Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 170
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 19(11): 1236-1247, 2018 11.
Article in English | MEDLINE | ID: mdl-30323345

ABSTRACT

Although neutrophils have been linked to the formation of the pre-metastatic niche, the mechanism of their migration to distant, uninvolved tissues has remained elusive. We report that bone marrow neutrophils from mice with early-stage cancer exhibited much more spontaneous migration than that of control neutrophils from tumor-free mice. These cells lacked immunosuppressive activity but had elevated rates of oxidative phosphorylation and glycolysis, and increased production of ATP, relative to that of control neutrophils. Their enhanced spontaneous migration was mediated by autocrine ATP signaling through purinergic receptors. In ectopic tumor models and late stages of cancer, bone marrow neutrophils demonstrated potent immunosuppressive activity. However, these cells had metabolic and migratory activity indistinguishable from that of control neutrophils. A similar pattern of migration was observed for neutrophils and polymorphonuclear myeloid-derived suppressor cells from patients with cancer. These results elucidate the dynamic changes that neutrophils undergo in cancer and demonstrate the mechanism of neutrophils' contribution to early tumor dissemination.


Subject(s)
Chemotaxis, Leukocyte/immunology , Neoplasms/immunology , Neoplasms/pathology , Neutrophil Infiltration/immunology , Neutrophils/immunology , Aged , Animals , Disease Progression , Female , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged
2.
Immunity ; 44(2): 303-15, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26885857

ABSTRACT

Recruitment of monocytic myeloid-derived suppressor cells (MDSCs) and differentiation of tumor-associated macrophages (TAMs) are the major factors contributing to tumor progression and metastasis. We demonstrated that differentiation of TAMs in tumor site from monocytic precursors was controlled by downregulation of the activity of the transcription factor STAT3. Decreased STAT3 activity was caused by hypoxia and affected all myeloid cells but was not observed in tumor cells. Upregulation of CD45 tyrosine phosphatase activity in MDSCs exposed to hypoxia in tumor site was responsible for downregulation of STAT3. This effect was mediated by the disruption of CD45 protein dimerization regulated by sialic acid. Thus, STAT3 has a unique function in the tumor environment in controlling the differentiation of MDSC into TAM, and its regulatory pathway could be a potential target for therapy.


Subject(s)
Hypoxia/immunology , Leukocyte Common Antigens/metabolism , Macrophages/immunology , Phosphoric Monoester Hydrolases/metabolism , STAT3 Transcription Factor/metabolism , Animals , Cell Differentiation , Cell Movement , Cells, Cultured , Dimerization , Female , Leukocyte Common Antigens/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/immunology , Phosphoric Monoester Hydrolases/genetics , STAT3 Transcription Factor/genetics , Sialic Acids/metabolism , Tumor Microenvironment
3.
Proc Natl Acad Sci U S A ; 119(8)2022 02 22.
Article in English | MEDLINE | ID: mdl-35177476

ABSTRACT

Cancer metabolism, including in mitochondria, is a disease hallmark and therapeutic target, but its regulation is poorly understood. Here, we show that many human tumors have heterogeneous and often reduced levels of Mic60, or Mitofilin, an essential scaffold of mitochondrial structure. Despite a catastrophic collapse of mitochondrial integrity, loss of bioenergetics, and oxidative damage, tumors with Mic60 depletion slow down cell proliferation, evade cell death, and activate a nuclear gene expression program of innate immunity and cytokine/chemokine signaling. In turn, this induces epithelial-mesenchymal transition (EMT), activates tumor cell movements through exaggerated mitochondrial dynamics, and promotes metastatic dissemination in vivo. In a small-molecule drug screen, compensatory activation of stress response (GCN2) and survival (Akt) signaling maintains the viability of Mic60-low tumors and provides a selective therapeutic vulnerability. These data demonstrate that acutely damaged, "ghost" mitochondria drive tumor progression and expose an actionable therapeutic target in metastasis-prone cancers.


