Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 216
Filter
Add more filters

Uruguay Oncology Collection
Country/Region as subject
Publication year range
1.
Cell ; 149(1): 63-74, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22464323

ABSTRACT

Osteoblasts are an important component of the hematopoietic microenvironment in bone. However, the mechanisms by which osteoblasts control hematopoiesis remain unknown. We show that augmented HIF signaling in osteoprogenitors results in HSC niche expansion associated with selective expansion of the erythroid lineage. Increased red blood cell production occurred in an EPO-dependent manner with increased EPO expression in bone and suppressed EPO expression in the kidney. In contrast, inactivation of HIF in osteoprogenitors reduced EPO expression in bone. Importantly, augmented HIF activity in osteoprogenitors protected mice from stress-induced anemia. Pharmacologic or genetic inhibition of prolyl hydroxylases1/2/3 in osteoprogenitors elevated EPO expression in bone and increased hematocrit. These data reveal an unexpected role for osteoblasts in the production of EPO and modulation of erythropoiesis. Furthermore, these studies demonstrate a molecular role for osteoblastic PHD/VHL/HIF signaling that can be targeted to elevate both HSCs and erythroid progenitors in the local hematopoietic microenvironment.


Subject(s)
Erythropoiesis , Erythropoietin/metabolism , Osteoblasts/metabolism , Signal Transduction , Anemia/prevention & control , Animals , Erythroid Precursor Cells/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney/metabolism , Mice , Sp7 Transcription Factor , Transcription Factors/genetics , Transcription Factors/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
2.
Nature ; 579(7799): 456, 2020 03.
Article in English | MEDLINE | ID: mdl-32188947

ABSTRACT

A Retraction to this paper has been published and can be accessed via a link at the top of the paper.

3.
Proc Natl Acad Sci U S A ; 117(35): 21441-21449, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32817424

ABSTRACT

Loss of the von Hippel-Lindau (VHL) tumor suppressor is a hallmark feature of renal clear cell carcinoma. VHL inactivation results in the constitutive activation of the hypoxia-inducible factors (HIFs) HIF-1 and HIF-2 and their downstream targets, including the proangiogenic factors VEGF and PDGF. However, antiangiogenic agents and HIF-2 inhibitors have limited efficacy in cancer therapy due to the development of resistance. Here we employed an innovative computational platform, Mining of Synthetic Lethals (MiSL), to identify synthetic lethal interactions with the loss of VHL through analysis of primary tumor genomic and transcriptomic data. Using this approach, we identified a synthetic lethal interaction between VHL and the m6A RNA demethylase FTO in renal cell carcinoma. MiSL identified FTO as a synthetic lethal partner of VHL because deletions of FTO are mutually exclusive with VHL loss in pan cancer datasets. Moreover, FTO expression is increased in VHL-deficient ccRCC tumors compared to normal adjacent tissue. Genetic inactivation of FTO using multiple orthogonal approaches revealed that FTO inhibition selectively reduces the growth and survival of VHL-deficient cells in vitro and in vivo. Notably, FTO inhibition reduced the survival of both HIF wild type and HIF-deficient tumors, identifying FTO as an HIF-independent vulnerability of VHL-deficient cancers. Integrated analysis of transcriptome-wide m6A-seq and mRNA-seq analysis identified the glutamine transporter SLC1A5 as an FTO target that promotes metabolic reprogramming and survival of VHL-deficient ccRCC cells. These findings identify FTO as a potential HIF-independent therapeutic target for the treatment of VHL-deficient renal cell carcinoma.


