Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Clin Obstet Gynecol ; 63(1): 109-119, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31876640

RESUMEN

In our proof-of-concept study of 1 patient with stage IIIC carcinosarcoma of the ovary, we discovered a rare mutation in the tumor suppressor, TP53, that results in the deletion of N131. Immunofluorescence imaging of the organoid culture revealed hyperstaining of p53 protein. Computational modeling suggests this residue is important for maintaining protein conformation. Drug screening identified the combination of a proteasome inhibitor with a histone deacetylase inhibitor as the most effective treatment. These data provide evidence for the successful culture of a patient tumor and analysis of drug response ex vivo.


Asunto(s)
Carcinoma Epitelial de Ovario/genética , Predisposición Genética a la Enfermedad , Neoplasias Ováricas/genética , Proteína p53 Supresora de Tumor/genética , Femenino , Humanos , Organoides/metabolismo , Modelación Específica para el Paciente
2.
Gynecol Oncol ; 155(2): 349-358, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31477281

RESUMEN

OBJECTIVE: Platinum compounds have been widely used as a primary treatment for many types of cancer. However, resistance is the major cause of therapeutic failure for patients with metastatic or recurrent disease, thus highlighting the need to identify novel factors driving resistance to Platinum compounds. Metadherin (MTDH, also known as AEG-1 and LYRIC), located in a frequently amplified region of chromosome 8, has been consistently associated with resistance to chemotherapeutic agents, though the precise mechanisms remain incompletely defined. METHODS: The mRNA of FANCD2 and FANCI was pulled down by RNA-binding protein immunoprecipitation. Pristimerin-loaded nanoparticles were prepared using the nanoprecipitation method. Immunocompromised mice bearing patient-derived xenograft tumors were treated with pristimerin-loaded nanoparticles, cisplatin and a combination of the two. RESULTS: MTDH, through its recently discovered role as an RNA binding protein, regulates expression of FANCD2 and FANCI, two components of the Fanconi anemia complementation group (FA) that play critical roles in interstrand crosslink damage induced by platinum compounds. Pristimerin, a quinonemethide triterpenoid extract from members of the Celastraceae family used to treat inflammation in traditional Chinese medicine, significantly decreased MTDH, FANCD2 and FANCI levels in cancer cells, thereby restoring sensitivity to platinum-based chemotherapy. Using a patient-derived xenograft model of endometrial cancer, we discovered that treatment with pristimerin in a novel nanoparticle formulation markedly inhibited tumor growth when combined with cisplatin. CONCLUSIONS: MTDH is involved in post-transcriptional regulation of FANCD2 and FANCI. Pristimerin can increase sensitivity to platinum-based agents in tumors with MTDH overexpression by inhibiting the FA pathway.


Asunto(s)
Antineoplásicos/farmacología , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/antagonistas & inhibidores , Proteínas del Grupo de Complementación de la Anemia de Fanconi/antagonistas & inhibidores , Proteínas de la Membrana/efectos de los fármacos , Triterpenos/farmacología , Animales , Cisplatino/farmacología , Cistadenocarcinoma Seroso/tratamiento farmacológico , Regulación hacia Abajo , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Masculino , Ratones Noqueados , Nanopartículas , Triterpenos Pentacíclicos , Proteínas de Unión al ARN , Neoplasias Uterinas/tratamiento farmacológico
3.
J Biomed Mater Res A ; 112(6): 931-940, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38230545

RESUMEN

Tumor hypoxia, resulting from rapid tumor growth and aberrant vascular proliferation, exacerbates tumor aggressiveness and resistance to treatments like radiation and chemotherapy. To increase tumor oxygenation, we developed solid oxygen gas-entrapping materials (O2-GeMs), which were modeled after clinical brachytherapy implants, for direct tumor implantation. The objective of this study was to investigate the impact different formulations of solid O2-GeMs have on the entrapment and delivery of oxygen. Using a Parr reactor, we fabricated solid O2-GeMs using carbohydrate-based formulations used in the confectionary industry. In evaluating solid O2-GeMs manufactured from different sugars, the sucrose-containing formulation exhibited the highest oxygen concentration at 1 mg/g, as well as the fastest dissolution rate. The addition of a surface coating to the solid O2-GeMs, especially polycaprolactone, effectively prolonged the dissolution of the solid O2-GeMs. In vivo evaluation confirmed robust insertion and positioning of O2-GeMs in a malignant peripheral nerve sheath tumor, highlighting potential clinical applications.


Asunto(s)
Neoplasias , Oxígeno , Humanos , Hipoxia Tumoral/fisiología , Neoplasias/tratamiento farmacológico
4.
Device ; 2(5)2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38911126

RESUMEN

Diabetic wound healing is uniquely challenging to manage due to chronic inflammation and heightened microbial growth from elevated interstitial glucose. Carbon monoxide (CO), widely acknowledged as a toxic gas, is also known to provide unique therapeutic immune modulating effects. To facilitate delivery of CO, we have designed hyaluronic acid-based CO-gas-entrapping materials (CO-GEMs) for topical and prolonged gas delivery to the wound bed. We demonstrate that CO-GEMs promote the healing response in murine diabetic wound models (full-thickness wounds and pressure ulcers) compared to N2-GEMs and untreated controls.

5.
Adv Sci (Weinh) ; 11(9): e2308346, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38084435

RESUMEN

Modulation of autophagy, specifically its inhibition, stands to transform the capacity to effectively treat a broad range of cancers. However, the clinical efficacy of autophagy inhibitors has been inconsistent. To delineate clinical and epidemiological features associated with autophagy inhibition and a positive oncological clinical response, a retrospective analysis of patients is conducted treated with hydroxychloroquine, a known autophagy inhibitor. A direct correlation between smoking status and inhibition of autophagy with hydroxychloroquine is identified. Recognizing that smoking is associated with elevated circulating levels of carbon monoxide (CO), it is hypothesized that supplemental CO can amplify autophagy inhibition. A novel, gas-entrapping material containing CO in a pre-clinical model is applied and demonstrated that CO can dramatically increase the cytotoxicity of autophagy inhibitors and significantly inhibit the growth of tumors when used in combination. These data support the notion that safe, therapeutic levels of CO can markedly enhance the efficacy of autophagy inhibitors, opening a promising new frontier in the quest to improve cancer therapies.


Asunto(s)
Hidroxicloroquina , Neoplasias Pulmonares , Masculino , Humanos , Hidroxicloroquina/efectos adversos , Neoplasias Pulmonares/tratamiento farmacológico , Monóxido de Carbono/farmacología , Próstata , Estudios Retrospectivos , Autofagia
6.
Spectrochim Acta A Mol Biomol Spectrosc ; 298: 122797, 2023 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-37150072

RESUMEN

Hydrogen peroxide (H2O2) acts as an important reactive oxygen species (ROS) and maintains the redox equilibrium in organisms. Imbalance of H2O2 concentration is associated with the development of many diseases. Traditional small molecular based fluorescent probes often show drawbacks of cytotoxicity and easily metabolic clearance. Herein, a chitosan-based two-photon fluorescent nanoprobe (DC-BI) was constructed and applied for H2O2 detection in live organisms. DC-BI was composed by chitosan nanoparticles and a two-photon fluorophore of naphthalimide analogues (BI) with H2O2-responsive property. The structure of DC-BI was characterized by NMR, FTIR, XPS, XRD, DLS and MLS analyses. As study shown, the nanoprobe DC-BI exhibited improved distribution stability and smaller cytotoxicity. In the presence of H2O2, both the absorption and emission spectra show dramatic changes, the fluorescence intensity at 580 nm obviously enhanced. Furthermore, fluorescence imaging results indicate that DC-BI is capable of imaging endogenous H2O2 in cells and zebrafish. The design and development of chitosan-based nanoprobe DC-BI has provided a general example of nanoprobe construction with excellent distribution stability, two-photon property, and biocompatibility.


Asunto(s)
Quitosano , Nanopartículas , Animales , Peróxido de Hidrógeno/análisis , Pez Cebra/metabolismo , Colorantes Fluorescentes/toxicidad , Colorantes Fluorescentes/química , Nanopartículas/química , Sondas Moleculares/química
7.
J Steroid Biochem Mol Biol ; 234: 106399, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37716459

RESUMEN

Progesterone prevents development of endometrial cancers through its receptor (PR) although the molecular mechanisms have yet to be fully characterized. In this study, we performed a global analysis of gene regulation by progesterone using human endometrial cancer cells that expressed PR endogenously or exogenously. We found progesterone strongly inhibits multiple components of the platelet derived growth factor receptor (PDGFR), Janus kinase (JAK), signal transducer and activator of transcription (STAT) pathway through PR. The PDGFR/JAK/STAT pathway signals to control numerous downstream targets including AP-1 transcription factors Fos and Jun. Treatment with inhibitors of the PDGFR/JAK/STAT pathway significantly blocked proliferation in multiple novel patient-derived organoid models of endometrial cancer, and activation of this pathway was found to be a poor prognostic signal for the survival of patients with endometrial cancer from The Cancer Genome Atlas. Our study identifies this pathway as central to the growth-limiting effects of progesterone in endometrial cancer and suggests that inhibitors of PDGFR/JAK/STAT should be considered for future therapeutic interventions.


Asunto(s)
Neoplasias Endometriales , Quinasas Janus , Femenino , Humanos , Progesterona/farmacología , Transducción de Señal , Factores de Transcripción STAT/genética , Neoplasias Endometriales/tratamiento farmacológico , Neoplasias Endometriales/genética
8.
Adv Sci (Weinh) ; 10(10): e2205995, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36727291

RESUMEN

Tumor hypoxia drives resistance to many cancer therapies, including radiotherapy and chemotherapy. Methods that increase tumor oxygen pressures, such as hyperbaric oxygen therapy and microbubble infusion, are utilized to improve the responses to current standard-of-care therapies. However, key obstacles remain, in particular delivery of oxygen at the appropriate dose and with optimal pharmacokinetics. Toward overcoming these hurdles, gas-entrapping materials (GeMs) that are capable of tunable oxygen release are formulated. It is shown that injection or implantation of these materials into tumors can mitigate tumor hypoxia by delivering oxygen locally and that these GeMs enhance responsiveness to radiation and chemotherapy in multiple tumor types. This paper also demonstrates, by comparing an oxygen (O2 )-GeM to a sham GeM, that the former generates an antitumorigenic and immunogenic tumor microenvironment in malignant peripheral nerve sheath tumors. Collectively the results indicate that the use of O2 -GeMs is promising as an adjunctive strategy for the treatment of solid tumors.


Asunto(s)
Oxigenoterapia Hiperbárica , Neoplasias , Humanos , Oxígeno , Neoplasias/tratamiento farmacológico , Hipoxia Tumoral , Microambiente Tumoral
9.
Talanta ; 240: 123194, 2022 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-34979463

RESUMEN

Carbon monoxide (CO) is one of the most significant signal molecules and plays an important role in regulating human physiological and pathological processes. In this study, a novel Pd-based complex (Pd-BNP-OH) was developed for endogenous CO detection. The structure and morphology of Pd-BNP-OH was characterized by SEM, XPS, and NMR analyses. When Pd-BNP-OH was reacted with CO, a strong fluorescence enhancement at 510 nm was observed. In addition, Pd-BNP-OH exhibited high stability and selectivity toward CO in PBS buffer. In biological experiments, Pd-BNP-OH exhibited little cytotoxicity in cellular environment, and a bright fluorescence turn on was observed in the presence of exogenous CO and endogenous generated CO. The probe was then applied for CO detection in live zebrafish by both one-photon and two-photon excitation. Significantly, Pd-BNP-OH has excellent two-photon property, controllable structure and high biocompatibility. These features enable the probe to detect endogenously generated carbon monoxide in live organisms successfully.


Asunto(s)
Colorantes Fluorescentes , Pez Cebra , Animales , Monóxido de Carbono , Fluorescencia , Células HeLa , Humanos , Fotones
10.
Cell Death Dis ; 13(1): 59, 2022 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-35039480

RESUMEN

Histone deacetylase (HDAC) inhibitors and proteasome inhibitors have been approved by the FDA for the treatment of multiple myeloma and lymphoma, respectively, but have not achieved similar activity as single agents in solid tumors. Preclinical studies have demonstrated the activity of the combination of an HDAC inhibitor and a proteasome inhibitor in a variety of tumor models. However, the mechanisms underlying sensitivity and resistance to this combination are not well-understood. This study explores the role of autophagy in adaptive resistance to dual HDAC and proteasome inhibition. Studies focus on ovarian and endometrial gynecologic cancers, two diseases with high mortality and a need for novel treatment approaches. We found that nanomolar concentrations of the proteasome inhibitor ixazomib and HDAC inhibitor romidepsin synergistically induce cell death in the majority of gynecologic cancer cells and patient-derived organoid (PDO) models created using endometrial and ovarian patient tumor tissue. However, some models were not sensitive to this combination, and mechanistic studies implicated autophagy as the main mediator of cell survival in the context of dual HDAC and proteasome inhibition. Whereas the combination of ixazomib and romidepsin reduces autophagy in sensitive gynecologic cancer models, autophagy is induced following drug treatment of resistant cells. Pharmacologic or genetic inhibition of autophagy in resistant cells reverses drug resistance as evidenced by an enhanced anti-tumor response both in vitro and in vivo. Taken together, our findings demonstrate a role for autophagic-mediated cell survival in proteasome inhibitor and HDAC inhibitor-resistant gynecologic cancer cells. These data reveal a new approach to overcome drug resistance by inhibiting the autophagy pathway.


Asunto(s)
Neoplasias de los Genitales Femeninos , Inhibidores de Histona Desacetilasas , Complejo de la Endopetidasa Proteasomal , Inhibidores de Proteasoma , Autofagia , Línea Celular Tumoral , Femenino , Neoplasias de los Genitales Femeninos/tratamiento farmacológico , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Humanos , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/farmacología
11.
Sci Transl Med ; 14(651): eabl4135, 2022 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-35767653

RESUMEN

Carbon monoxide (CO) has long been considered a toxic gas but is now a recognized bioactive gasotransmitter with potent immunomodulatory effects. Although inhaled CO is currently under investigation for use in patients with lung disease, this mode of administration can present clinical challenges. The capacity to deliver CO directly and safely to the gastrointestinal (GI) tract could transform the management of diseases affecting the GI mucosa such as inflammatory bowel disease or radiation injury. To address this unmet need, inspired by molecular gastronomy techniques, we have developed a family of gas-entrapping materials (GEMs) for delivery of CO to the GI tract. We show highly tunable and potent delivery of CO, achieving clinically relevant CO concentrations in vivo in rodent and swine models. To support the potential range of applications of foam GEMs, we evaluated the system in three distinct disease models. We show that a GEM containing CO dose-dependently reduced acetaminophen-induced hepatocellular injury, dampened colitis-associated inflammation and oxidative tissue injury, and mitigated radiation-induced gut epithelial damage in rodents. Collectively, foam GEMs have potential paradigm-shifting implications for the safe therapeutic use of CO across a range of indications.


Asunto(s)
Colitis , Enfermedades Inflamatorias del Intestino , Animales , Monóxido de Carbono/uso terapéutico , Colitis/tratamiento farmacológico , Gases , Inflamación/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Porcinos
12.
Cancers (Basel) ; 13(12)2021 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-34200645

RESUMEN

Developing reliable experimental models that can predict clinical response before treating the patient is a high priority in gynecologic cancer research, especially in advanced or recurrent endometrial and ovarian cancers. Patient-derived organoids (PDOs) represent such an opportunity. Herein, we describe our successful creation of 43 tumor organoid cultures and nine adjacent normal tissue organoid cultures derived from patients with endometrial or ovarian cancer. From an initial set of 45 tumor tissues and seven ascites fluid samples harvested at surgery, 83% grew as organoids. Drug sensitivity testing and organoid cell viability assays were performed in 19 PDOs, a process that was accomplished within seven days of obtaining the initial surgical tumor sample. Sufficient numbers of cells were obtained to facilitate testing of the most commonly used agents for ovarian and endometrial cancer. The models reflected a range of sensitivity to platinum-containing chemotherapy as well as other relevant agents. One PDO from a patient treated prior to surgery with neoadjuvant trastuzumab successfully predicted the patient's postoperative chemotherapy and trastuzumab resistance. In addition, the PDO drug sensitivity assay identified alternative treatment options that are currently used in the second-line setting. Our findings suggest that PDOs could be used as a preclinical platform for personalized cancer therapy for gynecologic cancer patients.

13.
Pharmaceuticals (Basel) ; 14(7)2021 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-34358108

RESUMEN

Angiogenesis plays a crucial role in tumor development and metastasis. Both bevacizumab and cediranib have demonstrated activity as single anti-angiogenic agents in endometrial cancer, though subsequent studies of bevacizumab combined with chemotherapy failed to improve outcomes compared to chemotherapy alone. Our objective was to compare the efficacy of cediranib and bevacizumab in endometrial cancer models. The cellular effects of bevacizumab and cediranib were examined in endometrial cancer cell lines using extracellular signal-related kinase (ERK) phosphorylation, ligand shedding, cell viability, and cell cycle progression as readouts. Cellular viability was also tested in eight patient-derived organoid models of endometrial cancer. Finally, we performed a phosphoproteomic array of 875 phosphoproteins to define the signaling changes related to bevacizumab versus cediranib. Cediranib but not bevacizumab blocked ligand-mediated ERK activation in endometrial cancer cells. In both cell lines and patient-derived organoids, neither bevacizumab nor cediranib alone had a notable effect on cell viability. Cediranib but not bevacizumab promoted marked cell death when combined with chemotherapy. Cell cycle analysis demonstrated an accumulation in mitosis after treatment with cediranib + chemotherapy, consistent with the abrogation of the G2/M checkpoint and subsequent mitotic catastrophe. Molecular analysis of key controllers of the G2/M cell cycle checkpoint confirmed its abrogation. Phosphoproteomic analysis revealed that bevacizumab and cediranib had both similar and unique effects on cell signaling that underlie their shared versus individual actions as anti-angiogenic agents. An anti-angiogenic tyrosine kinase inhibitor such as cediranib has the potential to be superior to bevacizumab in combination with chemotherapy.

14.
Cell Death Dis ; 10(10): 682, 2019 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-31527591

RESUMEN

Ferroptosis is an iron-dependent, non-apoptotic form of regulated cell death driven by lipid hydroperoxides within biological membranes. Although therapy-resistant mesenchymal-high cancers are particularly vulnerable to ferroptosis inducers, especially phospholipid glutathione peroxidase 4 (GPx4) inhibitors, the underlying mechanism is yet to be deciphered. As such, the full application of GPx4 inhibitors in cancer therapy remains challenging. Here we demonstrate that metadherin (MTDH) confers a therapy-resistant mesenchymal-high cell state and enhanced sensitivity to inducers of ferroptosis. Mechanistically, MTDH inhibited GPx4, as well as the solute carrier family 3 member 2 (SLC3A2, a system Xc- heterodimerization partner), at both the messenger RNA and protein levels. Our metabolomic studies demonstrated that MTDH reduced intracellular cysteine, but increased glutamate levels, ultimately decreasing levels of glutathione and setting the stage for increased vulnerability to ferroptosis. Finally, we observed an enhanced antitumor effect when we combined various ferroptosis inducers both in vitro and in vivo; the level of MTDH correlated with the ferroptotic effect. We have demonstrated for the first time that MTDH enhances the vulnerability of cancer cells to ferroptosis and may serve as a therapeutic biomarker for future ferroptosis-centered cancer therapy.


Asunto(s)
Muerte Celular/efectos de los fármacos , Ferroptosis/genética , Proteínas de la Membrana/metabolismo , Neoplasias/tratamiento farmacológico , Proteínas de Unión al ARN/metabolismo , Biomarcadores de Tumor , Línea Celular Tumoral , Humanos
15.
Dis Markers ; 2018: 3810108, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30647797

RESUMEN

Mutations in the "guardian of the genome" TP53 predominate in solid tumors. In addition to loss of tumor suppressor activity, a specific subset of missense mutations confers additional oncogenic properties. These "gain-of-function" (GOF) mutations portend poor prognosis across cancer types regardless of treatment. Our objective in this study was to identify novel therapeutic opportunities to overcome the deleterious effects of GOF TP53 mutants. Using gynecologic cancer cell lines with known TP53 mutational status, we established that treatment with a proteasome inhibitor induced cell death in cells with two recurrent GOF TP53 mutations (R175H and R248Q), and addition of a histone deacetylase inhibitor (HDACi) enhanced this effect. By contrast, p53-null cancer cells were relatively resistant to the combination. Proteasome inhibition promoted apoptosis of cells with TP53 GOF mutations, potentially through induction of the unfolded protein response. In line with the reported hyperstabilization of GOF p53 protein, cells treated with HDACi exhibited reduced levels of p53 protein. Together, these data form the basis for future clinical studies examining therapeutic efficacy in a preselected patient population with GOF TP53 mutations.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias Endometriales/genética , Inhibidores de Histona Desacetilasas/farmacología , Inhibidores de Proteasoma/farmacología , Proteína p53 Supresora de Tumor/genética , Bortezomib/farmacología , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular/efectos de los fármacos , Regulación hacia Abajo , Neoplasias Endometriales/tratamiento farmacológico , Neoplasias Endometriales/metabolismo , Femenino , Mutación con Ganancia de Función/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Mutación Missense , Panobinostat/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Respuesta de Proteína Desplegada
16.
Cancers (Basel) ; 10(5)2018 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-29783721

RESUMEN

Tumor suppressor p53 is responsible for enforcing cell cycle checkpoints at G1/S and G2/M in response to DNA damage, thereby allowing both normal and tumor cells to repair DNA before entering S and M. However, tumor cells with absent or mutated p53 are able to activate alternative signaling pathways that maintain the G2/M checkpoint, which becomes uniquely critical for the survival of such tumor cells. We hypothesized that abrogation of the G2 checkpoint might preferentially sensitize p53-defective tumor cells to DNA-damaging agents and spare normal cells with intact p53 function. The tyrosine kinase WEE1 regulates cdc2 activity at the G2/M checkpoint and prevents entry into mitosis in response to DNA damage or stalled DNA replication. AZD1775 is a WEE1 inhibitor that overrides and opens the G2/M checkpoint by preventing WEE1-mediated phosphorylation of cdc2 at tyrosine 15. In this study, we assessed the effect of AZD1775 on endometrial and ovarian cancer cells in the presence of two DNA damaging agents, the PARP1 inhibitor, olaparib, and the chemotherapeutic agent, gemcitabine. We show that AZD1775 alone is effective as a therapeutic agent against some p53 mutated cell models. Moreover, the combination of AZD1775 with olaparib or gemcitabine is synergistic in cells with mutant p53 and constitutes a new approach that should be considered in the treatment of advanced and recurrent gynecologic cancer.

17.
Oncotarget ; 8(26): 42272-42287, 2017 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-28178687

RESUMEN

Aquatic mammals, such as cetaceans experience various depths, with accordingly diverse oxygenation, thus, cetaceans have developed adaptations for hypoxia, but mechanisms underlying this tolerance to low oxygen are unclear. Here we analyzed VHL and HIF-2α, in the hypoxia signaling pathway. Variations in VHL are greater than HIF-2α between cetaceans and terrestrial mammals, and beluga whale VHL (BW-VHL) promotes HIF-2α degradation under hypoxia. BW-VHL catalyzes BW-HIF-2α to form K48-linked poly-ubiquitin chains mainly at the lysine 429 of BW-HIF-2α (K429) and induces BW-HIF-2α for proteasomal degradation. W100 within BW-VHL is a key site for BW-VHL functionally and BW-VHL enhances transcriptional activity of BW-HIF-2α under hypoxia. Our data therefore reveal that BW-VHL has a unique function that may contribute to hypoxic adaptation.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Hipoxia/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Animales , Ballena Beluga , Catálisis , Línea Celular , Regulación de la Expresión Génica , Humanos , Hipoxia/genética , Mutación , Estabilidad Proteica , Proteolisis , Activación Transcripcional , Ubiquitinación , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA