Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 299(10): 105186, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37611829

ABSTRACT

Loss of protein kinase Cδ (PKCδ) activity renders cells resistant to DNA damaging agents, including irradiation; however, the mechanism(s) underlying resistance is poorly understood. Here, we have asked if metabolic reprogramming by PKCδ contributes to radioprotection. Analysis of global metabolomics showed that depletion of PKCδ affects metabolic pathways that control energy production and antioxidant, nucleotide, and amino acid biosynthesis. Increased NADPH and nucleotide production in PKCδ-depleted cells is associated with upregulation of the pentose phosphate pathway (PPP) as evidenced by increased activation of G6PD and an increase in the nucleotide precursor, 5-phosphoribosyl-1-pyrophosphate. Stable isotope tracing with U-[13C6] glucose showed reduced utilization of glucose for glycolysis in PKCδ-depleted cells and no increase in U-[13C6] glucose incorporation into purines or pyrimidines. In contrast, isotope tracing with [13C5, 15N2] glutamine showed increased utilization of glutamine for synthesis of nucleotides, glutathione, and tricarboxylic acid intermediates and increased incorporation of labeled glutamine into pyruvate and lactate. Using a glycolytic rate assay, we confirmed that anaerobic glycolysis is increased in PKCδ-depleted cells; this was accompanied by a reduction in oxidative phosphorylation, as assayed using a mitochondrial stress assay. Importantly, pretreatment of cells with specific inhibitors of the PPP or glutaminase prior to irradiation reversed radioprotection in PKCδ-depleted cells, indicating that these cells have acquired codependency on the PPP and glutamine for survival. Our studies demonstrate that metabolic reprogramming to increase utilization of glutamine and nucleotide synthesis contributes to radioprotection in the context of PKCδ inhibition.

2.
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
3.
Cancers (Basel) ; 14(14)2022 Jul 21.
Article in English | MEDLINE | ID: mdl-35884604

ABSTRACT

Triple-negative breast cancer (TNBC) often undergoes at least partial epithelial-to-mesenchymal transition (EMT) to facilitate metastasis. Identifying EMT-associated characteristics can reveal novel dependencies that may serve as therapeutic vulnerabilities in this aggressive breast cancer subtype. We found that NPC1, which encodes the lysosomal cholesterol transporter Niemann-Pick type C1 is highly expressed in TNBC as compared to estrogen receptor-positive (ER+) breast cancer, and is significantly elevated in high-grade disease. We demonstrated that NPC1 is directly targeted by microRNA-200c (miR-200c), a potent suppressor of EMT, providing a mechanism for its differential expression in breast cancer subtypes. The silencing of NPC1 in TNBC causes an accumulation of cholesterol-filled lysosomes, and drives decreased growth in soft agar and invasive capacity. Conversely, overexpression of NPC1 in an ER+ cell line increases invasion and growth in soft agar. We further identified TNBC cell lines as cholesterol auxotrophs, however, they do not solely depend on NPC1 for adequate cholesterol supply. The silencing of NPC1 in TNBC cell lines led to altered mitochondrial function and morphology, suppression of mTOR signaling, and accumulation of autophagosomes. A small molecule inhibitor of NPC1, U18666A, decreased TNBC proliferation and synergized with the chemotherapeutic drug, paclitaxel. This work suggests that NPC1 promotes aggressive characteristics in TNBC, and identifies NPC1 as a potential therapeutic target.

4.
Front Cell Dev Biol ; 10: 849962, 2022.
Article in English | MEDLINE | ID: mdl-35356277

ABSTRACT

Mitochondria are highly dynamic organelles which can change their shape, via processes termed fission and fusion, in order to adapt to different environmental and developmental contexts. Due to the importance of these processes in maintaining a physiologically healthy pool of mitochondria, aberrant cycles of fission/fusion are often seen in pathological contexts. In this review we will discuss how dysregulated fission and fusion promote tumor progression. We focus on the molecular mechanisms involved in fission and fusion, discussing how altered mitochondrial fission and fusion change tumor cell growth, metabolism, motility, and invasion and, finally how changes to these tumor-cell intrinsic phenotypes directly and indirectly impact tumor progression to metastasis. Although this is an emerging field of investigation, the current consensus is that mitochondrial fission positively influences metastatic potential in a broad variety of tumor types. As mitochondria are now being investigated as vulnerable targets in a variety of cancer types, we underscore the importance of their dynamic nature in potentiating tumor progression.

5.
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
6.
Mol Cancer Res ; 20(4): 607-621, 2022 04 01.
Article in English | MEDLINE | ID: mdl-34992146

ABSTRACT

There is a continued need to identify novel therapeutic targets to prevent the mortality associated with prostate cancer. In this context, mitochondrial Rho GTPase 2 (MIRO2) mRNA was upregulated in metastatic prostate cancer compared with localized tumors, and higher MIRO2 levels were correlated with poor patient survival. Using human cell lines that represent androgen-independent or -sensitive prostate cancer, we showed that MIRO2 depletion impaired cell growth, colony formation, and tumor growth in mice. Network analysis of MIRO2's binding partners identified metabolism and cellular responses to extracellular stimuli as top overrepresented pathways. The top hit on our screen, General Control Nonderepressible 1 (GCN1), was overexpressed in prostate cancer, and interacted with MIRO2 in prostate cancer cell lines and in primary prostate cancer cells. Functional analysis of MIRO2 mutations present in patients with prostate cancer led to the identification of MIRO2 159L, which increased GCN1 binding. Importantly, MIRO2 was necessary for efficient GCN1-mediated GCN2 kinase signaling and induction of the transcription factor activating transcription factor 4 (ATF4) levels. Further, MIRO2's effect on regulating prostate cancer cell growth was mediated by ATF4. Finally, levels of activated GCN2 and ATF4 were correlated with MIRO2 expression in prostate cancer xenografts. Both MIRO2 and activated GCN2 levels were higher in hypoxic areas of prostate cancer xenografts. Overall, we propose that targeting the MIRO2-GCN1 axis may be a valuable strategy to halt prostate cancer growth. IMPLICATIONS: MIRO2/GCN1/GCN2 constitute a novel mitochondrial signaling pathway that controls androgen-independent and androgen-sensitive prostate cancer cell growth.


Subject(s)
Prostatic Neoplasms , Animals , Humans , Male , Mice , Peptide Elongation Factors/genetics , Peptide Elongation Factors/metabolism , Prostatic Neoplasms/genetics , Protein Serine-Threonine Kinases , RNA-Binding Proteins/metabolism , Signal Transduction , Trans-Activators/metabolism
7.
Dev Cell ; 56(14): 2029-2042.e5, 2021 07 26.
Article in English | MEDLINE | ID: mdl-34171288

ABSTRACT

Mitochondria are critical metabolic and signaling hubs, and dysregulated mitochondrial homeostasis is implicated in many diseases. Degradation of damaged mitochondria by selective GABARAP/LC3-dependent macro-autophagy (mitophagy) is critical for maintaining mitochondrial homeostasis. To identify alternate forms of mitochondrial quality control that functionally compensate if mitophagy is inactive, we selected for autophagy-dependent cancer cells that survived loss of LC3-dependent autophagosome formation caused by inactivation of ATG7 or RB1CC1/FIP200. We discovered rare surviving autophagy-deficient clones that adapted to maintain mitochondrial homeostasis after gene inactivation and identified two enhanced mechanisms affecting mitochondria including mitochondrial dynamics and mitochondrial-derived vesicles (MDVs). To further understand these mechanisms, we quantified MDVs via flow cytometry and confirmed an SNX9-mediated mechanism necessary for flux of MDVs to lysosomes. We show that the autophagy-dependent cells acquire unique dependencies on these processes, indicating that these alternate forms of mitochondrial homeostasis compensate for loss of autophagy to maintain mitochondrial health.


Subject(s)
Autophagy , Microtubule-Associated Proteins/metabolism , Mitochondria/pathology , Mitochondrial Dynamics , Mitophagy , Sorting Nexins/metabolism , Transport Vesicles/physiology , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein 7/metabolism , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/metabolism , Endosomes/metabolism , Humans , Lysosomes , Microtubule-Associated Proteins/genetics , Mitochondria/metabolism , Sorting Nexins/genetics
8.
Cancer Rep (Hoboken) ; 3(1): e1157, 2020 02.
Article in English | MEDLINE | ID: mdl-32671955

ABSTRACT

BACKGROUND: Mammalian cells must constantly reprogram the distribution of mitochondria in order to meet the local demands for energy, calcium, redox balance, and other mitochondrial functions. Mitochondrial localization inside the cell is a result of a combination of movement along the microtubule tracks plus anchoring to actin filaments. RECENT FINDINGS: Recent advances show that subcellular distribution of mitochondria can regulate tumor cell growth, proliferation/motility plasticity, metastatic competence, and therapy responses in tumors. In this review, we discuss our current understanding of the mechanisms by which mitochondrial subcellular distribution is regulated in tumor cells. CONCLUSIONS: Mitochondrial trafficking is dysregulated in tumors. Accumulation of mitochondria at the leading edge of the cell supports energy expensive processes of focal adhesion dynamics, cell membrane dynamics, migration, and invasion.