Subject(s)
Mitochondria/physiology , Neoplasm Metastasis/physiopathology , Neoplasms/genetics , Cell Death , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Epithelial-Mesenchymal Transition , Humans , Mitochondria/metabolism , Mitochondrial Dynamics/physiology , Mitochondrial Proteins/metabolism , Muscle Proteins/metabolism , Neoplasm Invasiveness/genetics , Neoplasms/metabolism , Neoplasms/physiopathology , Neoplastic Processes , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species , Signal Transduction
4.
J Biol Chem ; 299(6): 104774, 2023 06.
Article in English | MEDLINE | ID: mdl-37142218

ABSTRACT

Mitochondria are signaling organelles implicated in cancer, but the mechanisms are elusive. Here, we show that Parkin, an E3 ubiquitination (Ub) ligase altered in Parkinson's disease, forms a complex with the regulator of cell motility, Kindlin-2 (K2), at mitochondria of tumor cells. In turn, Parkin ubiquitinates Lys581 and Lys582 using Lys48 linkages, resulting in proteasomal degradation of K2 and shortened half-life from ∼5 h to ∼1.5 h. Loss of K2 inhibits focal adhesion turnover and ß1 integrin activation, impairs membrane lamellipodia size and frequency, and inhibits mitochondrial dynamics, altogether suppressing tumor cell-extracellular matrix interactions, migration, and invasion. Conversely, Parkin does not affect tumor cell proliferation, cell cycle transitions, or apoptosis. Expression of a Parkin Ub-resistant K2 Lys581Ala/Lys582Ala double mutant is sufficient to restore membrane lamellipodia dynamics, correct mitochondrial fusion/fission, and preserve single-cell migration and invasion. In a 3D model of mammary gland developmental morphogenesis, impaired K2 Ub drives multiple oncogenic traits of EMT, increased cell proliferation, reduced apoptosis, and disrupted basal-apical polarity. Therefore, deregulated K2 is a potent oncogene, and its Ub by Parkin enables mitochondria-associated metastasis suppression.


Subject(s)
Membrane Proteins , Ubiquitin-Protein Ligases , Cell Movement , Membrane Proteins/metabolism , Mitochondria/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Humans
5.
J Cell Sci ; 132(7)2019 04 04.
Article in English | MEDLINE | ID: mdl-30948431

ABSTRACT

Survivin (also known as BIRC5) is an evolutionarily conserved eukaryotic protein that is essential for cell division and can inhibit cell death. Normally it is only expressed in actively proliferating cells, but is upregulated in most, if not all cancers; consequently, it has received significant attention as a potential oncotherapeutic target. In this Cell Science at a Glance article and accompanying poster, we summarise our knowledge of survivin 21 years on from its initial discovery. We describe the structure, expression and function of survivin, highlight its interactome and conclude by describing anti-survivin strategies being trialled.


Subject(s)
Apoptosis , Mitosis , Neoplasms/metabolism , Survivin/metabolism , Humans , Molecular Targeted Therapy
6.
J Biol Chem ; 294(27): 10407-10414, 2019 07 05.
Article in English | MEDLINE | ID: mdl-31097545

ABSTRACT

The role of mitochondria in cancer continues to be debated, and whether exploitation of mitochondrial functions is a general hallmark of malignancy or a tumor- or context-specific response is still unknown. Using a variety of cancer cell lines and several technical approaches, including siRNA-mediated gene silencing, ChIP assays, global metabolomics and focused metabolite analyses, bioenergetics, and cell viability assays, we show that two oncogenic Myc proteins, c-Myc and N-Myc, transcriptionally control the expression of the mitochondrial chaperone TNFR-associated protein-1 (TRAP1) in cancer. In turn, this Myc-mediated regulation preserved the folding and function of mitochondrial oxidative phosphorylation (OXPHOS) complex II and IV subunits, dampened reactive oxygen species production, and enabled oxidative bioenergetics in tumor cells. Of note, we found that genetic or pharmacological targeting of this pathway shuts off tumor cell motility and invasion, kills Myc-expressing cells in a TRAP1-dependent manner, and suppresses primary and metastatic tumor growth in vivo We conclude that exploitation of mitochondrial functions is a general trait of tumorigenesis and that this reliance of cancer cells on mitochondrial OXPHOS pathways could offer an actionable therapeutic target in the clinic.


Subject(s)
HSP90 Heat-Shock Proteins/metabolism , Mitochondria/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Animals , Cell Line, Tumor , Cell Movement , Cell Survival/drug effects , Guanidines/pharmacology , Guanidines/therapeutic use , HSP90 Heat-Shock Proteins/genetics , Humans , Lactams, Macrocyclic/pharmacology , Lactams, Macrocyclic/therapeutic use , Liver Neoplasms/drug therapy , Liver Neoplasms/secondary , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Nude , Oxidative Phosphorylation , Promoter Regions, Genetic , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/genetics , RNA Interference , RNA, Small Interfering/metabolism , Transcription, Genetic
7.
Am J Pathol ; 189(6): 1180-1189, 2019 06.
Article in English | MEDLINE | ID: mdl-31079810

ABSTRACT

Easily accessible biomarkers that may inform on the metastatic potential of localized prostate cancer are urgently needed. Herein, we show that syntaphilin (SNPH), a molecule originally identified as a negative regulator of mitochondrial dynamics in neurons, is abundantly expressed in prostate cancer. SNPH distribution in prostate cancer is spatially biphasic, with high expression at the invasive front, correlating with increased proliferative rates, as determined by Ki-67 labeling, and reduced levels in the central tumor bulk, which are further decreased in patients with distant metastases. Higher levels of SNPH are observed with increasing Gleason grade. Prostate tumors predominantly express a novel, extraneuronal isoform of SNPH that accumulates in mitochondria and maintains oxidative metabolism and tumor cell proliferation. These data suggest that SNPH is a novel marker of high Gleason grade prostate cancer, differentially expressed at the invasive front compared with the central tumor bulk, and is potentially down-regulated in metastatic disease. This biphasic pattern of expression may reflect a dual function of SNPH in controlling the balance between cell proliferation and invasion in tumors.


Subject(s)
Biomarkers, Tumor/biosynthesis , Gene Expression Regulation , Membrane Proteins/biosynthesis , Neoplasm Proteins/biosynthesis , Nerve Tissue Proteins/biosynthesis , Prostatic Neoplasms/metabolism , Aged , Aged, 80 and over , Humans , Male , Middle Aged , Neoplasm Grading , Neoplasm Metastasis , Prostatic Neoplasms/pathology , Protein Isoforms/biosynthesis
8.
FASEB J ; 33(12): 13398-13411, 2019 12.
Article in English | MEDLINE | ID: mdl-31530011

ABSTRACT

The role of mitochondria in cancer continues to be debated and paradoxically implicated in opposing functions in tumor growth and tumor suppression. To understand this dichotomy, we explored the function of mitochondrial isocitrate dehydrogenase (IDH)2, a tricarboxylic acid cycle enzyme mutated in subsets of acute leukemias and gliomas, in cancer. Silencing of IDH2 in prostate cancer cells impaired oxidative bioenergetics, elevated reactive oxygen species (ROS) production, and promoted exaggerated mitochondrial dynamics. This was associated with increased subcellular mitochondrial trafficking, turnover of membrane focal adhesion complexes, and enhanced tumor cell migration and invasion, without changes in cell cycle progression. Mechanistically, loss of IDH2 caused ROS-dependent stabilization of hypoxia-inducible factor-1α in normoxia, which was required for increased mitochondrial trafficking and tumor cell movements. Therefore, IDH2 is a dual regulator of cancer bioenergetics and tumor cell motility. This pathway may reprogram mitochondrial dynamics to differentially adjust energy production or promote tumor cell invasion in response to microenvironment conditions.-Wang, Y., Agarwal, E., Bertolini, I., Ghosh, J. C., Seo, J. H., Altieri, D. C. IDH2 reprograms mitochondrial dynamics in cancer through a HIF-1α-regulated pseudohypoxic state.


Subject(s)
Energy Metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia/physiopathology , Isocitrate Dehydrogenase/metabolism , Mitochondria/metabolism , Mitochondrial Dynamics , Prostatic Neoplasms/pathology , Animals , Apoptosis , Cell Movement , Cell Proliferation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Isocitrate Dehydrogenase/genetics , Male , Mice , Mitochondria/pathology , Oxidation-Reduction , Prostatic Neoplasms/metabolism , Reactive Oxygen Species/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
Cell Mol Life Sci ; 76(5): 827-835, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30415375

ABSTRACT

Changes in cellular metabolism are now a recognized hallmark of cancer. Although this process is ripe with therapeutic potential in the clinic, its complexity and extraordinary plasticity have systematically defied dogmas and oversimplifications. Perhaps, best exemplifying this intricacy is the role of mitochondria in cancer, which in just a few years has gone from largely unnoticed to pivotal disease driver. The underlying mechanisms are only beginning to emerge. However, there is now clear evidence linking the dynamic nature of mitochondria to the machinery of tumor cell motility and metastatic spreading. These studies may open fresh therapeutic options for patients with disseminated cancer, currently an incurable and mostly lethal condition.


Subject(s)
Mitochondrial Dynamics/physiology , Neoplasm Metastasis , Animals , Cell Movement , Energy Metabolism , Humans , Neoplasms/pathology
10.
PLoS Biol ; 14(7): e1002507, 2016 07.
Article in English | MEDLINE | ID: mdl-27389535

ABSTRACT

Mitochondria must buffer the risk of proteotoxic stress to preserve bioenergetics, but the role of these mechanisms in disease is poorly understood. Using a proteomics screen, we now show that the mitochondrial unfoldase-peptidase complex ClpXP associates with the oncoprotein survivin and the respiratory chain Complex II subunit succinate dehydrogenase B (SDHB) in mitochondria of tumor cells. Knockdown of ClpXP subunits ClpP or ClpX induces the accumulation of misfolded SDHB, impairing oxidative phosphorylation and ATP production while activating "stress" signals of 5' adenosine monophosphate-activated protein kinase (AMPK) phosphorylation and autophagy. Deregulated mitochondrial respiration induced by ClpXP targeting causes oxidative stress, which in turn reduces tumor cell proliferation, suppresses cell motility, and abolishes metastatic dissemination in vivo. ClpP is universally overexpressed in primary and metastatic human cancer, correlating with shortened patient survival. Therefore, tumors exploit ClpXP-directed proteostasis to maintain mitochondrial bioenergetics, buffer oxidative stress, and enable metastatic competence. This pathway may provide a "drugable" therapeutic target in cancer.


Subject(s)
Endopeptidase Clp/metabolism , Energy Metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Endopeptidase Clp/genetics , Female , Humans , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Male , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Middle Aged , Mitochondria/genetics , Mitochondrial Proteins/genetics , Neoplasm Metastasis , Neoplasms/genetics , Neoplasms/pathology , Protein Subunits/genetics , Protein Subunits/metabolism , Proteomics/methods , RNA Interference , Succinate Dehydrogenase/genetics , Succinate Dehydrogenase/metabolism , Survivin , Transplantation, Heterologous
11.
Am J Respir Crit Care Med ; 198(1): 90-103, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29394093

ABSTRACT

RATIONALE: Pulmonary arterial hypertension (PAH) is a vascular remodeling disease with a poor prognosis and limited therapeutic options. Although the mechanisms contributing to vascular remodeling in PAH are still unclear, several features, including hyperproliferation and resistance to apoptosis of pulmonary artery smooth muscle cells (PASMCs), have led to the emergence of the cancer-like concept. The molecular chaperone HSP90 (heat shock protein 90) is directly associated with malignant growth and proliferation under stress conditions. In addition to being highly expressed in the cytosol, HSP90 exists in a subcellular pool compartmentalized in the mitochondria (mtHSP90) of tumor cells, but not in normal cells, where it promotes cell survival. OBJECTIVES: We hypothesized that mtHSP90 in PAH-PASMCs represents a protective mechanism against stress, promoting their proliferation and resistance to apoptosis. METHODS: Expression and localization of HSP90 were analyzed by Western blot, immunofluorescence, and immunogold electron microscopy. In vitro, effects of mtHSP90 inhibition on mitochondrial DNA integrity, bioenergetics, cell proliferation and resistance to apoptosis were assessed. In vivo, the therapeutic potential of Gamitrinib, a mitochondria-targeted HSP90 inhibitor, was tested in fawn-hooded and monocrotaline rats. MEASUREMENTS AND MAIN RESULTS: We demonstrated that, in response to stress, HSP90 preferentially accumulates in PAH-PASMC mitochondria (dual immunostaining, immunoblot, and immunogold electron microscopy) to ensure cell survival by preserving mitochondrial DNA integrity and bioenergetic functions. Whereas cytosolic HSP90 inhibition displays a lack of absolute specificity for PAH-PASMCs, Gamitrinib decreased mitochondrial DNA content and repair capacity and bioenergetic functions, thus repressing PAH-PASMC proliferation (Ki67 labeling) and resistance to apoptosis (Annexin V assay) without affecting control cells. In vivo, Gamitrinib improves PAH in two experimental rat models (monocrotaline and fawn-hooded rat). CONCLUSIONS: Our data show for the first time that accumulation of mtHSP90 is a feature of PAH-PASMCs and a key regulator of mitochondrial homeostasis contributing to vascular remodeling in PAH.


Subject(s)
Antihypertensive Agents/therapeutic use , HSP90 Heat-Shock Proteins/analysis , HSP90 Heat-Shock Proteins/metabolism , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/physiopathology , Mitochondria/metabolism , Vascular Remodeling/drug effects , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Cells, Cultured/drug effects , Disease Models, Animal , Humans , Muscle, Smooth, Vascular/drug effects , Rats
12.
Proc Natl Acad Sci U S A ; 112(28): 8638-43, 2015 Jul 14.
Article in English | MEDLINE | ID: mdl-26124089

ABSTRACT

Molecular therapies are hallmarks of "personalized" medicine, but how tumors adapt to these agents is not well-understood. Here we show that small-molecule inhibitors of phosphatidylinositol 3-kinase (PI3K) currently in the clinic induce global transcriptional reprogramming in tumors, with activation of growth factor receptors, (re)phosphorylation of Akt and mammalian target of rapamycin (mTOR), and increased tumor cell motility and invasion. This response involves redistribution of energetically active mitochondria to the cortical cytoskeleton, where they support membrane dynamics, turnover of focal adhesion complexes, and random cell motility. Blocking oxidative phosphorylation prevents adaptive mitochondrial trafficking, impairs membrane dynamics, and suppresses tumor cell invasion. Therefore, "spatiotemporal" mitochondrial respiration adaptively induced by PI3K therapy fuels tumor cell invasion, and may provide an important antimetastatic target.


Subject(s)
Enzyme Inhibitors/pharmacology , Mitochondria/drug effects , Neoplasm Invasiveness , Phosphoinositide-3 Kinase Inhibitors , Biological Transport , Cell Line, Tumor , Cell Movement/drug effects , Cytoskeleton/metabolism , Energy Metabolism , Humans , Mitochondria/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
13.
Semin Cell Dev Biol ; 39: 91-6, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25591986

ABSTRACT

Although technically a member of the Inhibitor of Apoptosis (IAP) gene family, survivin has consistently defied assumptions, refuted predictions and challenged paradigms. Despite its more than 5500 citations currently in Medline, the biology of survivin has remained fascinatingly complex, its exploitation in human disease, most notably cancer, tantalizing, and its regulation of cellular homeostasis unexpectedly far-reaching. An inconvenient outsider that resists schemes and dogmas, survivin continues to hold great promise to unlock fundamental circuitries of cellular functions in health and disease.


Subject(s)
Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Signal Transduction , Animals , Cell Division , Cell Survival , Humans , Inhibitor of Apoptosis Proteins/chemistry , Neoplasms/metabolism , Neoplasms/pathology , Protein Processing, Post-Translational , Survivin
14.
J Biol Chem ; 291(48): 25247-25254, 2016 Nov 25.
Article in English | MEDLINE | ID: mdl-27754870

ABSTRACT

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.


Subject(s)
Apoptosis , Cell Proliferation , HSP90 Heat-Shock Proteins/metabolism , Neoplasm Proteins/metabolism , Prostate/metabolism , Prostatic Neoplasms/metabolism , Animals , Cell Line, Tumor , HSP90 Heat-Shock Proteins/genetics , Humans , Male , Mice , Mice, Transgenic , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Prostate/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology
15.
Br J Cancer ; 117(3): 301-305, 2017 Jul 25.
Article in English | MEDLINE | ID: mdl-28677688

ABSTRACT

There is now a resurgent interest in the role of mitochondria in cancer. Long considered controversial or outright unimportant, mitochondrial biology is now increasingly recognised as an important tumour driver. The underlying mechanisms remain to be fully elucidated. But recent studies have uncovered a complex landscape where reprogramming of mitochondrial homoeostasis, including organelle dynamics, metabolic output, apoptosis control and redox status converge to promote tumour adaptation to an unfavourable microenvironment and inject new traits of aggressive disease. In particular, mechanisms of subcellular mitochondrial trafficking have unexpectedly emerged as central regulators of metastatic competence in disparate tumours. Some of these pathways are druggable, opening fresh therapeutic opportunities for advanced and disseminated disease.


Subject(s)
Mitochondria/physiology , Neoplasms/physiopathology , Adenosine Triphosphate/biosynthesis , Cell Movement , Cell Plasticity , Humans , Membrane Proteins , Mitochondria/metabolism , Neoplasm Metastasis/physiopathology , Neoplasms/genetics , Neoplasms/metabolism , Nerve Tissue Proteins/genetics , Neurons , Translational Research, Biomedical , Vesicular Transport Proteins/genetics
16.
Mol Cell ; 32(2): 159-60, 2008 Oct 24.
Article in English | MEDLINE | ID: mdl-18951080

ABSTRACT

Loss of endogenous tumor suppression is a critical step on the road to cancer. In a recent paper in Molecular Cell, Wang and colleagues provide evidence that inactivation of the pivotal BRCA1 tumor suppressor disrupts a safeguard gene network that opposes cell proliferation and cell survival.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Mammary Neoplasms, Experimental/genetics , Microtubule-Associated Proteins/physiology , Sirtuins/physiology , Animals , Cell Proliferation , Female , Genes, BRCA1 , Humans , Inhibitor of Apoptosis Proteins , Mammary Neoplasms, Experimental/metabolism , Mice , Microtubule-Associated Proteins/genetics , Repressor Proteins , Sirtuin 1 , Sirtuins/genetics , Survivin
17.
J Cell Physiol ; 230(1): 63-70, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24891298

ABSTRACT

Understanding the mechanisms that sustain pluripotency in human embryonic stem cells (hESCs) is an active area of research that may prove useful in regenerative medicine and will provide fundamental information relevant to development and cancer. hESCs and cancer cells share the unique ability to proliferate indefinitely and rapidly. Because the protein survivin is uniquely overexpressed in virtually all human cancers and in hESCs, we sought to investigate its role in supporting the distinctive capabilities of these cell types. Results presented here suggest that survivin contributes to the maintenance of pluripotency and that post-transcriptional control of survivin isoform expression is selectively regulated by microRNAs. miR-203 has been extensively studied in human tumors, but has not been characterized in hESCs. We show that miR-203 expression and activity is consistent with the expression and subcellular localization of survivin isoforms that in turn modulate expression of the Oct4 and Nanog transcription factors to sustain pluripotency. This study contributes to understanding of the complex regulatory mechanisms that govern whether hESCs proliferate or commit to lineages.


Subject(s)
Embryonic Stem Cells/physiology , Inhibitor of Apoptosis Proteins/metabolism , MicroRNAs/biosynthesis , Pluripotent Stem Cells/physiology , Cell Differentiation/genetics , Cell Proliferation , Cells, Cultured , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/biosynthesis , Humans , Inhibitor of Apoptosis Proteins/biosynthesis , Inhibitor of Apoptosis Proteins/genetics , MicroRNAs/genetics , Nanog Homeobox Protein , Octamer Transcription Factor-3/biosynthesis , Protein Isoforms/genetics , RNA Interference , RNA, Messenger/biosynthesis , RNA, Messenger/metabolism , RNA, Small Interfering , Survivin
18.
Pharmacol Res ; 102: 42-5, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26365877

ABSTRACT

Recent studies have demonstrated that tumor cells exposed to molecular therapy with PI3K antagonists redistribute their mitochondria to the peripheral cytoskeleton, fueling membrane dynamics, turnover of focal adhesion complexes and increased tumor cell motility and invasion. Although this process paradoxically increases metastatic propensity during molecular therapy, it also emphasizes a critical role of regional mitochondrial bioenergetics in tumor metabolic reprogramming and may offer prime therapeutic opportunities to prevent disseminated disease.


Subject(s)
Antineoplastic Agents/therapeutic use , Mitochondria/drug effects , Neoplasms/drug therapy , Cell Adhesion/drug effects , Cell Movement/drug effects , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Cytoskeleton/pathology , Focal Adhesions/drug effects , Focal Adhesions/metabolism , Focal Adhesions/pathology , Humans , Mitochondria/metabolism , Mitochondria/pathology , Neoplasms/metabolism , Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism
19.
J Biol Chem ; 288(8): 5553-61, 2013 Feb 22.
Article in English | MEDLINE | ID: mdl-23303179

ABSTRACT

Mitochondria control bioenergetics and cell fate decisions, but how they influence nuclear gene expression is understood poorly. Here, we show that deletion or reduction in the levels of cyclophilin D (CypD, also called Ppif), a mitochondrial matrix peptidyl prolyl isomerase and apoptosis regulator, results in increased cell proliferation and enhanced cell migration and invasion. These responses are associated with extensive transcriptional changes, modulation of a chemokine/chemokine receptor gene signature, and activation of the pleiotropic inflammatory mediator, STAT3. In the absence of CypD, active STAT3 enhances cell proliferation via accelerated entry into S-phase and stimulates autocrine/paracrine cell motility through Cxcl12-Cxcr4-directed chemotaxis. Therefore, CypD directs mitochondria-to-nuclei inflammatory gene expression in normal and tumor cells. This pathway may contribute to malignant traits under conditions of CypD modulation.


Subject(s)
Chemokines/metabolism , Cyclophilins/metabolism , Mitochondria/metabolism , Animals , Apoptosis , Cell Cycle , Cell Line, Tumor , Cell Lineage , Cell Movement , Cell Proliferation , Cell Survival , Peptidyl-Prolyl Isomerase F , Gene Expression Profiling , Gene Expression Regulation , Gene Expression Regulation, Neoplastic , Humans , Mice , NIH 3T3 Cells , RNA, Small Interfering/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction
20.
Breast Cancer Res ; 16(3): R55, 2014 May 30.
Article in English | MEDLINE | ID: mdl-24886669

ABSTRACT

INTRODUCTION: The molecular determinants of breast cancer resistance to first-line anthracycline-containing chemotherapy are unknown. METHODS: We examined the response to doxorubicin of organotypic cultures of primary human breast tumors ex vivo with respect to cell proliferation, DNA damage and modulation of apoptosis. Samples were analyzed for genome-wide modulation of cell death pathways, differential activation of p53, and the role of survivin family molecules in drug resistance. Rational drug combination regimens were explored by high-throughput screening, and validated in model breast cancer cell types. RESULTS: Doxorubicin treatment segregated organotypic human breast tumors into distinct Responder or Non Responder groups, characterized by differential proliferative index, stabilization of p53, and induction of apoptosis. Conversely, tumor histotype, hormone receptor or human epidermal growth factor receptor-2 (HER2) status did not influence chemotherapy sensitivity. Global analysis of cell death pathways identified survivin and its alternatively spliced form, survivin-ΔEx3 as uniquely overexpressed in Non Responder breast tumors. Forced expression of survivin-ΔEx3 preserved cell viability and prevented doxorubicin-induced apoptosis in breast cancer cell types. High-throughput pharmacologic targeting of survivin family proteins with a small-molecule survivin suppressant currently in the clinic (YM155) selectively potentiated the effect of doxorubicin, but not other chemotherapeutics in breast cancer cell types, and induced tumor cell apoptosis. CONCLUSIONS: Survivin family proteins are novel effectors of doxorubicin resistance in chemotherapy-naive breast cancer. The incorporation of survivin antagonist(s) in anthracycline-containing regimens may have improved clinical activity in these patients.


Subject(s)
Breast Neoplasms/drug therapy , Doxorubicin/therapeutic use , Drug Resistance, Neoplasm/genetics , Imidazoles/pharmacology , Inhibitor of Apoptosis Proteins/genetics , Naphthoquinones/pharmacology , Alternative Splicing , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Breast Neoplasms/genetics , Camptothecin/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival , DNA Damage/drug effects , Doxorubicin/pharmacology , Etoposide/pharmacology , Female , High-Throughput Screening Assays , Humans , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , MCF-7 Cells , Paclitaxel/pharmacology , Receptor, ErbB-2/metabolism , Survivin , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL