Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cureus ; 16(5): e59756, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38841042

RESUMEN

Renal cell carcinoma (RCC) has a high metastatic potential. While metastasis to common sites like the lungs, liver, bones, and brain is well-documented, metastasis to the colon, particularly the descending colon, remains an uncommon occurrence. When RCC does metastasize to the gastrointestinal tract, it commonly spreads to the small bowel and stomach. There are few cases reported in literature involving RCC metastasis to the colon. The commonly affected areas within the colon include the rectosigmoid colon, splenic flexure, and transverse colon. We describe an 87-year-old male with a history of stage III RCC diagnosed three years ago, followed by left-sided nephroureterectomy, partial adrenalectomy, and perinephric lymph node dissection. He presented to the emergency department (ED) with melena and generalized abdominal pain for one week. Stool occult blood was positive. Computed tomography (CT) of the abdomen was significant for stable postsurgical changes related to prior left nephrectomy and colonic mass at the proximal descending colon. A colonoscopy revealed a necrotic appearing friable mass in the descending colon. The pathology of the mass revealed proliferated atypical cells positive for paired box 8 (PAX8), a cluster of differentiation 10 (CD10), RCC, and pan-cytokeratin and negative for caudal-type homeobox 2 (CDX2), thyroid transcription factor-1 (TTF-1), and a cluster of differentiation 68 (CD68), consistent with metastatic RCC.

2.
Stem Cells Transl Med ; 13(7): 606-624, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38819251

RESUMEN

INTRODUCTION: Lung injuries, such as bronchopulmonary dysplasia (BPD), remain a major complication of preterm birth, with limited therapeutic options. One potential emerging therapy is umbilical cord blood (UCB)-derived therapy. OBJECTIVES: To systematically assess the safety and efficacy of UCB-derived therapy for preterm lung injury in preclinical and clinical studies. METHODS: A systematic search of MEDLINE, Embase, CENTRAL, ClinicalTrials.gov, and WHO International Trials Registry Platform was performed. A meta-analysis was conducted with Review Manager (5.4.1) using a random effects model. Data was expressed as standardized mean difference (SMD) for preclinical data and pooled relative risk (RR) for clinical data, with 95% confidence intervals (CI). Potential effect modifiers were investigated via subgroup analysis. Certainty of evidence was assessed using the GRADE system. RESULTS: Twenty-three preclinical studies and six clinical studies met eligibility criteria. Statistically significant improvements were seen across several preclinical outcomes, including alveolarization (SMD, 1.32, 95%CI [0.99, 1.65]), angiogenesis (SMD, 1.53, 95%CI [0.87, 2.18]), and anti-inflammatory cytokines (SMD, 1.68, 95%CI [1.03, 2.34]). In clinical studies, 103 preterm infants have received UCB-derived therapy for preterm lung injury and no significant difference was observed in the development of BPD (RR, 0.93, 95%CI [0.73, 1.18]). Across both preclinical and clinical studies, administration of UCB-derived therapy appeared safe. Certainty of evidence was assessed as "low." CONCLUSIONS: Administration of UCB-derived therapy was associated with statistically significant improvements across several lung injury markers in preclinical studies. Early clinical studies demonstrated the administration of UCB-derived therapy as safe and feasible but lacked data regarding efficacy.


Asunto(s)
Sangre Fetal , Humanos , Sangre Fetal/citología , Displasia Broncopulmonar/terapia , Recién Nacido , Recien Nacido Prematuro , Lesión Pulmonar/terapia , Animales , Trasplante de Células Madre de Sangre del Cordón Umbilical/métodos
3.
J Biol Chem ; 300(1): 105582, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38141762

RESUMEN

The intracellular parasite, Toxoplasma gondii, has developed sophisticated molecular strategies to subvert host processes and promote growth and survival. During infection, T. gondii replicates in a parasitophorous vacuole (PV) and modulates host functions through a network of secreted proteins. Of these, Mitochondrial Association Factor 1b (MAF1b) recruits host mitochondria to the PV, a process that confers an in vivo growth advantage, though the precise mechanisms remain enigmatic. To address this knowledge gap, we mapped the MAF1b interactome in human fibroblasts using a commercial Yeast-2-hybrid (Y2H) screen, which revealed several previously unidentified binding partners including the GAP domain of Ral GTPase Accelerating Protein α1 (RalGAPα1(GAP)). Recombinantly produced MAF1b and RalGAPα1(GAP) formed as a stable binary complex as shown by size exclusion chromatography with a Kd of 334 nM as measured by isothermal titration calorimetry (ITC). Notably, no binding was detected between RalGAPα1(GAP) and the structurally conserved MAF1b homolog, MAF1a, which does not recruit host mitochondria. Next, we used hydrogen deuterium exchange mass spectrometry (HDX-MS) to map the RalGAPα1(GAP)-MAF1b interface, which led to identification of the "GAP-binding loop" on MAF1b that was confirmed by mutagenesis and ITC to be necessary for complex formation. A high-confidence Alphafold model predicts the GAP-binding loop to lie at the RalGAPα1(GAP)-MAF1b interface further supporting the HDX-MS data. Mechanistic implications of a RalGAPα1(GAP)-MAF1b complex are discussed in the context of T. gondii infection and indicates that MAF1b may have evolved multiple independent functions to increase T. gondii fitness.


Asunto(s)
Proteínas Activadoras de GTPasa , Mitocondrias , Mapas de Interacción de Proteínas , Proteínas Protozoarias , Toxoplasma , Humanos , Sitios de Unión , Calorimetría , Cromatografía en Gel , Fibroblastos/metabolismo , Fibroblastos/parasitología , Proteínas Activadoras de GTPasa/química , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Espectrometría de Masas de Intercambio de Hidrógeno-Deuterio , Mitocondrias/metabolismo , Mitocondrias/parasitología , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Toxoplasma/química , Toxoplasma/genética , Toxoplasma/metabolismo , Técnicas del Sistema de Dos Híbridos
4.
PLoS Biol ; 21(5): e3002110, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37155705

RESUMEN

Toxoplasma gondii is a widespread apicomplexan parasite that can cause severe disease in its human hosts. The ability of T. gondii and other apicomplexan parasites to invade into, egress from, and move between cells of the hosts they infect is critical to parasite virulence and disease progression. An unusual and highly conserved parasite myosin motor (TgMyoA) plays a central role in T. gondii motility. The goal of this work was to determine whether the parasite's motility and lytic cycle can be disrupted through pharmacological inhibition of TgMyoA, as an approach to altering disease progression in vivo. To this end, we first sought to identify inhibitors of TgMyoA by screening a collection of 50,000 structurally diverse small molecules for inhibitors of the recombinant motor's actin-activated ATPase activity. The top hit to emerge from the screen, KNX-002, inhibited TgMyoA with little to no effect on any of the vertebrate myosins tested. KNX-002 was also active against parasites, inhibiting parasite motility and growth in culture in a dose-dependent manner. We used chemical mutagenesis, selection in KNX-002, and targeted sequencing to identify a mutation in TgMyoA (T130A) that renders the recombinant motor less sensitive to compound. Compared to wild-type parasites, parasites expressing the T130A mutation showed reduced sensitivity to KNX-002 in motility and growth assays, confirming TgMyoA as a biologically relevant target of KNX-002. Finally, we present evidence that KNX-002 can slow disease progression in mice infected with wild-type parasites, but not parasites expressing the resistance-conferring TgMyoA T130A mutation. Taken together, these data demonstrate the specificity of KNX-002 for TgMyoA, both in vitro and in vivo, and validate TgMyoA as a druggable target in infections with T. gondii. Since TgMyoA is essential for virulence, conserved in apicomplexan parasites, and distinctly different from the myosins found in humans, pharmacological inhibition of MyoA offers a promising new approach to treating the devastating diseases caused by T. gondii and other apicomplexan parasites.


Asunto(s)
Parásitos , Toxoplasma , Humanos , Animales , Ratones , Toxoplasma/genética , Miosinas , Mutación , Proteínas Protozoarias/genética
5.
Acta Crystallogr D Struct Biol ; 78(Pt 5): 623-632, 2022 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-35503210

RESUMEN

The structure of the antigen-binding fragment (Fab) of mouse monoclonal antibody 7H2.2 in complex with a 15-residue fragment from the metalloproteinase sperm acrosomal SLLP1 binding protein (SAS1B), which is a molecular and cellular candidate for both cancer therapy and female contraception, has been determined at 2.75 Šresolution by single-crystal X-ray diffraction. Although the crystallization conditions contained the final 148 C-terminal residues of SAS1B, the Fab was observed to crystallize in complex with a 15-residue fragment corresponding to one of only two elements of secondary structure that are predicted to be ordered within the C-terminal region of SAS1B. The antigen forms an amphipathic α-helix that binds the 7H2.2 combining site via hydrophilic residues in an epitope that spans the length of the antigen α-helix, with only two CH-π interactions observed along the edge of the interface between the antibody and antigen. Interestingly, the paratope contains two residues mutated away from the germline (YL32F and YH58R), as well as a ProH96-ThrH97-AspH98-AspH99 insertion within heavy chain CDR3. The intact 7H2.2 antibody exhibits high affinity for the SAS1B antigen, with 1:1 binding and nanomolar affinity for both the SAS1B C-terminal construct used for crystallization (3.38 ± 0.59 nM) and a 15-amino-acid synthetic peptide construct corresponding to the helical antigen observed within the crystal structure (1.60 ± 0.31 nM). The SAS1B-antibody structure provides the first structural insight into any portion of the subdomain architecture of the C-terminal region of the novel cancer-oocyte tumor surface neoantigen SAS1B and provides a basis for the targeted use of SAS1B.