Subject(s)
Mitochondria/physiology , Neoplasm Metastasis , Adenosine Triphosphate/metabolism , Animals , Cell Movement , Focal Adhesions , Humans , Membrane Proteins/physiology , Microtubules/physiology , Myosin Heavy Chains/physiology , Myosin Type V/physiology , Myosins/physiology , Neoplasm Invasiveness , Nerve Tissue Proteins/physiology , Signal Transduction
9.
Sci Signal ; 13(642)2020 07 28.
Article in English | MEDLINE | ID: mdl-32723812

ABSTRACT

Mitochondria are signaling hubs in eukaryotic cells. Here, we showed that the mitochondrial FUN14 domain-containing protein-1 (FUNDC1), an effector of Parkin-independent mitophagy, also participates in cellular plasticity by sustaining oxidative bioenergetics, buffering ROS production, and supporting cell proliferation. Targeting this pathway in cancer cells suppressed tumor growth but rendered transformed cells more motile and invasive in a manner dependent on ROS-mediated mitochondrial dynamics and mitochondrial repositioning to the cortical cytoskeleton. Global metabolomics and proteomics profiling identified a FUNDC1 interactome at the mitochondrial inner membrane, comprising the AAA+ protease, LonP1, and subunits of oxidative phosphorylation, complex V (ATP synthase). Independently of its previously identified role in mitophagy, FUNDC1 enabled LonP1 proteostasis, which in turn preserved complex V function and decreased ROS generation. Therefore, mitochondrial reprogramming by a FUNDC1-LonP1 axis controls tumor cell plasticity by switching between proliferative and invasive states in cancer.


Subject(s)
Membrane Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Mitophagy , Neoplasm Proteins/metabolism , Neoplasms/metabolism , A549 Cells , Animals , Humans , MCF-7 Cells , Membrane Proteins/genetics , Mice , Mitochondrial Proteins/genetics , NIH 3T3 Cells , Neoplasm Proteins/genetics , Neoplasms/genetics , PC-3 Cells
10.
Cancer Discov ; 10(9): 1282-1295, 2020 09.
Article in English | MEDLINE | ID: mdl-32499221

ABSTRACT

Older patients with melanoma (>50 years old) have poorer prognoses and response rates to targeted therapy compared with young patients (<50 years old), which can be driven, in part, by the aged microenvironment. Here, we show that aged dermal fibroblasts increase the secretion of neutral lipids, especially ceramides. When melanoma cells are exposed to the aged fibroblast lipid secretome, or cocultured with aged fibroblasts, they increase the uptake of lipids via the fatty acid transporter FATP2, which is upregulated in melanoma cells in the aged microenvironment and known to play roles in lipid synthesis and accumulation. We show that blocking FATP2 in melanoma cells in an aged microenvironment inhibits their accumulation of lipids and disrupts their mitochondrial metabolism. Inhibiting FATP2 overcomes age-related resistance to BRAF/MEK inhibition in animal models, ablates tumor relapse, and significantly extends survival time in older animals. SIGNIFICANCE: These data show that melanoma cells take up lipids from aged fibroblasts, via FATP2, and use them to resist targeted therapy. The response to targeted therapy is altered in aged individuals because of the influences of the aged microenvironment, and these data suggest FATP2 as a target to overcome resistance.See related commentary by Montal and White, p. 1255.This article is highlighted in the In This Issue feature, p. 1241.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Coenzyme A Ligases/metabolism , Fibroblasts/metabolism , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Cellular Senescence , Coculture Techniques , Coenzyme A Ligases/antagonists & inhibitors , Dermis/cytology , Dermis/pathology , Drug Resistance, Neoplasm/drug effects , Humans , Keratinocytes/metabolism , Lipid Metabolism , Melanoma/pathology , Molecular Targeted Therapy/methods , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Skin Neoplasms/pathology , Tumor Microenvironment
11.
Cancer Lett ; 471: 72-87, 2020 02 28.
Article in English | MEDLINE | ID: mdl-31838085

ABSTRACT

Androgen receptor (AR) signaling plays a central role in metabolic reprogramming for prostate cancer (PCa) growth and progression. Mitochondria are metabolic powerhouses of the cell and support several hallmarks of cancer. However, the molecular links between AR signaling and the mitochondria that support the metabolic demands of PCa cells are poorly understood. Here, we demonstrate increased levels of dynamin-related protein 1 (DRP1), a mitochondrial fission mediator, in androgen-sensitive and castration-resistant AR-driven PCa. AR signaling upregulates DRP1 to form the VDAC-MPC2 complex, increases pyruvate transport into mitochondria, and supports mitochondrial metabolism, including oxidative phosphorylation and lipogenesis. DRP1 inhibition activates the cellular metabolic stress response, which involves AMPK phosphorylation, induction of autophagy, and the ER unfolded protein response, and attenuates androgen-induced proliferation. Additionally, DRP1 expression facilitates PCa cell survival under diverse metabolic stress conditions, including hypoxia and oxidative stress. Moreover, we found that increased DRP1 expression was indicative of poor prognosis in patients with castration-resistant PCa. Collectively, our findings link androgen signaling-mediated mitochondrial dynamics to metabolic reprogramming; moreover, they have important implications for understanding PCa progression.


Subject(s)
Androgens/metabolism , Dynamins/biosynthesis , Mitochondria/metabolism , Prostatic Neoplasms, Castration-Resistant/metabolism , Cell Line, Tumor , Cell Proliferation/physiology , Citric Acid Cycle , Dihydrotestosterone/pharmacology , Dynamins/antagonists & inhibitors , Dynamins/genetics , Dynamins/metabolism , Gene Knockdown Techniques , Humans , Male , Mitochondrial Dynamics , Mitochondrial Membrane Transport Proteins/metabolism , Oxidative Phosphorylation , PC-3 Cells , Prostatic Neoplasms, Castration-Resistant/pathology , Pyruvates/metabolism , Receptors, Androgen/metabolism , Signal Transduction , Up-Regulation , Voltage-Dependent Anion Channels/metabolism
12.
Cancer Res ; 78(15): 4215-4228, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29898993

ABSTRACT

Syntaphilin (SNPH) inhibits the movement of mitochondria in tumor cells, preventing their accumulation at the cortical cytoskeleton and limiting the bioenergetics of cell motility and invasion. Although this may suppress metastasis, the regulation of the SNPH pathway is not well understood. Using a global proteomics screen, we show that SNPH associates with multiple regulators of ubiquitin-dependent responses and is ubiquitinated by the E3 ligase CHIP (or STUB1) on Lys111 and Lys153 in the microtubule-binding domain. SNPH ubiquitination did not result in protein degradation, but instead anchored SNPH on tubulin to inhibit mitochondrial motility and cycles of organelle fusion and fission, that is dynamics. Expression of ubiquitination-defective SNPH mutant Lys111→Arg or Lys153→Arg increased the speed and distance traveled by mitochondria, repositioned mitochondria to the cortical cytoskeleton, and supported heightened tumor chemotaxis, invasion, and metastasis in vivo Interference with SNPH ubiquitination activated mitochondrial dynamics, resulting in increased recruitment of the fission regulator dynamin-related protein-1 (Drp1) to mitochondria and Drp1-dependent tumor cell motility. These data uncover nondegradative ubiquitination of SNPH as a key regulator of mitochondrial trafficking and tumor cell motility and invasion. In this way, SNPH may function as a unique, ubiquitination-regulated suppressor of metastasis.Significance: These findings reveal a new mechanism of metastasis suppression by establishing the role of SNPH ubiquitination in inhibiting mitochondrial dynamics, chemotaxis, and metastasis. Cancer Res; 78(15); 4215-28. ©2018 AACR.


Subject(s)
Cell Movement/physiology , Mitochondria/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Nerve Tissue Proteins/metabolism , Ubiquitination/physiology , Vesicular Transport Proteins/metabolism , Animals , Cell Line , Cell Line, Tumor , Cytoskeleton/metabolism , Cytoskeleton/physiology , Dynamins/metabolism , Humans , Male , Membrane Proteins , Mice , Mice, Inbred C57BL , Mitochondria/pathology , Mitochondrial Dynamics/physiology , NIH 3T3 Cells , PC-3 Cells , Ubiquitin-Protein Ligases/metabolism
13.
Article in English | MEDLINE | ID: mdl-29755415

ABSTRACT

Mitochondria are bioenergetic, biosynthetic, and signaling organelles that control various aspects of cellular and organism homeostasis. Quality control mechanisms are in place to ensure maximal mitochondrial function and metabolic homeostasis at the cellular level. Dysregulation of these pathways is a common theme in human disease. In this mini-review, we discuss how alterations of the mitochondrial network influences mitochondrial function, focusing on the molecular regulators of mitochondrial dynamics (organelle's shape and localization). We highlight similarities and critical differences in the mitochondrial network of cancer and type 2 diabetes, which may be relevant for treatment of these diseases.

14.
J Clin Invest ; 127(10): 3755-3769, 2017 Oct 02.
Article in English | MEDLINE | ID: mdl-28891816

ABSTRACT

Tumors adapt to an unfavorable microenvironment by controlling the balance between cell proliferation and cell motility, but the regulators of this process are largely unknown. Here, we show that an alternatively spliced isoform of syntaphilin (SNPH), a cytoskeletal regulator of mitochondrial movements in neurons, is directed to mitochondria of tumor cells. Mitochondrial SNPH buffers oxidative stress and maintains complex II-dependent bioenergetics, sustaining local tumor growth while restricting mitochondrial redistribution to the cortical cytoskeleton and tumor cell motility. Conversely, introduction of stress stimuli to the microenvironment, including hypoxia, acutely lowered SNPH levels, resulting in bioenergetics defects and increased superoxide production. In turn, this suppressed tumor cell proliferation but increased tumor cell invasion via greater mitochondrial trafficking to the cortical cytoskeleton. Loss of SNPH or expression of an SNPH mutant lacking the mitochondrial localization sequence resulted in increased metastatic dissemination in xenograft or syngeneic tumor models in vivo. Accordingly, tumor cells that acquired the ability to metastasize in vivo constitutively downregulated SNPH and exhibited higher oxidative stress, reduced cell proliferation, and increased cell motility. Therefore, SNPH is a stress-regulated mitochondrial switch of the cell proliferation-motility balance in cancer, and its pathway may represent a therapeutic target.


Subject(s)
Cell Proliferation , Microtubule-Associated Proteins/metabolism , Mitochondria/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Nerve Tissue Proteins/metabolism , Vesicular Transport Proteins/metabolism , A549 Cells , Animals , Humans , MCF-7 Cells , Membrane Proteins , Mice , Microtubule-Associated Proteins/genetics , Mitochondria/genetics , Mitochondria/pathology , NIH 3T3 Cells , Neoplasm Metastasis , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology , Nerve Tissue Proteins/genetics , Vesicular Transport Proteins/genetics
15.
Nat Commun ; 7: 13730, 2016 12 19.
Article in English | MEDLINE | ID: mdl-27991488

ABSTRACT

The role of mitochondria in cancer is controversial. Using a genome-wide shRNA screen, we now show that tumours reprogram a network of mitochondrial dynamics operative in neurons, including syntaphilin (SNPH), kinesin KIF5B and GTPase Miro1/2 to localize mitochondria to the cortical cytoskeleton and power the membrane machinery of cell movements. When expressed in tumours, SNPH inhibits the speed and distance travelled by individual mitochondria, suppresses organelle dynamics, and blocks chemotaxis and metastasis, in vivo. Tumour progression in humans is associated with downregulation or loss of SNPH, which correlates with shortened patient survival, increased mitochondrial trafficking to the cortical cytoskeleton, greater membrane dynamics and heightened cell invasion. Therefore, a SNPH network regulates metastatic competence and may provide a therapeutic target in cancer.


Subject(s)
Kinesins/metabolism , Mitochondrial Dynamics/physiology , Mitochondrial Proteins/metabolism , Neoplasm Metastasis/physiopathology , Nerve Tissue Proteins/metabolism , Vesicular Transport Proteins/metabolism , rho GTP-Binding Proteins/metabolism , Down-Regulation , Gene Expression Regulation, Neoplastic , Humans , Kinesins/genetics , Membrane Proteins , Metabolic Networks and Pathways/physiology , Mitochondrial Proteins/genetics , rho GTP-Binding Proteins/genetics
16.
Cancer Cell ; 30(2): 257-272, 2016 08 08.
Article in English | MEDLINE | ID: mdl-27505672

ABSTRACT

Hypoxia is a universal driver of aggressive tumor behavior, but the underlying mechanisms are not completely understood. Using a phosphoproteomics screen, we now show that active Akt accumulates in the mitochondria during hypoxia and phosphorylates pyruvate dehydrogenase kinase 1 (PDK1) on Thr346 to inactivate the pyruvate dehydrogenase complex. In turn, this pathway switches tumor metabolism toward glycolysis, antagonizes apoptosis and autophagy, dampens oxidative stress, and maintains tumor cell proliferation in the face of severe hypoxia. Mitochondrial Akt-PDK1 signaling correlates with unfavorable prognostic markers and shorter survival in glioma patients and may provide an "actionable" therapeutic target in cancer.


Subject(s)
Cellular Reprogramming/physiology , Mitochondria/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Hypoxia/physiology , Cell Line, Tumor , Cell Proliferation/physiology , Female , Humans , Male , Mice , Mice, Inbred NOD , Mice, Nude , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Signal Transduction
17.
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
18.
Clin Cancer Res ; 22(3): 540-5, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26660517

ABSTRACT

Small-molecule inhibitors of the phosphoinositide 3-kinase (PI3K), Akt, and mTOR pathway currently in the clinic produce a paradoxical reactivation of the pathway they are intended to suppress. Furthermore, fresh experimental evidence with PI3K antagonists in melanoma, glioblastoma, and prostate cancer shows that mitochondrial metabolism drives an elaborate process of tumor adaptation culminating with drug resistance and metastatic competency. This is centered on reprogramming of mitochondrial functions to promote improved cell survival and to fuel the machinery of cell motility and invasion. Key players in these responses are molecular chaperones of the Hsp90 family compartmentalized in mitochondria, which suppress apoptosis via phosphorylation of the pore component, Cyclophilin D, and enable the subcellular repositioning of active mitochondria to membrane protrusions implicated in cell motility. An inhibitor of mitochondrial Hsp90s in preclinical development (gamitrinib) prevents adaptive mitochondrial reprogramming and shows potent antitumor activity in vitro and in vivo. Other therapeutic strategies to target mitochondria for cancer therapy include small-molecule inhibitors of mutant isocitrate dehydrogenase (IDH) IDH1 (AG-120) and IDH2 (AG-221), which opened new therapeutic prospects for patients with high-risk acute myelogenous leukemia (AML). A second approach of mitochondrial therapeutics focuses on agents that elevate toxic ROS levels from a leaky electron transport chain; nevertheless, the clinical experience with these compounds, including a quinone derivative, ARQ 501, and a copper chelator, elesclomol (STA-4783) is limited. In light of this evidence, we discuss how best to target a resurgence of mitochondrial bioenergetics for cancer therapy.


Subject(s)
Mitochondria/metabolism , Neoplasms/metabolism , Signal Transduction , Adaptation, Biological , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Clinical Trials as Topic , Disease Progression , Drug Evaluation, Preclinical , Humans , Mitochondria/drug effects , Molecular Targeted Therapy , Neoplasms/pathology , Neoplasms/therapy , Signal Transduction/drug effects , Translational Research, Biomedical
19.
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
20.
Sci Signal ; 8(389): ra80, 2015 Aug 11.
Article in English | MEDLINE | ID: mdl-26268608

ABSTRACT

Survivin promotes cell division and suppresses apoptosis in many human cancers, and increased abundance correlates with metastasis and poor prognosis. We showed that a pool of survivin that localized to the mitochondria of certain tumor cell lines enhanced the stability of oxidative phosphorylation complex II, which promoted cellular respiration. Survivin also supported the subcellular trafficking of mitochondria to the cortical cytoskeleton of tumor cells, which was associated with increased membrane ruffling, increased focal adhesion complex turnover, and increased tumor cell migration and invasion in cultured cells, and enhanced metastatic dissemination in vivo. Therefore, we found that mitochondrial respiration enhanced by survivin contributes to cancer metabolism, and relocalized mitochondria may provide a "regional" energy source to fuel tumor cell invasion and metastasis.


Subject(s)
Inhibitor of Apoptosis Proteins/metabolism , Mitochondria/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Oxidative Phosphorylation , Oxygen Consumption , Cell Movement , Female , Humans , Inhibitor of Apoptosis Proteins/genetics , MCF-7 Cells , Male , Mitochondria/genetics , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology , Survivin
SELECTION OF CITATIONS
SEARCH DETAIL
...