Subject(s)
Alpha-Ketoglutarate-Dependent Dioxygenase FTO/genetics , Carcinoma, Renal Cell/genetics , Kidney Neoplasms/genetics , Synthetic Lethal Mutations , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Amino Acid Transport System ASC/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Computer Simulation , Humans , Hypoxia-Inducible Factor 1/metabolism , Kidney Neoplasms/metabolism , Mice, Knockout , Minor Histocompatibility Antigens/metabolism
4.
Genes Dev ; 29(8): 817-31, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25846796

ABSTRACT

The bone microenvironment is composed of niches that house cells across variable oxygen tensions. However, the contribution of oxygen gradients in regulating bone and blood homeostasis remains unknown. Here, we generated mice with either single or combined genetic inactivation of the critical oxygen-sensing prolyl hydroxylase (PHD) enzymes (PHD1-3) in osteoprogenitors. Hypoxia-inducible factor (HIF) activation associated with Phd2 and Phd3 inactivation drove bone accumulation by modulating osteoblastic/osteoclastic cross-talk through the direct regulation of osteoprotegerin (OPG). In contrast, combined inactivation of Phd1, Phd2, and Phd3 resulted in extreme HIF signaling, leading to polycythemia and excessive bone accumulation by overstimulating angiogenic-osteogenic coupling. We also demonstrate that genetic ablation of Phd2 and Phd3 was sufficient to protect ovariectomized mice against bone loss without disrupting hematopoietic homeostasis. Importantly, we identify OPG as a HIF target gene capable of directing osteoblast-mediated osteoclastogenesis to regulate bone homeostasis. Here, we show that coordinated activation of specific PHD isoforms fine-tunes the osteoblastic response to hypoxia, thereby directing two important aspects of bone physiology: cross-talk between osteoblasts and osteoclasts and angiogenic-osteogenic coupling.


Subject(s)
Bone and Bones/enzymology , Homeostasis , Osteoprotegerin/metabolism , Oxygen/metabolism , Prolyl Hydroxylases/genetics , Prolyl Hydroxylases/metabolism , 3T3 Cells , Animals , Bone Resorption/genetics , Bone and Bones/cytology , Cell Communication , Cell Hypoxia/physiology , Cells, Cultured , Enzyme Activation , Female , Gene Silencing , Hypoxia-Inducible Factor 1/metabolism , Mice , Osteoblasts/metabolism , Osteoclasts/metabolism , Signal Transduction/genetics , Stem Cells/enzymology
5.
Int J Mol Sci ; 24(7)2023 Mar 26.
Article in English | MEDLINE | ID: mdl-37047232

ABSTRACT

Increased expression of NUSAP1 has been identified as a robust prognostic biomarker in prostate cancer and other malignancies. We have previously shown that NUSAP1 is positively regulated by E2F1 and promotes cancer invasion and metastasis. To further understand the biological function of NUSAP1, we used affinity purification and mass spectrometry proteomic analysis to identify NUSAP1 interactors. We identified 85 unique proteins in the NUSAP1 interactome, including ILF2, DHX9, and other RNA-binding proteins. Using proteomic approaches, we uncovered a function for NUSAP1 in maintaining R-loops and in DNA damage response through its interaction with ILF2. Co-immunoprecipitation and colocalization using confocal microscopy verified the interactions of NUSAP1 with ILF2 and DHX9, and RNA/DNA hybrids. We showed that the microtubule and charged helical domains of NUSAP1 were necessary for the protein-protein interactions. Depletion of ILF2 alone further increased camptothecin-induced R-loop accumulation and DNA damage, and NUSAP1 depletion abolished this effect. In human prostate adenocarcinoma, NUSAP1 and ILF2 mRNA expression levels are positively correlated, elevated, and associated with poor clinical outcomes. Our study identifies a novel role for NUSAP1 in regulating R-loop formation and accumulation in response to DNA damage through its interactions with ILF2 and hence provides a potential therapeutic target.


Subject(s)
Prostatic Neoplasms , R-Loop Structures , Humans , Male , DNA Damage , Microtubule-Associated Proteins/metabolism , Nuclear Factor 45 Protein/genetics , Nuclear Factor 45 Protein/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Proteomics
6.
Cancer Immunol Immunother ; 71(4): 839-850, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34435232

ABSTRACT

The expression of immune-related genes in cancer cells can alter the anti-tumor immune response and thereby impact patient outcomes. Radiotherapy has been shown to modulate immune-related genes dependent on the fractionation regimen. To identify long-term changes in gene expression after irradiation, PC3 (p53 deleted) and LNCaP (p53 wildtype) prostate cancer cells were irradiated with either a single dose (SD, 10 Gy) or a fractionated regimen (MF) of 10 fractions (1 Gy per fraction). Whole human genome arrays were used to determine gene expression at 24 h and 2 months after irradiation. Immune pathway activation was analyzed with Ingenuity Pathway Analysis software. Additionally, 3D colony formation assays and T-cell cytotoxicity assays were performed. LNCaP had a higher basal expression of immunogenic genes and was more efficiently killed by cytotoxic T-cells compared to PC3. In both cell lines, MF irradiation resulted in an increase in multiple immune-related genes immediately after irradiation, while at 2 months, SD irradiation had a more pronounced effect on radiation-induced gene expression. Both immunogenic and immunosuppressive genes were upregulated in the long term in PC3 cells by a 10 Gy SD irradiation but not in LNCaP. T-cell-mediated cytotoxicity was significantly increased in 10 Gy SD PC3 cells compared to the unirradiated control and could be further enhanced by treatment with immune checkpoint inhibitors. Irradiation impacts the expression of immune-related genes in cancer cells in a fractionation-dependent manner. Understanding and targeting these changes may be a promising strategy for primary prostate cancer and recurrent tumors.


Subject(s)
Neoplasm Recurrence, Local , Prostatic Neoplasms , Apoptosis , Cell Line, Tumor , Humans , Male , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/radiotherapy
7.
Biophys J ; 120(18): 3860-3868, 2021 09 21.
Article in English | MEDLINE | ID: mdl-34411578

ABSTRACT

We present a novel fiber finding algorithm (FFA) that will permit researchers to detect and return traces of individual biopolymers. Determining the biophysical properties and structural cues of biopolymers can permit researchers to assess the progression and severity of disease. Confocal microscopy images are a useful method for observing biopolymer structures in three dimensions, but their utility for identifying individual biopolymers is impaired by noise inherent in the acquisition process, including convolution from the point spread function (PSF). The new, iterative FFA we present here 1) measures a microscope's PSF and uses it as a metric for identifying fibers against the background; 2) traces each fiber within a cone angle; and 3) blots out the identified trace before identifying another fiber. Blotting out the identified traces in each iteration allows the FFA to detect and return traces of single fibers accurately and efficiently-even within fiber bundles. We used the FFA to trace unlabeled collagen type I fibers-a biopolymer used to mimic the extracellular matrix in in vitro cancer assays-imaged by confocal reflectance microscopy in three dimensions, enabling quantification of fiber contour length, persistence length, and three-dimensional (3D) mesh size. Based on 3D confocal reflectance microscopy images and the PSF, we traced and measured the fibers to confirm that colder gelation temperatures increased fiber contour length, persistence length, and 3D mesh size-thereby demonstrating the FFA's use in quantifying biopolymers' structural and physical cues from noisy microscope images.


Subject(s)
Algorithms , Imaging, Three-Dimensional , Biopolymers , Collagen Type I , Microscopy, Confocal
8.
Br J Cancer ; 124(3): 604-615, 2021 02.
Article in English | MEDLINE | ID: mdl-33139797

ABSTRACT

BACKGROUND: To circumvent Warburg effect, several clinical trials for different cancers are utilising a combinatorial approach using metabolic reprogramming and chemotherapeutic agents including metformin. The majority of these metabolic interventions work via indirectly activating AMP-activated protein kinase (AMPK) to alter cellular metabolism in favour of oxidative phosphorylation over aerobic glycolysis. The effect of these drugs is dependent on glycaemic and insulin conditions.  Therefore, development of small molecules, which can activate AMPK, irrespective of the energy state, may be a better approach for triple-negative breast cancer (TNBC) treatment. METHODS: Therapeutic effect of SU212 on TNBC cells was examined using in vitro and in vivo models. RESULTS: We developed and characterised the efficacy of novel AMPK activator (SU212) that selectively induces oxidative phosphorylation and decreases glycolysis in TNBC cells, while not affecting these pathways in normal cells.   SU212 accomplished this metabolic reprogramming by activating AMPK independent of energy stress and irrespective of the glycaemic/insulin state. This leads to mitotic phase arrest and apoptosis in TNBC cells. In vivo, SU212 inhibits tumour growth, cancer progression and metastasis. CONCLUSIONS: SU212 directly activates AMPK in TNBC cells, but does not hamper glucose metabolism in normal cells. Our study provides compelling preclinical data for further development of SU212 for the treatment of TNBC.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Cell Death , Oxidative Phosphorylation/drug effects , Podophyllotoxin/analogs & derivatives , Triple Negative Breast Neoplasms/metabolism , Animals , Apoptosis , Cell Line, Tumor , Cell Survival , Enzyme Activation/drug effects , Female , Glucose/metabolism , Glycolysis/drug effects , Humans , Lactic Acid/metabolism , Lipogenesis/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Neoplasm Proteins/metabolism , Random Allocation , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Warburg Effect, Oncologic
9.
Ann Surg ; 273(1): 173-180, 2021 01 01.
Article in English | MEDLINE | ID: mdl-30829705

ABSTRACT

OBJECTIVE: The aim of this study was to determine the interaction of full thickness excisional wounds and tumors in vivo. SUMMARY OF BACKGROUND DATA: Tumors have been described as wounds that do not heal due to similarities in stromal composition. On the basis of observations of slowed tumor growth after ulceration, we hypothesized that full thickness excisional wounds would inhibit tumor progression in vivo. METHODS: To determine the interaction of tumors and wounds, we developed a tumor xenograft/allograft (human head and neck squamous cell carcinoma SAS/mouse breast carcinoma 4T1) wound mouse model. We examined tumor growth with varying temporospatial placement of tumors and wounds or ischemic flap. In addition, we developed a tumor/wound parabiosis model to understand the ability of tumors and wounds to recruit circulating progenitor cells. RESULTS: Tumor growth inhibition by full thickness excisional wounds was dose-dependent, maintained by sequential wounding, and relative to distance. This effect was recapitulated by placement of an ischemic flap directly adjacent to a xenograft tumor. Using a parabiosis model, we demonstrated that a healing wound was able to recruit significantly more circulating progenitor cells than a growing tumor. Tumor inhibition by wound was unaffected by presence of an immune response in an immunocompetent model using a mammary carcinoma. Utilizing functional proteomics, we identified 100 proteins differentially expressed in tumors and wounds. CONCLUSION: Full thickness excisional wounds have the ability to inhibit tumor growth in vivo. Further research may provide an exact mechanism for this remarkable finding and new advances in wound healing and tumor biology.


Subject(s)
Neoplasms/pathology , Ulcer/pathology , Wounds and Injuries/pathology , Animals , Female , Mice , Neoplasms/complications , Ulcer/complications , Wounds and Injuries/complications
10.
Proc Natl Acad Sci U S A ; 115(42): 10756-10761, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30201710

ABSTRACT

Tumor hypoxia reduces the effectiveness of radiation therapy by limiting the biologically effective dose. An acute increase in tumor oxygenation before radiation treatment should therefore significantly improve the tumor cell kill after radiation. Efforts to increase oxygen delivery to the tumor have not shown positive clinical results. Here we show that targeting mitochondrial respiration results in a significant reduction of the tumor cells' demand for oxygen, leading to increased tumor oxygenation and radiation response. We identified an activity of the FDA-approved drug papaverine as an inhibitor of mitochondrial complex I. We also provide genetic evidence that papaverine's complex I inhibition is directly responsible for increased oxygenation and enhanced radiation response. Furthermore, we describe derivatives of papaverine that have the potential to become clinical radiosensitizers with potentially fewer side effects. Importantly, this radiosensitizing strategy will not sensitize well-oxygenated normal tissue, thereby increasing the therapeutic index of radiotherapy.


