Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
Add more filters










Publication year range
1.
iScience ; 27(2): 108958, 2024 Feb 16.
Article in English | MEDLINE | ID: mdl-38323010

ABSTRACT

The protein kinase D (PKD) family members regulate the fission of cargo vesicles at the Golgi complex and play a pro-oncogenic role in triple-negative breast cancer (TNBC). Whether PKD facilitates the secretion of tumor-promoting factors in TNBC, however, is still unknown. Using the pharmacological inhibition of PKD activity and siRNA-mediated depletion of PKD2 and PKD3, we identified the PKD-dependent secretome of the TNBC cell lines MDA-MB-231 and MDA-MB-468. Mass spectrometry-based proteomics and antibody-based assays revealed a significant downregulation of extracellular matrix related proteins and pro-invasive factors such as LIF, MMP-1, MMP-13, IL-11, M-CSF and GM-CSF in PKD-perturbed cells. Notably, secretion of these proteins in MDA-MB-231 cells was predominantly controlled by PKD2 and enhanced spheroid invasion. Consistently, PKD-dependent secretion of pro-invasive factors was more pronounced in metastatic TNBC cell lines. Our study thus uncovers a novel role of PKD2 in releasing a pro-invasive secretome.

2.
Front Oncol ; 13: 1190988, 2023.
Article in English | MEDLINE | ID: mdl-37305581

ABSTRACT

Introduction: Malignant pleural mesothelioma (MPM) is a neoplasm with dismal prognosis and notorious resistance to the standard therapeutics cisplatin and pemetrexed. Chalcone derivatives are efficacious anti-cancer agents with minimal toxicity and have, therefore, gained pharmaceutical interest. Here, we investigated the efficacy of CIT-026 and CIT-223, two indolyl-chalcones (CITs), to inhibit growth and viability of MPM cells and defined the mechanism by which the compounds induce cell death. Methods: The effects of CIT-026 and CIT-223 were analyzed in five MPM cell lines, using viability, immunofluorescence, real-time cell death monitoring, and tubulin polymerization assays, along with siRNA knockdown. Phospho-kinase arrays and immunoblotting were used to identify signaling molecules that contribute to cell death. Results: CIT-026 and CIT-223 were toxic in all cell lines at sub-micromolar concentrations, in particular in MPM cells resistant to cisplatin and pemetrexed, while normal fibroblasts were only modestly affected. Both CITs targeted tubulin polymerization via (1) direct interaction with tubulin and (2) phosphorylation of microtubule regulators STMN1, CRMP2 and WNK1. Formation of aberrant tubulin fibers caused abnormal spindle morphology, mitotic arrest and apoptosis. CIT activity was not reduced in CRMP2-negative and STMN1-silenced MPM cells, indicating that direct tubulin targeting is sufficient for toxic effects of CITs. Discussion: CIT-026 and CIT-223 are highly effective inducers of tumor cell apoptosis by disrupting microtubule assembly, with only modest effects on non-malignant cells. CITs are potent anti-tumor agents against MPM cells, in particular cells resistant to standard therapeutics, and thus warrant further evaluation as potential small-molecule therapeutics in MPM.

3.
Front Cell Dev Biol ; 11: 1173387, 2023.
Article in English | MEDLINE | ID: mdl-37293129

ABSTRACT

Protein kinase D (PKD) is a serine/threonine kinase family that controls important cellular functions, most notably playing a key role in the secretory pathway at the trans-Golgi network. Aberrant expression of PKD isoforms has been found mainly in breast cancer, where it promotes various cellular processes such as growth, invasion, survival and stem cell maintenance. In this review, we discuss the isoform-specific functions of PKD in breast cancer progression, with a particular focus on how the PKD controlled cellular processes might be linked to deregulated membrane trafficking and secretion. We further highlight the challenges of a therapeutic approach targeting PKD to prevent breast cancer progression.

4.
Mol Biol Cell ; 34(7): br8, 2023 06 01.
Article in English | MEDLINE | ID: mdl-36989034

ABSTRACT

Actin cytoskeleton predominantly regulates the formation and maintenance of synapses by controlling dendritic spine morphology and motility. To visualize actin dynamics, actin molecules can be labeled by genetically fusing fluorescent proteins to actin monomers, actin-binding proteins, or single-chain anti-actin antibodies. In the present study, we compared the dendritic effect of EGFP-actin, LifeAct-TagGFP2 (LifeAct-GFP), and Actin-Chromobody-TagGFP2 (AC-GFP) in mouse cultured hippocampal neurons using unbiased quantitative methods. The actin-binding probes LifeAct-GFP and AC-GFP showed similar affinity to F-actin, but in contrast to EGFP-actin, they did not reveal subtle changes in actin remodeling between mushroom-shaped spines and filopodia. All tested actin probes colocalized with phalloidin similarly; however, the enrichment of LifeAct-GFP in dendritic spines was remarkably lower compared with the other constructs. LifeAct-GFP expression was tolerated at a higher expression level compared with EGFP-actin and AC-GFP with only subtle differences identified in dendritic spine morphology and protrusion density. While EGFP-actin and LifeAct-GFP expression did not alter dendritic arborization, AC-GFP-expressing neurons displayed a reduced dendritic tree. Thus, although all tested actin probes may be suitable for actin imaging studies, certain limitations should be considered before performing experiments with a particular actin-labeling probe in primary neurons.


Subject(s)
Actins , Neurons , Mice , Animals , Actins/metabolism , Neurons/metabolism , Actin Cytoskeleton/metabolism , Microfilament Proteins/metabolism , Hippocampus/metabolism , Dendritic Spines/metabolism , Cells, Cultured
5.
Proc Natl Acad Sci U S A ; 120(7): e2212909120, 2023 02 14.
Article in English | MEDLINE | ID: mdl-36745811

ABSTRACT

Phosphorylation is a ubiquitous mechanism by which signals are transduced in cells. Protein kinases, enzymes that catalyze the phosphotransfer reaction are, themselves, often regulated by phosphorylation. Paradoxically, however, a substantial fraction of more than 500 human protein kinases are capable of catalyzing their own activation loop phosphorylation. Commonly, these kinases perform this autophosphorylation reaction in trans, whereby transient dimerization leads to the mutual phosphorylation of the activation loop of the opposing protomer. In this study, we demonstrate that protein kinase D (PKD) is regulated by the inverse mechanism of dimerization-mediated trans-autoinhibition, followed by activation loop autophosphorylation in cis. We show that PKD forms a stable face-to-face homodimer that is incapable of either autophosphorylation or substrate phosphorylation. Dissociation of this trans-autoinhibited dimer results in activation loop autophosphorylation, which occurs exclusively in cis. Phosphorylation serves to increase PKD activity and prevent trans-autoinhibition, thereby switching PKD on. Our findings not only reveal the mechanism of PKD regulation but also have profound implications for the regulation of many other eukaryotic kinases.


Subject(s)
Protein Kinase C , Humans , Phosphorylation/physiology , Protein Kinase C/metabolism
6.
Prog Mol Biol Transl Sci ; 196: 59-97, 2023.
Article in English | MEDLINE | ID: mdl-36813366

ABSTRACT

AMPA receptors are glutamate-gated ion channels, present in a wide range of neuron types and in glial cells. Their main role is to mediate fast excitatory synaptic transmission, and therefore, they are critical for normal brain function. In neurons, AMPA receptors undergo constitutive and activity-dependent trafficking between the synaptic, extrasynaptic and intracellular pools. The kinetics of AMPA receptor trafficking is crucial for the precise functioning of both individual neurons and neural networks involved in information processing and learning. Many of the neurological diseases evoked by neurodevelopmental and neurodegenerative malfunctions or traumatic injuries are caused by impaired synaptic function in the central nervous system. For example, attention-deficit/hyperactivity disorder (ADHD), Alzheimer's disease (AD), tumors, seizures, ischemic strokes, and traumatic brain injury are all characterized by impaired glutamate homeostasis and associated neuronal death, typically caused by excitotoxicity. Given the important role of AMPA receptors in neuronal function, it is not surprising that perturbations in AMPA receptor trafficking are associated with these neurological disorders. In this book chapter, we will first introduce the structure, physiology and synthesis of AMPA receptors, followed by an in-depth description of the molecular mechanisms that control AMPA receptor endocytosis and surface levels under basal conditions or synaptic plasticity. Finally, we will discuss how impairments in AMPA receptor trafficking, particularly endocytosis, contribute to the pathophysiology of various neurological disorders and what efforts are being made to therapeutically target this process.


Subject(s)
Nervous System Diseases , Receptors, AMPA , Humans , Receptors, AMPA/metabolism , Synaptic Transmission , Glutamic Acid/physiology , Endocytosis
7.
Cell Death Discov ; 8(1): 34, 2022 Jan 24.
Article in English | MEDLINE | ID: mdl-35075141

ABSTRACT

Prolonged ER stress and the associated unfolded protein response (UPR) can trigger programmed cell death. Studies in cancer cell lines demonstrated that the intracellular accumulation of TRAIL receptor-2 (TRAIL-R2) and the subsequent activation of caspase-8 contribute significantly to apoptosis induction upon ER stress. While this might motivate therapeutic strategies that promote cancer cell death through ER stress-induced caspase-8 activation, it could also support the unwanted demise of non-cancer cells. Here, we therefore investigated if TRAIL-R2 dependent signaling towards apoptosis can be induced in pancreatic ß cells, whose loss by prolonged ER stress is associated with the onset of diabetes. Interestingly, we found that elevated ER stress in these cells does not result in TRAIL-R2 transcriptional induction or elevated protein levels, and that the barely detectable expression of TRAIL-R2 is insufficient to allow TRAIL-induced apoptosis to proceed. Overall, this indicates that apoptotic cell death upon ER stress most likely proceeds independent of TRAIL-R2 in pancreatic ß cells. Our findings therefore point to differences in ER stress response and death decision-making between cancer cells and pancreatic ß cells and also have implications for future targeted treatment strategies that need to differentiate between ER stress susceptibility of cancer cells and pancreatic ß cells.

8.
Traffic ; 22(12): 454-470, 2021 12.
Article in English | MEDLINE | ID: mdl-34564930

ABSTRACT

α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) type glutamate receptors (AMPARs) mediate the majority of fast excitatory neurotransmission in the brain. The continuous trafficking of AMPARs into and out of synapses is a core feature of synaptic plasticity, which is considered as the cellular basis of learning and memory. The molecular mechanisms underlying the postsynaptic AMPAR trafficking, however, are still not fully understood. In this work, we demonstrate that the protein kinase D (PKD) family promotes basal and activity-induced AMPAR endocytosis in primary hippocampal neurons. Pharmacological inhibition of PKD increased synaptic levels of GluA1-containing AMPARs, slowed down their endocytic trafficking and increased neuronal network activity. By contrast, ectopic expression of constitutive active PKD decreased the synaptic level of AMPARs, while increasing their colocalization with early endosomes. Our results thus establish an important role for PKD in the regulation of postsynaptic AMPAR trafficking during synaptic plasticity.


Subject(s)
Hippocampus , Receptors, AMPA , Endocytosis/physiology , Hippocampus/metabolism , Neuronal Plasticity/physiology , Neurons/metabolism , Protein Kinase C , Receptors, AMPA/metabolism , Synapses/metabolism
9.
J Cell Biol ; 219(8)2020 08 03.
Article in English | MEDLINE | ID: mdl-32479594

ABSTRACT

Matrix metalloproteinases (MMPs) degrade several ECM components and are crucial modulators of cell invasion and tissue organization. Although much has been reported about their function in remodeling ECM in health and disease, their trafficking across the Golgi apparatus remains poorly understood. Here we report that the cis-Golgi protein nucleobindin-1 (NUCB1) is critical for MMP2 and MT1-MMP trafficking along the Golgi apparatus. This process is Ca2+-dependent and is required for invasive MDA-MB-231 cell migration as well as for gelatin degradation in primary human macrophages. Our findings emphasize the importance of NUCB1 as an essential component of MMP transport and its overall impact on ECM remodeling.


Subject(s)
Breast Neoplasms/enzymology , Extracellular Matrix/enzymology , Golgi Apparatus/enzymology , Macrophages/enzymology , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase 2/metabolism , Nucleobindins/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Calcium/metabolism , Calcium Signaling , Cell Movement , Extracellular Matrix/pathology , Female , Gelatin/metabolism , HEK293 Cells , HeLa Cells , Humans , Matrix Metalloproteinase 14/genetics , Matrix Metalloproteinase 2/genetics , Nucleobindins/genetics , Protein Transport , Proteolysis , Time Factors
10.
Hepatology ; 72(5): 1717-1734, 2020 11.
Article in English | MEDLINE | ID: mdl-32048304

ABSTRACT

BACKGROUND AND AIMS: Liver fibrosis (LF) is a central pathological process that occurs in most types of chronic liver diseases. Advanced LF causes cirrhosis, hepatocellular carcinoma, and liver failure. However, the exact molecular mechanisms underlying the initiation and progression of LF remain largely unknown. APPROACH AND RESULTS: This study was designed to investigate the role of protein kinase D3 (PKD3; gene name Prkd3) in the regulation of liver homeostasis. We generated global Prkd3 knockout (Prkd3-/- ) mice and myeloid-cell-specific Prkd3 knockout (Prkd3∆LysM ) mice, and we found that both Prkd3-/- mice and Prkd3∆LysM mice displayed spontaneous LF. PKD3 deficiency also aggravated CCl4 -induced LF. PKD3 is highly expressed in hepatic macrophages (HMs), and PKD3 deficiency skewed macrophage polarization toward a profibrotic phenotype. Activated profibrotic macrophages produced transforming growth factor beta that, in turn, activates hepatic stellate cells to become matrix-producing myofibroblasts. Moreover, PKD3 deficiency decreased the phosphatase activity of SH2-containing protein tyrosine phosphatase-1 (a bona-fide PKD3 substrate), resulting in sustained signal transducer and activator of transcription 6 activation in macrophages. In addition, we observed that PKD3 expression in HMs was down-regulated in cirrhotic human liver tissues. CONCLUSIONS: PKD3 deletion in mice drives LF through the profibrotic macrophage activation.


Subject(s)
Liver Cirrhosis, Experimental/pathology , Liver Cirrhosis/pathology , Protein Kinase C/deficiency , Animals , Carbon Tetrachloride/toxicity , Cells, Cultured , Disease Progression , Hepatic Stellate Cells/metabolism , Humans , Liver/cytology , Liver/pathology , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/diagnosis , Liver Cirrhosis, Experimental/genetics , Macrophages/metabolism , Mice , Mice, Knockout , Myofibroblasts/metabolism , Primary Cell Culture , Protein Kinase C/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Severity of Illness Index , Tissue Array Analysis , Transforming Growth Factor beta/metabolism
11.
Mol Cell ; 77(5): 970-984.e7, 2020 03 05.
Article in English | MEDLINE | ID: mdl-31982308

ABSTRACT

Cytosolic caspase-8 is a mediator of death receptor signaling. While caspase-8 expression is lost in some tumors, it is increased in others, indicating a conditional pro-survival function of caspase-8 in cancer. Here, we show that tumor cells employ DNA-damage-induced nuclear caspase-8 to override the p53-dependent G2/M cell-cycle checkpoint. Caspase-8 is upregulated and localized to the nucleus in multiple human cancers, correlating with treatment resistance and poor clinical outcome. Depletion of caspase-8 causes G2/M arrest, stabilization of p53, and induction of p53-dependent intrinsic apoptosis in tumor cells. In the nucleus, caspase-8 cleaves and inactivates the ubiquitin-specific peptidase 28 (USP28), preventing USP28 from de-ubiquitinating and stabilizing wild-type p53. This results in de facto p53 protein loss, switching cell fate from apoptosis toward mitosis. In summary, our work identifies a non-canonical role of caspase-8 exploited by cancer cells to override the p53-dependent G2/M cell-cycle checkpoint.


Subject(s)
Caspase 8/metabolism , Cell Nucleus/enzymology , Cell Proliferation , G2 Phase Cell Cycle Checkpoints , Neoplasms/enzymology , Tumor Suppressor Protein p53/metabolism , Ubiquitin Thiolesterase/metabolism , Antineoplastic Agents/pharmacology , Apoptosis , Caspase 8/genetics , Cell Nucleus/drug effects , Cell Nucleus/genetics , Cell Nucleus/pathology , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , HCT116 Cells , HeLa Cells , Humans , MCF-7 Cells , Male , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , PC-3 Cells , Protein Stability , Signal Transduction , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Ubiquitin Thiolesterase/genetics
12.
Int J Cancer ; 146(12): 3423-3434, 2020 06 15.
Article in English | MEDLINE | ID: mdl-31745977

ABSTRACT

Protein kinase D3 (PKD3) is upregulated in triple-negative breast cancer (TNBC) and associated with cell proliferation and metastasis development but its precise pro-oncogenic function is unknown. Here we show that PKD3 is required for the maintenance of the TNBC stem cell population. The depletion of PKD3 in MDA-MB-231 cells reduced the cancer stem cell frequency in vitro and tumor initiation potential in vivo. We further provide evidence that the RhoGEF GEF-H1 is upstream of PKD3 activation in TNBC stem cells. Most importantly, pharmacological PKD inhibition in combination with paclitaxel synergistically decreased oncosphere and colony formation efficiency in vitro and tumor recurrence in vivo. Based on our results we propose that targeting the GEF-H1/PKD3 signaling pathway in combination with chemotherapy might provide an effective therapeutic option for TNBC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Neoplastic Stem Cells/pathology , Protein Kinase C/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Triple Negative Breast Neoplasms/pathology , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Survival , Drug Synergism , Female , Gene Knockdown Techniques , Humans , Mice , Neoplastic Stem Cells/drug effects , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Signal Transduction/drug effects , Signal Transduction/genetics , Triple Negative Breast Neoplasms/drug therapy , Xenograft Model Antitumor Assays
13.
Cells ; 8(12)2019 11 21.
Article in English | MEDLINE | ID: mdl-31766364

ABSTRACT

As membrane-associated master regulators of cytoskeletal remodeling, Rho GTPases coordinate a wide range of biological processes such as cell adhesion, motility, and polarity. In the last years, Rho GTPases have also been recognized to control intracellular membrane sorting and trafficking steps directly; however, how Rho GTPase signaling is regulated at endomembranes is still poorly understood. In this review, we will specifically address the local Rho GTPase pools coordinating intracellular membrane trafficking with a focus on the endo- and exocytic pathways. We will further highlight the spatiotemporal molecular regulation of Rho signaling at endomembrane sites through Rho regulatory proteins, the GEFs and GAPs. Finally, we will discuss the contribution of dysregulated Rho signaling emanating from endomembranes to the development and progression of cancer.


Subject(s)
Intracellular Membranes/metabolism , rho GTP-Binding Proteins/metabolism , rho GTP-Binding Proteins/physiology , Cell Adhesion , Cell Movement , Cell Polarity , Cytoskeleton/metabolism , Endocytosis/physiology , Exocytosis/physiology , Guanine Nucleotide Exchange Factors/metabolism , Humans , Protein Transport/physiology , Signal Transduction
14.
FEBS Lett ; 593(17): 2412-2427, 2019 09.
Article in English | MEDLINE | ID: mdl-31344259

ABSTRACT

In eukaryotic cells, the trans-Golgi network (TGN) serves as a platform for secretory cargo sorting and trafficking. In recent years, it has become evident that a complex network of lipid-lipid and lipid-protein interactions contributes to these key functions. This review addresses the role of lipids at the TGN with a particular emphasis on sphingolipids and diacylglycerol. We further highlight how these lipids couple secretory cargo sorting and trafficking for spatiotemporal coordination of protein transport to the plasma membrane.


Subject(s)
Lipid Metabolism , trans-Golgi Network/metabolism , Animals , Biological Transport , Humans
15.
Small GTPases ; 10(6): 419-432, 2019 11.
Article in English | MEDLINE | ID: mdl-28628388

ABSTRACT

Synaptic connections in the brain are continuously weakened or strengthened in response to changes in neuronal activity. This process, known as synaptic plasticity, is the cellular basis for learning and memory, and is thought to be altered in several neuronal disorders. An important aspect of synaptic plasticity is the tightly controlled trafficking and synaptic targeting of the AMPA-type glutamate receptors, which are the major mediators of fast excitatory transmission in the brain. This review addresses the role of Rab GTPases in AMPA receptor trafficking in neurons under basal conditions and during activity-induced synaptic plasticity, especially during long-term potentiation (LTP) and long-term depression (LTD). We highlight the importance of the tight spatio-temporal control of Rab activity and suggest that this is critical for proper neuronal functions. We also discuss how abnormal AMPA receptor trafficking and malfunctioning of Rabs can lead to neurologic disorders or memory problems.


Subject(s)
Receptors, AMPA/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Neuronal Plasticity , Neurons/physiology , Protein Transport
16.
J Biol Chem ; 293(37): 14407-14416, 2018 09 14.
Article in English | MEDLINE | ID: mdl-30045871

ABSTRACT

Many newly synthesized cellular proteins pass through the Golgi complex from where secretory transport carriers sort them to the plasma membrane and the extracellular environment. The formation of these secretory carriers at the trans-Golgi network is promoted by the protein kinase D (PKD) family of serine/threonine kinases. Here, using mathematical modeling and experimental validation of the PKD activation and substrate phosphorylation kinetics, we reveal that the expression level of the PKD substrate deleted in liver cancer 1 (DLC1), a Rho GTPase-activating protein that is inhibited by PKD-mediated phosphorylation, determines PKD activity at the Golgi membranes. RNAi-mediated depletion of DLC1 reduced PKD activity in a Rho-Rho-associated protein kinase (ROCK)-dependent manner, impaired the exocytosis of the cargo protein horseradish peroxidase, and was associated with the accumulation of the small GTPase RAB6 on Golgi membranes, indicating a protein-trafficking defect. In summary, our findings reveal that DLC1 maintains basal activation of PKD at the Golgi and Golgi secretory activity, in part by down-regulating Rho-ROCK signaling. We propose that PKD senses cytoskeletal changes downstream of DLC1 to coordinate Rho signaling with Golgi secretory function.


Subject(s)
GTPase-Activating Proteins/metabolism , Protein Kinase C/metabolism , Tumor Suppressor Proteins/metabolism , trans-Golgi Network/metabolism , Cell Line, Tumor , Enzyme Activation , Exocytosis , GTPase-Activating Proteins/genetics , HEK293 Cells , Humans , Intracellular Membranes/metabolism , Models, Biological , Phosphorylation , RNA Interference , Signal Transduction , Substrate Specificity , Tumor Suppressor Proteins/genetics , rab GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism
17.
Elife ; 72018 07 20.
Article in English | MEDLINE | ID: mdl-30028295

ABSTRACT

Protein kinase D (PKD) is a family of serine/threonine kinases that is required for the structural integrity and function of the Golgi complex. Despite its importance in the regulation of Golgi function, the molecular mechanisms regulating PKD activity are still incompletely understood. Using the genetically encoded PKD activity reporter G-PKDrep we now uncover a Rho signaling network comprising GEF-H1, the RhoGAP DLC3, and the Rho effector PLCε that regulate the activation of PKD at trans-Golgi membranes. We further show that this molecular network coordinates the formation of TGN-derived Rab6-positive transport carriers delivering cargo for localized exocytosis at focal adhesions.


Subject(s)
Focal Adhesions/physiology , Microtubules/metabolism , Protein Kinase C/metabolism , Signal Transduction , trans-Golgi Network/metabolism , Cytoskeleton , HEK293 Cells , HeLa Cells , Humans , Phosphoinositide Phospholipase C/genetics , Phosphoinositide Phospholipase C/metabolism , Protein Transport , Rho Guanine Nucleotide Exchange Factors/genetics , Rho Guanine Nucleotide Exchange Factors/metabolism , TRPP Cation Channels/genetics , TRPP Cation Channels/metabolism , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
18.
J Neurochem ; 142(6): 948-961, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28722750

ABSTRACT

Members of the protein kinase D (PKD) family of serine/threonine kinases are known to exert diverse roles in neuronal stress responses. Here, we show the transient activation and nuclear translocation of endogenous PKD upon oxidative stress induced by H2 O2 treatment in primary neuronal cultures. Using pharmacological inhibition, we show that PKD activity protects neurons from oxidative stress-induced cell death. Although members of the canonical nuclear factor kappa-light-chain-enhancer of activated B cells (NF kappaB) pathway were phosphorylated upon H2 O2 treatment, it was found that the neuronal response to oxidative stress is not executed through the nuclear translocation and activity of RelA. On the other hand, we demonstrate for the first time in neuronal cells, the association of green fluorescent protein-tagged kinase inactive PKD1 with mitochondrial membranes in vivo and the presence of PKD activity in the close vicinity of mitochondria in vitro. Our findings thus support the notion that the neuroprotective role of PKD is exerted independently from NF kappaB signaling and suggest a potential mitochondrial function for PKD in cultured neurons.

19.
EMBO J ; 36(12): 1736-1754, 2017 06 14.
Article in English | MEDLINE | ID: mdl-28495678

ABSTRACT

Sphingolipids are membrane lipids globally required for eukaryotic life. The sphingolipid content varies among endomembranes with pre- and post-Golgi compartments being poor and rich in sphingolipids, respectively. Due to this different sphingolipid content, pre- and post-Golgi membranes serve different cellular functions. The basis for maintaining distinct subcellular sphingolipid levels in the presence of membrane trafficking and metabolic fluxes is only partially understood. Here, we describe a homeostatic regulatory circuit that controls sphingolipid levels at the trans-Golgi network (TGN). Specifically, we show that sphingomyelin production at the TGN triggers a signalling pathway leading to PtdIns(4)P dephosphorylation. Since PtdIns(4)P is required for cholesterol and sphingolipid transport to the trans-Golgi network, PtdIns(4)P consumption interrupts this transport in response to excessive sphingomyelin production. Based on this evidence, we envisage a model where this homeostatic circuit maintains a constant lipid composition in the trans-Golgi network and post-Golgi compartments, thus counteracting fluctuations in the sphingolipid biosynthetic flow.


Subject(s)
Phosphatidylinositols/metabolism , Sphingolipids/metabolism , trans-Golgi Network/metabolism , HeLa Cells , Homeostasis , Humans , Models, Biological
20.
Biotechnol Bioeng ; 114(6): 1310-1318, 2017 06.
Article in English | MEDLINE | ID: mdl-28165157

ABSTRACT

The dynamics of protein folding and secretion are key issues in improving the productivity and robustness of Chinese hamster ovary (CHO) producer cells. High recombinant protein secretion in CHO producer clones triggers the activation of the unfolded protein response (UPR), an intracellular response to the accumulation of unfolded and misfolded proteins in the endoplasmic reticulum (ER). We previously reported that the human microRNA (miRNA) miR-1287 enhances productivity in IgG-expressing CHO cells (CHO-IgG). Here, through next-generation sequencing (NGS), we identified the activating transcription factor 6 beta (ATF6ß), a repressor of the pro-survival and UPR promoting factor ATF6α, as a direct target gene of miR-1287 in CHO-IgG cells. We show that the transient depletion of ATF6ß resulted in enhanced specific productivity comparable to that of miR-1287-expressing CHO-IgG cells. Strikingly, stable ATF6ß knockdown in CHO-IgG cells significantly improved antibody titer and viable cell density under fed-batch conditions. This was associated with the elevated expression of the UPR genes glucose-regulated protein 78 (GRP78), homocysteine inducible ER protein with ubiquitin like domain 1 (Herpud1) and CCAAT/enhancer-binding protein homologous protein (CHOP). We hence demonstrate that ATF6ß-based cell line engineering is a promising strategy to improve the productivity of CHO producer cells by activating an optimally balanced UPR program. Biotechnol. Bioeng. 2017;114: 1310-1318. © 2017 Wiley Periodicals, Inc.


Subject(s)
Antibodies, Monoclonal/metabolism , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/physiology , Genetic Enhancement/methods , Recombinant Proteins/biosynthesis , Unfolded Protein Response/genetics , Activating Transcription Factor 6 , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/isolation & purification , CHO Cells , Cricetulus , Endoplasmic Reticulum Chaperone BiP , Recombinant Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...