Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 117(29): 16938-16948, 2020 07 21.
Article in English | MEDLINE | ID: mdl-32616570

ABSTRACT

Despite nearly four decades of effort, broad inhibition of oncogenic RAS using small-molecule approaches has proven to be a major challenge. Here we describe the development of a pan-RAS biologic inhibitor composed of the RAS-RAP1-specific endopeptidase fused to the protein delivery machinery of diphtheria toxin. We show that this engineered chimeric toxin irreversibly cleaves and inactivates intracellular RAS at low picomolar concentrations terminating downstream signaling in receptor-bearing cells. Furthermore, we demonstrate in vivo target engagement and reduction of tumor burden in three mouse xenograft models driven by either wild-type or mutant RAS Intracellular delivery of a potent anti-RAS biologic through a receptor-mediated mechanism represents a promising approach to developing RAS therapeutics against a broad array of cancers.


Subject(s)
Diphtheria Toxin/metabolism , Endopeptidases/metabolism , Neoplasms, Experimental/drug therapy , Proteolysis , rap1 GTP-Binding Proteins/metabolism , ras Proteins/metabolism , Animals , Antineoplastic Agents/therapeutic use , Cells, Cultured , Diphtheria Toxin/chemistry , Diphtheria Toxin/genetics , Endopeptidases/chemistry , Endopeptidases/genetics , Female , HCT116 Cells , Humans , Male , Mice , Mice, Nude , Mutation , Protein Sorting Signals , Recombinant Proteins/therapeutic use , ras Proteins/genetics
2.
Cell Signal ; 70: 109574, 2020 06.
Article in English | MEDLINE | ID: mdl-32084531

ABSTRACT

KLF4 plays an important role in orchestrating a variety of cellular events, including cell-fate decision, genome stability and apoptosis. Its deregulation is correlated with human diseases such as breast cancer and gastrointestinal cancer. Results from recent biochemical studies have revealed that KLF4 is tightly regulated by posttranslational modifications. Here we report a new finding that KLF4 orchestrates estrogen receptor signaling and facilitates endocrine resistance. We also uncovered the underlying mechanism that alteration of KLF4 by posttranslational modifications such as phosphorylation and ubiquitylation changes tumor cell response to endocrine therapy drugs. IHC analyses using based on human breast cancer specimens showed the accumulation of KLF4 protein in ER-positive breast cancer tissues. Elevated KLF4 expression significantly correlated with prognosis and endocrine resistance. Our drug screening for suppressing KLF4 protein expression led to identification of Src kinase to be a critical player in modulating KLF4-mediated tamoxifen resistance. Depletion of VHL (von Hippel-Lindau tumor suppressor), a ubiquitin E3 ligase for KLF4, reduces tumor cell sensitivity to tamoxifen. We demonstrated phosphorylation of VHL by Src enhances proteolysis of VHL that in turn leads to upregulation of KLF4 and increases endocrine resistance. Suppression of Src-VHL-KLF4 cascade by Src inhibitor or enhancement of VHL-KLF4 ubiquitination by TAT-KLF4 (371-420AAa) peptides re-sensitizes tamoxifen-resistant breast cancer cells to tamoxifen treatment. Taken together, our findings demonstrate a novel role for KLF4 in modulating endocrine resistance via the Src-VHL-KLF4 axis.


Subject(s)
Breast Neoplasms/metabolism , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Kruppel-Like Transcription Factors/physiology , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , HEK293 Cells , Humans , Kruppel-Like Factor 4 , MCF-7 Cells
3.
Cell Metab ; 29(2): 443-456.e5, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30595481

ABSTRACT

During wound injury, efferocytosis fills the macrophage with a metabolite load nearly equal to the phagocyte itself. A timely question pertains to how metabolic phagocytic signaling regulates the signature anti-inflammatory macrophage response. Here we report the metabolome of activated macrophages during efferocytosis to reveal an interleukin-10 (IL-10) cytokine escalation that was independent of glycolysis yet bolstered by apoptotic cell fatty acids and mitochondrial ß-oxidation, the electron transport chain, and heightened coenzyme NAD+. Loss of IL-10 due to mitochondrial complex III defects was remarkably rescued by adding NAD+ precursors. This activated a SIRTUIN1 signaling cascade, largely independent of ATP, that culminated in activation of IL-10 transcription factor PBX1. Il-10 activation by the respiratory chain was also important in vivo, as efferocyte mitochondrial dysfunction led to cardiac rupture after myocardial injury. These findings highlight a new paradigm whereby macrophages leverage efferocytic metabolites and electron transport for anti-inflammatory reprogramming that culminates in organ repair.


Subject(s)
Fatty Acids/metabolism , Interleukin-10/metabolism , Macrophages/metabolism , Mitochondria/metabolism , NAD/metabolism , Animals , Cytophagocytosis , Electron Transport , Humans , Inflammation/metabolism , Jurkat Cells , Macrophages/cytology , Mice , Mice, Inbred C57BL , Oxidation-Reduction , Wound Healing
4.
Mol Pharm ; 15(9): 3625-3633, 2018 09 04.
Article in English | MEDLINE | ID: mdl-29771531

ABSTRACT

Exosomes play a pivotal role in mediating intercellular communications and package delivery. They have recently been discovered to serve as diagnostic biomarkers as well as a possible drug delivery vehicle based on their nanometer size range and capability to transfer biological materials to recipient cells. Their unique biocompatibility, high stability, preferred tumor homing, and adjustable targeting efficiency can make exosomes an attractive and potentially effective tool of drug delivery in cancer therapy. While exosomes possess properties that make them uniquely suitable for delivery of bioactive molecules, there remains a to-be-filled gap between the current understanding about exosome biology and the ideal application scenarios. In this review, we summarize the characteristics enabling the potential of exosomes for drug delivery as well as the outstanding questions related to exosome composition and function, production and purification, bioengineering and targeting, uptake and biodistribution, efficacy and immune regulation, etc. Advanced technologies are demanded to visualize, characterize, and sort heterogeneous exosome populations. We are positive that the deeper and more comprehensive understanding of exosome biology as well as advanced nanotechnology will certainly accelerate its therapeutic applications.


Subject(s)
Drug Delivery Systems/methods , Exosomes/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Bioengineering/methods , Cell Communication/physiology , Humans , Neoplasms/pathology
5.
J Bioenerg Biomembr ; 49(4): 325-333, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28616679

ABSTRACT

It is becoming increasingly clear that mitochondria drive cellular functions and in vivo phenotypes by directing the production rate and abundance of metabolites that are proposed to function as signaling molecules (Chandel 2015; Selak et al. 2005; Etchegaray and Mostoslavsky 2016). Many of these metabolites are intermediates that make up cellular metabolism, part of which occur in mitochondria (i.e. the TCA and urea cycles), while others are produced "on demand" mainly in response to alterations in the microenvironment in order to participate in the activation of acute adaptive responses (Mills et al. 2016; Go et al. 2010). Reactive oxygen species (ROS) are well suited for the purpose of executing rapid and transient signaling due to their short lived nature (Bae et al. 2011). Hydrogen peroxide (H2O2), in particular, possesses important characteristics including diffusibility and faster reactivity with specific residues such as methionine, cysteine and selenocysteine (Bonini et al. 2014). Therefore, it is reasonable to propose that H2O2 functions as a relatively specific redox signaling molecule. Even though it is now established that mtH2O2 is indispensable, at least for hypoxic adaptation and energetic and/or metabolic homeostasis (Hamanaka et al. 2016; Guzy et al. 2005), the question of how H2O2 is produced and regulated in the mitochondria is only partially answered. In this review, some roles of this indispensable signaling molecule in driving cellular metabolism will be discussed. In addition, we will discuss how H2O2 formation in mitochondria depends on and is controlled by MnSOD. Finally, we will conclude this manuscript by highlighting why a better understanding of redox hubs in the mitochondria will likely lead to new and improved therapeutics of a number of diseases, including cancer.


Subject(s)
Mitochondria/metabolism , Signal Transduction , Superoxide Dismutase/physiology , Animals , Humans , Hydrogen Peroxide/metabolism , Oxidation-Reduction
6.
J Am Soc Nephrol ; 27(5): 1362-78, 2016 05.
Article in English | MEDLINE | ID: mdl-26424786

ABSTRACT

Obesity and diabetes mellitus are the leading causes of renal disease. In this study, we determined the regulation and role of the G protein-coupled bile acid receptor TGR5, previously shown to be regulated by high glucose and/or fatty acids, in obesity-related glomerulopathy (ORG) and diabetic nephropathy (DN). Treatment of diabetic db/db mice with the selective TGR5 agonist INT-777 decreased proteinuria, podocyte injury, mesangial expansion, fibrosis, and CD68 macrophage infiltration in the kidney. INT-777 also induced renal expression of master regulators of mitochondrial biogenesis, inhibitors of oxidative stress, and inducers of fatty acid ß-oxidation, including sirtuin 1 (SIRT1), sirtuin 3 (SIRT3), and Nrf-1. Increased activity of SIRT3 was evidenced by normalization of the increased acetylation of mitochondrial superoxide dismutase 2 (SOD2) and isocitrate dehydrogenase 2 (IDH2) observed in untreated db/db mice. Accordingly, INT-777 decreased mitochondrial H2O2 generation and increased the activity of SOD2, which associated with decreased urinary levels of H2O2 and thiobarbituric acid reactive substances. Furthermore, INT-777 decreased renal lipid accumulation. INT-777 also prevented kidney disease in mice with diet-induced obesity. In human podocytes cultured with high glucose, INT-777 induced mitochondrial biogenesis, decreased oxidative stress, and increased fatty acid ß-oxidation. Compared with normal kidney biopsy specimens, kidney specimens from patients with established ORG or DN expressed significantly less TGR5 mRNA, and levels inversely correlated with disease progression. Our results indicate that TGR5 activation induces mitochondrial biogenesis and prevents renal oxidative stress and lipid accumulation, establishing a role for TGR5 in inhibiting kidney disease in obesity and diabetes.


Subject(s)
Cholic Acids/pharmacology , Cholic Acids/therapeutic use , Diabetic Nephropathies/etiology , Diabetic Nephropathies/prevention & control , Kidney Diseases/etiology , Kidney Diseases/prevention & control , Obesity/complications , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/physiology , Animals , Bile Acids and Salts , Humans , Hydrogen Peroxide , Male , Mice , Oxidative Stress , Podocytes , Signal Transduction , Superoxide Dismutase
7.
Infect Immun ; 83(11): 4392-403, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26351282

ABSTRACT

The multifunctional-autoprocessing repeats-in-toxin (MARTXVv) toxin of Vibrio vulnificus plays a significant role in the pathogenesis of this bacterium through delivery of up to five effector domains to the host cells. Previous studies have established that the MARTXVv toxin is linked to V. vulnificus dependent induction of apoptosis, but the region of the large multifunction protein essential for this activity was not previously identified. Recently, we showed that the Makes Caterpillar Floppy-like MARTX effector domain (MCFVv) is an autoproteolytic cysteine protease that induces rounding of various cell types. In this study, we demonstrate that cell rounding induced by MCFVv is coupled to reduced metabolic rate and inhibition of cellular proliferation. Moreover, delivery of MCFVv into host cells either as a fusion to the N-terminal fragment of anthrax toxin lethal factor or when naturally delivered as a V. vulnificus MARTX toxin led to loss of mitochondrial membrane potential, release of cytochrome c, activation of Bax and Bak, and processing of caspases and poly-(ADP-ribose) polymerase (PARP-γ). These studies specifically link the MCFVv effector domain to induction of the intrinsic apoptosis pathway by V. vulnificus.


Subject(s)
Apoptosis , Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , Mitochondria/metabolism , Vibrio Infections/physiopathology , Vibrio vulnificus/metabolism , Bacterial Toxins/genetics , Caspases/metabolism , Cytochromes c/metabolism , Host-Pathogen Interactions , Humans , Membrane Potential, Mitochondrial , Mitochondria/enzymology , Protein Structure, Tertiary , Vibrio Infections/metabolism , Vibrio Infections/microbiology , Vibrio vulnificus/chemistry , Vibrio vulnificus/genetics
8.
Mol Cancer Ther ; 14(9): 2090-102, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26141949

ABSTRACT

Tumor adaptive resistance to therapeutic radiation remains a barrier for further improvement of local cancer control. SIRT3, a member of the sirtuin family of NAD(+)-dependent protein deacetylases in mitochondria, promotes metabolic homeostasis through regulation of mitochondrial protein deacetylation and plays a key role in prevention of cell aging. Here, we demonstrate that SIRT3 expression is induced in an array of radiation-treated human tumor cells and their corresponding xenograft tumors, including colon cancer HCT-116, glioblastoma U87, and breast cancer MDA-MB231 cells. SIRT3 transcriptional activation is due to SIRT3 promoter activation controlled by the stress transcription factor NF-κB. Posttranscriptionally, SIRT3 enzymatic activity is further enhanced via Thr150/Ser159 phosphorylation by cyclin B1-CDK1, which is also induced by radiation and relocated to mitochondria together with SIRT3. Cells expressing Thr150Ala/Ser159Ala-mutant SIRT3 show a reduction in mitochondrial protein lysine deacetylation, Δψm, MnSOD activity, and mitochondrial ATP generation. The clonogenicity of Thr150Ala/Ser159Ala-mutant transfectants is lower and significantly decreased under radiation. Tumors harboring Thr150Ala/Ser159Ala-mutant SIRT3 show inhibited growth and increased sensitivity to in vivo local irradiation. These results demonstrate that enhanced SIRT3 transcription and posttranslational modifications in mitochondria contribute to adaptive radioresistance in tumor cells. CDK1-mediated SIRT3 phosphorylation is a potential effective target to sensitize tumor cells to radiotherapy.


Subject(s)
Cyclin-Dependent Kinases/metabolism , Mitochondria/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Radiation Tolerance/genetics , Sirtuin 3/genetics , Transcriptional Activation , Acetylation , Animals , CDC2 Protein Kinase , Cell Line, Tumor , Disease Models, Animal , Enzyme Activation , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Mitochondria/radiation effects , Mitochondrial Proteins/metabolism , Mutation , NF-kappa B/metabolism , Neoplasms/pathology , Neoplasms/radiotherapy , Phosphorylation , Sirtuin 3/metabolism , Transcription, Genetic
9.
Surgery ; 158(3): 827-36, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26088922

ABSTRACT

BACKGROUND: Reactive oxygen species (ROS) contribute to adult tumorigenesis; however, their roles in pediatric solid tumors are unknown. Here, we sought to define the steady-state ROS levels in neuroblastoma and to examine whether aggressive cellular behavior, which may predict treatment failure, is regulated by ROS. METHODS: Neuroblastoma sections were assessed for 4-hydroxynonenal (4-HNE), a marker of intracellular lipid peroxidation and a byproduct of increased levels of ROS. Human neuroblastoma cell lines, MYCN-amplified BE(2)-C and MYCN-nonamplified SK-N-SH, were examined in our study. Superoxide and hydroperoxide oxidation products were detected by staining for dihydroethidium (DHE) and 5, 6-carboxy-2', 7'-dichlorodihydrofluorescein diacetate (CDCFH2), using the oxidation-insensitive analog CDCF as a negative control. Cells were treated with N-acetylcysteine (NAC; 10 mmol/L) daily for 5 days and analyzed. RESULTS: Greater expression of 4-HNE was observed in undifferentiated tumor sections as compared with the more differentiated tumors. Interestingly, increased levels of ROS were detected in MYCN-amplified BE(2)-C cells. Moreover, gastrin-releasing peptide receptor-induced ROS production stimulated upregulation of the hypoxia inducible factor (HIF)-1α/vascular endothelial growth factor (VEGF) pathway and an increase in cell growth. Antioxidant NAC decreased HIF-1α/VEGF expression and inhibited BE(2)-C cell growth. CONCLUSION: We report a novel observation that shifting the redox balance toward greater ROS levels results in a more aggressive neuroblastoma phenotype. Our data suggest that ROS play a critical role in refractory neuroblastoma.


Subject(s)
Acetylcysteine/pharmacology , Antioxidants/pharmacology , Biomarkers, Tumor/metabolism , Cell Proliferation/drug effects , Neuroblastoma/metabolism , Reactive Oxygen Species/antagonists & inhibitors , Blotting, Western , Cell Line, Tumor , Humans , Immunohistochemistry , Reactive Oxygen Species/metabolism
10.
Nat Commun ; 6: 7396, 2015 Jun 08.
Article in English | MEDLINE | ID: mdl-26051945

ABSTRACT

Ras (Rat sarcoma) protein is a central regulator of cell growth and proliferation. Mutations in the RAS gene are known to occur in human cancers and have been shown to contribute to carcinogenesis. In this study, we show that the multifunctional-autoprocessing repeats-in-toxin (MARTX) toxin-effector domain DUF5(Vv) from Vibrio vulnificus to be a site-specific endopeptidase that cleaves within the Switch 1 region of Ras and Rap1. DUF5(Vv) processing of Ras, which occurs both biochemically and in mammalian cell culture, inactivates ERK1/2, thereby inhibiting cell proliferation. The ability to cleave Ras and Rap1 is shared by DUF5(Vv) homologues found in other bacteria. In addition, DUF5(Vv )can cleave all Ras isoforms and KRas with mutations commonly implicated in malignancies. Therefore, we speculate that this new family of Ras/Rap1-specific endopeptidases (RRSPs) has potential to inactivate both wild-type and mutant Ras proteins expressed in malignancies.


Subject(s)
Bacterial Toxins/metabolism , Vibrio vulnificus/metabolism , rap1 GTP-Binding Proteins/metabolism , ras Proteins/metabolism , Amino Acid Sequence , HeLa Cells , Humans , Molecular Sequence Data , Sequence Homology, Amino Acid , rap1 GTP-Binding Proteins/chemistry , ras Proteins/chemistry
11.
Curr Opin Oncol ; 25(6): 630-6, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24048020

ABSTRACT

PURPOSE OF REVIEW: The purpose of this review is to highlight recent studies on mammalian sirtuins that coordinately regulate cellular metabolic homeostasis upon fasting and to summarize the beneficial effects of fasting on carcinogenesis and cancer therapy. RECENT FINDINGS: Recent studies have demonstrated that fasting may protect normal cells and mice from the metabolic conditions that are harmful as well as decrease the incidence of carcinogenesis. Fasting could also slow the tumor growth and augment the efficacy of certain systemic agents/chemotherapy drugs in various cancers. The mechanism behind this proposed idea may be due to, at least in some part, the metabolic regulation by Sirtuin family proteins whose functions are involved in specific aspects of longevity, stress response and metabolism. Sirtuins, particularly SIRT1 and SIRT3, can be activated by fasting and further exhibit their effects in insulin response, antioxidant defense, and glycolysis. Therefore, sirtuins may have anticancer effects by shifting metabolism to a less proliferative cell phenotype as well as less prone to oxidative stress attack. SUMMARY: The in-depth understanding of the essential role of sirtuins in fasting process may have significant implications in developing a new metabolic diagram of cancer prevention or treatment.


Subject(s)
Antioxidants/metabolism , Energy Metabolism , Fasting/metabolism , Neoplasms/metabolism , Oxidative Stress , Sirtuin 1/metabolism , Sirtuin 3/metabolism , Animals , Female , Glucose/metabolism , Glycolysis , Homeostasis , Humans , Insulin/metabolism , Male , Neoplasms/prevention & control , Neoplasms/therapy , Receptor Cross-Talk , Sirtuins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...