Asunto(s)
Anticuerpos Monoclonales , Neoplasias , Animales , Anticuerpos Monoclonales/química , Sitios de Unión de Anticuerpos , Cristalografía por Rayos X , Femenino , Fragmentos Fab de Inmunoglobulinas/química , Ratones , Oocitos/metabolismo , Conformación Proteica
6.
Proc Natl Acad Sci U S A ; 118(12)2021 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-33723040

RESUMEN

Host mitochondrial association (HMA) is a well-known phenomenon during Toxoplasma gondii infection of the host cell. The T. gondii locus mitochondrial association factor 1 (MAF1) is required for HMA and MAF1 encodes distinct paralogs of secreted dense granule effector proteins, some of which mediate the HMA phenotype (MAF1b paralogs drive HMA; MAF1a paralogs do not). To identify host proteins required for MAF1b-mediated HMA, we performed unbiased, label-free quantitative proteomics on host cells infected with type II parasites expressing MAF1b, MAF1a, and an HMA-incompetent MAF1b mutant. Across these samples, we identified ∼1,360 MAF1-interacting proteins, but only 13 that were significantly and uniquely enriched in MAF1b pull-downs. The gene products include multiple mitochondria-associated proteins, including those that traffic to the mitochondrial outer membrane. Based on follow-up endoribonuclease-prepared short interfering RNA (esiRNA) experiments targeting these candidate MAF1b-targeted host factors, we determined that the mitochondrial receptor protein TOM70 and mitochondria-specific chaperone HSPA9 were essential mediators of HMA. Additionally, the enrichment of TOM70 at the parasitophorous vacuole membrane interface suggests parasite-driven sequestration of TOM70 by the parasite. These results show that the interface between the T. gondii vacuole and the host mitochondria is characterized by interactions between a single parasite effector and multiple target host proteins, some of which are critical for the HMA phenotype itself. The elucidation of the functional members of this complex will permit us to explain the link between HMA and changes in the biology of the host cell.


Asunto(s)
Interacciones Huésped-Parásitos , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Toxoplasma/fisiología , Toxoplasmosis/metabolismo , Toxoplasmosis/parasitología , Proteínas Portadoras , Expresión Génica Ectópica , Técnica del Anticuerpo Fluorescente , Interacciones Huésped-Parásitos/genética , Espectrometría de Masas , Mitocondrias/genética , Proteínas Mitocondriales/genética , Unión Proteica , Mapeo de Interacción de Proteínas , Mapas de Interacción de Proteínas , Transporte de Proteínas , Interferencia de ARN , ARN Interferente Pequeño/genética , Vacuolas/metabolismo , Virulencia
7.
EMBO Rep ; 21(2): e48441, 2020 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-31829496

RESUMEN

The lipid kinase PI4KB, which generates phosphatidylinositol 4-phosphate (PI4P), is a key enzyme in regulating membrane transport and is also hijacked by multiple picornaviruses to mediate viral replication. PI4KB can interact with multiple protein binding partners, which are differentially manipulated by picornaviruses to facilitate replication. The protein c10orf76 is a PI4KB-associated protein that increases PI4P levels at the Golgi and is essential for the viral replication of specific enteroviruses. We used hydrogen-deuterium exchange mass spectrometry to characterize the c10orf76-PI4KB complex and reveal that binding is mediated by the kinase linker of PI4KB, with formation of the heterodimeric complex modulated by PKA-dependent phosphorylation. Complex-disrupting mutations demonstrate that PI4KB is required for membrane recruitment of c10orf76 to the Golgi, and that an intact c10orf76-PI4KB complex is required for the replication of c10orf76-dependent enteroviruses. Intriguingly, c10orf76 also contributed to proper Arf1 activation at the Golgi, providing a putative mechanism for the c10orf76-dependent increase in PI4P levels at the Golgi.


Asunto(s)
Enterovirus , Animales , Enterovirus/genética , Enterovirus/metabolismo , Aparato de Golgi/metabolismo , Fosfatos de Fosfatidilinositol , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Unión Proteica , Células Sf9 , Replicación Viral
8.
Structure ; 28(2): 145-156.e5, 2020 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-31831213

RESUMEN

The class I phosphoinositide 3-kinases (PI3Ks) are key signaling enzymes composed of a heterodimer of a p110 catalytic subunit and a p85 regulatory subunit, with PI3K mutations being causative of multiple human diseases including cancer, primary immunodeficiencies, and developmental disorders. Mutations in the p85α regulatory subunit encoded by PIK3R1 can both activate PI3K through oncogenic truncations in the iSH2 domain, or inhibit PI3K through developmental disorder mutations in the cSH2 domain. Using a combined biochemical and hydrogen deuterium exchange mass spectrometry approach we have defined the molecular basis for how these mutations alter the activity of p110α/p110δ catalytic subunits. We find that the oncogenic Q572∗ truncation of PIK3R1 disrupts all p85-inhibitory inputs, with p110α being hyper-activated compared with p110δ. In addition, we find that the R649W mutation in the cSH2 of PIK3R1 decreases sensitivity to activation by receptor tyrosine kinases. This work reveals unique insight into isoform-specific regulation of p110s by p85α.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Fosfatidilinositol 3-Quinasa Clase Ia/genética , Mutación , Dominio Catalítico , Fosfatidilinositol 3-Quinasa Clase I/química , Fosfatidilinositol 3-Quinasa Clase Ia/química , Activación Enzimática , Humanos , Espectrometría de Masas de Intercambio de Hidrógeno-Deuterio , Modelos Moleculares , Conformación Proteica , Dominios Proteicos
9.
Proc Natl Acad Sci U S A ; 115(45): E10548-E10555, 2018 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-30348763

RESUMEN

Parasites of the phylum Apicomplexa are responsible for significant morbidity and mortality on a global scale. Central to the virulence of these pathogens are the phylum-specific, unconventional class XIV myosins that power the essential processes of parasite motility and host cell invasion. Notably, class XIV myosins differ from human myosins in key functional regions, yet they are capable of fast movement along actin filaments with kinetics rivaling previously studied myosins. Toward establishing a detailed molecular mechanism of class XIV motility, we determined the 2.6-Å resolution crystal structure of the Toxoplasma gondii MyoA (TgMyoA) motor domain. Structural analysis reveals intriguing strategies for force transduction and chemomechanical coupling that rely on a divergent SH1/SH2 region, the class-defining "HYAG"-site polymorphism, and the actin-binding surface. In vitro motility assays and hydrogen-deuterium exchange coupled with MS further reveal the mechanistic underpinnings of phosphorylation-dependent modulation of TgMyoA motility whereby localized regions of increased stability and order correlate with enhanced motility. Analysis of solvent-accessible pockets reveals striking differences between apicomplexan class XIV and human myosins. Extending these analyses to high-confidence homology models of Plasmodium and Cryptosporidium MyoA motor domains supports the intriguing potential of designing class-specific, yet broadly active, apicomplexan myosin inhibitors. The successful expression of the functional TgMyoA complex combined with our crystal structure of the motor domain provides a strong foundation in support of detailed structure-function studies and enables the development of small-molecule inhibitors targeting these devastating global pathogens.


Asunto(s)
Miosina Tipo IIA no Muscular/química , Toxoplasma/metabolismo , Secuencia de Aminoácidos , Antiprotozoarios/química , Antiprotozoarios/farmacología , Sitios de Unión , Diseño de Fármacos , Imitación Molecular , Mutación , Miosina Tipo IIA no Muscular/antagonistas & inhibidores , Miosina Tipo IIA no Muscular/genética , Miosina Tipo IIA no Muscular/metabolismo , Unión Proteica , Conformación Proteica , Estabilidad Proteica , Homología de Secuencia de Aminoácido , Toxoplasma/efectos de los fármacos
10.
Mol Microbiol ; 108(5): 519-535, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29505111

RESUMEN

The Toxoplasma gondii locus mitochondrial association factor 1 (MAF1) encodes multiple paralogs, some of which mediate host mitochondrial association (HMA). Previous work showed that HMA was a trait that arose in T. gondii through neofunctionalization of an ancestral MAF1 ortholog. Structural analysis of HMA-competent and incompetent MAF1 paralogs (MAF1b and MAF1a, respectively) revealed that both paralogs harbor an ADP ribose binding macro-domain, with comparatively low (micromolar) affinity for ADP ribose. Replacing the 16 C-terminal residues of MAF1b with those of MAF1a abrogated HMA, and we also show that only three residues in the C-terminal helix are required for MAF1-mediated HMA. Importantly these same three residues are also required for the in vivo growth advantage conferred by MAF1b, providing a definitive link between in vivo proliferation and manipulation of host mitochondria. Co-immunoprecipitation assays reveal that the ability to interact with the mitochondrial MICOS complex is shared by HMA-competent and incompetent MAF1 paralogs and mutants. The weak ADPr coordination and ability to interact with the MICOS complex shared between divergent paralogs may represent modular ancestral functions for this tandemly expanded and diversified T. gondii locus.


Asunto(s)
Mitocondrias/metabolismo , Proteínas Protozoarias/química , Proteínas Protozoarias/metabolismo , Toxoplasma/fisiología , Toxoplasmosis/parasitología , Adenosina Difosfato Ribosa/química , Adenosina Difosfato Ribosa/genética , Adenosina Difosfato Ribosa/metabolismo , Animales , Femenino , Fibroblastos/citología , Fibroblastos/parasitología , Prepucio/citología , Sitios Genéticos , Interacciones Huésped-Parásitos/fisiología , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Proteínas Protozoarias/genética , Toxoplasma/genética
11.
J Biol Chem ; 292(47): 19469-19477, 2017 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-28972141

RESUMEN

Apicomplexan parasites such as Toxoplasma gondii rely on a unique form of locomotion known as gliding motility. Generating the mechanical forces to support motility are divergent class XIV myosins (MyoA) coordinated by accessory proteins known as light chains. Although the importance of the MyoA-light chain complex is well-established, the detailed mechanisms governing its assembly and regulation are relatively unknown. To establish a molecular blueprint of this dynamic complex, we first mapped the adjacent binding sites of light chains MLC1 and ELC1 on the MyoA neck (residues 775-818) using a combination of hydrogen-deuterium exchange mass spectrometry and isothermal titration calorimetry. We then determined the 1.85 Å resolution crystal structure of MLC1 in complex with its cognate MyoA peptide. Structural analysis revealed a bilobed architecture with MLC1 clamping tightly around the helical MyoA peptide, consistent with the stable 10 nm Kd measured by isothermal titration calorimetry. We next showed that coordination of calcium by an EF-hand in ELC1 and prebinding of MLC1 to the MyoA neck enhanced the affinity of ELC1 for the MyoA neck 7- and 8-fold, respectively. When combined, these factors enhanced ELC1 binding 49-fold (to a Kd of 12 nm). Using the full-length MyoA motor (residues 1-831), we then showed that, in addition to coordinating the neck region, ELC1 appears to engage the MyoA converter subdomain, which couples the motor domain to the neck. These data support an assembly model where staged binding events cooperate to yield high-affinity complexes that are able to maximize force transduction.


Asunto(s)
Miosina Tipo IIA no Muscular/química , Proteínas Protozoarias/química , Toxoplasma/metabolismo , Animales , Calcio/metabolismo , Movimiento Celular , Cristalografía por Rayos X , Miosina Tipo IIA no Muscular/metabolismo , Unión Proteica , Conformación Proteica , Proteínas Protozoarias/metabolismo , Toxoplasma/crecimiento & desarrollo
12.
Protein Sci ; 26(9): 1878-1885, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28681555

RESUMEN

Plasmodium falciparum, the causative agent of malaria, employs a diverse array of surface displayed proteins to promote dissemination and establish infection in the human host. Of these, Pf3D7_0606800 is highly immunogenic and has been designated a potential top 10 candidate for inclusion in a multicomponent malarial vaccine. The role of Pf3D7_0606800 in parasite biology, however, is unknown and its characterization has been complicated by a lack of sequence identity with proteins of known structure or function. Towards elucidating Pf3D7_0606800 function, we determined its structure to a resolution of 2.35 Å using selenium single wavelength anomalous dispersion. A bi-lobed architecture displays the core structural hallmarks of Venus Flytrap (VFT) proteins prompting us to re-annotate Pf3D7_0606800 as PfVFT1. Structural analysis further revealed an extended inter-lobe groove that, when interrogated by molecular docking, appears well suited to bind peptide-based ligands. Collectively, our structural characterization of the highly antigenic P. falciparum surface protein PfVFT1 provides intriguing functional insight and establishes a structural template that could prove valuable for malaria vaccine engineering studies.


Asunto(s)
Plasmodium falciparum/química , Proteínas Protozoarias/química , Cristalografía por Rayos X , Droseraceae/química , Simulación del Acoplamiento Molecular , Proteínas de Plantas/química , Plasmodium falciparum/genética , Conformación Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...