Subject(s)
Cell Hypoxia/drug effects , Lung Neoplasms/radiotherapy , Mitochondria/drug effects , NADH Dehydrogenase/antagonists & inhibitors , Oxygen/metabolism , Papaverine/pharmacology , Radiation-Sensitizing Agents/pharmacology , Animals , CRISPR-Cas Systems , Cell Hypoxia/radiation effects , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Electron Transport Complex I , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondria/radiation effects , NADH Dehydrogenase/genetics , Phosphodiesterase Inhibitors/pharmacology , Radiation Tolerance , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
11.
Circulation ; 139(4): 502-517, 2019 01 22.
Article in English | MEDLINE | ID: mdl-30586708

ABSTRACT

BACKGROUND: Hypoxia-inducible factors (HIFs), especially HIF-1α and HIF-2α, are key mediators of the adaptive response to hypoxic stress and play essential roles in maintaining lung homeostasis. Human and animal genetics studies confirm that abnormal HIF correlates with pulmonary vascular pathology and chronic lung diseases, but it remains unclear whether endothelial cell HIF production is essential for microvascular health. The large airway has an ideal circulatory bed for evaluating histological changes and physiology in genetically modified rodents. METHODS: The tracheal microvasculature of mice, with conditionally deleted or overexpressed HIF-1α or HIF-2α, was evaluated for anatomy, perfusion, and permeability. Angiogenic signaling studies assessed vascular changes attributable to dysregulated HIF expression. An orthotopic tracheal transplantation model further evaluated the contribution of individual HIF isoforms in airway endothelial cells. RESULTS: The genetic deletion of Hif-2α but not Hif-1α caused tracheal endothelial cell apoptosis, diminished pericyte coverage, reduced vascular perfusion, defective barrier function, overlying epithelial abnormalities, and subepithelial fibrotic remodeling. HIF-2α promoted microvascular integrity in airways through endothelial angiopoietin-1/TIE2 signaling and Notch activity. In functional tracheal transplants, HIF-2α deficiency in airway donors accelerated graft microvascular loss, whereas HIF-2α or angiopoietin-1 overexpression prolonged transplant microvascular perfusion. Augmented endothelial HIF-2α in transplant donors promoted airway microvascular integrity and diminished alloimmune inflammation. CONCLUSIONS: Our findings reveal that the constitutive expression of endothelial HIF-2α is required for airway microvascular health.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Endothelial Cells/metabolism , Microvessels/metabolism , Trachea/blood supply , Angiopoietin-1/metabolism , Animals , Apoptosis , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Cells, Cultured , Endothelial Cells/pathology , Endothelial Cells/transplantation , Female , Graft Survival , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Microvessels/pathology , Microvessels/transplantation , Neovascularization, Physiologic , Receptor, TIE-2/metabolism , Signal Transduction , Trachea/transplantation
12.
Sens Actuators B Chem ; 3062020 Mar 01.
Article in English | MEDLINE | ID: mdl-32265579

ABSTRACT

Hypoxia (pO2 ≤ ~1.5%) is an important characteristic of tumor microenvironments that directly correlates with resistance against first-line therapies and tumor proliferation/infiltration. The ability to accurately identify hypoxic tumor cells/tissue could afford tailored therapeutic regimens for personalized treatment, development of more-effective therapies, and discerning the mechanisms underlying disease progression. Fluorogenic constructs identifying aforesaid cells/tissue operate by targeting the bioreductive activity of primarily nitroreductases (NTRs), but collectively present photophysical and/or physicochemical shortcomings that could limit effectiveness. To overcome these limitations, we present the rational design, development, and evaluation of the first activatable ultracompact xanthene core-based molecular probe (NO 2 -Rosol) for selectively imaging NTR activity that affords an "OFF-ON" near-infrared (NIR) fluorescence response (> 700 nm) alongside a remarkable Stokes shift (> 150 nm) via NTR activity-facilitated modulation to its energetics whose resultant interplay discontinues an intramolecular d-PET fluorescence-quenching mechanism transpiring between directly-linked electronically-uncoupled π-systems comprising its components. DFT calculations guided selection of a suitable fluorogenic scaffold and nitroaromatic moiety candidate that when adjoined could (i) afford such photophysical response upon bioreduction by upregulated NTR activity in hypoxic tumor cells/tissue and (ii) employ a retention mechanism strategy that capitalizes on an inherent physical property of the NIR fluorogenic scaffold for achieving signal amplification. NO 2 -Rosol demonstrated 705 nm NIR fluorescence emission and 157 nm Stokes shift, selectivity for NTR over relevant bioanalytes, and a 28-/12-fold fluorescence enhancement in solution and between cells cultured under different oxic conditions, respectively. In establishing feasibility for NO 2 -Rosol to provide favorable contrast levels in solutio/vitro, we anticipate NO 2 -Rosol doing so in preclinical studies.

13.
Sensors (Basel) ; 20(15)2020 Aug 01.
Article in English | MEDLINE | ID: mdl-32752216

ABSTRACT

There has been strong demand for the development of an accurate but simple method to assess the freshness of food. In this study, we demonstrated a system to determine food freshness by analyzing the spectral response from a portable visible/near-infrared (VIS/NIR) spectrometer using the Convolutional Neural Network (CNN)-based machine learning algorithm. Spectral response data from salmon, tuna, and beef incubated at 25 °C were obtained every minute for 30 h and then categorized into three states of "fresh", "likely spoiled", and "spoiled" based on time and pH. Using the obtained spectral data, a CNN-based machine learning algorithm was built to evaluate the freshness of experimental objects. In addition, a CNN-based machine learning algorithm with a shift-invariant feature can minimize the effect of the variation caused using multiple devices in a real environment. The accuracy of the obtained machine learning model based on the spectral data in predicting the freshness was approximately 85% for salmon, 88% for tuna, and 92% for beef. Therefore, our study demonstrates the practicality of a portable spectrometer in food freshness assessment.


Subject(s)
Red Meat , Salmon , Tuna , Algorithms , Animals , Cattle , Neural Networks, Computer , Seafood
14.
Nucleic Acids Res ; 45(7): 3674-3692, 2017 04 20.
Article in English | MEDLINE | ID: mdl-28073943

ABSTRACT

The p53 tumor suppressor protein plays a critical role in orchestrating the genomic response to various stress signals by acting as a master transcriptional regulator. Differential gene activity is controlled by transcription factors but also dependent on the underlying chromatin structure, especially on covalent histone modifications. After screening different histone lysine methyltransferases and demethylases, we identified JMJD2B/KDM4B as a p53-inducible gene in response to DNA damage. p53 directly regulates JMJD2B gene expression by binding to a canonical p53-consensus motif in the JMJD2B promoter. JMJD2B induction attenuates the transcription of key p53 transcriptional targets including p21, PIG3 and PUMA, and this modulation is dependent on the catalytic capacity of JMJD2B. Conversely, JMJD2B silencing led to an enhancement of the DNA-damage driven induction of p21 and PIG3. These findings indicate that JMJD2B acts in an auto-regulatory loop by which p53, through JMJD2B activation, is able to influence its own transcriptional program. Functionally, exogenous expression of JMJD2B enhanced subcutaneous tumor growth of colon cancer cells in a p53-dependent manner, and genetic inhibition of JMJD2B impaired tumor growth in vivo. These studies provide new insights into the regulatory effect exerted by JMJD2B on tumor growth through the modulation of p53 target genes.


Subject(s)
DNA Damage , Epigenesis, Genetic , Jumonji Domain-Containing Histone Demethylases/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line , Cells, Cultured , Histone Demethylases/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/biosynthesis , Jumonji Domain-Containing Histone Demethylases/metabolism , Mice , Mutagens/toxicity , Neoplasms/pathology , Promoter Regions, Genetic , Transcriptional Activation
15.
Proc Natl Acad Sci U S A ; 113(46): 13120-13125, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27799523

ABSTRACT

Duchenne muscular dystrophy (DMD) is an incurable X-linked genetic disease that is caused by a mutation in the dystrophin gene and affects one in every 3,600 boys. We previously showed that long telomeres protect mice from the lethal cardiac disease seen in humans with the same genetic defect, dystrophin deficiency. By generating the mdx4cv/mTRG2 mouse model with "humanized" telomere lengths, the devastating dilated cardiomyopathy phenotype seen in patients with DMD was recapitulated. Here, we analyze the degenerative sequelae that culminate in heart failure and death in this mouse model. We report progressive telomere shortening in developing mouse cardiomyocytes after postnatal week 1, a time when the cells are no longer dividing. This proliferation-independent telomere shortening is accompanied by an induction of a DNA damage response, evident by p53 activation and increased expression of its target gene p21 in isolated cardiomyocytes. The consequent repression of Pgc1α/ß leads to impaired mitochondrial biogenesis, which, in conjunction with the high demands of contraction, leads to increased oxidative stress and decreased mitochondrial membrane potential. As a result, cardiomyocyte respiration and ATP output are severely compromised. Importantly, treatment with a mitochondrial-specific antioxidant before the onset of cardiac dysfunction rescues the metabolic defects. These findings provide evidence for a link between short telomere length and metabolic compromise in the etiology of dilated cardiomyopathy in DMD and identify a window of opportunity for preventive interventions.


Subject(s)
Cardiomyopathy, Dilated , Muscular Dystrophy, Animal , Myocytes, Cardiac/physiology , Telomere Shortening , Animals , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/physiopathology , Cell Cycle , Cell Proliferation , DNA Damage , Male , Membrane Potential, Mitochondrial , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Heart/metabolism , Mitochondria, Heart/physiology , Mitosis , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Animal/physiopathology , Muscular Dystrophy, Duchenne , Reactive Oxygen Species/metabolism
16.
Proc Natl Acad Sci U S A ; 112(14): 4441-6, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25792458

ABSTRACT

Scaffold proteins are critical hubs within cells that have the ability to modulate upstream signaling molecules and their downstream effectors to fine-tune biological responses. Although they can serve as focal points for association of signaling molecules and downstream pathways that regulate tumorigenesis, little is known about how the tumor microenvironment affects the expression and activity of scaffold proteins. This study demonstrates that hypoxia, a common element of solid tumors harboring low oxygen levels, regulates expression of a specific variant of the scaffold protein AKAP12 (A-kinase anchor protein 12), AKAP12v2, in metastatic melanoma. In turn, through a kinome-wide phosphoproteomic and MS study, we demonstrate that this scaffolding protein regulates a shift in protein kinase A (PKA)-mediated phosphorylation events under hypoxia, causing alterations in tumor cell invasion and migration in vitro, as well as metastasis in an in vivo orthotopic model of melanoma. Mechanistically, the shift in AKAP12-dependent PKA-mediated phosphorylations under hypoxia is due to changes in AKAP12 localization vs. structural differences between its two variants. Importantly, our work defines a mechanism through which a scaffold protein can be regulated by the tumor microenvironment and further explains how a tumor cell can coordinate many critical signaling pathways that are essential for tumor growth through one individual scaffolding protein.


Subject(s)
A Kinase Anchor Proteins/metabolism , Cell Cycle Proteins/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation, Neoplastic , Melanoma/pathology , Skin Neoplasms/metabolism , A Kinase Anchor Proteins/genetics , Amino Acid Sequence , Animals , Cell Cycle Proteins/genetics , Cell Hypoxia , Cell Line, Tumor , Cell Movement , Humans , Melanoma/metabolism , Mice , Molecular Sequence Data , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Transplantation , Oxygen/metabolism , Phosphoproteins/metabolism , Phosphorylation , Proteomics , Signal Transduction
18.
Mol Cell ; 35(6): 856-67, 2009 Sep 24.
Article in English | MEDLINE | ID: mdl-19782034

ABSTRACT

Previous studies have suggested that the HIF transcription factors can both activate and inhibit gene expression. Here we show that HIF1 regulates the expression of mir-210 in a variety of tumor types through a hypoxia-responsive element. Expression analysis in primary head and neck tumor samples indicates that mir-210 may serve as an in vivo marker for tumor hypoxia. By Argonaute protein immunoprecipitation, we identified 50 potential mir-210 targets and validated randomly selected ones. The majority of these 50 genes are not classical hypoxia-inducible genes, suggesting mir-210 represses genes expressed under normoxia that are no longer necessary to adapt and survive in a hypoxic environment. When human head and neck or pancreatic tumor cells ectopically expressing mir-210 were implanted into immunodeficient mice, mir-210 repressed initiation of tumor growth. Taken together, these data implicate an important role for mir-210 in regulating the hypoxic response of tumor cells and tumor growth.


Subject(s)
Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Gene Silencing , Head and Neck Neoplasms/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , MicroRNAs/metabolism , Pancreatic Neoplasms/genetics , Stress, Physiological/genetics , Animals , Base Sequence , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Eukaryotic Initiation Factor-2/metabolism , Gene Expression Profiling/methods , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunoprecipitation , Male , Mice , Mice, Nude , Molecular Sequence Data , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Protein Binding , Receptor, Fibroblast Growth Factor, Type 5/genetics , Receptor, Fibroblast Growth Factor, Type 5/metabolism , Reproducibility of Results , Response Elements , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic , Transduction, Genetic , Up-Regulation
19.
Curr Osteoporos Rep ; 15(4): 231-238, 2017 08.
Article in English | MEDLINE | ID: mdl-28597139

ABSTRACT

PURPOSE OF REVIEW: This review highlights our current knowledge of oxygen tensions in the bone marrow, and how low oxygen tensions (hypoxia) regulate tumor metastasis to and colonization of the bone marrow. RECENT FINDINGS: The bone marrow is a relatively hypoxic microenvironment, but oxygen tensions fluctuate throughout the marrow cavity and across the endosteal and periosteal surfaces. Recent advances in imaging have made it possible to better characterize these fluctuations in bone oxygenation, but technical challenges remain. We have compiled evidence from multiple groups that suggests that hypoxia or hypoxia inducible factor (HIF) signaling may induce spontaneous metastasis to the bone and promote tumor colonization of bone, particularly in the case of breast cancer dissemination to the bone marrow. We are beginning to understand oxygenation patterns within the bone compartment and the role for hypoxia and HIF signaling in tumor cell dissemination to the bone marrow, but further studies are warranted.


Subject(s)
Bone Marrow/metabolism , Bone Neoplasms/secondary , Breast Neoplasms/pathology , Hypoxia/metabolism , Bone Neoplasms/metabolism , Bone and Bones/metabolism , Female , Humans , Hypoxia-Inducible Factor 1/metabolism , Neoplasm Metastasis , Signal Transduction
20.
Proc Natl Acad Sci U S A ; 111(37): 13373-8, 2014 Sep 16.
Article in English | MEDLINE | ID: mdl-25187556

ABSTRACT

Dysregulation of the von Hippel-Lindau/hypoxia-inducible transcription factor (HIF) signaling pathway promotes clear cell renal cell carcinoma (ccRCC) progression and metastasis. The protein kinase GAS6/AXL signaling pathway has recently been implicated as an essential mediator of metastasis and receptor tyrosine kinase crosstalk in cancer. Here we establish a molecular link between HIF stabilization and induction of AXL receptor expression in metastatic ccRCC. We found that HIF-1 and HIF-2 directly activate the expression of AXL by binding to the hypoxia-response element in the AXL proximal promoter. Importantly, genetic and therapeutic inactivation of AXL signaling in metastatic ccRCC cells reversed the invasive and metastatic phenotype in vivo. Furthermore, we define a pathway by which GAS6/AXL signaling uses lateral activation of the met proto-oncogene (MET) through SRC proto-oncogene nonreceptor tyrosine kinase to maximize cellular invasion. Clinically, AXL expression in primary tumors of ccRCC patients correlates with aggressive tumor behavior and patient lethality. These findings provide an alternative model for SRC and MET activation by growth arrest-specific 6 in ccRCC and identify AXL as a therapeutic target driving the aggressive phenotype in renal clear cell carcinoma.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Kidney Neoplasms/secondary , Proto-Oncogene Proteins c-met/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , src-Family Kinases/metabolism , Carcinoma, Renal Cell/enzymology , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/secondary , Cell Hypoxia , Cell Line, Tumor , Enzyme Activation , Hepatocyte Growth Factor/pharmacology , Humans , Kidney Neoplasms/enzymology , Kidney Neoplasms/pathology , Models, Biological , Neoplasm Invasiveness , Phenotype , Proto-Oncogene Mas , Signal Transduction , Treatment Outcome , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Axl Receptor Tyrosine Kinase
SELECTION OF CITATIONS
SEARCH DETAIL