Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Immunol ; 205(6): 1593-1600, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32769122

RESUMEN

We previously reported sex differences in innate susceptibility to Staphylococcus aureus skin infection and that bone marrow neutrophils (BMN) from female mice have an enhanced ability to kill S. aureus ex vivo compared with those of male mice. However, the mechanism(s) driving this sex bias in neutrophil killing have not been reported. Given the role of opsonins such as complement, as well as their receptors, in S. aureus recognition and clearance, we investigated their contribution to the enhanced bactericidal capacity of female BMN. We found that levels of C3 in the serum and CR3 (CD11b/CD18) on the surface of BMN were higher in female compared with male mice. Consistent with increased CR3 expression following TNF-α priming, production of reactive oxygen species (ROS), an important bactericidal effector, was also increased in female versus male BMN in response to serum-opsonized S. aureus Furthermore, blocking CD11b reduced both ROS levels and S. aureus killing by murine BMN from both sexes. However, at the same concentration of CD11b blocking Ab, S. aureus killing by female BMN was greatly reduced compared with those from male mice, suggesting CR3-dependent differences in bacterial killing between sexes. Overall, this work highlights the contributions of CR3, C3, and ROS to innate sex bias in the neutrophil response to S. aureus Given that neutrophils are crucial for S. aureus clearance, understanding the mechanism(s) driving the innate sex bias in neutrophil bactericidal capacity could identify novel host factors important for host defense against S. aureus.


Asunto(s)
Antígeno de Macrófago-1/metabolismo , Neutrófilos/fisiología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/fisiología , Animales , Anticuerpos Bloqueadores/metabolismo , Antígeno CD11b/inmunología , Antígeno CD11b/metabolismo , Complemento C3/metabolismo , Citotoxicidad Inmunológica , Femenino , Interacciones Huésped-Patógeno , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/metabolismo , Caracteres Sexuales , Factores Sexuales
2.
Wound Repair Regen ; 28(4): 470-479, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32428975

RESUMEN

Sex differences in susceptibility to ischemia/reperfusion injury have been documented in humans. Premenopausal women have a lower risk of ischemic heart disease than age-matched men, whereas after menopause, the risk is similar or even higher in women. However, little is known about the effects of sex on myocutaneous ischemia/reperfusion. To explore sex differences in wound revascularization, we utilized a murine myocutaneous flap model of graded ischemia. A cranial-based, peninsular-shaped, myocutaneous flap was surgically created on the dorsum of male and female mice. Physiological, pathological, immunohistochemical, and molecular parameters were analyzed. Flaps created on female mice were re-attached to the recipient site resulting in nearly complete viability at post-operative day 10. In contrast, distal full-thickness myocutaneous necrosis was evident at 10 days post-surgery in male mice. Over the 10 day study interval, laser speckle imaging documented functional revascularization in all flap regions in female mice, but minimal distal flap reperfusion in male mice. Day 10 immunostained histologic sections confirmed significant increases in distal flap vessel count and vascular surface area in female compared to male mice. RT-PCR demonstrated significant differences in growth factor and metabolic gene expression between female and male mice at day 10. In conclusion, in a graded-ischemia wound healing model, flap revascularization was more effective in female mice. The recognition and identification of sex-specific wound healing differences may lead to a better understanding of the underlying mechanisms of myocutaneous revascularization and drive novel discovery to improve soft tissue wound healing following tissue transfer for traumatic injury and cancer resection.


Asunto(s)
Colgajo Miocutáneo/irrigación sanguínea , Colgajo Miocutáneo/patología , Neovascularización Fisiológica/fisiología , Daño por Reperfusión/patología , Caracteres Sexuales , Cicatrización de Heridas/fisiología , Animales , Carnitina O-Palmitoiltransferasa/genética , Femenino , Factor 2 de Crecimiento de Fibroblastos/genética , Proteína Forkhead Box O1/genética , Hexoquinasa/genética , Factores de Transcripción de Tipo Kruppel/genética , Imágenes de Contraste de Punto Láser , Masculino , Ratones , Necrosis , Neovascularización Fisiológica/genética , Fosfofructoquinasa-2/genética , Receptor Notch1/genética , Daño por Reperfusión/genética , Daño por Reperfusión/metabolismo , Transcriptoma , Factor A de Crecimiento Endotelial Vascular/genética , Cicatrización de Heridas/genética
3.
J Immunol ; 200(2): 657-668, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29222165

RESUMEN

Numerous studies have reported sex bias in infectious diseases, with bias direction dependent on pathogen and site of infection. Staphylococcus aureus is the most common cause of skin and soft tissue infections (SSTIs), yet sex bias in susceptibility to S. aureus SSTI has not been described. A search of electronic health records revealed an odds ratio of 2.4 for S. aureus SSTI in males versus females. To investigate the physiological basis of this bias, we compared outcomes between male and female mice in a model of S. aureus dermonecrosis. Consistent with the epidemiological data, female mice were better protected against SSTI, with reduced dermonecrosis followed later by increased bacterial clearance. Protection in females was disrupted by ovariectomy and restored by short-term estrogen administration. Importantly, this sex bias was mediated by a sex-specific response to the S. aureus-secreted virulence factor α-hemolysin (Hla). Infection with wild-type S. aureus suppressed inflammatory cytokine production in the skin of female, but not male, mice when compared with infection with an isogenic hla deletion mutant. This differential response was conserved following injection with Hla alone, demonstrating a direct response to Hla independent of bacterial burden. Additionally, neutrophils, essential for clearing S. aureus, demonstrated sex-specific S. aureus bactericidal capacity ex vivo. This work suggests that sex-specific skin innate responsiveness to Hla and neutrophil bactericidal capacity play important roles in limiting S. aureus SSTI in females. Understanding the molecular mechanisms controlling this sex bias may reveal novel targets to promote host innate defense against S. aureus skin infection.


Asunto(s)
Toxinas Bacterianas/metabolismo , Proteínas Hemolisinas/metabolismo , Infecciones Cutáneas Estafilocócicas/microbiología , Staphylococcus aureus/patogenicidad , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Resistencia a la Enfermedad , Estrógenos/metabolismo , Femenino , Expresión Génica , Inmunidad Innata , Inflamasomas/metabolismo , Mediadores de Inflamación , Masculino , Ratones , Viabilidad Microbiana/inmunología , Neutrófilos/inmunología , Neutrófilos/metabolismo , Neutrófilos/microbiología , Factores Sexuales , Infecciones Cutáneas Estafilocócicas/genética , Infecciones Cutáneas Estafilocócicas/inmunología , Infecciones Cutáneas Estafilocócicas/metabolismo , Virulencia , Factores de Virulencia
4.
Am J Pathol ; 188(2): 515-524, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29169987

RESUMEN

Epidemiologic studies report improved breast cancer survival in women who receive ketorolac (Toradol) for postoperative pain relief compared with other analgesic agents. Ketorolac is a racemic drug. The S-enantiomer inhibits cyclooxygenases; R-ketorolac is a selective inhibitor of the small GTPases Ras-related C3 botulinum toxin substrate 1 (Rac1) and cell division control protein 42 (Cdc42), which are signaling molecules up-regulated during breast cancer progression and metastasis. The goal of this study was to determine whether R-ketorolac altered breast cancer development in the mouse mammary tumor virus-polyoma middle T-antigen model. Mice were administered ketorolac orally at 1 mg/kg twice daily to approximate the typical human dose. Mammary glands were analyzed for tumor number and immunohistochemical markers of proliferation and differentiation. R-ketorolac treatment significantly reduced mammary epithelial proliferation, based on Ki67 staining, and suppressed tumor development. Proliferative mammary epithelium from R-ketorolac-treated mice displayed greater differentiation, based on significantly higher total E-cadherin and decreased keratin 5 staining than epithelium of placebo-treated mice. No differences were detected in estrogen receptor, progesterone receptor, ß-catenin, or vimentin expression between placebo and R-ketorolac treatment groups. These findings indicate that R-ketorolac treatment slows tumor progression in an aggressive model of breast cancer. R-ketorolac may thus represent a novel therapeutic approach for breast cancer prevention or treatment based on its pharmacologic activity as a Rac1 and Cdc42 inhibitor.


Asunto(s)
Antineoplásicos/uso terapéutico , Ketorolaco Trometamina/uso terapéutico , Neoplasias Mamarias Animales/prevención & control , Administración Oral , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Progresión de la Enfermedad , Esquema de Medicación , Evaluación Preclínica de Medicamentos/métodos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Femenino , Ketorolaco Trometamina/administración & dosificación , Ketorolaco Trometamina/farmacología , Neoplasias Mamarias Animales/patología , Virus del Tumor Mamario del Ratón , Ratones Transgénicos , Poliomavirus
5.
Bioorg Med Chem Lett ; 24(3): 725-30, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24418777

RESUMEN

The purpose of this study was to examine whether the introduction of D-Phe could improve the GnRH receptor binding affinities of DOTA-conjugated D-Lys(6)-GnRH peptides. Building upon the construct of DOTA-Ahx-(D-Lys(6)-GnRH1) we previously reported, an aromatic amino acid of D-Phe was inserted either between the DOTA and Ahx or between the Ahx and D-Lys(6) to generate new DOTA-D-Phe-Ahx-(D-Lys(6)-GnRH) or DOTA-Ahx-D-Phe-(D-Lys(6)-GnRH) peptides. Compared to DOTA-Ahx-(D-Lys(6)-GnRH1) (36.1 nM), the introduction of D-Phe improved the GnRH receptor binding affinities of DOTA-D-Phe-Ahx-(D-Lys(6)-GnRH) (16.3 nM) and DOTA-Ahx-D-Phe-(D-Lys(6)-GnRH) (7.6 nM). The tumor targeting and pharmacokinetic properties of (111)In-DOTA-Ahx-D-Phe-(D-Lys(6)-GnRH) was determined in MDA-MB-231 human breast cancer-xenografted nude mice. Compared to (111)In-DOTA-Ahx-(D-Lys(6)-GnRH1), (111)In-DOTA-Ahx-D-Phe-(D-Lys(6)-GnRH) exhibited comparable tumor uptake with faster renal and liver clearance. The MDA-MB-231 human breast cancer-xenografted tumors were clearly visualized by single photon emission computed tomography (SPECT) using (111)In-DOTA-Ahx-D-Phe-(D-Lys(6)-GnRH) as an imaging probe, providing a new insight into the design of new GnRH peptides in the future.


Asunto(s)
Neoplasias de la Mama/diagnóstico , Hormona Liberadora de Gonadotropina/química , Hormona Liberadora de Gonadotropina/metabolismo , Fenilalanina/química , Fenilalanina/metabolismo , Receptores LHRH/metabolismo , Animales , Unión Competitiva , Cromatografía Líquida de Alta Presión , Femenino , Xenoinjertos , Humanos , Concentración 50 Inhibidora , Ratones , Ratones Desnudos , Estructura Molecular , Imagen Óptica , Péptidos/química , Péptidos/metabolismo , Unión Proteica , Tomografía Computarizada de Emisión de Fotón Único
6.
J Steroid Biochem Mol Biol ; 244: 106607, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39197539

RESUMEN

Long-term administration of exogenous estrogen is known to cause urinary retention and marked, often fatal, bladder distention in both male and female mice. Estrogen-treated mice have increased bladder pressure and decreased urine flow, suggesting that urinary retention in estrogen-treated mice is due to infravesicular obstruction to urine outflow. Thus, the condition is commonly referred to as bladder outlet obstruction (BOO). Obesity can also lead to urinary retention. As the effects of estrogen are mediated by multiple receptors, including estrogen receptors ERα and ERß and the G protein-coupled estrogen receptor (GPER), we sought to determine whether GPER plays a role in estrogen-induced BOO, particularly in the context of obesity. Wild type and GPER knockout (KO) mice fed a high-fat diet were ovariectomized or left ovary-intact (sham surgery) and supplemented with slow-release estrogen or vehicle-only pellets. Supplementing both GPER KO and wild type obese mice with estrogen for 8 weeks resulted in weight loss, splenic enlargement, and thymic atrophy, as expected. However, estrogen-treated obese GPER KO mice developed abdominal distension, debilitation, and ulceration of the skin surrounding the urogenital opening. At necropsy, these mice had prominently distended bladders and hydronephrosis. In contrast, estrogen-treated obese wild type mice only rarely displayed these signs. Our results suggest that, under conditions of obesity, estrogen induces BOO as a result of ERα-driven pathways and that GPER expression is protective against BOO.


Asunto(s)
Estrógenos , Ratones Noqueados , Obesidad , Receptores de Estrógenos , Receptores Acoplados a Proteínas G , Retención Urinaria , Animales , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores de Estrógenos/metabolismo , Receptores de Estrógenos/genética , Estrógenos/metabolismo , Femenino , Obesidad/metabolismo , Obesidad/complicaciones , Obesidad/genética , Ratones , Retención Urinaria/metabolismo , Retención Urinaria/genética , Ratones Endogámicos C57BL , Ratones Obesos , Dieta Alta en Grasa/efectos adversos , Ovariectomía , Masculino , Obstrucción del Cuello de la Vejiga Urinaria/metabolismo , Obstrucción del Cuello de la Vejiga Urinaria/patología , Obstrucción del Cuello de la Vejiga Urinaria/genética
7.
Biomedicines ; 12(2)2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38397942

RESUMEN

RNA binding proteins (RBPs) post-transcriptionally regulate gene expression by associating with regulatory sequences in the untranslated regions of mRNAs. Cold-inducible RBP (CIRP) is a stress-induced RBP that was recently shown to modulate inflammation in response to cellular stress, where it increases or decreases pro-tumorigenic (proinflammatory) cytokines in different contexts. CIRP expression is altered in several cancers, including breast cancer, but the effects of CIRP on inflammation in breast cancer is not known. Here, we investigate if CIRP alters growth and the inflammatory profile of breast tumors. Transgenic mice overexpressing CIRP in the mammary epithelium were crossed with the PyMT mouse model of breast cancer, and the effects on both early and late tumorigenesis and inflammation were assessed. The effects of CIRP knockdown were also assessed in Py2T cell grafts. Overexpression of CIRP led to decreased tumorigenesis in the PyMT mouse model. Conversely, the knockdown of CIRP in Py2T cell grafts led to increased tumor growth. Luminex cytokine assays assessed the effects on the inflammatory environment. CIRP/PyMT mammary glands/mammary tumors and serum had decreased cytokines that promote inflammation, angiogenesis, and metastasis compared to PyMT mammary glands and serum, documenting a shift towards an environment less supportive of tumorigenesis. CIRP overexpression also decreased CD4+ helper T cells and increased CD8+ cytotoxic T cells in mammary tumors. Overall, these data support a role for CIRP as a potent antitumor molecule that suppresses both local and systemic pro-tumorigenic inflammation.

8.
Sci Rep ; 14(1): 18400, 2024 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-39117675

RESUMEN

Estrogens regulate numerous physiological and pathological processes, including wide-ranging effects in wound healing. The effects of estrogens are mediated through multiple estrogen receptors (ERs), including the classical nuclear ERs (ERα and ER ß ), that typically regulate gene expression, and the 7-transmembrane G protein-coupled estrogen receptor (GPER), that predominantly mediates rapid "non-genomic" signaling. Estrogen modulates the expression of various genes involved in epidermal function and regeneration, inflammation, matrix production, and protease inhibition, all critical to wound healing. Our previous work demonstrated improved myocutaneous wound healing in female mice compared to male mice. In the current study, we employed male and female GPER knockout mice to investigate the role of this estrogen receptor in wound revascularization and tissue viability. Using a murine myocutaneous flap model of graded ischemia, we measured real-time flap perfusion via laser speckle perfusion imaging. We conducted histologic and immunohistochemical analyses to assess skin and muscle viability, microvascular density and vessel morphology. Our results demonstrate that GPER is crucial in wound healing, mediating effects that are both dependent and independent of sex. Lack of GPER expression is associated with increased skin necrosis, reduced flap perfusion and altered vessel morphology. These findings contribute to understanding GPER signaling in wound healing and suggest possible therapeutic opportunities by targeting GPER.


Asunto(s)
Ratones Noqueados , Neovascularización Fisiológica , Receptores de Estrógenos , Receptores Acoplados a Proteínas G , Cicatrización de Heridas , Animales , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores de Estrógenos/metabolismo , Receptores de Estrógenos/genética , Masculino , Ratones , Femenino , Piel/metabolismo , Piel/irrigación sanguínea , Isquemia/metabolismo , Colgajos Quirúrgicos
9.
Bioorg Med Chem Lett ; 23(20): 5484-7, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24018188

RESUMEN

Three new DOTA-conjugated GnRH peptides with various hydrocarbon linkers were synthesized to evaluate the influences of the linkers on their receptor binding affinities. The hydrocarbon linker displayed a profound impact on the receptor binding affinities of DOTA-conjugated GnRH peptides. The Aun linker was better than Gaba, Ahx and Aoc linkers in retaining strong receptor binding affinity of the GnRH peptide. DOTA-Aun-(D-Lys(6)-GnRH) displayed 22.8 nM GnRH receptor binding affinity. (111)In-DOTA-Aun-(D-Lys(6)-GnRH) exhibited fast tumor uptake and urinary clearance in MDA-MB-231 human breast cancer-xenografted nude mice. The cellular and biological results provided an insight into the design of new GnRH peptides in the future.


Asunto(s)
Hormona Liberadora de Gonadotropina/metabolismo , Hidrocarburos/química , Péptidos/metabolismo , Aminocaproatos/química , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Caprilatos/química , Línea Celular Tumoral , Ácidos Grasos/química , Femenino , Hormona Liberadora de Gonadotropina/química , Compuestos Heterocíclicos con 1 Anillo/química , Humanos , Ratones , Ratones Desnudos , Péptidos/síntesis química , Péptidos/orina , Unión Proteica , Receptores LHRH/metabolismo , Distribución Tisular , Trasplante Heterólogo , Ácido gamma-Aminobutírico/química
10.
Breast Cancer Res ; 13(5): R108, 2011 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-22035507

RESUMEN

INTRODUCTION: Breast cancer detection using mammography has improved clinical outcomes for many women, because mammography can detect very small (5 mm) tumors early in the course of the disease. However, mammography fails to detect 10 - 25% of tumors, and the results do not distinguish benign and malignant tumors. Reducing the false positive rate, even by a modest 10%, while improving the sensitivity, will lead to improved screening, and is a desirable and attainable goal. The emerging application of magnetic relaxometry, in particular using superconducting quantum interference device (SQUID) sensors, is fast and potentially more specific than mammography because it is designed to detect tumor-targeted iron oxide magnetic nanoparticles. Furthermore, magnetic relaxometry is theoretically more specific than MRI detection, because only target-bound nanoparticles are detected. Our group is developing antibody-conjugated magnetic nanoparticles targeted to breast cancer cells that can be detected using magnetic relaxometry. METHODS: To accomplish this, we identified a series of breast cancer cell lines expressing varying levels of the plasma membrane-expressed human epidermal growth factor-like receptor 2 (Her2) by flow cytometry. Anti-Her2 antibody was then conjugated to superparamagnetic iron oxide nanoparticles using the carbodiimide method. Labeled nanoparticles were incubated with breast cancer cell lines and visualized by confocal microscopy, Prussian blue histochemistry, and magnetic relaxometry. RESULTS: We demonstrated a time- and antigen concentration-dependent increase in the number of antibody-conjugated nanoparticles bound to cells. Next, anti Her2-conjugated nanoparticles injected into highly Her2-expressing tumor xenograft explants yielded a significantly higher SQUID relaxometry signal relative to unconjugated nanoparticles. Finally, labeled cells introduced into breast phantoms were measured by magnetic relaxometry, and as few as 1 million labeled cells were detected at a distance of 4.5 cm using our early prototype system. CONCLUSIONS: These results suggest that the antibody-conjugated magnetic nanoparticles are promising reagents to apply to in vivo breast tumor cell detection, and that SQUID-detected magnetic relaxometry is a viable, rapid, and highly sensitive method for in vitro nanoparticle development and eventual in vivo tumor detection.


Asunto(s)
Neoplasias de la Mama/diagnóstico , Espectroscopía de Resonancia Magnética/métodos , Nanopartículas de Magnetita , Animales , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Membrana Celular/inmunología , Membrana Celular/metabolismo , Femenino , Compuestos Férricos , Humanos , Inmunoconjugados , Ratones , Ratones Desnudos , Fantasmas de Imagen , Receptor ErbB-2/inmunología , Receptor ErbB-2/metabolismo , Sensibilidad y Especificidad , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Breast Cancer Res Treat ; 127(2): 397-406, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20607601

RESUMEN

The behavior of breast epithelial cells is influenced by their microenvironment which includes stromal cells and extracellular matrix (ECM). During cancer progression, the tissue microenvironment fails to control proliferation and differentiation, resulting in uncontrolled growth and invasion. Upon invasion, the ECM encountered by breast cancer cells changes from primarily laminin and collagen IV to primarily collagen I. We show here that culturing invasive breast cancer cells in 3-dimensional (3D) collagen I inhibits proliferation through direct regulation of cyclin E1, a G(1)/S regulator that is overexpressed in breast cancer. When the breast cancer cell line MDA-MB-231 was cultured within 3D collagen I gels, the G(1)/S transition was inhibited as compared to cells cultured on conventional 2D collagen or plastic dishes. Cells in 3D collagen downregulated cyclin E1 protein and mRNA, with no change in cyclin D1 level. Cyclin D1 was primarily cytoplasmic in 3D cultures, and this was accompanied by decreased phosphorylation of Rb, a nuclear target for both cyclin E1- and cyclin D1-associated kinases. Positive regulators of cyclin E1 expression, the transcription factor c-Myc and cold-inducible RNA binding protein (CIRP), were decreased in 3D collagen cultures, while the collagen I receptor ß1 integrin was greatly increased. Inhibition of ß1 integrin function rescued proliferation and cyclin E1 expression as well as c-Myc expression and Rb phosphorylation, but cyclin D1 remained cytoplasmic. We conclude that cyclin E1 is repressed independent of effects on cyclin D1 in a 3D collagen environment and dependent on ß1 integrin interaction with collagen I, reducing proliferation of invasive breast cancer cells.


Asunto(s)
Neoplasias de la Mama/patología , Colágeno/farmacología , Ciclina E/metabolismo , Integrina beta1/metabolismo , Proteínas Oncogénicas/metabolismo , Neoplasias de la Mama/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Colágeno/metabolismo , Ciclina D1/metabolismo , Citoplasma/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Femenino , Humanos , Fosforilación/efectos de los fármacos , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteína de Retinoblastoma/metabolismo
12.
Bioconjug Chem ; 22(8): 1682-9, 2011 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-21749045

RESUMEN

The purpose of this study was to develop novel radiolabeled gonadotropin-releasing hormone (GnRH) receptor-targeting peptides for breast cancer imaging. Three novel 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA)-conjugated GnRH peptides were designed and synthesized. The radiometal chelator DOTA was conjugated to the epsilon or alpha amino group of D-lysine, or the epsilon amino group of L-lysine via an Ahx {aminohexanoic acid} linker to generate DOTA-Ahx-(D-Lys(6)-GnRH1), DOTA-Ahx-(D-Lys(6)-GnRH2) and DOTA-Ahx-(L-Lys(6)-GnRH3), respectively. The conjugation of the DOTA to the epsilon amino group of D-lysine (rather than alpha amino group of D-lysine nor epsilon amino group of L-lysine) maintained the nanomolar GnRH receptor binding affinity. The IC(50) values of DOTA-Ahx-(D-Lys(6)-GnRH1), DOTA-Ahx-(D-Lys(6)-GnRH2) and DOTA-Ahx-(L-Lys(6)-GnRH3) were 36.1 nM, 10.6 mM and 4.3 mM, respectively. Since only DOTA-Ahx-(D-Lys(6)-GnRH1) displayed nanomolar receptor binding affinity, the specific GnRH receptor binding of (111)In-DOTA-Ahx-(D-Lys(6)-GnRH1) was determined in human GnRH receptor membrane preparations. Furthermore, the biodistribution and tumor imaging properties of (111)In-DOTA-Ahx-(D-Lys(6)-GnRH1) were examined in MDA-MB-231 human breast cancer-xenografted nude mice. (111)In-DOTA-Ahx-(D-Lys(6)-GnRH1) exhibited specific GnRH receptor binding and rapid tumor uptake (1.76 ± 0.58% ID/g at 0.5 h postinjection) coupled with fast whole-body clearance through the urinary system. The MDA-MB-231 human breast cancer-xenografted tumor lesions were clearly visualized by single photon emission computed tomography (SPECT)/CT at 1 h postinjection of (111)In-DOTA-Ahx-(D-Lys(6)-GnRH1). The profound impact of DOTA position on the binding affinity of the GnRH peptide provided a new insight into the design of novel radiolabeled GnRH peptides. The successful imaging of MDA-MB-231 human breast cancer-xenografted tumor lesions using (111)In-DOTA-Ahx-(D-Lys(6)-GnRH1) suggested its potential as a novel imaging probe for human breast cancer imaging.


Asunto(s)
Neoplasias de la Mama/diagnóstico por imagen , Péptidos , Receptores LHRH/metabolismo , Animales , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/patología , Diagnóstico por Imagen/métodos , Femenino , Humanos , Radioisótopos de Indio , Ratones , Ratones Desnudos , Péptidos/síntesis química , Péptidos/química , Radiofármacos/síntesis química , Distribución Tisular , Tomografía Computarizada de Emisión de Fotón Único/métodos , Trasplante Heterólogo
13.
Nat Chem Biol ; 5(6): 421-7, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19430488

RESUMEN

Estrogen is central to many physiological processes throughout the human body. We have previously shown that the G protein-coupled receptor GPR30 (also known as GPER), in addition to classical nuclear estrogen receptors (ER and ER), activates cellular signaling pathways in response to estrogen. In order to distinguish between the actions of classical estrogen receptors and GPR30, we have previously characterized G-1 (1), a selective agonist of GPR30. To complement the pharmacological properties of G-1, we sought to identify an antagonist of GPR30 that displays similar selectivity against the classical estrogen receptors. Here we describe the identification and characterization of G15 (2), a G-1 analog that binds to GPR30 with high affinity and acts as an antagonist of estrogen signaling through GPR30. In vivo administration of G15 revealed that GPR30 contributes to both uterine and neurological responses initiated by estrogen. The identification of this antagonist will accelerate the evaluation of the roles of GPR30 in human physiology.


Asunto(s)
Receptores de Estrógenos/fisiología , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Animales , Células COS , Chlorocebus aethiops , Estrógenos/metabolismo , Femenino , Humanos , Ligandos , Masculino , Ratones , Ratones Endogámicos ICR , Resonancia Magnética Nuclear Biomolecular , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/fisiología , Transducción de Señal
14.
Sci Transl Med ; 12(528)2020 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-31996464

RESUMEN

Human obesity has become a global health epidemic, with few safe and effective pharmacological therapies currently available. The systemic loss of ovarian estradiol (E2) in women after menopause greatly increases the risk of obesity and metabolic dysfunction, revealing the critical role of E2 in this setting. The salutary effects of E2 are traditionally attributed to the classical estrogen receptors ERα and ERß, with the contribution of the G protein-coupled estrogen receptor (GPER) still largely unknown. Here, we used ovariectomy- and diet-induced obesity (DIO) mouse models to evaluate the preclinical activity of GPER-selective small-molecule agonist G-1 (also called Tespria) against obesity and metabolic dysfunction. G-1 treatment of ovariectomized female mice (a model of postmenopausal obesity) reduced body weight and improved glucose homeostasis without changes in food intake, fuel source usage, or locomotor activity. G-1-treated female mice also exhibited increased energy expenditure, lower body fat content, and reduced fasting cholesterol, glucose, insulin, and inflammatory markers but did not display feminizing effects on the uterus (imbibition) or beneficial effects on bone health. G-1 treatment of DIO male mice did not elicit weight loss but prevented further weight gain and improved glucose tolerance, indicating that G-1 improved glucose homeostasis independently of its antiobesity effects. However, in ovariectomized DIO female mice, G-1 continued to elicit weight loss, reflecting possible sex differences in the mechanisms of G-1 action. In conclusion, this work demonstrates that GPER-selective agonism is a viable therapeutic approach against obesity, diabetes, and associated metabolic abnormalities in multiple preclinical male and female models.


Asunto(s)
Diabetes Mellitus/tratamiento farmacológico , Obesidad/tratamiento farmacológico , Receptores Acoplados a Proteínas G/agonistas , Tejido Adiposo/patología , Adiposidad/efectos de los fármacos , Animales , Respiración de la Célula , Modelos Animales de Enfermedad , Metabolismo Energético , Estrógenos/deficiencia , Femenino , Genes Mitocondriales , Glucosa/metabolismo , Homeostasis , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/genética , Obesidad/complicaciones , Ovariectomía , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Resultado del Tratamiento , Regulación hacia Arriba , Aumento de Peso
15.
Cell Rep ; 30(6): 1798-1810.e4, 2020 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-32049011

RESUMEN

The reliance of many cancers on aerobic glycolysis has stimulated efforts to develop lactate dehydrogenase (LDH) inhibitors. However, despite significant efforts, LDH inhibitors (LDHi) with sufficient specificity and in vivo activity to determine whether LDH is a feasible drug target are lacking. We describe an LDHi with potent, on-target, in vivo activity. Using hyperpolarized magnetic resonance spectroscopic imaging (HP-MRSI), we demonstrate in vivo LDH inhibition in two glycolytic cancer models, MIA PaCa-2 and HT29, and we correlate depth and duration of LDH inhibition with direct anti-tumor activity. HP-MRSI also reveals a metabolic rewiring that occurs in vivo within 30 min of LDH inhibition, wherein pyruvate in a tumor is redirected toward mitochondrial metabolism. Using HP-MRSI, we show that inhibition of mitochondrial complex 1 rapidly redirects tumor pyruvate toward lactate. Inhibition of both mitochondrial complex 1 and LDH suppresses metabolic plasticity, causing metabolic quiescence in vitro and tumor growth inhibition in vivo.


Asunto(s)
Quimioterapia Combinada/métodos , L-Lactato Deshidrogenasa/antagonistas & inhibidores , Neoplasias/inmunología , Animales , Humanos , Ratones , Neoplasias/tratamiento farmacológico
16.
J Med Chem ; 63(19): 10984-11011, 2020 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-32902275

RESUMEN

Lactate dehydrogenase (LDH) catalyzes the conversion of pyruvate to lactate, with concomitant oxidation of reduced nicotinamide adenine dinucleotide as the final step in the glycolytic pathway. Glycolysis plays an important role in the metabolic plasticity of cancer cells and has long been recognized as a potential therapeutic target. Thus, potent, selective inhibitors of LDH represent an attractive therapeutic approach. However, to date, pharmacological agents have failed to achieve significant target engagement in vivo, possibly because the protein is present in cells at very high concentrations. We report herein a lead optimization campaign focused on a pyrazole-based series of compounds, using structure-based design concepts, coupled with optimization of cellular potency, in vitro drug-target residence times, and in vivo PK properties, to identify first-in-class inhibitors that demonstrate LDH inhibition in vivo. The lead compounds, named NCATS-SM1440 (43) and NCATS-SM1441 (52), possess desirable attributes for further studying the effect of in vivo LDH inhibition.


Asunto(s)
Inhibidores Enzimáticos/farmacología , L-Lactato Deshidrogenasa/antagonistas & inhibidores , Pirazoles/farmacología , Animales , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacocinética , Semivida , Humanos , Ratones , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Gynecol Oncol ; 114(3): 465-71, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19501895

RESUMEN

OBJECTIVES: GPR30 is a 7-transmembrane G protein-coupled estrogen receptor that functions alongside traditional estrogen receptors to regulate cellular responses to estrogen. Recent studies suggest that GPR30 expression is linked to lower survival rates in endometrial and breast cancer. This study was conducted to evaluate GPR30 expression in ovarian tumors. METHODS: GPR30 expression was analyzed using immunohistochemistry and archival specimens from 45 patients with ovarian tumors of low malignant potential (LMP) and 89 patients with epithelial ovarian cancer (EOC). Expression, defined as above or below the median (intensity times the percentage of positive epithelial cells) was correlated with predictors of adverse outcome and survival. RESULTS: GPR30 expression above the median was observed more frequently in EOC than in LMP tumors (48.3% vs. 20%, p=0.002), and in EOC was associated with lower 5-year survival rates (44.2% vs. 82.6%, Log-rank p<0.001). Tumor grade and FIGO stage, the other significant predictors of survival, were used to stratify cases into "high risk" and "low risk" groups. The 5-year survival rate for "low risk" EOC (all grade 1 and Stage I/II, grade 2) was 100%. In "high risk" EOC (all grade 3 and Stage III/IV, grade 2), the difference in 5-year survival by GPR 30 expression was significant (33.3% vs. 72.4%, p=0.001). CONCLUSIONS: The novel estrogen-responsive receptor GPR30 is preferentially expressed in "high risk" EOC and is associated with lower survival rates. Further investigation of GPR30 as a potential target for therapeutic intervention in high risk EOC is warranted.


Asunto(s)
Biomarcadores de Tumor/biosíntesis , Neoplasias Ováricas/metabolismo , Receptores Acoplados a Proteínas G/biosíntesis , Células Epiteliales/patología , Femenino , Humanos , Inmunohistoquímica , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias Ováricas/patología , Pronóstico , Receptores de Estrógenos , Tasa de Supervivencia
18.
J Magn Magn Mater ; 321(10): 1459-1464, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-20161153

RESUMEN

Magnetite nanoparticles (Chemicell SiMAG-TCL) were characterized by SQUID-relaxometry, susceptometry, and TEM. The magnetization detected by SQUID-relaxometry was 0.33% of that detected by susceptometry, indicating that the sensitivity of SQUID-relaxometry could be significantly increased through improved control of nanoparticle size. The relaxometry data were analyzed by the moment superposition model (MSM) to determine the distribution of nanoparticle moments. Analysis of the binding of CD34-conjugated nanoparticles to U937 leukemia cells revealed 60,000 nanoparticles per cell, which were collected from whole blood using a prototype magnetic biopsy needle, with a capture efficiency of >65% from a 750 µl sample volume in 1 minute.

19.
Sci Rep ; 9(1): 1343, 2019 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-30718654

RESUMEN

Sex bias in innate defense against Staphylococcus aureus skin and soft tissue infection (SSTI) is dependent on both estrogen production by the host and S. aureus secretion of the virulence factor, α-hemolysin (Hla). The impact of estrogen signaling on the immune system is most often studied in terms of the nuclear estrogen receptors ERα and ERß. However, the potential contribution of the G protein-coupled estrogen receptor (GPER) to innate defense against infectious disease, particularly with respect to skin infection, has not been addressed. Using a murine model of SSTI, we found that GPER activation with the highly selective agonist G-1 limits S. aureus SSTI and Hla-mediated pathogenesis, effects that were absent in GPER knockout mice. Specifically, G-1 reduced Hla-mediated skin lesion formation and pro-inflammatory cytokine production, while increasing bacterial clearance. In vitro, G-1 reduced surface expression of the Hla receptor, ADAM10, in a human keratinocyte cell line and increased resistance to Hla-mediated permeability barrier disruption. This novel role for GPER activation in skin innate defense against infectious disease suggests that G-1 may have clinical utility in patients with epithelial permeability barrier dysfunction or who are otherwise at increased risk of S. aureus infection, including those with atopic dermatitis or cancer.


Asunto(s)
Toxinas Bacterianas/genética , Receptor alfa de Estrógeno/genética , Receptor beta de Estrógeno/genética , Proteínas Hemolisinas/genética , Receptores de Estrógenos/genética , Receptores Acoplados a Proteínas G/genética , Infecciones Estafilocócicas/genética , Proteína ADAM10/genética , Animales , Toxinas Bacterianas/metabolismo , Células Epiteliales/microbiología , Células Epiteliales/patología , Proteínas Hemolisinas/metabolismo , Interacciones Huésped-Patógeno/genética , Humanos , Inmunidad Innata/genética , Queratinocitos/microbiología , Ratones , Ratones Noqueados , Transducción de Señal/genética , Piel/inmunología , Piel/microbiología , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/patología , Staphylococcus aureus/genética , Staphylococcus aureus/patogenicidad
20.
Cell Chem Biol ; 26(12): 1692-1702.e5, 2019 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-31706983

RESUMEN

Estrogen exerts extensive and diverse effects throughout the body of women. In addition to the classical nuclear estrogen receptors (ERα and ERß), the G protein-coupled estrogen receptor GPER is an important mediator of estrogen action. Existing ER-targeted therapeutic agents act as GPER agonists. Here, we report the identification of a small molecule, named AB-1, with the previously unidentified activity of high selectivity for binding classical ERs over GPER. AB-1 also possesses a unique functional activity profile as an agonist of transcriptional activity but an antagonist of rapid signaling through ERα. Our results define a class of small molecules that discriminate between the classical ERs and GPER, as well as between modes of signaling within the classical ERs. Such an activity profile, if developed into an ER antagonist, could represent an opportunity for the development of first-in-class nuclear hormone receptor-targeted therapeutics for breast cancer exhibiting reduced acquired and de novo resistance.


Asunto(s)
Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Ligandos , Transducción de Señal , Animales , Proliferación Celular/efectos de los fármacos , Estradiol/farmacología , Receptor alfa de Estrógeno/antagonistas & inhibidores , Receptor beta de Estrógeno/antagonistas & inhibidores , Femenino , Proteína Forkhead Box O3/genética , Proteína Forkhead Box O3/metabolismo , Humanos , Células MCF-7 , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Útero/efectos de los fármacos , Útero/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA