Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 14(1): 3765, 2024 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-38355600

RESUMEN

Homozygous Apolipoprotein L1 (APOL1) variants G1 and G2 cause APOL1-mediated kidney disease, purportedly acting as surface cation channels in podocytes. APOL1-G0 exhibits various single nucleotide polymorphisms, most commonly haplotype E150K, M228I and R255K ("KIK"; the Reference Sequence is "EMR"), whereas variants G1 and G2 are mostly found in a single "African" haplotype background ("EIK"). Several labs reported cytotoxicity with risk variants G1 and G2 in KIK or EIK background haplotypes, but used HEK-293 cells and did not verify equal surface expression. To see if haplotype matters in a more relevant cell type, we induced APOL1-G0, G1 and G2 EIK, KIK and EMR at comparable surface levels in immortalized podocytes. G1 and G2 risk variants (but not G0) caused dose-dependent podocyte death within 48h only in their native African EIK haplotype and correlated with K+ conductance (thallium FLIPR). We ruled out differences in localization and trafficking, except for possibly greater surface clustering of cytotoxic haplotypes. APOL1 surface expression was required, since Brefeldin A rescued cytotoxicity; and cytoplasmic isoforms vB3 and vC were not cytotoxic. Thus, APOL1-EIK risk variants kill podocytes in a dose and haplotype-dependent manner (as in HEK-293 cells), whereas unlike in HEK-293 cells the KIK risk variants did not.


Asunto(s)
Podocitos , Humanos , Podocitos/metabolismo , Haplotipos , Apolipoproteína L1/genética , Apolipoproteína L1/metabolismo , Células HEK293 , Variación Genética
2.
PLoS Genet ; 19(8): e1010609, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37585454

RESUMEN

Diabetic retinopathy (DR) is a common complication of diabetes. Approximately 20% of DR patients have diabetic macular edema (DME) characterized by fluid leakage into the retina. There is a genetic component to DR and DME risk, but few replicable loci. Because not all DR cases have DME, we focused on DME to increase power, and conducted a multi-ancestry GWAS to assess DME risk in a total of 1,502 DME patients and 5,603 non-DME controls in discovery and replication datasets. Two loci reached GWAS significance (p<5x10-8). The strongest association was rs2239785, (K150E) in APOL1. The second finding was rs10402468, which co-localized to PLVAP and ANKLE1 in vascular / endothelium tissues. We conducted multiple sensitivity analyses to establish that the associations were specific to DME status and did not reflect diabetes status or other diabetic complications. Here we report two novel loci for risk of DME which replicated in multiple clinical trial and biobank derived datasets. One of these loci, containing the gene APOL1, is a risk factor in African American DME and DKD patients, indicating that this locus plays a broader role in diabetic complications for multiple ancestries. Trial Registration: NCT00473330, NCT00473382, NCT03622580, NCT03622593, NCT04108156.


Asunto(s)
Diabetes Mellitus , Retinopatía Diabética , Edema Macular , Humanos , Edema Macular/genética , Edema Macular/complicaciones , Retinopatía Diabética/genética , Retinopatía Diabética/complicaciones , Estudio de Asociación del Genoma Completo , Apolipoproteína L1/genética , Factores de Riesgo
3.
Nat Chem Biol ; 19(1): 55-63, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36577875

RESUMEN

Engineered destruction of target proteins by recruitment to the cell's degradation machinery has emerged as a promising strategy in drug discovery. The majority of molecules that facilitate targeted degradation do so via a select number of ubiquitin ligases, restricting this therapeutic approach to tissue types that express the requisite ligase. Here, we describe a new strategy of targeted protein degradation through direct substrate recruitment to the 26S proteasome. The proteolytic complex is essential and abundantly expressed in all cells; however, proteasomal ligands remain scarce. We identify potent peptidic macrocycles that bind directly to the 26S proteasome subunit PSMD2, with a 2.5-Å-resolution cryo-electron microscopy complex structure revealing a binding site near the 26S pore. Conjugation of this macrocycle to a potent BRD4 ligand enabled generation of chimeric molecules that effectively degrade BRD4 in cells, thus demonstrating that degradation via direct proteasomal recruitment is a viable strategy for targeted protein degradation.


Asunto(s)
Proteínas Nucleares , Factores de Transcripción , Proteínas Nucleares/metabolismo , Microscopía por Crioelectrón , Factores de Transcripción/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
4.
Commun Biol ; 4(1): 916, 2021 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-34316015

RESUMEN

Apolipoprotein L1 (ApoL1) is a circulating innate immunity protein protecting against trypanosome infection. However, two ApoL1 coding variants are associated with a highly increased risk of chronic kidney disease. Here we present X-ray and NMR structures of the N-terminal domain (NTD) of ApoL1 and of its closest relative ApoL2. In both proteins, four of the five NTD helices form a four-helix core structure which is different from the classical four-helix bundle and from the pore-forming domain of colicin A. The reactivity with a conformation-specific antibody and structural models predict that this four-helix motif is also present in the NTDs of ApoL3 and ApoL4, suggesting related functions within the small ApoL family. The long helix 5 of ApoL1 is conformationally flexible and contains the BH3-like region. This BH3-like α-helix resembles true BH3 domains only in sequence and structure but not in function, since it does not bind to the pro-survival members of the Bcl-2 family, suggesting a Bcl-2-independent role in cytotoxicity. These findings should expedite a more comprehensive structural and functional understanding of the ApoL immune protein family.


Asunto(s)
Apolipoproteína L1/química , Apolipoproteínas L/química , Dominios Proteicos , Apolipoproteína L1/genética , Apolipoproteína L1/metabolismo , Apolipoproteínas L/genética , Apolipoproteínas L/metabolismo , Humanos
5.
J Am Soc Nephrol ; 31(9): 2065-2082, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32764138

RESUMEN

BACKGROUND: Circulating APOL1 lyses trypanosomes, protecting against human sleeping sickness. Two common African gene variants of APOL1, G1 and G2, protect against infection by species of trypanosomes that resist wild-type APOL1. At the same time, the protection predisposes humans to CKD, an elegant example of balanced polymorphism. However, the exact mechanism of APOL1-mediated podocyte damage is not clear, including APOL1's subcellular localization, topology, and whether the damage is related to trypanolysis. METHODS: APOL1 topology in serum (HDL particles) and in kidney podocytes was mapped with flow cytometry, immunoprecipitation, and trypanolysis assays that tracked 170 APOL1 domain-specific monoclonal antibodies. APOL1 knockout podocytes confirmed antibody specificity. RESULTS: APOL1 localizes to the surface of podocytes, with most of the pore-forming domain (PFD) and C terminus of the Serum Resistance Associated-interacting domain (SRA-ID), but not the membrane-addressing domain (MAD), being exposed. In contrast, differential trypanolytic blocking activity reveals that the MAD is exposed in serum APOL1, with less of the PFD accessible. Low pH did not detectably alter the gross topology of APOL1, as determined by antibody accessibility, in serum or on podocytes. CONCLUSIONS: Our antibodies highlighted different conformations of native APOL1 topology in serum (HDL particles) and at the podocyte surface. Our findings support the surface ion channel model for APOL1 risk variant-mediated podocyte injury, as well as providing domain accessibility information for designing APOL1-targeted therapeutics.


Asunto(s)
Apolipoproteína L1/análisis , Membrana Celular/química , Podocitos/química , Animales , Anticuerpos/inmunología , Especificidad de Anticuerpos , Apolipoproteína L1/sangre , Apolipoproteína L1/química , Apolipoproteína L1/inmunología , Células CHO , Cricetulus , Humanos , Concentración de Iones de Hidrógeno , Podocitos/ultraestructura , Dominios Proteicos
6.
J Am Soc Nephrol ; 31(9): 2044-2064, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32764142

RESUMEN

BACKGROUND: APOL1 is found in human kidney podocytes and endothelia. Variants G1 and G2 of the APOL1 gene account for the high frequency of nondiabetic CKD among African Americans. Proposed mechanisms of kidney podocyte cytotoxicity resulting from APOL1 variant overexpression implicate different subcellular compartments. It is unclear where endogenous podocyte APOL1 resides, because previous immunolocalization studies utilized overexpressed protein or commercially available antibodies that crossreact with APOL2. This study describes and distinguishes the locations of both APOLs. METHODS: Immunohistochemistry, confocal and immunoelectron microscopy, and podocyte fractionation localized endogenous and transfected APOL1 using a large panel of novel APOL1-specific mouse and rabbit monoclonal antibodies. RESULTS: Both endogenous podocyte and transfected APOL1 isoforms vA and vB1 (and a little of isoform vC) localize to the luminal face of the endoplasmic reticulum (ER) and to the cell surface, but not to mitochondria, endosomes, or lipid droplets. In contrast, APOL2, isoform vB3, and most vC of APOL1 localize to the cytoplasmic face of the ER and are consequently absent from the cell surface. APOL1 knockout podocytes do not stain for APOL1, attesting to the APOL1-specificity of the antibodies. Stable re-transfection of knockout podocytes with inducible APOL1-G0, -G1, and -G2 showed no differences in localization among variants. CONCLUSIONS: APOL1 is found in the ER and plasma membrane, consistent with either the ER stress or surface cation channel models of APOL1-mediated cytotoxicity. The surface localization of APOL1 variants potentially opens new therapeutic targeting avenues.


Asunto(s)
Apolipoproteína L1/análisis , Membrana Celular/química , Retículo Endoplásmico/química , Podocitos/química , Animales , Anticuerpos/inmunología , Apolipoproteína L1/inmunología , Apolipoproteínas L/análisis , Células COS , Células Cultivadas , Chlorocebus aethiops , Reacciones Cruzadas , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Podocitos/ultraestructura
7.
J Immunol Methods ; 480: 112767, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32119889

RESUMEN

IgG antibodies have been used to treat many diseases including cancer. IgG antibody-drug conjugates (ADCs) deliver cytotoxic drugs to target cells for cell elimination, but they have dose limiting toxicity due to target-independent uptake, including pinocytotic uptake. Neonatal Fc receptor (FcRn) recycles pinocytosed IgG in a pH-dependent manner and is the receptor responsible for the long half-life of IgG. Use of IgG variants with stronger FcRn binding at pH 6.0 for ADCs might improve recycling efficiency and reduce toxicity. However, these variants have residual FcRn binding at pH 7.4, which could lead to FcRn-mediated uptake and higher toxicity. Thus, the uptake of such variants at pH 7.4 needs to be evaluated. Here we report a reproducible and quantitative assay using an inducible HM7 colorectal cancer cell line to measure IgG uptake at endogenous and overexpressed FcRn levels. Our assay had comparable reproducibility at pH 6.0, 6.8 and 7.4. The wild type (WT) IgG had similar uptake at endogenous and overexpressed FcRn levels, as expected for pinocytotic uptake. We found similar uptake of a WT IgG and a stronger FcRn binding T307Q/N434A variant (QA variant) at endogenous FcRn levels at pH 7.4, although the QA variant had higher uptake at overexpressed FcRn levels. The QA variant also had higher uptake than the WT IgG at overexpressed FcRn levels at pH 6.8. Our assay can be used to characterize the stronger FcRn binding variants to aid in selection of suitable variants with low uptake at pH 7.4 for use as ADCs.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Inmunoglobulina G/metabolismo , Pinocitosis , Receptores Fc/metabolismo , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Concentración de Iones de Hidrógeno , Cinética , Unión Proteica , Receptores Fc/genética , Regulación hacia Arriba
8.
Bioconjug Chem ; 30(12): 3046-3056, 2019 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-31726009

RESUMEN

Disulfide-linked bioconjugates allow the delivery of pharmacologically active or other cargo to specific tissues in a redox-sensitive fashion. However, an understanding of the kinetics, subcellular distribution, and mechanism of disulfide cleavage in such bioconjugates is generally lacking. Here, we report a modular disulfide-linked TAMRA-BODIPY based FRET probe that can be readily synthesized, modified, and conjugated to a cysteine-containing biomolecule to enable real-time monitoring of disulfide cleavage during receptor-mediated endocytosis in cells. We demonstrate the utility of this probe to study disulfide reduction during HER2 receptor-mediated uptake of a Cys-engineered anti-HER2 THIOMAB antibody. We found that introduction of positive, but not negative, charges in the probe improved retention of the BODIPY catabolite. This permitted the observation of significant disulfide cleavage in endosomes or lysosomes on par with proteolytic cleavage of a similarly charged valine-citrulline peptide-based probe. In general, the FRET probe we describe should enable real-time cellular monitoring of disulfide cleavage in other targeted delivery systems for mechanistic or diagnostic applications. Furthermore, modifications to the released BODIPY moiety permit evaluation of physicochemical properties that govern lysosomal egress or retention, which may have implications for the development of next-generation antibody-drug conjugates.


Asunto(s)
Cisteína/química , Disulfuros/química , Transferencia Resonante de Energía de Fluorescencia , Colorantes Fluorescentes/química , Animales , Compuestos de Boro , Monitoreo de Drogas/métodos , Endocitosis , Endosomas/metabolismo , Transferencia Resonante de Energía de Fluorescencia/tendencias , Humanos , Inmunoconjugados , Lisosomas/metabolismo , Receptor ErbB-2/inmunología , Rodaminas
9.
MAbs ; 11(6): 996-1011, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31156033

RESUMEN

Agonism of members of the tumor necrosis factor receptor superfamily (TNFRSF) with monoclonal antibodies is of high therapeutic interest due to their role in immune regulation and cell proliferation. A major hurdle for pharmacologic activation of this receptor class is the requirement for high-order clustering, a mechanism that imposes a reliance in vivo on Fc receptor-mediated crosslinking. This extrinsic dependence represents a potential limitation of virtually the entire pipeline of agonist TNFRSF antibody drugs, of which none have thus far been approved or reached late-stage clinical trials. We show that tetravalent biepitopic targeting enables robust intrinsic antibody agonism for two members of this family, OX40 and DR5, that is superior to extrinsically crosslinked native parental antibodies. Tetravalent biepitopic anti-OX40 engagement co-stimulated OX40low cells, obviated the requirement for CD28 co-signal for T cell activation, and enabled superior pharmacodynamic activity relative to native IgG in a murine vaccination model. This work establishes a proof of concept for an engineering approach that addresses a major gap for the therapeutic activation of this important receptor class.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Recubrimiento Inmunológico , Ligando OX40/agonistas , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/agonistas , Transducción de Señal/inmunología , Linfocitos T/inmunología , Animales , Antígenos CD28/inmunología , Células CHO , Cricetulus , Humanos , Células Jurkat , Ratones , Ratones SCID , Ratones Transgénicos , Ligando OX40/inmunología , Receptores Fc/inmunología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/inmunología , Linfocitos T/citología
10.
MAbs ; 11(5): 942-955, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30982394

RESUMEN

A cell-based assay employing Madin-Darby canine kidney cells stably expressing human neonatal Fc receptor (FcRn) heavy chain and ß2-microglobulin genes was developed to measure transcytosis of monoclonal antibodies (mAbs) under conditions relevant to the FcRn-mediated immunoglobulin G (IgG) salvage pathway. The FcRn-dependent transcytosis assay is modeled to reflect combined effects of nonspecific interactions between mAbs and cells, cellular uptake via pinocytosis, pH-dependent interactions with FcRn, and dynamics of intracellular trafficking and sorting mechanisms. Evaluation of 53 mAbs, including 30 marketed mAb drugs, revealed a notable correlation between the transcytosis readouts and clearance in humans. FcRn was required to promote efficient transcytosis of mAbs and contributed directly to the observed correlation. Furthermore, the transcytosis assay correctly predicted rank order of clearance of glycosylation and Fv charge variants of Fc-containing proteins. These results strongly support the utility of this assay as a cost-effective and animal-sparing screening tool for evaluation of mAb-based drug candidates during lead selection, optimization, and process development for desired pharmacokinetic properties.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacocinética , Antígenos de Histocompatibilidad Clase I/inmunología , Receptores Fc/inmunología , Transcitosis/inmunología , Animales , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Monoclonales Humanizados/metabolismo , Bioensayo/métodos , Perros , Glicosilación , Humanos , Inmunoglobulina G/metabolismo , Células de Riñón Canino Madin Darby , Ratones
11.
Mol Cancer Ther ; 16(1): 134-142, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27760836

RESUMEN

Antibody-drug conjugates (ADC) use monoclonal antibodies (mAb) as vehicles to deliver potent cytotoxic drugs selectively to tumor cells expressing the target. Molecular imaging with zirconium-89 (89Zr)-labeled mAbs recapitulates similar targeting biology and might help predict the efficacy of these ADCs. An anti-mesothelin antibody (AMA, MMOT0530A) was used to make comparisons between its efficacy as an ADC and its tumor uptake as measured by 89Zr immunoPET imaging. Mesothelin-targeted tumor growth inhibition by monomethyl auristatin E (MMAE), ADC AMA-MMAE (DMOT4039A), was measured in mice bearing xenografts of ovarian cancer OVCAR-3×2.1, pancreatic cancers Capan-2, HPAC, AsPC-1, and HPAF-II, or mesothelioma MSTO-211H. Ex vivo analysis of mesothelin expression was performed using immunohistochemistry. AMA-MMAE showed the greatest growth inhibition in OVCAR-3×2.1, Capan-2, and HPAC tumors, which showed target-specific tumor uptake of 89Zr-AMA. The less responsive xenografts (AsPC-1, HPAF-II, and MSTO-211H) did not show 89Zr-AMA uptake despite confirmed mesothelin expression. ImmunoPET can demonstrate the necessary delivery, binding, and internalization of an ADC antibody in vivo and this correlates with the efficacy of mesothelin-targeted ADC in tumors vulnerable to the cytotoxic drug delivered. Mol Cancer Ther; 16(1); 134-42. ©2016 AACR.


Asunto(s)
Antineoplásicos/farmacología , Proteínas Ligadas a GPI/antagonistas & inhibidores , Inmunoconjugados/farmacología , Tomografía de Emisión de Positrones , Radiofármacos , Circonio , Animales , Antineoplásicos/farmacocinética , Biomarcadores de Tumor , Línea Celular Tumoral , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Femenino , Citometría de Flujo , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Expresión Génica , Humanos , Inmunoconjugados/farmacocinética , Mesotelina , Ratones , Terapia Molecular Dirigida , Neoplasias/diagnóstico , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Carga Tumoral/efectos de los fármacos , Carga Tumoral/efectos de la radiación , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Mol Cancer Ther ; 15(3): 439-47, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26823490

RESUMEN

DMOT4039A, a humanized anti-mesothelin mAb conjugated to the antimitotic agent monomethyl auristatin E (MMAE), was given to patients with pancreatic and ovarian cancer every 3 weeks (0.2-2.8 mg/kg; q3w) or weekly (0.8-1.2 mg/kg). A 3+3 design was used for dose escalation followed by expansion at the recommended phase II dose (RP2D) to evaluate safety and pharmacokinetics. Antitumor response was evaluated per RECIST 1.1 and serum CA19-9 or CA125 declines. Tumor mesothelin expression was determined by IHC. Seventy-one patients (40 pancreatic cancer; 31 ovarian cancer) were treated with DMOT4039A. For the q3w schedule (n = 54), the MTD and RP2D was 2.4 mg/kg, with dose-limiting toxicities of grade 3 hyperglycemia and grade 3 hypophosphatemia at 2.8 mg/kg. For the weekly schedule (n = 17), the maximum assessed dose was 1.2 mg/kg, with further dose escalations deferred because of toxicities limiting scheduled retreatment in later cycles, and therefore the RP2D level for the weekly regimen was determined to be 1 mg/kg. Across both schedules, the most common toxicities were gastrointestinal and constitutional. Treatment-related serious adverse events occurred in 6 patients; 4 patients continued treatment following dose reductions. Drug exposure as measured by antibody-conjugated MMAE and total antibody was generally dose proportional over all dose levels on both schedules. A total of 6 patients had confirmed partial responses (4 ovarian; 2 pancreatic) with DMOT4039A at 2.4 to 2.8 mg/kg i.v. q3w. DMOT4039A administered at doses up to 2.4 mg/kg q3w and 1.0 mg/kg weekly has a tolerable safety profile and antitumor activity in both pancreatic and ovarian cancer.


Asunto(s)
Antineoplásicos/uso terapéutico , Resistencia a Antineoplásicos , Proteínas Ligadas a GPI/antagonistas & inhibidores , Inmunoconjugados/uso terapéutico , Terapia Molecular Dirigida , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/metabolismo , Adulto , Anciano , Antineoplásicos/farmacología , Biomarcadores , Progresión de la Enfermedad , Esquema de Medicación , Femenino , Humanos , Inmunoconjugados/farmacología , Inmunohistoquímica , Mesotelina , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias Ováricas/mortalidad , Neoplasias Ováricas/patología , Platino (Metal)/uso terapéutico , Retratamiento , Tomografía Computarizada por Rayos X , Resultado del Tratamiento
13.
J Biol Chem ; 290(49): 29663-75, 2015 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-26451044

RESUMEN

The Hedgehog (Hh) signaling pathway plays an essential role in vertebrate embryonic tissue patterning of many developing organs. Signaling occurs predominantly in primary cilia and is initiated by the entry of the G protein-coupled receptor (GPCR)-like protein Smoothened into cilia and culminates in gene transcription via the Gli family of transcription factors upon their nuclear entry. Here we identify an orphan GPCR, Gpr175 (also known as Tpra1 or Tpra40: transmembrane protein, adipocyte associated 1 or of 40 kDa), which also localizes to primary cilia upon Hh stimulation and positively regulates Hh signaling. Interaction experiments place Gpr175 at the level of PKA and upstream of the Gαi component of heterotrimeric G proteins, which itself localizes to cilia and can modulate Hh signaling. Gpr175 or Gαi1 depletion leads to increases in cellular cAMP levels and in Gli3 processing into its repressor form. Thus we propose that Gpr175 coupled to Gαi1 normally functions to inhibit the production of cAMP by adenylyl cyclase upon Hh stimulation, thus maximizing signaling by turning off PKA activity and hence Gli3 repressor formation. Taken together our data suggest that Gpr175 is a novel positive regulator of the Hh signaling pathway.


Asunto(s)
AMP Cíclico/metabolismo , Proteínas Hedgehog/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Células 3T3 , Secuencia de Aminoácidos , Animales , Células COS , Línea Celular , Chlorocebus aethiops , Cilios/metabolismo , ADN Complementario/metabolismo , Humanos , Ratones , Ratones Endogámicos C3H , Microscopía Fluorescente , Datos de Secuencia Molecular , ARN Interferente Pequeño/metabolismo , Homología de Secuencia de Aminoácido , Transducción de Señal , Receptor Smoothened , Pez Cebra , Proteína Gli3 con Dedos de Zinc
14.
Nucleic Acids Res ; 43(2): 1189-203, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25550431

RESUMEN

Delivery of siRNA is a key hurdle to realizing the therapeutic promise of RNAi. By targeting internalizing cell surface antigens, antibody-siRNA complexes provide a possible solution. However, initial reports of antibody-siRNA complexes relied on non-specific charged interactions and have not been broadly applicable. To assess and improve this delivery method, we built on an industrial platform of therapeutic antibodies called THIOMABs, engineered to enable precise covalent coupling of siRNAs. We report that such coupling generates monomeric antibody-siRNA conjugates (ARCs) that retain antibody and siRNA activities. To broadly assess this technology, we generated a battery of THIOMABs against seven targets that use multiple internalization routes, enabling systematic manipulation of multiple parameters that impact delivery. We identify ARCs that induce targeted silencing in vitro and extend tests to target prostate carcinoma cells following systemic administration in mouse models. However, optimal silencing was restricted to specific conditions and only observed using a subset of ARCs. Trafficking studies point to ARC entrapment in endocytic compartments as a limiting factor, independent of the route of antigen internalization. Our broad characterization of multiple parameters using therapeutic-grade conjugate technology provides a thorough assessment of this delivery technology, highlighting both examples of success as well as remaining challenges.


Asunto(s)
Anticuerpos , ARN Interferente Pequeño/administración & dosificación , Animales , Anticuerpos/genética , Anticuerpos/inmunología , Anticuerpos/metabolismo , Línea Celular , Endosomas/metabolismo , Ratones , Neoplasias/genética , Ingeniería de Proteínas , Interferencia de ARN , ARN Interferente Pequeño/química , ARN Interferente Pequeño/metabolismo
15.
Dev Cell ; 31(3): 345-357, 2014 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-25453830

RESUMEN

The genetic networks that govern vertebrate development are well studied, but how the interactions of trans-acting factors with cis-regulatory modules (CRMs) are integrated into spatiotemporal regulation of gene expression is not clear. The transcriptional regulator HAND2 is required during limb, heart, and branchial arch development. Here, we identify the genomic regions enriched in HAND2 chromatin complexes from mouse embryos and limb buds. Then we analyze the HAND2 target CRMs in the genomic landscapes encoding transcriptional regulators required in early limb buds. HAND2 controls the expression of genes functioning in the proximal limb bud and orchestrates the establishment of anterior and posterior polarity of the nascent limb bud mesenchyme by impacting Gli3 and Tbx3 expression. TBX3 is required downstream of HAND2 to refine the posterior Gli3 expression boundary. Our analysis uncovers the transcriptional circuits that function in establishing distinct mesenchymal compartments downstream of HAND2 and upstream of SHH signaling.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Extremidades/embriología , Regulación del Desarrollo de la Expresión Génica/fisiología , Esbozos de los Miembros/metabolismo , Mesodermo/metabolismo , Animales , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Transactivadores/metabolismo
16.
Mol Cancer Ther ; 13(11): 2630-40, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25249555

RESUMEN

Mesothelin (MSLN) is an attractive target for antibody-drug conjugate therapy because it is highly expressed in various epithelial cancers, with normal expression limited to nondividing mesothelia. We generated novel antimesothelin antibodies and conjugated an internalizing one (7D9) to the microtubule-disrupting drugs monomethyl auristatin E (MMAE) and MMAF, finding the most effective to be MMAE with a lysosomal protease-cleavable valine-citrulline linker. The humanized (h7D9.v3) version, αMSLN-MMAE, specifically targeted mesothelin-expressing cells and inhibited their proliferation with an IC50 of 0.3 nmol/L. Because the antitumor activity of an antimesothelin immunotoxin (SS1P) in transfected mesothelin models did not translate to the clinic, we carefully selected in vivo efficacy models endogenously expressing clinically relevant levels of mesothelin, after scoring mesothelin levels in ovarian, pancreatic, and mesothelioma tumors by immunohistochemistry. We found that endogenous mesothelin in cancer cells is upregulated in vivo and identified two suitable xenograft models for each of these three indications. A single dose of αMSLN-MMAE profoundly inhibited or regressed tumor growth in a dose-dependent manner in all six models, including two patient-derived tumor xenografts. The robust and durable efficacy of αMSLN-MMAE in preclinical models of ovarian, mesothelioma, and pancreatic cancers justifies the ongoing phase I clinical trial.


Asunto(s)
Proteínas Ligadas a GPI/inmunología , Inmunotoxinas/farmacología , Mesotelioma/tratamiento farmacológico , Oligopéptidos/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Pancreáticas/tratamiento farmacológico , Animales , Femenino , Proteínas Ligadas a GPI/biosíntesis , Humanos , Inmunohistoquímica , Inmunotoxinas/química , Inmunotoxinas/inmunología , Mesotelina , Ratones , Oligopéptidos/química , Distribución Aleatoria , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Traffic ; 15(3): 255-72, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24329939

RESUMEN

CD22 is a cell surface glycoprotein restricted to normal and malignant B-cells and is the target of several anti-CD22 antibody-based cancer therapies. For therapeutic antibody-payload conjugates, it is important to understand the subcellular trafficking of anti-CD22 antibodies to optimize antibody and/or linker-drug properties to maximize antitumor efficacy. It is agreed that anti-CD22 antibodies rapidly internalize, but controversial whether they recycle or are degraded in lysosomes, and it is unclear if trafficking is antibody or cell-type dependent. No studies examined anti-CD22 trafficking to either pathway in B-cells over time by dual immunofluorescence microscopy, likely partly because multiple samples of suspension cells are tedious to stain. We overcame this by using DropArray™, a novel wall-less 96-well plate technology allowing rapid simultaneous staining of suspension or adherent cells in small (10-20 µL) volumes. We examined the time-course of trafficking of five different anti-CD22 antibodies in eight B-cell lines representing four B-cell cancer types and show that in all cases antibodies internalize within 5 min and recycle, with only small amounts eventually trafficking to lysosomes. CD22 also localizes to recycling endosomes at steady state in the absence of antibody. Our data may help explain the differential efficacies of anti-CD22 antibodies conjugated to different therapeutic payloads.


Asunto(s)
Endocitosis , Microscopía Fluorescente/métodos , Lectina 2 Similar a Ig de Unión al Ácido Siálico/metabolismo , Análisis de Matrices Tisulares/métodos , Linfocitos B/metabolismo , Línea Celular Tumoral , Humanos
18.
J Cell Biol ; 200(6): 709-20, 2013 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-23479743

RESUMEN

Cilia project from the surface of most vertebrate cells and are important for several physiological and developmental processes. Ciliary defects are linked to a variety of human diseases, named ciliopathies, underscoring the importance of understanding signaling pathways involved in cilia formation and maintenance. In this paper, we identified Rer1p as the first endoplasmic reticulum/cis-Golgi-localized membrane protein involved in ciliogenesis. Rer1p, a protein quality control receptor, was highly expressed in zebrafish ciliated organs and regulated ciliary structure and function. Both in zebrafish and mammalian cells, loss of Rer1p resulted in the shortening of cilium and impairment of its motile or sensory function, which was reflected by hearing, vision, and left-right asymmetry defects as well as decreased Hedgehog signaling. We further demonstrate that Rer1p depletion reduced ciliary length and function by increasing γ-secretase complex assembly and activity and, consequently, enhancing Notch signaling as well as reducing Foxj1a expression.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Factores de Transcripción Forkhead/biosíntesis , Regulación de la Expresión Génica/fisiología , Glicoproteínas de Membrana/metabolismo , Transducción de Señal/fisiología , Proteínas Adaptadoras del Transporte Vesicular , Secretasas de la Proteína Precursora del Amiloide/genética , Animales , Línea Celular , Cilios/genética , Cilios/metabolismo , Factores de Transcripción Forkhead/genética , Humanos , Glicoproteínas de Membrana/genética , Receptores Notch/genética , Receptores Notch/metabolismo , Porcinos , Pez Cebra , Proteínas de Pez Cebra
19.
Cell ; 152(1-2): 210-23, 2013 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-23332756

RESUMEN

The primary cilium is required for Sonic hedgehog (Shh) signaling in vertebrates. In contrast to mutants affecting ciliary assembly, mutations in the intraflagellar transport complex A (IFT-A) paradoxically cause increased Shh signaling. We previously showed that the IFT-A complex, in addition to its canonical role in retrograde IFT, binds to the tubby-like protein, Tulp3, and recruits it to cilia. Here, we describe a conserved vertebrate G-protein-coupled receptor, Gpr161, which localizes to primary cilia in a Tulp3/IFT-A-dependent manner. Complete loss of Gpr161 in mouse causes midgestation lethality and increased Shh signaling in the neural tube, phenocopying Tulp3/IFT-A mutants. Constitutive Gpr161 activity increases cAMP levels and represses Shh signaling by determining the processing of Gli3 to its repressor form. Conversely, Shh signaling directs Gpr161 to be internalized from cilia, preventing its activity. Thus, Gpr161 defines a morphogenetic pathway coupling protein kinase A activation to Shh signaling during neural tube development.


Asunto(s)
Cilios/metabolismo , Embrión de Mamíferos/metabolismo , Tubo Neural/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Secuencia de Aminoácidos , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular , Péptidos y Proteínas de Señalización Intracelular , Ratones , Datos de Secuencia Molecular , Filogenia , Proteínas/metabolismo , Receptores Acoplados a Proteínas G/química , Alineación de Secuencia
20.
Mol Cancer Ther ; 11(10): 2222-32, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22807577

RESUMEN

Fc receptor-like 5 (FcRL5/FcRH5/IRTA2/CD307) is a surface protein expressed selectively on B cells and plasma cells. We found that FcRL5 was expressed at elevated levels on the surface of plasma cells from the bone marrow of patients diagnosed with multiple myeloma. This prevalence in multiple myeloma and narrow pattern of normal expression indicate that FcRL5 could be a target for antibody-based therapies for multiple myeloma, particularly antibody-drug conjugates (ADC), potent cytotoxic drugs linked to antibodies via specialized chemical linkers, where limited expression on normal tissues is a key component to their safety. We found that FcRL5 is internalized upon antibody binding, indicating that ADCs to FcRL5 could be effective. Indeed, we found that FcRL5 ADCs were efficacious in vitro and in vivo but the unconjugated antibody was not. The two most effective consisted of our anti-FcRL5 antibody conjugated through cysteines to monomethylauristatin E (MMAE) by a maleimidocaproyl-valine-citrulline-p-aminobenzyloxycarbonyl (MC-vcPAB) linker (anti-FcRL5-MC-vcPAB-MMAE) or conjugated via lysines to the maytansinoid DM4 through a disulfide linker (anti-FcRL5-SPDB-DM4). These two ADCs were highly effective in vivo in combination with bortezomib or lenalidomide, drugs in use for the treatment of multiple myeloma. These data show that the FcRL5 ADCs described herein show promise as an effective treatment for multiple myeloma.


Asunto(s)
Anticuerpos Antineoplásicos/inmunología , Antineoplásicos/uso terapéutico , Inmunoconjugados/uso terapéutico , Terapia Molecular Dirigida , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/inmunología , Receptores de Superficie Celular/inmunología , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Endocitosis/efectos de los fármacos , Humanos , Inmunoconjugados/química , Inmunoconjugados/farmacología , Ratones , Ratones SCID , Receptores Fc , Reproducibilidad de los Resultados , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA