Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(7): e2212940120, 2023 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-36749725

RESUMEN

Missense mutations that inactivate p53 occur commonly in cancer, and germline mutations in TP53 cause Li Fraumeni syndrome, which is associated with early-onset cancer. In addition, there are over two hundred germline missense variants of p53 that remain uncharacterized. In some cases, these germline variants have been shown to encode lesser-functioning, or hypomorphic, p53 protein, and these alleles are associated with increased cancer risk in humans and mouse models. However, most hypomorphic p53 variants remain un- or mis-classified in clinical genetics databases. There thus exists a significant need to better understand the behavior of p53 hypomorphs and to develop a functional assay that can distinguish hypomorphs from wild-type p53 or benign variants. We report the surprising finding that two different African-centric genetic hypomorphs of p53 that occur in distinct functional domains of the protein share common activities. Specifically, the Pro47Ser variant, located in the transactivation domain, and the Tyr107His variant, located in the DNA binding domain, both share increased propensity to misfold into a conformation specific for mutant, misfolded p53. Additionally, cells and tissues containing these hypomorphic variants show increased NF-κB activity. We identify a common gene expression signature from unstressed lymphocyte cell lines that is shared between multiple germline hypomorphic variants of TP53, and which successfully distinguishes wild-type p53 and a benign variant from lesser-functioning hypomorphic p53 variants. Our findings will allow us to better understand the contribution of p53 hypomorphs to disease risk and should help better inform cancer risk in the carriers of p53 variants.


Asunto(s)
Síndrome de Li-Fraumeni , Proteína p53 Supresora de Tumor , Animales , Ratones , Humanos , Proteína p53 Supresora de Tumor/metabolismo , Predisposición Genética a la Enfermedad , Síndrome de Li-Fraumeni/genética , Genes p53 , Heterocigoto , Mutación de Línea Germinal
2.
Genes Dev ; 30(8): 918-30, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-27034505

RESUMEN

A nonsynonymous single-nucleotide polymorphism at codon 47 in TP53 exists in African-descent populations (P47S, rs1800371; referred to here as S47). Here we report that, in human cell lines and a mouse model, the S47 variant exhibits a modest decrease in apoptosis in response to most genotoxic stresses compared with wild-type p53 but exhibits a significant defect in cell death induced by cisplatin. We show that, compared with wild-type p53, S47 has nearly indistinguishable transcriptional function but shows impaired ability to transactivate a subset of p53 target genes, including two involved in metabolism:Gls2(glutaminase 2) and Sco2 We also show that human and mouse cells expressing the S47 variant are markedly resistant to cell death by agents that induce ferroptosis (iron-mediated nonapoptotic cell death). We show that mice expressing S47 in homozygous or heterozygous form are susceptible to spontaneous cancers of diverse histological types. Our data suggest that the S47 variant may contribute to increased cancer risk in individuals of African descent, and our findings highlight the need to assess the contribution of this variant to cancer risk in these populations. These data also confirm the potential relevance of metabolism and ferroptosis to tumor suppression by p53.


Asunto(s)
Genes p53/genética , Polimorfismo de Nucleótido Simple , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Animales , Población Negra/genética , Carcinoma Hepatocelular/genética , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Línea Celular , Cisplatino/farmacología , Codón/química , Codón/genética , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos C57BL , Neoplasias/genética , Unión Proteica/genética , Factores de Riesgo , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/genética
3.
Genes Dev ; 25(6): 634-45, 2011 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-21406557

RESUMEN

The cJun NH(2)-terminal kinase (JNK) signal transduction pathway has been implicated in the growth of carcinogen-induced hepatocellular carcinoma. However, the mechanism that accounts for JNK-regulated tumor growth is unclear. Here we demonstrate that compound deficiency of the two ubiquitously expressed JNK isoforms (JNK1 and JNK2) in hepatocytes does not prevent hepatocellular carcinoma development. Indeed, JNK deficiency in hepatocytes increased the tumor burden. In contrast, compound JNK deficiency in hepatocytes and nonparenchymal cells reduced both hepatic inflammation and tumorigenesis. These data indicate that JNK plays a dual role in the development of hepatocellular carcinoma. JNK promotes an inflammatory hepatic environment that supports tumor development, but also functions in hepatocytes to reduce tumor development.


Asunto(s)
Carcinoma Hepatocelular/fisiopatología , Hepatocitos , Neoplasias Hepáticas/fisiopatología , Hígado/enzimología , Proteína Quinasa 8 Activada por Mitógenos , Animales , Proliferación Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Citocinas/metabolismo , Hepatocitos/enzimología , Hepatocitos/patología , Interleucina-6/metabolismo , Hígado/metabolismo , Hígado/patología , Regeneración Hepática/genética , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa 8 Activada por Mitógenos/deficiencia , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Proteína Quinasa 9 Activada por Mitógenos/deficiencia , Proteína Quinasa 9 Activada por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Transducción de Señal , Células Tumorales Cultivadas
4.
J Biol Chem ; 291(48): 25247-25254, 2016 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-27754870

RESUMEN

Protein homeostasis, or proteostasis, is required for mitochondrial function, but its role in cancer is controversial. Here we show that transgenic mice expressing the mitochondrial chaperone TNFR-associated protein 1 (TRAP1) in the prostate develop epithelial hyperplasia and cellular atypia. When examined on a Pten+/- background, a common alteration in human prostate cancer, TRAP1 transgenic mice showed accelerated incidence of invasive prostatic adenocarcinoma, characterized by increased cell proliferation and reduced apoptosis, in situ Conversely, homozygous deletion of TRAP1 delays prostatic tumorigenesis in Pten+/- mice without affecting hyperplasia or prostatic intraepithelial neoplasia. Global profiling of Pten+/--TRAP1 transgenic mice by RNA sequencing and reverse phase protein array reveals modulation of oncogenic networks of cell proliferation, apoptosis, cell motility, and DNA damage. Mechanistically, reconstitution of Pten+/- prostatic epithelial cells with TRAP1 increases cell proliferation, reduces apoptosis, and promotes cell invasion without changes in mitochondrial bioenergetics. Therefore, TRAP1 is a driver of prostate cancer in vivo and an "actionable" therapeutic target.


Asunto(s)
Apoptosis , Proliferación Celular , Proteínas HSP90 de Choque Térmico/metabolismo , Proteínas de Neoplasias/metabolismo , Próstata/metabolismo , Neoplasias de la Próstata/metabolismo , Animales , Línea Celular Tumoral , Proteínas HSP90 de Choque Térmico/genética , Humanos , Masculino , Ratones , Ratones Transgénicos , Invasividad Neoplásica , Proteínas de Neoplasias/genética , Próstata/patología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología
5.
J Immunol ; 192(7): 3043-56, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24591372

RESUMEN

Although therapies targeting distinct cellular pathways (e.g., anticytokine versus anti-B cell therapy) have been found to be an effective strategy for at least some patients with inflammatory arthritis, the mechanisms that determine which pathways promote arthritis development are poorly understood. We have used a transgenic mouse model to examine how variations in the CD4(+) T cell response to a surrogate self-peptide can affect the cellular pathways that are required for arthritis development. CD4(+) T cells that are highly reactive with the self-peptide induce inflammatory arthritis that affects male and female mice equally. Arthritis develops by a B cell-independent mechanism, although it can be suppressed by an anti-TNF treatment, which prevented the accumulation of effector CD4(+) Th17 cells in the joints of treated mice. By contrast, arthritis develops with a significant female bias in the context of a more weakly autoreactive CD4(+) T cell response, and B cells play a prominent role in disease pathogenesis. In this setting of lower CD4(+) T cell autoreactivity, B cells promote the formation of autoreactive CD4(+) effector T cells (including Th17 cells), and IL-17 is required for arthritis development. These studies show that the degree of CD4(+) T cell reactivity for a self-peptide can play a prominent role in determining whether distinct cellular pathways can be targeted to prevent the development of inflammatory arthritis.


Asunto(s)
Artritis/inmunología , Autoinmunidad/inmunología , Linfocitos T CD4-Positivos/inmunología , Transducción de Señal/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/farmacología , Artritis/genética , Artritis/prevención & control , Autoinmunidad/genética , Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas , Femenino , Citometría de Flujo , Miembro Anterior/inmunología , Miembro Anterior/metabolismo , Miembro Anterior/patología , Factores de Transcripción Forkhead/inmunología , Factores de Transcripción Forkhead/metabolismo , Miembro Posterior/inmunología , Miembro Posterior/metabolismo , Miembro Posterior/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Factores Sexuales , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
6.
J Immunol ; 190(12): 6115-25, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23667113

RESUMEN

We examined the formation, participation, and functional specialization of virus-reactive Foxp3(+) regulatory T cells (Tregs) in a mouse model of influenza virus infection. "Natural" Tregs generated intrathymically, based on interactions with a self-peptide, proliferated in response to a homologous viral Ag in the lungs and, to a lesser extent, in the lung-draining mediastinal lymph nodes (medLNs) of virus-infected mice. In contrast, conventional CD4(+) T cells with identical TCR specificity underwent little or no conversion to become "adaptive" Tregs. The virus-reactive Tregs in the medLNs and the lungs of infected mice upregulated a variety of molecules associated with Treg activation, as well as acquired expression of molecules (T-bet, Blimp-1, and IL-10) that confer functional specialization to Tregs. Notably, however, the phenotypes of the T-bet(+) Tregs obtained from these sites were distinct, because Tregs isolated from the lungs expressed significantly higher levels of T-bet, Blimp-1, and IL-10 than did Tregs from the medLNs. Adoptive transfer of Ag-reactive Tregs led to decreased proliferation of antiviral CD4(+) and CD8(+) effector T cells in the lungs of infected hosts, whereas depletion of Tregs had a reciprocal effect. These studies demonstrate that thymically generated Tregs can become activated by a pathogen-derived peptide and acquire discrete T-bet(+) Treg phenotypes while participating in and modulating an antiviral immune response.


Asunto(s)
Activación de Linfocitos/inmunología , Infecciones por Orthomyxoviridae/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Animales , Antígenos Virales/inmunología , Diferenciación Celular/inmunología , Citometría de Flujo , Factores de Transcripción Forkhead/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Orthomyxoviridae/inmunología , Fenotipo , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Subgrupos de Linfocitos T/citología , Linfocitos T Reguladores/citología
7.
J Cardiol ; 83(3): 184-190, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37684005

RESUMEN

BACKGROUND: As the catheter-based device closure of the patent foramen ovale (PFO) is expanding, novel devices aim to address the limitations of first-generation occluders (e.g. bulk, erosion, dislodgment). The second-generation device from Encore Medical (Eagan, MN, USA) features an articulating frame structure which allows the device to better conform to atrial anatomies, has lower disc thickness and metal mass/surface area, and is fully retrievable at any point in the procedure. The aim of the study was to evaluate the feasibility and safety of a novel low-profile, fully retrievable, Encore PFO closure device in the animal model. METHODS: Six swine underwent implantation of the novel PFO occluder under fluoroscopic and intra-cardiac echocardiography guidance and survived for 140 days. Interim transthoracic echocardiography (TTE) was conducted on Day 29. Following terminal angiography and TTE at 140 days, the hearts were subjected to gross and histopathologic analysis. RESULTS: All animals were successfully implanted and survived for 140 days. Interim TTE revealed proper device retention with no blood flow across the septum or thrombus in any of the animals. X-ray and pathology results showed preserved implant integrity with no fractures, and complete integration of the devices into the septum with complete re-endothelialization and nearly complete coverage by a mature, relatively thin neoendocardium. No surface fibrin deposition or thrombosis was reported. CONCLUSIONS: In the standard porcine model, device retention and biocompatibility remained favorable following structural and functional device modifications exemplified by the second-generation PFO occluder from Encore Medical, including marked reduction of metal mass.


Asunto(s)
Foramen Oval Permeable , Dispositivo Oclusor Septal , Porcinos , Animales , Resultado del Tratamiento , Cateterismo Cardíaco/métodos , Ecocardiografía , Foramen Oval Permeable/diagnóstico por imagen , Foramen Oval Permeable/cirugía , Fluoroscopía
8.
J Cell Physiol ; 228(7): 1601-9, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23359252

RESUMEN

This study was carried out to dissect the mechanism by which ß1 integrins promote resistance to radiation. For this purpose, we conditionally ablated ß1 integrins in the prostatic epithelium of transgenic adenocarcinoma of mouse prostate (TRAMP) mice. The ability of ß1 to promote resistance to radiation was also analyzed by using an inhibitory antibody to ß1 , AIIB2, in a xenograft model. The role of ß1 integrins and of a ß1 downstream target, c-Jun amino-terminal kinase 1 (JNK1), in regulating radiation-induced apoptosis in vivo and in vitro was studied. We show that ß1 integrins promote prostate cancer (PrCa) progression and resistance to radiation in vivo. Mechanistically, ß1 integrins are shown here to suppress activation of JNK1 and, consequently apoptosis, in response to irradiation. Downregulation of JNK1 is necessary to preserve the effect of ß1 on resistance to radiation in vitro and in vivo. Finally, given the established crosstalk between ß1 integrins and type1 insulin-like growth factor receptor (IGF-IR), we analyzed the ability of IGF-IR to modulate ß1 integrin levels. We report that IGF-IR regulates the expression of ß1 integrins, which in turn confer resistance to radiation in PrCa cells. In conclusion, this study demonstrates that ß1 integrins mediate resistance to ionizing radiation through inhibition of JNK1 activation.


Asunto(s)
Integrina beta1/metabolismo , Proteína Quinasa 8 Activada por Mitógenos/antagonistas & inhibidores , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/radioterapia , Tolerancia a Radiación/fisiología , Animales , Apoptosis/fisiología , Apoptosis/efectos de la radiación , Caspasa 3/metabolismo , Línea Celular Tumoral , Humanos , Integrina beta1/genética , Masculino , Ratones , Ratones Noqueados , Ratones Desnudos , Ratones Transgénicos , Proteína Quinasa 8 Activada por Mitógenos/genética , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Neoplasias de la Próstata/patología , ARN Interferente Pequeño/genética , Receptor IGF Tipo 1/antagonistas & inhibidores , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Trasplante Heterólogo
9.
Adv Healthc Mater ; 12(29): e2301944, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37565378

RESUMEN

Porous tissue-engineered 3D-printed scaffolds are a compelling alternative to autografts for the treatment of large periorbital bone defects. Matching the defect-specific geometry has long been considered an optimal strategy to restore pre-injury anatomy. However, studies in large animal models have revealed that biomaterial-induced bone formation largely occurs around the scaffold periphery. Such ectopic bone formation in the periorbital region can affect vision and cause disfigurement. To enhance anatomic reconstruction, geometric mismatches are introduced in the scaffolds used to treat full thickness zygomatic defects created bilaterally in adult Yucatan minipigs. 3D-printed, anatomically-mirrored scaffolds are used in combination with autologous stromal vascular fraction of cells (SVF) for treatment. An advanced image-registration workflow is developed to quantify the post-surgical geometric mismatch and correlate it with the spatial pattern of the regenerating bone. Osteoconductive bone growth on the dorsal and ventral aspect of the defect enhances scaffold integration with the native bone while medio-lateral bone growth leads to failure of the scaffolds to integrate. A strong positive correlation is found between geometric mismatch and orthotopic bone deposition at the defect site. The data suggest that strategic mismatch >20% could improve bone scaffold design to promote enhanced regeneration, osseointegration, and long-term scaffold survivability.


Asunto(s)
Impresión Tridimensional , Andamios del Tejido , Porcinos , Animales , Porcinos Enanos , Materiales Biocompatibles/farmacología , Regeneración Ósea , Osteogénesis
10.
Biomaterials ; 282: 121392, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35134701

RESUMEN

Critical-sized midfacial bone defects present a unique clinical challenge due to their complex three-dimensional shapes and intimate associations with sensory organs. To address this challenge, a point-of-care treatment strategy for functional, long-term regeneration of 2 cm full-thickness segmental defects in the zygomatic arches of Yucatan minipigs is evaluated. A digital workflow is used to 3D-print anatomically precise, porous, biodegradable scaffolds from clinical-grade poly-ε-caprolactone and decellularized bone composites. The autologous stromal vascular fraction of cells (SVF) is isolated from adipose tissue extracts and infused into the scaffolds that are implanted into the zygomatic ostectomies. Bone regeneration is assessed up to 52 weeks post-operatively in acellular (AC) and SVF groups (BV/DV = 0.64 ± 0.10 and 0.65 ± 0.10 respectively). In both treated groups, bone grows from the adjacent tissues and restores the native anatomy. Significantly higher torque is required to fracture the bone-scaffold interface in the SVF (7.11 ± 2.31 N m) compared to AC groups (2.83 ± 0.23 N m). Three-dimensional microcomputed tomography analysis reveals two distinct regenerative patterns: osteoconduction along the periphery of scaffolds to form dense lamellar bone and small islands of woven bone deposits growing along the struts in the scaffold interior. Overall, this study validates the efficacy of using 3D-printed bioactive scaffolds with autologous SVF to restore geometrically complex midfacial bone defects of clinically relevant sizes while also highlighting remaining challenges to be addressed prior to clinical translation.


Asunto(s)
Fracción Vascular Estromal , Andamios del Tejido , Animales , Regeneración Ósea , Osteogénesis , Sistemas de Atención de Punto , Impresión Tridimensional , Porcinos , Porcinos Enanos , Microtomografía por Rayos X
11.
EBioMedicine ; 48: 353-363, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31542392

RESUMEN

BACKGROUND: Mitochondrial functions are exploited in cancer and provide a validated therapeutic target. However, how this process is regulated has remained mostly elusive and the identification of new pathways that control mitochondrial integrity in cancer is an urgent priority. METHODS: We studied clinically-annotated patient series of primary and metastatic prostate cancer, representative cases of multiple myeloma (MM) and publicly available genetic databases. Gene regulation studies involved chromatin immunoprecipitation, PCR amplification and Western blotting of conditional Myc-expressing cell lines. Transient or stable gene silencing was used to quantify mitochondrial functions in bioenergetics, outer membrane permeability, Ca2+ homeostasis, redox balance and cell death. Tumorigenicity was assessed by cell proliferation, colony formation and xenograft tumour growth. FINDINGS: We identified Mitochondrial Fission Factor (MFF) as a novel transcriptional target of oncogenic Myc overexpressed in primary and metastatic cancer, compared to normal tissues. Biochemically, MFF isoforms, MFF1 and MFF2 associate with the Voltage-Dependent Anion Channel-1 (VDAC1) at the mitochondrial outer membrane, in vivo. Disruption of this complex by MFF silencing induces general collapse of mitochondrial functions with increased outer membrane permeability, loss of inner membrane potential, Ca2+ unbalance, bioenergetics defects and activation of cell death pathways. In turn, this inhibits tumour cell proliferation, suppresses colony formation and reduces xenograft tumour growth in mice. INTERPRETATION: An MFF-VDAC1 complex is a novel regulator of mitochondrial integrity and actionable therapeutic target in cancer.


Asunto(s)
Mitocondrias/genética , Mitocondrias/metabolismo , Dinámicas Mitocondriales/genética , Neoplasias/genética , Neoplasias/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Línea Celular Tumoral , Permeabilidad de la Membrana Celular , Proliferación Celular , Humanos , Potencial de la Membrana Mitocondrial/genética , Proteínas Mitocondriales/genética , Permeabilidad
12.
Kidney Blood Press Res ; 31(6): 421-32, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19142019

RESUMEN

BACKGROUND AND AIMS: Glomerular diseases are the third leading cause of kidney failure worldwide, behind only diabetes and hypertension. The molecular mechanisms underlying the cause of glomerular diseases are still largely unknown. The identification and characterization of new molecules associated with glomerular function should provide new insights into understanding the diverse group of glomerular diseases. The Chd2 protein belongs to a family of enzymes involved in ATP-dependent chromatin remodeling, suggesting that it likely functions as an epigenetic regulator of gene expression via the modification of chromatin structure. METHODS: In this study, we present a detailed histomorphologic characterization of mice containing a mutation in the chromodomain helicase DNA-binding protein 2 (Chd2). RESULTS: We show that Chd2-mutant mice present with glomerulopathy, proteinuria, and significantly impaired kidney function. Additionally, serum analysis revealed decreased hemoglobin and hematocrit levels in Chd2-mutant mice, suggesting that the glomerulopathy observed in these mice is associated with anemia. CONCLUSION: Collectively, the data suggest a role for the Chd2 protein in the maintenance of kidney function.


Asunto(s)
Ensamble y Desensamble de Cromatina , Proteínas de Unión al ADN/genética , Enfermedades Renales/genética , Mutación , Animales , Proteínas de Unión al ADN/fisiología , Epigénesis Genética , Glomerulonefritis Membranosa , Riñón/fisiología , Riñón/fisiopatología , Ratones , Fenotipo , Proteinuria
13.
Elife ; 72018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29856313

RESUMEN

Breast cancer is the most commonly diagnosed malignancy in women. Analysis of breast cancer genomic DNA indicates frequent loss-of-function mutations in components of the cJUN NH2-terminal kinase (JNK) signaling pathway. Since JNK signaling can promote cell proliferation by activating the AP1 transcription factor, this apparent association of reduced JNK signaling with tumor development was unexpected. We examined the effect of JNK deficiency in the murine breast epithelium. Loss of JNK signaling caused genomic instability and the development of breast cancer. Moreover, JNK deficiency caused widespread early neoplasia and rapid tumor formation in a murine model of breast cancer. This tumor suppressive function was not mediated by a role of JNK in the growth of established tumors, but by a requirement of JNK to prevent tumor initiation. Together, these data identify JNK pathway defects as 'driver' mutations that promote genome instability and tumor initiation.


Asunto(s)
Carcinogénesis/genética , Carcinogénesis/patología , Inestabilidad Genómica , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Sistema de Señalización de MAP Quinasas , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proliferación Celular , Supervivencia Celular , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Células Epiteliales/patología , Exoma/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/deficiencia , Ratones Endogámicos C57BL , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Fenotipo
14.
J Biomed Mater Res B Appl Biomater ; 81(2): 516-23, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17034012

RESUMEN

This study investigated the bone-regenerative potential of a demineralized bone and acellular matrix (DBM/AM) composite (AlloCraft DBM) in comparison with autologous bone using an in vivo model. Critical-sized calvarial defects (5 mm) were created in athymic rats. The defects were grafted with either the DBM/AM composite or the acellular human dermal matrix (AM), and compared with the defects filled with autologous bone (positive control) and the empty defect (negative control). Histological and radiographic assessments were carried out at 4 and 8 weeks after surgery to determine the biological healing, the amount and type of new bone formation and the percentage of new bone filled in the critical defects. At 4 weeks, DBM/AM composite group had the highest percentage of the defect filled with new bone (84%), which was significantly greater than autologous bone (62%), AM (41%), and untreated control (32%) groups. At 8 weeks, the DBM/AM continued to have the highest percentage of the defect filled with new bone (91%). The autologous bone group increased the percentage of bone fill to 83%. The defects either filled with AM or left untreated still had less of the defect filled with new bone, 57% and 33%, respectively. The total healing of defects grafted with DBM/AM was comparable with autologous bone group at 8 weeks. The results demonstrated that the DBM/AM composite promoted new bone formation more rapidly than autologous bone at calvarial defect in athymic rats. The study supports that DBM/AM is a potential substitute of autologous bone for bone repair.


Asunto(s)
Regeneración Ósea , Sustitutos de Huesos , Animales , Trasplante Óseo , Humanos , Ensayo de Materiales , Ratas , Ratas Desnudas , Cráneo/lesiones , Cráneo/fisiología , Cráneo/cirugía , Factores de Tiempo , Trasplante Autólogo
15.
Cancer Res ; 65(9): 3531-4, 2005 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15867343

RESUMEN

Gene signatures that predict aggressive tumor behavior at the earliest stages of disease, ideally before overt tissue abnormalities, are urgently needed. To search for such genes, we generated a transgenic model of survivin, an essential regulator of cell division and apoptosis overexpressed in cancer. Transgenic expression of survivin in the urinary bladder did not cause histologic abnormalities of the urothelium. However, microarray analysis revealed that survivin-expressing bladders exhibited profound changes in gene expression profile affecting extracellular matrix and inflammatory genes. Following exposure to a bladder carcinogen, N-butyl-N-(4-hydroxybutyl) nitrosamine (OH-BBN), survivin transgenic animals exhibited accelerated tumor progression, preferential incidence of tumors as compared with premalignant lesions, and dramatically abbreviated survival. Conversely, transgenic expression of a survivin Thr34-->Ala dominant-negative mutant did not cause changes in gene expression or accelerated tumor progression after OH-BBN treatment. Therefore, survivin expression induces global transcriptional changes in the tissue microenvironment that may promote tumorigenesis. Detection of survivin or its associated gene signature may provide an early biomarker of aggressive tumor behavior before the appearance of tissue abnormalities.


Asunto(s)
Proteínas Asociadas a Microtúbulos/genética , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/patología , Animales , Butilhidroxibutilnitrosamina , Carcinógenos , Progresión de la Enfermedad , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas Inhibidoras de la Apoptosis , Proteínas de la Membrana/genética , Ratones , Ratones Transgénicos , Proteínas Asociadas a Microtúbulos/biosíntesis , Proteínas de Neoplasias , Reacción en Cadena de la Polimerasa , Survivin , Transgenes , Proteína p53 Supresora de Tumor/genética , Vejiga Urinaria/efectos de los fármacos , Vejiga Urinaria/metabolismo , Vejiga Urinaria/patología , Vejiga Urinaria/fisiología , Neoplasias de la Vejiga Urinaria/inducido químicamente , Neoplasias de la Vejiga Urinaria/metabolismo , Uroplaquina II
16.
Comp Med ; 66(3): 225-34, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27298248

RESUMEN

Five birds in a captive zebra finch research colony were diagnosed with systemic amyloidosis within a 7-mo period by means of postmortem Congo red staining and green birefringence under polarized light. The liver was the most frequently and usually the most seriously affected organ, followed by the spleen and then the kidney. All 5 birds had been clinically affected with various inflammatory, infectious, and neoplastic conditions associated with amyloid A (AA) amyloidosis in humans and animals. Immunohistochemistry using antisera against duck AA protein revealed that tissues from 2 of the 5 birds were positive for the presence of AA protein and systemic inflammation-associated amyloidosis. Although the development of AA amyloidosis has been associated with chronic inflammation, trauma, and various infectious and neoplastic diseases as well as possible genetic predispositions and stresses linked to overcrowding, the root causes for individual cases of AA amyloidosis are incompletely understood. As far as we know, this report is the first description of AA amyloidosis in captive, research zebra finches.


Asunto(s)
Amiloidosis/veterinaria , Enfermedades de las Aves/patología , Pinzones , Amiloidosis/patología , Animales , Femenino , Inmunohistoquímica , Riñón/patología , Hígado/patología , Masculino , Proteína Amiloide A Sérica/metabolismo , Bazo/patología
17.
Hand (N Y) ; 10(3): 482-8, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26330782

RESUMEN

BACKGROUND: Reduction of peritendinous adhesions after injury and repair has been the subject of extensive prior investigation. The application of a circumferential barrier at the repair site may limit the quantity of peritendinous adhesions while preserving the tendon's innate ability to heal. The authors compare the effectiveness of a type I/III collagen membrane and a collagen-glycosaminoglycan (GAG) resorbable matrix in reducing tendon adhesions in an experimental chicken model of a "zone II" tendon laceration and repair. METHODS: In Leghorn chickens, flexor tendons were sharply divided using a scalpel and underwent repair in a standard fashion (54 total repairs). The sites were treated with a type I/III collagen membrane, collagen-GAG resorbable matrix, or saline in a randomized fashion. After 3 weeks, qualitative and semiquantitative histological analysis was performed to evaluate the "extent of peritendinous adhesions" and "nature of tendon healing." The data was evaluated with chi-square analysis and unpaired Student's t test. RESULTS: For both collagen materials, there was a statistically significant improvement in the degree of both extent of peritendinous adhesions and nature of tendon healing relative to the control group. There was no significant difference seen between the two materials. There was one tendon rupture observed in each treatment group. Surgical handling characteristics were subjectively favored for type I/III collagen membrane over the collagen-GAG resorbable matrix. CONCLUSION: The ideal method of reducing clinically significant tendon adhesions after injury remains elusive. Both materials in this study demonstrate promise in reducing tendon adhesions after flexor tendon repair without impeding tendon healing in this model.

18.
Virology ; 462-463: 254-65, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24999050

RESUMEN

The response to influenza virus (IAV) infection and severity of disease is highly variable in humans. We hypothesized that one factor contributing to this variability is the presence of specific respiratory tract (RT) microbes. One such microbe is Streptococcus pneumoniae (Sp) that is carried asymptomatically in the RT of many humans. In a mouse co-infection model we found that in contrast to secondary bacterial infection that exacerbates disease, Sp colonization 10 days prior to IAV protects from virus-induced morbidity and lung pathology. Using mutant Sp strains, we identified a critical role for the bacterial virulence factor pneumolysin (PLY) in mediating this protection. Colonization with the PLY-sufficient Sp strain induces expression of the immune-suppressive enzyme arginase 1 in alveolar macrophages (aMø) and correlates with attenuated recruitment and function of pulmonary inflammatory cells. Our study demonstrates a novel role for PLY in Sp-mediated protection by maintaining aMø as "gatekeepers" against virus-induced immunopathology.


Asunto(s)
Infecciones por Orthomyxoviridae/inmunología , Orthomyxoviridae/inmunología , Streptococcus pneumoniae/enzimología , Estreptolisinas/inmunología , Estreptolisinas/metabolismo , Animales , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Modelos Animales de Enfermedad , Factores Inmunológicos/inmunología , Factores Inmunológicos/metabolismo , Pulmón/patología , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/prevención & control , Análisis de Supervivencia
19.
Cancer Res ; 73(8): 2695-705, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23440423

RESUMEN

Metastatic traits seem to be acquired by transformed cells with progenitor-like cancer-initiating properties, but there remains little mechanistic insight into this linkage. In this report, we show that the polarity protein Numbl, which is expressed normally in neuronal progenitors, becomes overexpressed and mislocalized in cancer cells from a variety of human tumors. Numbl overexpression relies on loss of the tumor suppressor miRNA-296-5p (miR-296), which actively represses translation of Numbl in normal cells. In turn, deregulated expression of Numbl mediates random tumor cell migration and invasion, blocking anoikis and promoting metastatic dissemination. In clinical specimens of non-small cell lung cancer, we found that Numbl overexpression correlated with a reduction in overall patient survival. Mechanistically, Numbl-mediated tumorigenesis involved suppression of a "stemness" transcriptional program driven by the stem cell programming transcription factor Klf4, thereby preserving a pool of progenitor-like cells in lung cancer. Our results reveal that Numbl-Klf4 signaling is critical to maintain multiple nodes of metastatic progression, including persistence of cancer-initiating cells, rationalizing its therapeutic exploitation to improve the treatment of advanced lung cancer.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/genética , Factores de Transcripción de Tipo Kruppel/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Transducción de Señal , Animales , Secuencia de Bases , Línea Celular Tumoral , Movimiento Celular/genética , Análisis por Conglomerados , Progresión de la Enfermedad , Femenino , Perfilación de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/metabolismo , Neoplasias Pulmonares/metabolismo , Ratones , MicroARNs/química , MicroARNs/genética , Metástasis de la Neoplasia , Células Madre Neoplásicas/metabolismo , Interferencia de ARN , Transcripción Genética
20.
J Am Assoc Lab Anim Sci ; 51(6): 832-41, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23294892

RESUMEN

Perioperative treatment of several rats in our facility with ketoprofen (5 mg/kg SC) resulted in blood loss, peritonitis, and death within a day to a little more than a week after surgery that was not related to the gastrointestinal tract. Published reports have established the 5-mg/kg dose as safe and effective for rats. Because ketoprofen is a nonselective nonsteroidal antiinflammatory drug that can damage the gastrointestinal tract, the putative diagnosis for these morbidities and mortalities was gastrointestinal toxicity caused by ketoprofen (5 mg/kg). We conducted a prospective study evaluating the effect of this therapeutic dose of ketoprofen on the rat gastrointestinal tract within 24 h. Ketoprofen (5 mg/kg SC) was administered to one group of rats that then received gas anesthesia for 30 min and to another group without subsequent anesthesia. A third group was injected with saline followed by 30 min of gas anesthesia. Our primary hypothesis was that noteworthy gastrointestinal bleeding and lesions would occur in both groups treated with ketoprofen but not in rats that received saline and anesthesia. Our results showed marked gastrointestinal bleeding, erosions, and small intestinal ulcers in the ketoprofen-treated rats and minimal damages in the saline-treated group. The combination of ketoprofen and anesthesia resulted in worse clinical signs than did ketoprofen alone. We conclude that a single 5-mg/kg dose of ketoprofen causes acute mucosal damage to the rat small intestine.


Asunto(s)
Antiinflamatorios no Esteroideos/efectos adversos , Enfermedades Gastrointestinales/veterinaria , Cetoprofeno/efectos adversos , Dolor Postoperatorio/veterinaria , Ratas , Enfermedades de los Roedores/inducido químicamente , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Heces/química , Femenino , Enfermedades Gastrointestinales/inducido químicamente , Enfermedades Gastrointestinales/patología , Cetoprofeno/administración & dosificación , Dolor Postoperatorio/complicaciones , Dolor Postoperatorio/tratamiento farmacológico , Estudios Prospectivos , Enfermedades de los Roedores/patología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda