Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Theranostics ; 12(18): 7646-7667, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36451861

RESUMEN

Rationale: High mortality in pancreatic cancer (PDAC) and triple negative breast cancer (TNBC) highlight the need to capitalize on nanoscale-design advantages for multifunctional diagnostics and therapies. DNA/RNA-therapies can provide potential breakthroughs, however, to date, there is no FDA-approved systemic delivery system to solid tumors. Methods: Here, we report a Janus-nanoparticle (jNP)-system with modular targeting, payload-delivery, and targeted-imaging capabilities. Our jNP-system consists of 10 nm ultrasmall superparamagnetic iron oxide nanoparticles (USPION) with opposing antibody-targeting and DNA/RNA payload-protecting faces, directionally self-assembled with commercially available zwitterionic microbubbles (MBs) and DNA/RNA payloads. Results: Sonoporation of targeted jNP-payload-MBs delivers functional reporter-DNA imparting tumor-fluorescence, and micro-RNA126 reducing non-druggable KRAS in PDAC-Panc1 and TNBC-MB231 xenografted tumors. The targeting jNP-system enhances ultrasound-imaging of intra-tumoral microvasculature using less MBs/body weight (BW). The jNP-design enhances USPION's T2*-magnetic resonance (MR) and MR-imaging of PDAC-peritoneal metastases using less Fe/BW. Conclusion: Altogether, data advance the asymmetric jNP-design as a potential theranostic Janus-USPION Modular Platform - a JUMP forward.


Asunto(s)
MicroARNs , Neoplasias de la Mama Triple Negativas , Humanos , Medicina de Precisión , Neoplasias de la Mama Triple Negativas/diagnóstico por imagen , Neoplasias de la Mama Triple Negativas/terapia , Diagnóstico por Imagen , ADN , Neoplasias Pancreáticas
2.
Sci Rep ; 12(1): 5583, 2022 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-35379853

RESUMEN

Neutrophil-mediated secondary tissue injury underlies acute respiratory distress syndrome (ARDS) and progression to multi-organ-failure (MOF) and death, processes linked to COVID-19-ARDS. This secondary tissue injury arises from dysregulated neutrophils and neutrophil extracellular traps (NETs) intended to kill pathogens, but instead cause cell-injury. Insufficiency of pleiotropic therapeutic approaches delineate the need for inhibitors of dysregulated neutrophil-subset(s) that induce subset-specific apoptosis critical for neutrophil function-shutdown. We hypothesized that neutrophils expressing the pro-survival dual endothelin-1/VEGF-signal peptide receptor, DEspR, are apoptosis-resistant like DEspR+ cancer-cells, hence comprise a consequential pathogenic neutrophil-subset in ARDS and COVID-19-ARDS. Here, we report the significant association of increased peripheral DEspR+CD11b+ neutrophil-counts with severity and mortality in ARDS and COVID-19-ARDS, and intravascular NET-formation, in contrast to DEspR[-] neutrophils. We detect DEspR+ neutrophils and monocytes in lung tissue patients in ARDS and COVID-19-ARDS, and increased neutrophil RNA-levels of DEspR ligands and modulators in COVID-19-ARDS scRNA-seq data-files. Unlike DEspR[-] neutrophils, DEspR+CD11b+ neutrophils exhibit delayed apoptosis, which is blocked by humanized anti-DEspR-IgG4S228P antibody, hu6g8, in ex vivo assays. Ex vivo live-cell imaging of Rhesus-derived DEspR+CD11b+ neutrophils showed hu6g8 target-engagement, internalization, and induction of apoptosis. Altogether, data identify DEspR+CD11b+ neutrophils as a targetable 'rogue' neutrophil-subset associated with severity and mortality in ARDS and COVID-19-ARDS.


Asunto(s)
COVID-19 , Trampas Extracelulares , Síndrome de Dificultad Respiratoria , Humanos , Inmunofenotipificación , Neutrófilos
3.
BMC Cancer ; 21(1): 407, 2021 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-33853558

RESUMEN

BACKGROUND: Pancreatic peritoneal carcinomatosis (PPC), with the worst median overall-survival (mOS), epitomizes the incurability of metastatic cancer. Cancer stem cells (CSCs) underpin this incurability. However, inhibitors of CSC-stemness fail to increase mOS in cancer patients despite preclinical tumor-reduction. This shortfall reinforces that preclinical efficacy should be defined by increased mOS in the presence of cancer comorbidities, CSC-heterogeneity and plasticity. The primary objectives of this study are: to test the dual endothelin-1/signal peptide receptor, DEspR, as a nodal therapeutic target in PPC, given DEspR induction in anoikis-resistant pancreatic CSCs, and to validate humanized anti-DEspR antibody, hu-6g8, as a potential therapeutic for PPC. METHODS: We used heterogeneous pools of CSCs selected for anoikis resistance from reprogrammed Panc1 and MiaPaCa2 tumor cells (TCs), and adherent TCs reprogrammed from CSCs (cscTCs). We used multiple anti-DEspR blocking antibodies (mAbs) with different epitopes, and a humanized anti-DEspR recombinant mAb cross-reactive in rodents and humans, to test DEspR inhibition effects. We measured DEspR-inhibition efficacy on multiple prometastatic CSC-functions in vitro, and on tumorigenesis and overall survival in a CSC-derived xenograft (CDX) nude rat model of PPC with comorbidities. RESULTS: Here we show that DEspR, a stress-survival receptor, is present on subsets of PDAC Panc1-TCs, TC-derived CSCs, and CSC-differentiated TCs (cscTCs), and that DESpR-inhibition decreases apoptosis-resistance and pro-metastatic mesenchymal functions of CSCs and cscTCs in vitro. We resolve the DNA-sequence/protein-function discordance by confirming ADAR1-RNA editing-dependent DEspR-protein expression in Panc1 and MiaPaCa2 TCs. To advance DEspR-inhibition as a nodal therapeutic approach for PPC, we developed and show improved functionality of a recombinant, humanized anti-DEspR IgG4S228P antibody, hu-6g8, over murine precursor anti-DEspR mabs. Hu-6g8 internalizes and translocates to the nucleus colocalized with cyto-nuclear shuttling galectins-1/3, and induces apoptotic cell changes. DEspR-inhibition blocks transperitoneal dissemination and progression to peritoneal carcinomatosis of heterogeneous DEspR±/CD133 ± Panc1-derived CSCs in xenografted nude rats, improving mOS without chemotherapy-like adverse effects. Lastly, we show DEspR expression in Stage II-IV primary and invasive TCs in the stroma in PDAC-patient tumor arrays. CONCLUSION: Collectively, the data support humanized anti-DEspR hu-6g8 as a potential targeted antibody-therapeutic with promising efficacy, safety and prevalence profiles for PPC patients.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Antineoplásicos Inmunológicos/farmacología , Inmunoglobulina G/farmacología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Neoplasias Peritoneales/tratamiento farmacológico , Neoplasias Peritoneales/secundario , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales Humanizados/química , Antineoplásicos Inmunológicos/química , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Inmunoglobulina G/química , Inmunohistoquímica , Inmunofenotipificación , Neoplasias Pancreáticas/patología , Ratas , Receptor de Endotelina A , Ensayos Antitumor por Modelo de Xenoinjerto
4.
ACS Nano ; 11(2): 1466-1477, 2017 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-28099801

RESUMEN

A current challenge in the treatment of peritoneal carcinomatosis is the inability to detect, visualize, and resect small or microscopic tumors of pancreatic, ovarian, or mesothelial origin. In these diseases, the completeness of primary tumor resection is directly correlated with patient survival, and hence, identifying small sub-millimeter tumors (i.e., disseminated disease) is critical. Thus, new imaging techniques and probes are needed to improve cytoreductive surgery and patient outcomes. Highly fluorescent rhodamine-labeled expansile nanoparticles (HFR-eNPs) are described for use as a visual aid during cytoreductive surgery of pancreatic carcinomatosis. The covalent incorporation of rhodamine into ∼30 nm eNPs increases the fluorescent signal compared to free rhodamine, thereby affording a brighter and more effective probe than would be achieved by a single rhodamine molecule. Using the intraperitoneal route of administration, HFR-eNPs localize to regions of large (∼1 cm), sub-centimeter, and sub-millimeter intraperitoneal tumor in three different animal models, including pancreatic, mesothelioma, and ovarian carcinoma. Tumoral localization of the HFR-eNPs depends on both the material property (i.e., eNP polymer) as well as the surface chemistry (anionic surfactant vs PEGylated noncharged surfactant). In a rat model of pancreatic carcinomatosis, HFR-eNP identification of tumor is validated against gold-standard histopathological analysis to reveal that HFR-eNPs possess high specificity (99%) and sensitivity (92%) for tumors, in particular, sub-centimeter and microscopic sub-millimeter tumors, with an overall accuracy of 95%. Finally, as a proof-of-concept, HFR-eNPs are used to guide the resection of pancreatic tumors in a rat model of peritoneal carcinomatosis.


Asunto(s)
Neoplasias Pulmonares/diagnóstico por imagen , Mesotelioma/diagnóstico por imagen , Imagen Óptica , Neoplasias Ováricas/diagnóstico por imagen , Neoplasias Pancreáticas/diagnóstico por imagen , Neoplasias Peritoneales/diagnóstico por imagen , Animales , Supervivencia Celular/efectos de los fármacos , Terapia Combinada , Procedimientos Quirúrgicos de Citorreducción , Modelos Animales de Enfermedad , Femenino , Colorantes Fluorescentes/administración & dosificación , Colorantes Fluorescentes/química , Colorantes Fluorescentes/farmacocinética , Inyecciones Intraperitoneales , Neoplasias Pulmonares/cirugía , Mesotelioma/cirugía , Mesotelioma Maligno , Ratones , Ratones Desnudos , Nanopartículas/administración & dosificación , Nanopartículas/química , Neoplasias Ováricas/cirugía , Neoplasias Pancreáticas/cirugía , Neoplasias Peritoneales/cirugía , Ratas , Rodaminas/administración & dosificación , Rodaminas/química , Rodaminas/farmacocinética , Tensoactivos/química , Distribución Tisular , Células Tumorales Cultivadas
5.
BMC Mol Biol ; 17(1): 15, 2016 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-27301377

RESUMEN

BACKGROUND: In contrast to rat and mouse databases, the NCBI gene database lists the human dual-endothelin1/VEGFsp receptor (DEspR, formerly Dear) as a unitary transcribed pseudogene due to a stop [TGA]-codon at codon#14 in automated DNA and RNA sequences. However, re-analysis is needed given prior single gene studies detected a tryptophan [TGG]-codon#14 by manual Sanger sequencing, demonstrated DEspR translatability and functionality, and since the demonstration of actual non-translatability through expression studies, the standard-of-excellence for pseudogene designation, has not been performed. Re-analysis must meet UNIPROT criteria for demonstration of a protein's existence at the highest (protein) level, which a priori, would override DNA- or RNA-based deductions. METHODS: To dissect the nucleotide sequence discrepancy, we performed Maxam-Gilbert sequencing and reviewed 727 RNA-seq entries. To comply with the highest level multiple UNIPROT criteria for determining DEspR's existence, we performed various experiments using multiple anti-DEspR monoclonal antibodies (mAbs) targeting distinct DEspR epitopes with one spanning the contested tryptophan [TGG]-codon#14, assessing: (a) DEspR protein expression, (b) predicted full-length protein size, (c) sequence-predicted protein-specific properties beyond codon#14: receptor glycosylation and internalization, (d) protein-partner interactions, and (e) DEspR functionality via DEspR-inhibition effects. RESULTS: Maxam-Gilbert sequencing and some RNA-seq entries demonstrate two guanines, hence a tryptophan [TGG]-codon#14 within a compression site spanning an error-prone compression sequence motif. Western blot analysis using anti-DEspR mAbs targeting distinct DEspR epitopes detect the identical glycosylated 17.5 kDa pull-down protein. Decrease in DEspR-protein size after PNGase-F digest demonstrates post-translational glycosylation, concordant with the consensus-glycosylation site beyond codon#14. Like other small single-transmembrane proteins, mass spectrometry analysis of anti-DEspR mAb pull-down proteins do not detect DEspR, but detect DEspR-protein interactions with proteins implicated in intracellular trafficking and cancer. FACS analyses also detect DEspR-protein in different human cancer stem-like cells (CSCs). DEspR-inhibition studies identify DEspR-roles in CSC survival and growth. Live cell imaging detects fluorescently-labeled anti-DEspR mAb targeted-receptor internalization, concordant with the single internalization-recognition sequence also located beyond codon#14. CONCLUSIONS: Data confirm translatability of DEspR, the full-length DEspR protein beyond codon#14, and elucidate DEspR-specific functionality. Along with detection of the tryptophan [TGG]-codon#14 within an error-prone compression site, cumulative data demonstrating DEspR protein existence fulfill multiple UNIPROT criteria, thus refuting its pseudogene designation.


Asunto(s)
Biosíntesis de Proteínas , Seudogenes/genética , Animales , Anoicis , Línea Celular Tumoral , Codón , Galectina 1/análisis , Galectina 1/metabolismo , Humanos , Ratones , Neoplasias/genética , Neoplasias/metabolismo , Mapas de Interacción de Proteínas , Ratas , Triptófano/genética
6.
Nanomedicine (Lond) ; 11(9): 1001-15, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27078118

RESUMEN

AIM: To evaluate the tumor localization and efficacy pH-responsive expansile nanoparticles (eNPs) as a drug delivery system for pancreatic peritoneal carcinomatosis (PPC) modeled in nude rats. METHODS & MATERIALS: A Panc-1-cancer stem cell xeno1graft model of PPC was validated in vitro and in vivo. Tumor localization was tracked via in situ imaging of fluorescent eNPs. Survival of animals treated with paclitaxel-loaded eNPs (PTX-eNPs) was evaluated in vivo. RESULTS: The Panc-1-cancer stem cell xenograft model recapitulates significant features of PPC. Rhodamine-labeled eNPs demonstrate tumor-specific, dose- and time-dependent localization to macro- and microscopic tumors following intraperitoneal injection. PTX-eNPs are as effective as free PTX in treating established PPC; but, PTX-eNPs result in fewer side effects. CONCLUSION: eNPs are a promising tool for the detection and treatment of PPC.


Asunto(s)
Carcinoma/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , Nanopartículas/administración & dosificación , Neoplasias Pancreáticas/tratamiento farmacológico , Animales , Carcinoma/patología , Modelos Animales de Enfermedad , Humanos , Nanopartículas/química , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Paclitaxel/administración & dosificación , Paclitaxel/química , Neoplasias Pancreáticas/patología , Peritoneo/efectos de los fármacos , Peritoneo/patología , Ratas , Rodaminas/química , Ensayos Antitumor por Modelo de Xenoinjerto
7.
PLoS One ; 10(1): e0116724, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25615575

RESUMEN

Identification of susceptibility genes for essential hypertension in humans has been a challenge due to its multifactorial pathogenesis complicated by gene-gene and gene-environment interactions, developmental programing and sex specific differences. These concurrent features make identification of causal hypertension susceptibility genes with a single approach difficult, thus requiring multiple lines of evidence involving genetic, biochemical and biological experimentation to establish causal functional mutations. Here we report experimental evidence encompassing genetic, biochemical and in vivo modeling that altogether support ATP1A1 as a hypertension susceptibility gene in males in Sardinia, Italy. ATP1A1 encodes the α1Na,K-ATPase isoform, the sole sodium pump in vascular endothelial and renal tubular epithelial cells. DNA-sequencing detected a 12-nucleotide long thymidine (12T) insertion(ins)/deletion(del) polymorphism within a poly-T sequence (38T vs 26T) in the ATP1A1 5'-regulatory region associated with hypertension in a male Sardinian population. The 12T-insertion allele confers decreased susceptibility to hypertension (P = 0.035; OR = 0.50 [0.28-0.93]) accounting for 12.1 mmHg decrease in systolic BP (P = 0.02) and 6.6 mmHg in diastolic BP (P = 0.046). The ATP1A1 promoter containing the 12T-insertion exhibited decreased transcriptional activity in in vitro reporter-assay systems, indicating decreased α1Na,K-ATPase expression with the 12T-insertion, compared with the 12T-deletion ATP1A1 promoter. To test the effects of decreased α1Na,K-ATPase expression on blood pressure, we measured blood pressure by radiotelemetry in three month-old, highly inbred heterozygous knockout ATP1A1+/- male mice with resultant 58% reduction in ATP1A1 protein levels. Male ATP1A1+/- mice showed significantly lower blood pressure (P < 0.03) than age-matched male wild-type littermate controls. Concordantly, lower ATP1A1 expression is expected to lower Na-reabsorption in the kidney thereby decreasing sodium-associated risk for hypertension and sodium-induced endothelial stiffness and dysfunction. Altogether, data support ATP1A1 as a hypertension susceptibility gene in a male Sardinian population, and mandate further investigation of its involvement in hypertension in the general population.


Asunto(s)
Hipertensión/genética , Hipertensión/prevención & control , Mutación INDEL , ATPasa Intercambiadora de Sodio-Potasio/genética , Población Blanca/genética , Adulto , Anciano , Animales , Modelos Animales de Enfermedad , Hipertensión Esencial , Predisposición Genética a la Enfermedad , Humanos , Italia , Masculino , Ratones , Persona de Mediana Edad , Regiones Promotoras Genéticas , Análisis de Secuencia de ADN , Factores Sexuales , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Timidina/metabolismo
8.
PLoS One ; 9(1): e85821, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24465725

RESUMEN

A priori, a common receptor induced in tumor microvessels, cancer cells and cancer stem-like cells (CSCs) that is involved in tumor angiogenesis, invasiveness, and CSC anoikis resistance and survival, could underlie contemporaneous coordination of these events rather than assume stochasticity. Here we show that functional analysis of the dual endothelin1/VEGFsignal peptide receptor, DEspR, (formerly named Dear, Chr.4q31.2) supports the putative common receptor paradigm in pancreatic ductal adenocarcinoma (PDAC) and glioblastoma (GBM) selected for their invasiveness, CD133+CSCs, and polar angiogenic features. Unlike normal tissue, DEspR is detected in PDAC and GBM microvessels, tumor cells, and CSCs isolated from PDAC-Panc1 and GBM-U87 cells. DEspR-inhibition decreased angiogenesis, invasiveness, CSC-survival and anoikis resistance in vitro, and decreased Panc1-CSC and U87-CSC xenograft tumor growth, vasculo-angiogenesis and invasiveness in nude(nu/nu) rats, suggesting that DEspR activation would coordinate these tumor progression events. As an accessible, cell-surface 'common receptor coordinator', DEspR-inhibition defines a novel targeted-therapy paradigm for pancreatic cancer and glioblastoma.


Asunto(s)
Anoicis , Neoplasias Encefálicas/irrigación sanguínea , Glioblastoma/irrigación sanguínea , Células Madre Neoplásicas/patología , Neovascularización Patológica/metabolismo , Neoplasias Pancreáticas/irrigación sanguínea , Receptores de Superficie Celular/metabolismo , Animales , Neoplasias Encefálicas/patología , Células COS , Línea Celular Tumoral , Supervivencia Celular , Chlorocebus aethiops , Glioblastoma/metabolismo , Glioblastoma/patología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Ligandos , Microvasos/metabolismo , Microvasos/patología , Invasividad Neoplásica , Células Madre Neoplásicas/metabolismo , Neovascularización Patológica/patología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Seudogenes , Ratas , Ratas Desnudas , Receptores de Superficie Celular/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto , Neoplasias Pancreáticas
9.
PLoS One ; 8(8): e72143, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23967281

RESUMEN

Although two major breast cancer susceptibility genes, BRCA1 and BRCA2, have been identified accounting for 20% of breast cancer genetic risk, identification of other susceptibility genes accounting for 80% risk remains a challenge due to the complex, multi-factorial nature of breast cancer. Complexity derives from multiple genetic determinants, permutations of gene-environment interactions, along with presumptive low-penetrance of breast cancer predisposing genes, and genetic heterogeneity of human populations. As with other complex diseases, dissection of genetic determinants in animal models provides key insight since genetic heterogeneity and environmental factors can be experimentally controlled, thus facilitating the detection of quantitative trait loci (QTL). We therefore, performed the first genome-wide scan for loci contributing to radiation-induced mammary tumorigenesis in female F2-(Dahl S x R)-intercross rats. Tumorigenesis was measured as tumor burden index (TBI) after induction of rat mammary tumors at forty days of age via ¹²7Cs-radiation. We observed a spectrum of tumor latency, size-progression, and pathology from poorly differentiated ductal adenocarcinoma to fibroadenoma, indicating major effects of gene-environment interactions. We identified two mammary tumorigenesis susceptibility quantitative trait loci (Mts-QTLs) with significant linkage: Mts-1 on chromosome-9 (LOD-2.98) and Mts-2 on chromosome-1 (LOD-2.61), as well as two Mts-QTLs with suggestive linkage: Mts-3 on chromosome-5 (LOD-1.93) and Mts-4 on chromosome-18 (LOD-1.54). Interestingly, Chr9-Mts-1, Chr5-Mts-3 and Chr18-Mts-4 QTLs are unique to irradiation-induced mammary tumorigenesis, while Chr1-Mts-2 QTL overlaps with a mammary cancer susceptibility QTL (Mcs 3) reported for 7,12-dimethylbenz-[α]antracene (DMBA)-induced mammary tumorigenesis in F2[COP x Wistar-Furth]-intercross rats. Altogether, our results suggest at least three distinct susceptibility QTLs for irradiation-induced mammary tumorigenesis not detected in genetic studies of chemically-induced and hormone-induced mammary tumorigenesis. While more study is needed to identify the specific Mts-gene variants, elucidation of specific variant(s) could establish causal gene pathways involved in mammary tumorigenesis in humans, and hence novel pathways for therapy.


Asunto(s)
Carcinogénesis/efectos de la radiación , Sitios Genéticos , Hibridación Genética , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Neoplasias Inducidas por Radiación/genética , Neoplasias Inducidas por Radiación/patología , Animales , Carcinogénesis/genética , Cesio/efectos adversos , Femenino , Predisposición Genética a la Enfermedad/genética , Genómica , Humanos , Masculino , Sitios de Carácter Cuantitativo/genética , Ratas
10.
Physiol Genomics ; 43(1): 32-42, 2011 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-20923861

RESUMEN

Arginine vasopressin (AVP) and angiotensin II (ANG II) are distinct peptide hormones involved in multiple organs modulating renal, cardiovascular, and brain functions. They achieve these functions via specific G protein-coupled receptors, respectively. The AVR/NAVR locus encodes two overlapping V2-type vasopressin isoreceptors: angiotensin-vasopressin receptor (AVR) responding to ANG II and AVP equivalently, and nonangiotensin vasopressin receptor (NAVR), which binds vasopressin exclusively. AVR and NAVR are expressed from a single gene by alternative promoter usage that is synergistically upregulated by testosterone and estrogen. This study tested the hypothesis that AVR/NAVR modulates urinary concentrating ability, blood pressure, and cognitive performance in vivo in a sex-specific manner. We developed a C57BL/6 inbred AVR/NAVR(-/-) knockout mouse that showed lower blood pressure in both male and female subjects and a urinary-concentrating defect restricted to male mice. We also detected sex-specific effects on cognitive and anxiety-like behaviors. AVR/NAVR(-/-) male mice exhibited impaired visuospatial and associative learning, while female mice showed improved performance in both type of cognition. AVR/NAVR deficiency produced an anxiolytic-like effect in female mice, while males were unaffected. Analysis of AVR- and NAVR-mediated phosphorylation/dephosphorylation of signaling proteins revealed activation/deactivation of known modulators of cognitive function. Our studies identify AVR/NAVR as key receptors involved in blood pressure regulation and sex-specific modulation of renal water homeostasis, cognitive function, and anxiety-like behavior. As such, the AVR/NAVR receptor system provides a molecular mechanism for sexually diergic traits and a putative common pathway for the emerging association of hypertension and cognitive decline and dementia.


Asunto(s)
Ansiedad/fisiopatología , Presión Sanguínea/fisiología , Cognición/fisiología , Capacidad de Concentración Renal/fisiología , Receptores de Angiotensina/deficiencia , Receptores Acoplados a Proteínas G/deficiencia , Receptores de Vasopresinas/deficiencia , Animales , Ansiedad/genética , Presión Sanguínea/genética , Femenino , Capacidad de Concentración Renal/genética , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Fosforilación , Receptores de Angiotensina/genética , Receptores Acoplados a Proteínas G/genética , Receptores de Vasopresinas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
Mol Imaging Biol ; 13(6): 1096-106, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20972637

RESUMEN

PURPOSE: Given that carotid vasa vasorum neovascularization is associated with increased risk for stroke and cardiac events, the present in vivo study was designed to investigate molecular imaging of carotid artery vasa vasorum neovascularization via target-specific contrast-enhanced ultrasound (CEU) micro-imaging. PROCEDURES: Molecular imaging was performed in male transgenic rats with carotid artery disease and non-transgenic controls using dual endothelin1/VEGFsp receptor (DEspR)-targeted microbubbles (MB(D)) and the Vevo770 micro-imaging system and CEU imaging software. RESULTS: DEspR-targeted CEU-positive imaging exhibited significantly higher contrast intensity signal (CIS)-levels and pre-/post-destruction CIS-differences in seven of 13 transgenic rats, in contrast to significantly lower CIS-levels and differences in control isotype-targeted microbubble (MB(C))-CEU imaging (n = 8) and in MB(D) CEU-imaging of five non-transgenic control rats (P < 0.0001). Ex vivo immunofluorescence analysis demonstrated binding of MB(D) to DEspR-positive endothelial cells; and association of DEspR-targeted increased contrast intensity signals with DEspR expression in vasa vasorum neovessel and intimal lesions. In vitro analysis demonstrated dose-dependent binding of MB(D) to DEspR-positive human endothelial cells with increasing %cells bound and number of MB(D) per cell, in contrast to MB(C) or non-labeled microbubbles (P < 0.0001). CONCLUSION: In vivo DEspR-targeted molecular imaging detected increased DEspR-expression in carotid artery lesions and in expanded vasa vasorum neovessels in transgenic rats with carotid artery disease. Future studies are needed to determine predictive value for stroke or heart disease in this transgenic atherosclerosis rat model and translational applications.


Asunto(s)
Aterosclerosis/diagnóstico por imagen , Medios de Contraste , Microburbujas , Imagen Molecular/métodos , Neovascularización Patológica/diagnóstico por imagen , Receptor de Endotelina A/metabolismo , Receptores de Endotelina/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Vasa Vasorum/patología , Animales , Arterias Carótidas/diagnóstico por imagen , Arterias Carótidas/patología , Modelos Animales de Enfermedad , Células Endoteliales/patología , Técnica del Anticuerpo Fluorescente , Humanos , Microscopía Fluorescente , Microscopía de Contraste de Fase , Ratas , Ratas Transgénicas , Ultrasonografía , Vasa Vasorum/diagnóstico por imagen
12.
Physiol Genomics ; 35(3): 316-29, 2008 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-18780760

RESUMEN

Aside from abnormal angiogenesis, dual endothelin-1/VEGF signal peptide-activated receptor deficiency (DEspR(-/-)) results in aberrant neuroepithelium and neural tube differentiation, thus elucidating DEspR's role in neurogenesis. With the emerging importance of neurogenesis in adulthood, we tested the hypothesis that nonembryonic-lethal DEspR haploinsufficiency (DEspR(+/-)) perturbs neuronal homeostasis, thereby facilitating aging-associated neurodegeneration. Here we show that, in male mice only, DEspR-haploinsufficiency impaired hippocampus-dependent visuospatial and associative learning and induced noninflammatory spongiform changes, neuronal vacuolation, and loss in the hippocampus, cerebral cortex, and subcortical regions, consistent with autophagic cell death. In contrast, DEspR(+/-) females exhibited better cognitive performance than wild-type females and showed absence of neuropathological changes. Signaling pathway analysis revealed DEspR-mediated phosphorylation of activators of autophagy inhibitor mammalian target of rapamycin (mTOR) and dephosphorylation of known autophagy inducers. Altogether, the data demonstrate DEspR-mediated diametrical, sex-specific modulation of cognitive performance and autophagy, highlight cerebral neuronal vulnerability to autophagic dysregulation, and causally link DEspR haploinsufficiency with increased neuronal autophagy, spongiosis, and cognitive decline in mice.


Asunto(s)
Autofagia/fisiología , Trastornos del Conocimiento/genética , Hipocampo/metabolismo , Neuronas/metabolismo , Animales , Autofagia/genética , Conducta Animal/fisiología , Trastornos del Conocimiento/patología , Trastornos del Conocimiento/fisiopatología , Femenino , Hipocampo/patología , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Neuronas/patología , Receptores de Angiotensina/genética , Receptores de Endotelina/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores Sexuales , Conducta Social
13.
Physiol Genomics ; 34(1): 65-77, 2008 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-18413781

RESUMEN

The angiotensin-vasopressin receptor (AVR) responds with equivalent affinities to angiotensin II (ANG II) and vasopressin and is coupled to adenylate cyclase and hence a V2-type vasopressin receptor. AVR maps to the Nalp6 locus and overlaps with the larger Nalp6/PYPAF5 reported to be a T cell/granulocyte-specific, cytoplasmic-specific proapoptotic protein, thus questioning the existence of AVR. Here we confirm, through different experimental modalities, that AVR is distinct from Nalp6/PYPAF5 based on different mRNA and protein sizes, subcellular localization, and tissue-specific expression patterns. Binding studies of PYPAF5-specific Cos1 transfectants detect high-affinity binding to vasopressin but not ANG II, thus assigning PYPAF5 as a non-AVR (NAVR). Signaling array analysis reveals that AVP stimulation of AVR- and NAVR-specific Cos1 transfectants results in diametrical activation as well as coactivation of signaling pathways known to mediate renal sodium and water balance. Likewise, ANG II stimulation of Cos1-AVR transfectants reveals a signaling profile distinct from that of AVP-stimulated Cos1-AVR transfectants. Analysis of genomic organization of the AVR/NAVR locus shows an overlapping gene arrangement with alternative promoter usage resulting in different NH(2) termini for NAVR and AVR. In addition to core promoter elements, androgen and estrogen response elements are detected. Promoter analysis of NAVR/AVR 5'-regulatory region detects transcriptional upregulation by testosterone and synergistic upregulation by testosterone and estrogen, thus suggesting that AVR and/or NAVR contribute to sex-specific V2-type vasopressin-mediated effects. Altogether, confirmation of AVR and identification of NAVR as vasopressin receptors are concordant with emerging vasopressin functions not attributable to V1a, V1b, or V2 receptors and add molecular bases for the multifunctional complexity of vasopressin-mediated functions and regulation.


Asunto(s)
Receptores de Angiotensina/genética , Receptores de Vasopresinas/genética , Angiotensina II/metabolismo , Animales , Western Blotting , Células COS , Chlorocebus aethiops , Regulación de la Expresión Génica/efectos de los fármacos , Genoma/genética , Hormonas Esteroides Gonadales/farmacología , Ligandos , Microscopía Confocal , Especificidad de Órganos/efectos de los fármacos , Péptidos/metabolismo , Unión Proteica/efectos de los fármacos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Receptores de Angiotensina/metabolismo , Receptores de Vasopresinas/metabolismo , Transducción de Señal/efectos de los fármacos , Sitio de Iniciación de la Transcripción , Transcripción Genética/efectos de los fármacos , Transfección , Vasopresinas/metabolismo
14.
Circ Res ; 100(10): 1522-9, 2007 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-17446437

RESUMEN

Essential hypertension remains a major risk factor for cardiovascular and cerebrovascular diseases. As a complex multifactorial disease, elucidation of susceptibility loci remains elusive. ATP1A1 and Dear are candidate genes for 2 closely linked rat chromosome-2 blood pressure quantitative trait loci. Because corresponding human syntenic regions are on different chromosomes, investigation of ATP1A1 (chromosome [chr]-1p21) and Dear (chr-4q31.3) facilitates genetic analyses of each blood pressure quantitative trait locus in human hypertension. Here we report the association of human ATP1A1 (P<0.000005) and Dear (P<0.03) with hypertension in a relatively isolated, case/control hypertension cohort from northern Sardinia by single-nucleotide polymorphism haplotype analysis. Sex-specific haplotype analyses detected stronger association of both loci with hypertension in males than in females. Haplotype trend-regression analyses support ATP1A1 and Dear as independent susceptibility loci and reveal haplotype-specific association with hypertension and normotension, thus delineating haplotype-specific subsets of hypertension. Although investigation in other cohorts needs to be performed to determine genetic effects in other populations, haplotype subtyping already allows systematic stratification of susceptibility and, hence, clinical heterogeneity, a prerequisite for unraveling the polygenic etiology and polygene-environment interactions in essential hypertension. As hypertension susceptibility genes, coexpression of ATP1A1 and Dear in both renal tubular cells and vascular endothelium suggest a cellular pathogenic scaffold for polygenic mechanisms of hypertension, as well as the hypothesis that ATP1A1 and/or Dear could contribute to the known renal and vascular endothelial dysfunction associated with essential (polygenic) hypertension.


Asunto(s)
Haplotipos , Hipertensión/genética , Polimorfismo de Nucleótido Simple , Receptores de Angiotensina/genética , Receptores de Endotelina/genética , ATPasa Intercambiadora de Sodio-Potasio/genética , Frecuencia de los Genes , Humanos , Desequilibrio de Ligamiento , Análisis de Regresión , Caracteres Sexuales
15.
Physiol Genomics ; 23(3): 257-68, 2005 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-16293765

RESUMEN

The dual endothelin-1/angiotensin II receptor (Dear) binds endothelin-1 (ET-1) and angiotensin II (ANG II) with equal affinities in the Dahl S/JRHS rat strain. To elucidate its physiological significance within the context of multiple receptor isoforms and diverse ET-1 and ANG II functions spanning blood pressure regulation, tumor proliferation, and angiogenesis, we characterized mouse Dear and Dear-deficient mice. Unlike null mutant models of ET-1, ANG II, and all other ET-1 and ANG II receptors, Dear(-/-) deficiency results in impaired angiogenesis, dysregulated neuroepithelial development, and embryonic lethality by embryonic day 12.5. Interestingly, mouse Dear does not bind ANG II, similar to Dahl R/JRHS rat Dear, but binds ET-1 and vascular endothelial growth factor (VEGF) signal peptide (VEGFsp) with equal affinities, suggesting a putative novel multifunction for VEGFsp and a parsimonious mechanism for coordination of VEGF-induced and Dear-mediated pathways. Consistent with its developmental angiogenic role, Dear inhibition results in decreased tumor growth in B16-F10 melanoma cell-induced subcutaneous tumor in female Dear(+/-)/C57BL6BC10 mice, but not in males (age 3.5 mo), and in 127Cs radiation-induced orthotopic mammary tumors in Sprague-Dawley female rats (age range 3-6.5 mo). Altogether, the data identify Dear as a new player in angiogenesis during development downstream to, and nonredundant with, VEGF-mediated pathways, as well as a putative modulator of tumor angiogenesis acting within a gender-specific paradigm.


Asunto(s)
Desarrollo Embrionario/genética , Neovascularización Fisiológica/genética , Receptores de Angiotensina/genética , Receptores de Endotelina/genética , Animales , Clonación Molecular , Cartilla de ADN , Femenino , Regulación de la Expresión Génica , Genotipo , Masculino , Ratones , Ratones Mutantes , Fenotipo , Embarazo , Mapeo Restrictivo
16.
J Hypertens ; 23(4): 745-52, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15775778

RESUMEN

OBJECTIVE: Multifactorial analyses support the hypothesis that alpha1Na,K-ATPase is a hypertension susceptibility gene in Dahl S rats. However, two studies report non-detection of the A1079T transversion underlying the Q276L substitution in Dahl S alpha1Na,K-ATPase questioning the validity of ATP1A1 as a hypertension susceptibility gene. To resolve this discordance, we investigated the issue at the protein level. DESIGN AND METHODS: We employed protein blot analysis using Q276L- and Q276-specific; antipeptide-specific antibodies; tested differential chymotrypsin cleavage efficiency, measured differential Na and K affinities of alpha1Na,K-ATPases in Dahl S and Dahl R renal membranes and determined amino acid sequences of purified Dahl S alpha1Na,K-ATPase chymotryptic-digest peptides. RESULTS: We detected Q276L variant protein in Dahl S rats; and Q276 wild-type variant in Dahl R, spontaneously hypertensive (SHR), Lewis and Wistar-Kyoto (WKY) rat kidney membranes. Q276L variant exhibits less chymotrypsin cleavage efficiency than the Q276 wild-type variant, consistent with the substitution of hydrophobic L for hydrophilic Q. Kinetic studies of kidney membranes detect increased Na affinity and decreased K affinity in renal Dahl S alpha1Na,K-ATPase compared with Dahl R. Protein sequencing of high pressure liquid chromatography (HPLC)-purified chymotrypsin digested 77 kDa peptide confirms Q276L substitution in the Dahl S alpha1Na,K-ATPase. CONCLUSIONS: Data demonstrate the existence and functional significance of the Q276L variant in Dahl S rats.


Asunto(s)
Hipertensión/genética , Hipertensión/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/genética , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Sustitución de Aminoácidos/genética , Animales , Especificidad de Anticuerpos , Western Blotting , Inmunohistoquímica , Riñón/enzimología , Cinética , Ligandos , Ratas , Ratas Endogámicas Dahl , Ratas Endogámicas Lew , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Análisis de Secuencia de Proteína , ATPasa Intercambiadora de Sodio-Potasio/inmunología
17.
Mol Med ; 8(1): 24-32, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11984003

RESUMEN

BACKGROUND: Essential hypertension is a prevalent complex polygenic disease and a major risk factor for cardiovascular disease, the leading cause of death in developed countries. Because of its complex and multifactorial nature, its genetic determinants still remain largely unknown. The Dahl salt-sensitive hypertensive rat model exhibits impaired sodium handling, which is hypothesized to play a key role in the pathophysiology of polygenic hypertension. Thus, genes associated with renal regulation of salt and water balance are a priori likely candidates for a causative role in hypertension pathogenesis. The functional properties and renal-specific expression of the recently characterized AngII/AVP receptor suggest a putative modulator role in tubular sodium and fluid reabsorption. Based on these observations, we investigated the potential involvement of the AngII/AVP receptor in salt-sensitive hypertension. MATERIALS AND METHODS: We performed cosegregation analysis of the AngII/AVP receptor locus with salt-sensitive hypertension in an F2 (Dahl S X Dahl salt-resistant [R]) hybrid male cohort characterized for blood pressure by radiotelemetry after 8 weeks of high salt challenge. Further molecular analysis was done to identify putative AngII/AVP receptor molecular variants that could account for the AngII/ AVP receptor involvement in salt-sensitive hypertension pathogenesis. RESULTS: The AngII/AVP receptor was mapped to rat chromosome 1, 1.7 cM centromeric to the D1Rat188 marker by radiation hybrid mapping analysis. Quantitative trait locus (QTL) analysis detected a highly significant linkage of the AngII/AVP receptor locus with high blood pressure (LRS = 13.8, p= 0.0002). Molecular characterization of the Dahl S and Dahl R AngII/AVP receptor cDNAs revealed two amino acid substitutions in the Dahl S AngII/AVP receptor (N119S, C163R) when compared to the Dahl R AngII/AVP receptor. These mutations are associated with an increased receptor affinity for both ligands (AVP and AngII) and an enhanced G(s)-coupling by the receptor resulting in increased activation of adenylate cyclase with concomitant increase in cAMP production. CONCLUSIONS: The observed molecular dysfunction in the Dahl S AngII/AVP receptor is consistent with increased tubular sodium and fluid reabsorption observed in Dahl S rats. Interestingly, the AngII/AVPr locus is within the narrowed chromosome 1 QTL region for blood pressure detected in different rat intercross linkage analyses. Altogether, the data strongly suggest that the AngII/AVP receptor is a hypertension susceptibility gene in the Dahl S rat model, as well as raises the hypothesis that it too underlies the chromosome 1 blood pressure QTL identified in other hypertension rat models.


Asunto(s)
Genes , Hipertensión/genética , Ratas Endogámicas Dahl/genética , Receptores de Angiotensina/genética , Receptores de Vasopresinas/genética , Regulación Alostérica , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Angiotensina II/metabolismo , Animales , Arginina Vasopresina/metabolismo , Presión Sanguínea/genética , Mapeo Cromosómico , Cruzamientos Genéticos , AMP Cíclico/metabolismo , ADN Complementario/genética , Marcadores Genéticos , Predisposición Genética a la Enfermedad , Hipertensión/inducido químicamente , Hipertensión/metabolismo , Riñón/química , Ligandos , Escala de Lod , Masculino , Datos de Secuencia Molecular , Carácter Cuantitativo Heredable , Ratas , Ratas Endogámicas Dahl/metabolismo , Receptores de Angiotensina/química , Receptores de Angiotensina/fisiología , Receptores de Vasopresinas/química , Receptores de Vasopresinas/fisiología , Sistemas de Mensajero Secundario , Homología de Secuencia , Sodio/farmacología , Cloruro de Sodio/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA