variant strains creates the need for flexibility to correctly and efficiently diagnose the presence of substrains. To address these needs we developed the diagnostic test ADESSO (Accurate Detection of Evolving SARS-CoV-2 through SHERLOCK (Specific High Sensitivity Enzymatic Reporter UnLOCKing) Optimization) which employs Cas13 to diagnose patients in 1 h without sophisticated equipment. Using an extensive panel of clinical samples, we demonstrate that ADESSO correctly identifies infected individuals at a sensitivity and specificity comparable to RT-qPCR on extracted RNA and higher than antigen tests for unextracted samples. Altogether, ADESSO is a fast, sensitive and cheap method that can be applied in a point of care setting to diagnose COVID-19 and can be quickly adjusted to detect new variants.


Asunto(s)
COVID-19 , COVID-19/diagnóstico , Prueba de COVID-19 , Humanos , Pandemias , ARN Viral/análisis , ARN Viral/genética , SARS-CoV-2 , Sensibilidad y Especificidad
6.
PLoS Pathog ; 17(9): e1008768, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34559857

RESUMEN

Trypanosome Lytic Factor (TLF) is a primate-specific high-density lipoprotein (HDL) complex that, through the cation channel-forming protein apolipoprotein L-1 (APOL1), provides innate immunity to select kinetoplastid parasites. The immunoprotective effects of TLF have been extensively investigated in the context of its interaction with the extracellular protozoan Trypanosoma brucei brucei, to which it confers sterile immunity. We previously showed that TLF could act against an intracellular pathogen Leishmania, and here we dissected the role of TLF and its synergy with host-immune cells. Leishmania major is transmitted by Phlebotomine sand flies, which deposit the parasite intradermally into mammalian hosts, where neutrophils are the predominant phagocytes recruited to the site of infection. Once in the host, the parasites are phagocytosed and shed their surface glycoconjugates during differentiation to the mammalian-resident amastigote stage. Our data show that mice producing TLF have reduced parasite burdens when infected intradermally with metacyclic promastigotes of L. major, the infective, fly-transmitted stage. This TLF-mediated reduction in parasite burden was lost in neutrophil-depleted mice, suggesting that early recruitment of neutrophils is required for TLF-mediated killing of L. major. In vitro we find that only metacyclic promastigotes co-incubated with TLF in an acidic milieu were lysed. However, amastigotes were not killed by TLF at any pH. These findings correlated with binding experiments, revealing that labeled TLF binds specifically to the surface of metacyclic promastigotes, but not to amastigotes. Metacyclic promastigotes of L. major deficient in the synthesis of surface glycoconjugates LPG and/or PPG (lpg1- and lpg5A-/lpg5B- respectively) whose absence mimics the amastigote surface, were resistant to TLF-mediated lysis. We propose that TLF binds to the outer surface glycoconjugates of metacyclic promastigotes, whereupon it kills the parasite in the acidic phagosome of phagocytes. We hypothesize that resistance to TLF requires shedding of the surface glycoconjugates, which occurs several hours after phagocytosis by immune cells, creating a relatively short-lived but effective window for TLF to act against Leishmania.


Asunto(s)
Interacciones Huésped-Parásitos/fisiología , Inmunidad Innata , Leishmaniasis Cutánea , Lipoproteínas HDL/metabolismo , Animales , Humanos , Leishmania major , Leishmaniasis Cutánea/inmunología , Leishmaniasis Cutánea/metabolismo , Leishmaniasis Cutánea/patología , Lipoproteínas HDL/inmunología , Ratones
7.
PLoS Negl Trop Dis ; 15(9): e0009814, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34587165

RESUMEN

Anemia caused by trypanosome infection is poorly understood. Autoimmunity during Trypanosoma brucei infection was proposed to have a role during anemia, but the mechanisms involved during this pathology have not been elucidated. In mouse models and human patients infected with malaria parasites, atypical B-cells promote anemia through the secretion of autoimmune anti-phosphatidylserine (anti-PS) antibodies that bind to uninfected erythrocytes and facilitate their clearance. Using mouse models of two trypanosome infections, Trypanosoma brucei and Trypanosoma cruzi, we assessed levels of autoantibodies and anemia. Our results indicate that acute T. brucei infection, but not T. cruzi, leads to early increased levels of plasma autoantibodies against different auto antigens tested (PS, DNA and erythrocyte lysate) and expansion of atypical B cells (ABCs) that secrete these autoantibodies. In vitro studies confirmed that a lysate of T. brucei, but not T. cruzi, could directly promote the expansion of these ABCs. PS exposure on erythrocyte plasma membrane seems to be an important contributor to anemia by delaying erythrocyte recovery since treatment with an agent that prevents binding to it (Annexin V) ameliorated anemia in T. brucei-infected mice. Analysis of the plasma of patients with human African trypanosomiasis (HAT) revealed high levels of anti-PS antibodies that correlated with anemia. Altogether these results suggest a relation between autoimmunity against PS and anemia in both mice and patients infected with T. brucei.


Asunto(s)
Anemia/etiología , Autoinmunidad , Fosfatidilserinas/inmunología , Tripanosomiasis Africana/inmunología , Adolescente , Adulto , Animales , Autoanticuerpos/inmunología , Eritrocitos/inmunología , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Trypanosoma , Trypanosoma brucei brucei/inmunología , Tripanosomiasis Africana/complicaciones , Adulto Joven
8.
Nat Microbiol ; 6(3): 392-400, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33462435

RESUMEN

Suramin has been a primary early-stage treatment for African trypanosomiasis for nearly 100 yr. Recent studies revealed that trypanosome strains that express the variant surface glycoprotein (VSG) VSGsur possess heightened resistance to suramin. Here, we show that VSGsur binds tightly to suramin but other VSGs do not. By solving high-resolution crystal structures of VSGsur and VSG13, we also demonstrate that these VSGs define a structurally divergent subgroup of the coat proteins. The co-crystal structure of VSGsur with suramin reveals that the chemically symmetric drug binds within a large cavity in the VSG homodimer asymmetrically, primarily through contacts of its central benzene rings. Structure-based, loss-of-contact mutations in VSGsur significantly decrease the affinity to suramin and lead to a loss of the resistance phenotype. Altogether, these data show that the resistance phenotype is dependent on the binding of suramin to VSGsur, establishing that the VSG proteins can possess functionality beyond their role in antigenic variation.


Asunto(s)
Resistencia a Medicamentos/inmunología , Suramina/metabolismo , Trypanosoma brucei rhodesiense/inmunología , Glicoproteínas Variantes de Superficie de Trypanosoma/química , Glicoproteínas Variantes de Superficie de Trypanosoma/metabolismo , Variación Antigénica/efectos de los fármacos , Variación Antigénica/inmunología , Sitios de Unión , Cristalografía por Rayos X , Resistencia a Medicamentos/genética , Endocitosis/genética , Evasión Inmune , Mutación , Unión Proteica , Conformación Proteica , Suramina/toxicidad , Tripanocidas/metabolismo , Tripanocidas/toxicidad , Trypanosoma brucei rhodesiense/química , Trypanosoma brucei rhodesiense/efectos de los fármacos , Trypanosoma brucei rhodesiense/metabolismo , Tripanosomiasis Africana/parasitología , Glicoproteínas Variantes de Superficie de Trypanosoma/genética
9.
J Biol Chem ; 295(38): 13138-13149, 2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-32727852

RESUMEN

The human innate immunity factor apolipoprotein L-I (APOL1) protects against infection by several protozoan parasites, including Trypanosoma brucei brucei Endocytosis and acidification of high-density lipoprotein-associated APOL1 in trypanosome endosomes leads to eventual lysis of the parasite due to increased plasma membrane cation permeability, followed by colloid-osmotic swelling. It was previously shown that recombinant APOL1 inserts into planar lipid bilayers at acidic pH to form pH-gated nonselective cation channels that are opened upon pH neutralization. This corresponds to the pH changes encountered during endocytic recycling, suggesting APOL1 forms a cytotoxic cation channel in the parasite plasma membrane. Currently, the mechanism and domains required for channel formation have yet to be elucidated, although a predicted helix-loop-helix (H-L-H) was suggested to form pores by virtue of its similarity to bacterial pore-forming colicins. Here, we compare recombinant human and baboon APOL1 orthologs, along with interspecies chimeras and individual amino acid substitutions, to identify regions required for channel formation and pH gating in planar lipid bilayers. We found that whereas neutralization of glutamates within the H-L-H may be important for pH-dependent channel formation, there was no evidence of H-L-H involvement in either pH gating or ion selectivity. In contrast, we found two residues in the C-terminal domain, tyrosine 351 and glutamate 355, that influence pH gating properties, as well as a single residue, aspartate 348, that determines both cation selectivity and pH gating. These data point to the predicted transmembrane region closest to the APOL1 C terminus as the pore-lining segment of this novel channel-forming protein.


Asunto(s)
Apolipoproteína L1/química , Inmunidad Innata , Animales , Apolipoproteína L1/genética , Apolipoproteína L1/inmunología , Secuencias Hélice-Asa-Hélice , Humanos , Concentración de Iones de Hidrógeno , Papio hamadryas , Trypanosoma brucei brucei/inmunología
10.
Cell Host Microbe ; 28(1): 79-88.e4, 2020 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-32416060

RESUMEN

Trypanosomiasis is a devastating neglected tropical disease affecting livestock and humans. Humans are susceptible to two Trypanosoma brucei subspecies but protected from other trypanosomes by circulating high-density lipoprotein (HDL) complexes called trypanosome lytic factors (TLFs) 1 and 2. TLFs contain apolipoprotein L-1 contributing to lysis and haptoglobin-related protein (HPR), which can function as a ligand for a parasite receptor. TLF2 also uniquely contains non-covalently associated immunoglobin M (IgM) antibodies, the role and origin of which remain unclear. Here, we show that these TLF2-associated IgMs interact with both HPR and alternate trypanosome surface proteins, including variant surface glycoprotein, likely facilitating complex biogenesis and TLF uptake into parasites. TLF2-IgMs are germline antibodies that, while present at basal concentrations in healthy individuals, are elicited by trypanosome infection in both murine models and human sleeping sickness patients. These data suggest that poly- and self-reactive germline antibodies such as TLF2-associated IgMs play a role in antimicrobial immunity.


Asunto(s)
Anticuerpos Antiprotozoarios/inmunología , Antígenos de Neoplasias/inmunología , Apolipoproteína L1/inmunología , Haptoglobinas/inmunología , Inmunoglobulina M/inmunología , Lipoproteínas HDL/inmunología , Tripanosomiasis Africana/inmunología , Adolescente , Adulto , Anciano , Animales , Línea Celular , Niño , Femenino , Células Germinativas/inmunología , Interacciones Huésped-Parásitos , Humanos , Masculino , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Noqueados , Persona de Mediana Edad , Modelos Animales , Parásitos , Trypanosoma brucei brucei , Adulto Joven
11.
Methods Mol Biol ; 2116: 463-483, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32221937

RESUMEN

Interest in trypanosome lytic factors (TLFs) and apolipoprotein L1, the ion channel-forming protein component of TLFs, has increased tenfold since 2010. This is due to the association of African variants of APOL1 with kidney disease such that interest has reached circles beyond parasitology. We have extensive experience purifying and working with these proteins and protein complexes. Herein we describe our detailed purification protocols to aid the new burgeoning field by providing an opportunity for consistency in reagents used across laboratories. We emphasize that it is imperative to maintain APOL1 protein intact (~42 kDa) to analyze the active ion channel-forming component/protein.


Asunto(s)
Apolipoproteína L1/aislamiento & purificación , Lipoproteínas HDL/aislamiento & purificación , Tripanosomiasis Africana/sangre , Apolipoproteína L1/sangre , Apolipoproteína L1/química , Apolipoproteína L1/metabolismo , Humanos , Enfermedades Renales/sangre , Enfermedades Renales/inmunología , Lipoproteínas HDL/química , Lipoproteínas HDL/metabolismo , Peso Molecular , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Trypanosoma/inmunología , Tripanosomiasis Africana/complicaciones , Tripanosomiasis Africana/inmunología , Tripanosomiasis Africana/parasitología
12.
Nat Microbiol ; 3(8): 932-938, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29988048

RESUMEN

The African trypanosome Trypanosoma brucei spp. is a paradigm for antigenic variation, the orchestrated alteration of cell surface molecules to evade host immunity. The parasite elicits robust antibody-mediated immune responses to its variant surface glycoprotein (VSG) coat, but evades immune clearance by repeatedly accessing a large genetic VSG repertoire and 'switching' to antigenically distinct VSGs. This persistent immune evasion has been ascribed exclusively to amino-acid variance on the VSG surface presented by a conserved underlying protein architecture. We establish here that this model does not account for the scope of VSG structural and biochemical diversity. The 1.4-Å-resolution crystal structure of the variant VSG3 manifests divergence in the tertiary fold and oligomeric state. The structure also reveals an O-linked carbohydrate on the top surface of VSG3. Mass spectrometric analysis indicates that this O-glycosylation site is heterogeneously occupied in VSG3 by zero to three hexose residues and is also present in other VSGs. We demonstrate that this O-glycosylation increases parasite virulence by impairing the generation of protective immunity. These data alter the paradigm of antigenic variation by the African trypanosome, expanding VSG variability beyond amino-acid sequence to include surface post-translational modifications with immunomodulatory impact.


Asunto(s)
Anticuerpos Antiprotozoarios/metabolismo , Trypanosoma brucei brucei/patogenicidad , Glicoproteínas Variantes de Superficie de Trypanosoma/química , Glicoproteínas Variantes de Superficie de Trypanosoma/genética , Sitios de Unión , Cristalografía por Rayos X , Variación Genética , Glicosilación , Modelos Moleculares , Conformación Proteica , Dominios Proteicos , Trypanosoma brucei brucei/inmunología , Glicoproteínas Variantes de Superficie de Trypanosoma/inmunología
13.
J Cell Biochem ; 118(2): 237-251, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27302072

RESUMEN

Mammalian NUMB is alternatively spliced generating four isoforms NUMB1-NUMB4 that can function as tumor suppressors. NUMB1-NUMB4 proteins, which normally determine how different cell types develop, are reduced in 21% of primary breast tumors. Our previous work has, however, indicated that two novel NUMB isoforms, NUMB5 and NUMB6 have the pro-oncogenic functions. Herein, we address a novel function of human NUMB isoform 6 (NUMB6) in promoting cancer cell migration and invasion. We found that NUMB6 induced expression of embryonic transcription factor Slug, which in turn actively repressed E-cadherin, prompting cells to undergo epithelial-mesenchymal transition (EMT). Low-metastatic breast cancer cells DB-7 stably expressing NUMB6, lost their epithelial phenotype, exhibited migratory and pro-invasive behavior, and ultimately elevated expression of mesenchymal markers. Among these markers, increased vimentin, ß-catenin, and fibronectin expression elicited metalloproteinase 9 (MMP9) production. Our results revealed that NUMB6-DB-7 cells have significantly increased level of Akt1 and Akt2 phosphorylation. Therefore, antagonizing Akt signaling using a chemical inhibitor LY294002, we found that NUMB6-induced Slug expression was reduced, and ultimately accompanied with decreased cell migration and invasion. In summary, this study identified a novel molecular determinant of breast cancer progression, uncovering a potential oncogenic role for the NUMB6 protein in cancer cell migration and invasion, coupled to the maintenance of mesenchymal-like cells. J. Cell. Biochem. 118: 237-251, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Neoplasias de la Mama/metabolismo , Movimiento Celular , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Células Madre Neoplásicas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Humanos , Proteínas de la Membrana/genética , Invasividad Neoplásica , Células Madre Neoplásicas/patología , Proteínas del Tejido Nervioso/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
14.
PLoS One ; 6(7): e20659, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21789165

RESUMEN

In normal growth and development, apoptosis is necessary to shape the central nervous system and to eliminate excess neurons which are not required for innervation. In some diseases, however, apoptosis can be either overactive as in some neurodegenerative disorders or severely attenuated as in the spread of certain cancers. Bone morphogenetic proteins (BMPs) transmit signals for regulating cell growth, differentiation, and apoptosis. Responding to BMP receptors stimulated from BMP ligands, neurotrophin receptor-mediated MAGE homolog (NRAGE) binds and functions with the XIAP-TAK1-TAB1 complex to activate p38(MAPK) and induces apoptosis in cortical neural progenitors. NRAGE contains a unique repeat domain that is only found in human, mouse, and rat homologs that we theorize is pivotal in its BMP MAPK role. Previously, we showed that deletion of the repeat domain inhibits apoptosis, p38(MAPK) phosphorylation, and caspase-3 cleavage in P19 neural progenitor cells. We also showed that the XIAP-TAB1-TAK1 complex is dependent on NRAGE for IKK-α/ß phosphorylation and NF-κB activation. XIAP is a major inhibitor of caspases, the main executioners of apoptosis. Although it has been shown previously that NRAGE binds to the RING domain of XIAP, it has not been determined which NRAGE domain binds to XIAP. Here, we used fluorescence resonance energy transfer (FRET) to determine that there is a strong likelihood of a direct interaction between NRAGE and XIAP occurring at NRAGE's unique repeat domain which we also attribute to be the domain responsible for downstream signaling of NF-κB and activating IKK subunits. From these results, we designed a small peptide modeled after the NRAGE repeat domain which we have determined inhibits NF-κB activation and apoptosis in P19 cells. These intriguing results illustrate that the paradigm of the NRAGE repeat domain may hold promising therapeutic strategies in developing pharmaceutical solutions for combating harmful diseases involving excessive downstream BMP signaling, including apoptosis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Apoptosis/efectos de los fármacos , Quinasas Quinasa Quinasa PAM/metabolismo , FN-kappa B/metabolismo , Proteínas de Neoplasias/química , Péptidos/farmacología , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Compartimento Celular/efectos de los fármacos , Citoplasma/efectos de los fármacos , Citoplasma/metabolismo , ADN/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Ratones , Microscopía Confocal , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Datos de Secuencia Molecular , Proteínas de Neoplasias/metabolismo , Péptidos/química , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Transporte de Proteínas/efectos de los fármacos , Secuencias Repetitivas de Aminoácido , Transducción de Señal/efectos de los fármacos
15.
J Cell Physiol ; 226(11): 3064-75, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21302306

RESUMEN

FGF1, a widely expressed proangiogenic factor involved in tissue repair and carcinogenesis, is released from cells through a non-classical pathway independent of endoplasmic reticulum and Golgi. Although several proteins participating in FGF1 export were identified, genetic mechanisms regulating this process remained obscure. We found that FGF1 export and expression are regulated through Notch signaling mediated by transcription factor CBF1 and its partner MAML. The expression of a dominant negative (dn) form of CBF1 in 3T3 cells induces transcription of FGF1 and sphingosine kinase 1 (SphK1), which is a component of FGF1 export pathway. dnCBF1 expression stimulates the stress-independent release of transduced FGF1 from NIH 3T3 cells and endogenous FGF1 from A375 melanoma cells. NIH 3T3 cells transfected with dnCBF1 form colonies in soft agar and produce rapidly growing highly angiogenic tumors in nude mice. The transformed phenotype of dnCBF1 transfected cells is efficiently blocked by dn forms of FGF receptor 1 and S100A13, which is a component of FGF1 export pathway. FGF1 export and acceleration of cell growth induced by dnCBF1 depend on SphK1. Similar to dnCBF1, dnMAML transfection induces FGF1 expression and release, and accelerates cell proliferation. The latter effect is strongly decreased in FGF1 null cells. We suggest that the regulation of FGF1 expression and release by CBF1-mediated Notch signaling can play an important role in tumor formation.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Factor 1 de Crecimiento de Fibroblastos/metabolismo , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Receptores Notch/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/genética , Humanos , Melanoma/tratamiento farmacológico , Melanoma/metabolismo , Ratones , Ratones Desnudos , Células 3T3 NIH , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Proteínas Nucleares/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteínas S100/farmacología , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/metabolismo , Transfección
16.
Neural Dev ; 5: 31, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21122105

RESUMEN

We previously identified four functionally distinct human NUMB isoforms. Here, we report the identification of two additional isoforms and propose a link between the expression of these isoforms and cancer. These novel isoforms, NUMB5 and NUMB6, lack exon 10 and are expressed in cells known for polarity and migratory behavior, such as human amniotic fluid cells, glioblastoma and metastatic tumor cells. RT-PCR and luciferase assays demonstrate that NUMB5 and NUMB6 are less antagonistic to NOTCH signaling than other NUMB isoforms. Immunocytochemistry analyses show that NUMB5 and NUMB6 interact and complex with CDC42, vimentin and the CDC42 regulator IQGAP1 (IQ (motif) GTPase activating protein 1). Furthermore, the ectopic expression of NUMB5 and NUMB6 induces the formation of lamellipodia (NUMB5) and filopodia (NUMB6) in a CDC42- and RAC1-dependent manner. These results are complemented by in vitro and in vivo studies, demonstrating that NUMB5 and NUMB6 alter the migratory behavior of cells. Together, these novel isoforms may play a role in further understanding the NUMB function in development and cancer.


Asunto(s)
Desarrollo Fetal/fisiología , Regulación Neoplásica de la Expresión Génica , Proteínas de la Membrana/genética , Neoplasias/genética , Proteínas del Tejido Nervioso/genética , Transducción de Señal/fisiología , Animales , Western Blotting , Movimiento Celular/genética , Polaridad Celular/genética , Embrión de Pollo , Perfilación de la Expresión Génica , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Neoplasias/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales , Neurogénesis/fisiología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
BMC Biol ; 8: 7, 2010 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-20100315

RESUMEN

BACKGROUND: Previous studies have linked neurotrophin receptor-interacting MAGE protein to the bone morphogenic protein signaling pathway and its effect on p38 mediated apoptosis of neural progenitor cells via the XIAP-Tak1-Tab1 complex. Its effect on NF-kappaB has yet to be explored. RESULTS: Herein we report that NRAGE, via the same XIAP-Tak1-Tab1 complex, is required for the phosphorylation of IKK -alpha/beta and subsequent transcriptional activation of the p65 subunit of NF-kappaB. Ablation of endogenous NRAGE by siRNA inhibited NF-kappaB pathway activation, while ablation of Tak1 and Tab1 by morpholino inhibited overexpression of NRAGE from activating NF-kappaB. Finally, cytokine profiling of an NRAGE over-expressing stable line revealed the expression of macrophage migration inhibitory factor. CONCLUSION: Modulation of NRAGE expression revealed novel roles in regulating NF-kappaB activity in the non-canonical bone morphogenic protein signaling pathway. The expression of macrophage migration inhibitory factor by bone morphogenic protein -4 reveals novel crosstalk between an immune cytokine and a developmental pathway.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas de Neoplasias/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Transcripción ReIA/metabolismo , Animales , Antígenos de Neoplasias/genética , Western Blotting , Proteína Morfogenética Ósea 4/farmacología , Proteínas Morfogenéticas Óseas/genética , Línea Celular , Línea Celular Tumoral , Humanos , Quinasa I-kappa B/metabolismo , Inmunohistoquímica , Inmunoprecipitación , Riñón/metabolismo , Factores Inhibidores de la Migración de Macrófagos/genética , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Ratones , Ratones Transgénicos , Proteínas de Neoplasias/genética , Fosforilación/efectos de los fármacos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/fisiología , Transducción de Señal/genética , Factor de Transcripción ReIA/genética
18.
Apoptosis ; 15(1): 63-70, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19937275

RESUMEN

Bone morphogenetic signaling (BMP) is a key pathway during neurogenesis and depends on many downstream intermediators to carry out its signaling. One such signaling pathway utilizes neurotrophin receptor-interacting MAGE protein (NRAGE), a member of the melanoma-associated antigen (MAGE) family, to upregulate p38 mitogen activated protein kinase (p38(MAPK)) in response to cellular stress and activate caspases which are critical in leading cells to death. NRAGE consists of two conserved MAGE homology domains separated by a unique hexapeptide repeat domain. Although we have previously implicated NRAGE in inducing apoptosis in neural progenitors and P19 cells, a model system for neural progenitors, its domains have yet to be explored in determining which one may be responsible for setting up the signaling for apoptosis. Here, we overexpressed a series of deletion mutations in P19 cells to show that only those with at least half of the repeat domain, activated p38(MAPK) and underwent apoptosis offering intriguing incite into NRAGE's contribution in BMP apoptotic signaling.


Asunto(s)
Apoptosis , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas de Neoplasias/química , Transducción de Señal , Animales , Proteínas Morfogenéticas Óseas/genética , Línea Celular , Supervivencia Celular , Femenino , Humanos , Ratones , Ratones Endogámicos ICR , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Estructura Terciaria de Proteína , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
19.
Mech Dev ; 126(5-6): 337-49, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19268530

RESUMEN

Branching morphogenesis is a developmental process characteristic of many organ systems. Specifically, during renal branching morphogenesis, its been postulated that the final number of nephrons formed is one key clinical factor in the development of hypertension in adulthood. As it has been established that BMPs regulate, in part, renal activity of p38 MAP kinase (p38(MAPK)) and it has demonstrated that the cytoplasmic protein Neurotrophin Receptor MAGE homologue (NRAGE) augments p38(MAPK) activation, it was hypothesized that a decrease in the expression of NRAGE during renal branching would result in decreased branching of the UB that correlated with changes in p38(MAPK) activation. To verify this, the expression of NRAGE was reduced in ex vivo kidney explants cultures using antisense morpholino. Morpholino treated ex vivo kidney explants expression were severely stunted in branching, a trait that was rescued with the addition of exogenous GDNF. Renal explants also demonstrated a precipitous drop in p38(MAPK) activation that too was reversed in the presence of recombinant GDNF. RNA profiling of NRAGE diminished ex vivo kidney explants resulted in altered expression of GDNF, Ret, BMP7 and BMPRIb mRNAs. Our results suggested that in early kidney development NRAGE might have multiple roles during renal branching morphogenesis through association with both the BMP and GDNF signaling pathways.


Asunto(s)
Riñón/embriología , Morfogénesis , Proteínas de Neoplasias/metabolismo , Animales , Apoptosis/efectos de los fármacos , Proteínas Morfogenéticas Óseas/metabolismo , Proliferación Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Factor Neurotrófico Derivado de la Línea Celular Glial/farmacología , Proteínas de Homeodominio/metabolismo , Inmunoprecipitación , Riñón/citología , Riñón/efectos de los fármacos , Riñón/enzimología , Ratones , Ratones Transgénicos , Modelos Biológicos , Morfogénesis/efectos de los fármacos , Oligonucleótidos Antisentido/farmacología , Técnicas de Cultivo de Órganos , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Uréter/efectos de los fármacos , Uréter/embriología , Uréter/enzimología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
20.
Mol Biol Cell ; 19(11): 4863-74, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18784255

RESUMEN

Angiogenesis is controlled by several regulatory mechanisms, including the Notch and fibroblast growth factor (FGF) signaling pathways. FGF1, a prototype member of FGF family, lacks a signal peptide and is released through an endoplasmic reticulum-Golgi-independent mechanism. A soluble extracellular domain of the Notch ligand Jagged1 (sJ1) inhibits Notch signaling and induces FGF1 release. Thrombin, a key protease of the blood coagulation cascade and a potent inducer of angiogenesis, stimulates rapid FGF1 release through a mechanism dependent on the major thrombin receptor protease-activated receptor (PAR) 1. This study demonstrates that thrombin cleaves Jagged1 in its extracellular domain. The sJ1 form produced as a result of thrombin cleavage inhibits Notch-mediated CBF1/Suppressor of Hairless [(Su(H)]/Lag-1-dependent transcription and induces FGF1 expression and release. The overexpression of Jagged1 in PAR1 null cells results in a rapid thrombin-induced export of FGF1. These data demonstrate the existence of novel cross-talk between thrombin, FGF, and Notch signaling pathways, which play important roles in vascular formation and remodeling.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Fármacos Cardiovasculares/metabolismo , Factor 1 de Crecimiento de Fibroblastos/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/metabolismo , Fragmentos de Péptidos/metabolismo , Trombina/farmacología , Animales , Proteínas de Unión al Calcio/química , Línea Celular , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Factor 1 de Crecimiento de Fibroblastos/genética , Humanos , Péptidos y Proteínas de Señalización Intercelular/química , Proteína Jagged-1 , Proteínas de la Membrana/química , Ratones , Peso Molecular , Células Madre Multipotentes/citología , Células Madre Multipotentes/efectos de los fármacos , Células Madre Multipotentes/metabolismo , Cresta Neural/citología , Estructura Terciaria de Proteína , Transporte de Proteínas/efectos de los fármacos , Receptor PAR-1/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Receptores Notch/metabolismo , Receptores de Trombina , Proteínas Serrate-Jagged , Transducción de Señal/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA