Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 19 de 19
1.
Front Immunol ; 15: 1341804, 2024.
Article En | MEDLINE | ID: mdl-38515757

IL-15 has shown preclinical activity by enhancing the functional maturation of natural killer (NK) cells. Clinical evaluation of the potential anticancer activity of most cytokines, including IL-15, has been limited by low tolerability and rapid in vivo clearance. Efbalropendekin Alfa (XmAb24306) is a soluble IL15/IL15-receptor alpha heterodimer complex fused to a half-life extended Fc domain (IL15/IL15Rα-Fc), engineered with mutations to reduce IL-15 affinity for CD122. Reduced affinity drives lower potency, leading to prolonged pharmacodynamic response in cynomolgus monkeys. We show that in vitro, human NK cells treated with XmAb24306 demonstrate enhanced cytotoxicity against various tumor cell lines. XmAb24306-treated NK cells also exhibit enhanced killing of 3D colorectal cancer spheroids. Daratumumab (dara), a monoclonal antibody (mAb) that targets CD38 results in antibody-dependent cellular cytotoxicity (ADCC) of both multiple myeloma (MM) cells and NK cells. Addition of XmAb24306 increases dara-mediated NK cell ADCC against various MM cell lines in vitro. Because NK cells express CD38, XmAb24306 increases dara-mediated NK cell fratricide, but overall does not negatively impact the ADCC activity against a MM cell line likely due to increased NK cell activity of the surviving cells. These data show that XmAb24306 increases direct and ADCC-mediated human NK cell cytotoxicity in vitro.


Antineoplastic Agents , Interleukin-15 , Humans , Interleukin-15/pharmacology , Interleukin-15/metabolism , Antineoplastic Agents/pharmacology , Cytokines/metabolism , Immunologic Factors/metabolism , Killer Cells, Natural , Cell Line, Tumor
2.
J Crohns Colitis ; 2024 Jan 24.
Article En | MEDLINE | ID: mdl-38267224

BACKGROUND AND AIMS: The goal was to identify microbial drivers of IBD, by investigating mucosal-associated bacteria and their detrimental products in IBD patients. METHODS: We directly cultured bacterial communities from mucosal biopsies from pediatric gastrointestinal patients and examined for pathogenicity-associated traits. Upon identifying C. perfringens as toxigenic bacteria present in mucosal biopsies, we isolated strains and further characterized toxicity and prevalence. RESULTS: Mucosal biopsy microbial composition differed from corresponding stool samples. C. perfringens was present in 8 of 9 patients' mucosal biopsies, correlating with hemolytic activity, while not in all corresponding stool samples. Large IBD datasets showed higher C. perfringens prevalence in stool samples of IBD adults (18.7-27.1%) versus healthy (5.1%). In vitro, C. perfringens supernatants were toxic to cell types beneath the intestinal epithelial barrier, including endothelial, neuroblasts, and neutrophils, while impact on epithelial cells was less pronounced, suggesting C. perfringens may be damaging particularly when barrier integrity is compromised. Further characterization using purified toxins and genetic insertion mutants confirmed PFO toxin was sufficient for toxicity. Toxin RNA signatures were found in the original patient biopsies by PCR, suggesting intestinal production. C. perfringens supernatants also induced activation of neuroblast and dorsal root ganglion neurons in vitro, suggesting C. perfringens in inflamed mucosal tissue may directly contribute to abdominal pain, a frequent IBD symptom. CONCLUSIONS: Gastrointestinal carriage of certain toxigenic C. perfringens may have an important pathogenic impact on IBD patients. These findings support routine monitoring of C. perfringens and PFO toxins and potential treatment in patients.

3.
Cell Rep Methods ; 4(1): 100673, 2024 Jan 22.
Article En | MEDLINE | ID: mdl-38171361

While antisense oligonucleotides (ASOs) are used in the clinic, therapeutic development is hindered by the inability to assay ASO delivery and activity in vivo. Accordingly, we developed a dual-fluorescence, knockin mouse model that constitutively expresses mKate2 and an engineered EGFP that is alternatively spliced in the presence of ASO to induce expression. We first examined free ASO activity in the brain following intracerebroventricular injection revealing EGFP splice-switching is both ASO concentration and time dependent in major central nervous system cell types. We then assayed the impact of lipid nanoparticle delivery on ASO activity after intravenous administration. Robust EGFP fluorescence was observed in the liver and EGFP+ cells were successfully isolated using fluorescence-activated cell sorting. Together, these results show the utility of this animal model in quantifying both cell-type- and organ-specific ASO delivery, which can be used to advance ASO therapeutics for many disease indications.


Oligonucleotides, Antisense , Oligonucleotides , Mice , Animals , Liver/metabolism , Administration, Intravenous , Coloring Agents/metabolism
4.
Nat Commun ; 14(1): 5945, 2023 09 23.
Article En | MEDLINE | ID: mdl-37741832

Microsatellite-stable colorectal cancer (MSS-CRC) is highly refractory to immunotherapy. Understanding tumor-intrinsic determinants of immunotherapy resistance is critical to improve MSS-CRC patient outcomes. Here, we demonstrate that high tumor expression of the core autophagy gene ATG16L1 is associated with poor clinical response to anti-PD-L1 therapy in KRAS-mutant tumors from IMblaze370 (NCT02788279), a large phase III clinical trial of atezolizumab (anti-PD-L1) in advanced metastatic MSS-CRC. Deletion of Atg16l1 in engineered murine colon cancer organoids inhibits tumor growth in primary (colon) and metastatic (liver and lung) niches in syngeneic female hosts, primarily due to increased sensitivity to IFN-γ-mediated immune pressure. ATG16L1 deficiency enhances programmed cell death of colon cancer organoids induced by IFN-γ and TNF, thus increasing their sensitivity to host immunity. In parallel, ATG16L1 deficiency reduces tumor stem-like populations in vivo independently of adaptive immune pressure. This work reveals autophagy as a clinically relevant mechanism of immune evasion and tumor fitness in MSS-CRC and provides a rationale for autophagy inhibition to boost immunotherapy responses in the clinic.


Colonic Neoplasms , Colorectal Neoplasms , Animals , Female , Humans , Mice , Autophagy/genetics , Autophagy-Related Proteins/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Genes, Regulator , Liver , Clinical Trials, Phase III as Topic
5.
Heliyon ; 9(3): e14238, 2023 Mar.
Article En | MEDLINE | ID: mdl-36950615

The ability of stem cells to rapidly proliferate and differentiate is integral to the steady-state maintenance of tissues with high turnover such as the blood and intestine. Mutations that alter these processes can cause primary immunodeficiencies, malignancies and defects in barrier function. The Rho-kinases, Rock1 and Rock2, regulate cell shape and cytoskeletal rearrangement, activities essential to mitosis. Here, we use inducible gene targeting to ablate Rock1 and Rock2 in adult mice, and identify an obligate requirement for these enzymes in the preservation of the hematopoietic and gastrointestinal systems. Hematopoietic cell progenitors devoid of Rho-kinases display cell cycle arrest, blocking the differentiation to mature blood lineages. Similarly, these mice exhibit impaired epithelial cell renewal in the small intestine, which is ultimately fatal. Our data reveal a novel role for these kinases in the proliferation and viability of stem cells and their progenitors, which is vital to maintaining the steady-state integrity of these organ systems.

6.
Eur Respir J ; 61(4)2023 04.
Article En | MEDLINE | ID: mdl-36585256

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a progressive fibrotic disease characterised by aberrant fibroblast/myofibroblast accumulation and excessive collagen matrix deposition in the alveolar areas of lungs. As the first approved IPF medication, pirfenidone (PFD) significantly decelerates lung function decline while its underlying anti-fibrotic mechanism remains elusive. METHODS: We performed transcriptomic and immunofluorescence analyses of primary human IPF tissues. RESULTS: We showed that myocardin-related transcription factor (MRTF) signalling is activated in myofibroblasts accumulated in IPF lungs. Furthermore, we showed that PFD inhibits MRTF activation in primary human lung fibroblasts at clinically achievable concentrations (half-maximal inhibitory concentration 50-150 µM, maximal inhibition >90%, maximal concentration of PFD in patients <100 µM). Mechanistically, PFD appears to exert its inhibitory effects by promoting the interaction between MRTF and actin indirectly. Finally, PFD-treated IPF lungs exhibit significantly less MRTF activation in fibroblast foci areas than naïve IPF lungs. CONCLUSIONS: Our results suggest MRTF signalling as a direct target for PFD and implicate that some of the anti-fibrotic effects of PFD may be due to MRTF inhibition in lung fibroblasts.


Idiopathic Pulmonary Fibrosis , Transcription Factors , Humans , Fibrosis , Trans-Activators/pharmacology , Lung/pathology , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/pathology , Fibroblasts , Myofibroblasts
7.
Cell Metab ; 34(9): 1377-1393.e8, 2022 09 06.
Article En | MEDLINE | ID: mdl-35987202

Fibrosis is the major risk factor associated with morbidity and mortality in patients with non-alcoholic steatohepatitis (NASH)-driven chronic liver disease. Although numerous efforts have been made to identify the mediators of the initiation of liver fibrosis, the molecular underpinnings of fibrosis progression remain poorly understood, and therapies to arrest liver fibrosis progression are elusive. Here, we identify a pathway involving WNT1-inducible signaling pathway protein 1 (WISP1) and myocardin-related transcription factor (MRTF) as a central mechanism driving liver fibrosis progression through the integrin-dependent transcriptional reprogramming of myofibroblast cytoskeleton and motility. In mice, WISP1 deficiency protects against fibrosis progression, but not fibrosis onset. Moreover, the therapeutic administration of a novel antibody blocking WISP1 halted the progression of existing liver fibrosis in NASH models. These findings implicate the WISP1-MRTF axis as a crucial determinant of liver fibrosis progression and support targeting this pathway by antibody-based therapy for the treatment of NASH fibrosis.


Non-alcoholic Fatty Liver Disease , Transcription Factors , Animals , Liver/metabolism , Liver Cirrhosis/metabolism , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/metabolism , Nuclear Proteins , Signal Transduction , Trans-Activators , Transcription Factors/metabolism
8.
Sci Transl Med ; 14(627): eabf8188, 2022 01 12.
Article En | MEDLINE | ID: mdl-35020406

Exacerbations of symptoms represent an unmet need for people with asthma. Bacterial dysbiosis and opportunistic bacterial infections have been observed in, and may contribute to, more severe asthma. However, the molecular mechanisms driving these exacerbations remain unclear. We show here that bacterial lipopolysaccharide (LPS) induces oncostatin M (OSM) and that airway biopsies from patients with severe asthma present with an OSM-driven transcriptional profile. This profile correlates with activation of inflammatory and mucus-producing pathways. Using primary human lung tissue or human epithelial and mesenchymal cells, we demonstrate that OSM is necessary and sufficient to drive pathophysiological features observed in severe asthma after exposure to LPS or Klebsiella pneumoniae. These findings were further supported through blockade of OSM with an OSM-specific antibody. Single-cell RNA sequencing from human lung biopsies identified macrophages as a source of OSM. Additional studies using Osm-deficient murine macrophages demonstrated that macrophage-derived OSM translates LPS signals into asthma-associated pathologies. Together, these data provide rationale for inhibiting OSM to prevent bacterial-associated progression and exacerbation of severe asthma.


Asthma , Oncostatin M/metabolism , Animals , Asthma/pathology , Humans , Lung/pathology , Macrophages/metabolism , Mice , Mucus , Oncostatin M/genetics
9.
PLoS One ; 16(12): e0260800, 2021.
Article En | MEDLINE | ID: mdl-34879110

The cancer-associated fibroblast (CAF) marker podoplanin (PDPN) is generally correlated with poor clinical outcomes in cancer patients and thus represents a promising therapeutic target. Despite its biomedical relevance, basic aspects of PDPN biology such as its cellular functions and cell surface ligands remain poorly uncharacterized, thus challenging drug development. Here, we utilize a high throughput platform to elucidate the PDPN cell surface interactome, and uncover the neutrophil protein CD177 as a new binding partner. Quantitative proteomics analysis of the CAF phosphoproteome reveals a role for PDPN in cell signaling, growth and actomyosin contractility, among other processes. Moreover, cellular assays demonstrate that CD177 is a functional antagonist, recapitulating the phenotype observed in PDPN-deficient CAFs. In sum, starting from the unbiased elucidation of the PDPN co-receptome, our work provides insights into PDPN functions and reveals the PDPN/CD177 axis as a possible modulator of fibroblast physiology in the tumor microenvironment.


Biomarkers, Tumor/metabolism , Cancer-Associated Fibroblasts/pathology , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Isoantigens/metabolism , Membrane Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , Tumor Microenvironment , Apoptosis , Biomarkers, Tumor/genetics , Cancer-Associated Fibroblasts/immunology , Cancer-Associated Fibroblasts/metabolism , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Humans , Isoantigens/genetics , Membrane Glycoproteins/genetics , Neutrophils/immunology , Neutrophils/metabolism , Prognosis , Receptors, Cell Surface/genetics , Survival Rate , Tumor Cells, Cultured
10.
Cancer Discov ; 10(2): 232-253, 2020 02.
Article En | MEDLINE | ID: mdl-31699795

With only a fraction of patients responding to cancer immunotherapy, a better understanding of the entire tumor microenvironment is needed. Using single-cell transcriptomics, we chart the fibroblastic landscape during pancreatic ductal adenocarcinoma (PDAC) progression in animal models. We identify a population of carcinoma-associated fibroblasts (CAF) that are programmed by TGFß and express the leucine-rich repeat containing 15 (LRRC15) protein. These LRRC15+ CAFs surround tumor islets and are absent from normal pancreatic tissue. The presence of LRRC15+ CAFs in human patients was confirmed in >80,000 single cells from 22 patients with PDAC as well as by using IHC on samples from 70 patients. Furthermore, immunotherapy clinical trials comprising more than 600 patients across six cancer types revealed elevated levels of the LRRC15+ CAF signature correlated with poor response to anti-PD-L1 therapy. This work has important implications for targeting nonimmune elements of the tumor microenvironment to boost responses of patients with cancer to immune checkpoint blockade therapy. SIGNIFICANCE: This study describes the single-cell landscape of CAFs in pancreatic cancer during in vivo tumor evolution. A TGFß-driven, LRRC15+ CAF lineage is associated with poor outcome in immunotherapy trial data comprising multiple solid-tumor entities and represents a target for combinatorial therapy.This article is highlighted in the In This Issue feature, p. 161.


Cancer-Associated Fibroblasts/immunology , Carcinoma, Pancreatic Ductal/drug therapy , Drug Resistance, Neoplasm/genetics , Immune Checkpoint Inhibitors/pharmacology , Membrane Proteins/metabolism , Myofibroblasts/immunology , Pancreatic Neoplasms/drug therapy , Animals , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/metabolism , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/immunology , Cell Line, Tumor , Cell Lineage/genetics , Cell Lineage/immunology , Clinical Trials as Topic , Computational Biology , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/immunology , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/immunology , Humans , Immune Checkpoint Inhibitors/therapeutic use , Mice , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/immunology , RNA-Seq , Single-Cell Analysis , Transforming Growth Factor beta/metabolism , Treatment Outcome , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
11.
J Immunol ; 203(1): 137-147, 2019 07 01.
Article En | MEDLINE | ID: mdl-31092639

PI3K is one of the most frequently mutated genes in cancers and has been the target of numerous anticancer therapies. With the additional development of therapeutics that mobilize the immune system, such as Abs with effector functions, bispecific Abs, and checkpoint inhibitors, many small molecule inhibitors that target PI3K are being combined with these immunomodulatory treatments. However, the PI3K pathway is also essential for lymphocyte function, and the presence of the PI3K inhibitor may render the immunomodulatory therapeutic ineffective in these combinatorial treatments. Therefore, therapeutics with enhanced activity, such as afucosylated Abs, which promote signaling and function, may be ideal in these types of treatments to offset the negative effect of PI3K inhibitors on immune cell function. Indeed, we show that afucosylated Abs can counterbalance these inhibitory effects on FcγRIIIa-driven signaling in human NK cells to produce signals similar to cells treated only with fucosylated Ab. Furthermore, NK cell activation, degranulation, chemokine/cytokine production, and Ab-dependent cellular cytotoxicity were similar between inhibitor-treated, afucosylated Ab-stimulated NK cells and cells activated only with its fucosylated counterpart. To our knowledge, these studies also identified a previously undefined role for phospho-S6 in human NK cells. In this study, a kinetic delay in PI3K-driven phosphorylation of S6 was observed to control transcription of the temporally regulated production of IFN-γ and TNF-α but not MIP-1α, MIP-1ß, and RANTES. Together, these studies demonstrate the importance of the PI3K pathway for S6 phosphorylation in human NK cells and the need to combine PI3K inhibitors with therapeutic molecules that enhance immunomodulatory function for anticancer therapies.


Antineoplastic Agents/pharmacology , Killer Cells, Natural/immunology , Neoplasms/drug therapy , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Receptors, IgG/metabolism , Ribosomal Protein S6 Kinases/metabolism , Antibodies/chemistry , Antibodies/metabolism , Antibody-Dependent Cell Cytotoxicity , Cells, Cultured , Cytokines/metabolism , Fucose/chemistry , Humans , Immunomodulation , Lymphocyte Activation , Phosphorylation , Signal Transduction
12.
PLoS One ; 14(4): e0214110, 2019.
Article En | MEDLINE | ID: mdl-30951545

Peg10 (paternally expressed gene 10) is an imprinted gene that is essential for placental development. It is thought to derive from a Ty3-gyspy LTR (long terminal repeat) retrotransposon and retains Gag and Pol-like domains. Here we show that the Gag domain of PEG10 can promote vesicle budding similar to the HIV p24 Gag protein. Expressed in a subset of mouse endocrine organs in addition to the placenta, PEG10 was identified as a substrate of the deubiquitinating enzyme USP9X. Consistent with PEG10 having a critical role in placental development, PEG10-deficient trophoblast stem cells (TSCs) exhibited impaired differentiation into placental lineages. PEG10 expressed in wild-type, differentiating TSCs was bound to many cellular RNAs including Hbegf (Heparin-binding EGF-like growth factor), which is known to play an important role in placentation. Expression of Hbegf was reduced in PEG10-deficient TSCs suggesting that PEG10 might bind to and stabilize RNAs that are critical for normal placental development.


Cell Differentiation/genetics , Heparin-binding EGF-like Growth Factor/genetics , Nuclear Proteins/genetics , Placentation/genetics , Transcription Factors/genetics , Animals , Apoptosis Regulatory Proteins , Cell Lineage/genetics , DNA Transposable Elements/genetics , DNA-Binding Proteins , Female , Gene Expression Regulation, Developmental , Gene Products, gag/genetics , Genomic Imprinting/genetics , Humans , Mice , Placenta/metabolism , Pregnancy , RNA-Binding Proteins/genetics , Stem Cells/cytology , Stem Cells/metabolism , Trophoblasts/cytology , Trophoblasts/metabolism
13.
Structure ; 24(1): 179-186, 2016 Jan 05.
Article En | MEDLINE | ID: mdl-26724994

The Hippo signaling pathway is responsible for regulating the function of TEAD family transcription factors in metazoans. TEADs, with their co-activators YAP/TAZ, are critical for controlling cell differentiation and organ size through their transcriptional activation of genes involved in cell growth and proliferation. Dysregulation of the Hippo pathway has been implicated in multiple forms of cancer. Here, we identify a novel form of regulation of TEAD family proteins. We show that human TEADs are palmitoylated at a universally conserved cysteine, and report the crystal structures of the human TEAD2 and TEAD3 YAP-binding domains in their palmitoylated forms. These structures show a palmitate bound within a highly conserved hydrophobic cavity at each protein's core. Our findings also demonstrate that this modification is required for proper TEAD folding and stability, indicating a potential new avenue for pharmacologically regulating the Hippo pathway through the modulation of TEAD palmitoylation.


DNA-Binding Proteins/chemistry , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Transcription Factors/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Cysteine/metabolism , DNA-Binding Proteins/metabolism , HeLa Cells , Hippo Signaling Pathway , Humans , Lipoylation , Molecular Sequence Data , Nuclear Envelope/metabolism , Phosphoproteins/metabolism , Protein Binding , Protein Folding , Protein Stability , TEA Domain Transcription Factors , Transcription Factors/metabolism , YAP-Signaling Proteins
14.
Breast Cancer Res ; 17: 59, 2015 Apr 23.
Article En | MEDLINE | ID: mdl-25902869

INTRODUCTION: Breast cancer, the most common cause of cancer-related deaths worldwide among women, is a molecularly and clinically heterogeneous disease. Extensive genetic and epigenetic profiling of breast tumors has recently revealed novel putative driver genes, including p21-activated kinase (PAK)1. PAK1 is a serine/threonine kinase downstream of small GTP-binding proteins, Rac1 and Cdc42, and is an integral component of growth factor signaling networks and cellular functions fundamental to tumorigenesis. METHODS: PAK1 dysregulation (copy number gain, mRNA and protein expression) was evaluated in two cohorts of breast cancer tissues (n=980 and 1,108). A novel small molecule inhibitor, FRAX1036, and RNA interference were used to examine PAK1 loss of function and combination with docetaxel in vitro. Mechanism of action for the therapeutic combination, both cellular and molecular, was assessed via time-lapse microscopy and immunoblotting. RESULTS: We demonstrate that focal genomic amplification and overexpression of PAK1 are associated with poor clinical outcome in the luminal subtype of breast cancer (P=1.29×10(-4) and P=0.015, respectively). Given the role for PAK1 in regulating cytoskeletal organization, we hypothesized that combination of PAK1 inhibition with taxane treatment could be combined to further interfere with microtubule dynamics and cell survival. Consistent with this, administration of docetaxel with either a novel small molecule inhibitor of group I PAKs, FRAX1036, or PAK1 small interfering RNA oligonucleotides dramatically altered signaling to cytoskeletal-associated proteins, such as stathmin, and induced microtubule disorganization and cellular apoptosis. Live-cell imaging revealed that the duration of mitotic arrest mediated by docetaxel was significantly reduced in the presence of FRAX1036, and this was associated with increased kinetics of apoptosis. CONCLUSIONS: Taken together, these findings further support PAK1 as a potential target in breast cancer and suggest combination with taxanes as a viable strategy to increase anti-tumor efficacy.


Apoptosis/drug effects , Breast Neoplasms/metabolism , Microtubules/metabolism , Protein Kinase Inhibitors/pharmacology , Tubulin Modulators/pharmacology , p21-Activated Kinases/antagonists & inhibitors , Apoptosis/genetics , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , DNA Copy Number Variations , Docetaxel , Drug Synergism , Female , Gene Amplification , Gene Expression , Humans , Prognosis , Signal Transduction/drug effects , Taxoids/pharmacology , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism
15.
Nat Cell Biol ; 16(6): 561-73, 2014 Jun.
Article En | MEDLINE | ID: mdl-24859005

Turnover of integrin-based focal adhesions (FAs) with the extracellular matrix (ECM) is essential for coordinated cell movement. In collectively migrating human keratinocytes, FAs assemble near the leading edge, grow and mature as a result of contractile forces and disassemble underneath the advancing cell body. We report that clustering of microtubule-associated CLASP1 and CLASP2 proteins around FAs temporally correlates with FA turnover. CLASPs and LL5ß (also known as PHLDB2), which recruits CLASPs to FAs, facilitate FA disassembly. CLASPs are further required for FA-associated ECM degradation, and matrix metalloprotease inhibition slows FA disassembly similarly to CLASP or PHLDB2 (LL5ß) depletion. Finally, CLASP-mediated microtubule tethering at FAs establishes an FA-directed transport pathway for delivery, docking and localized fusion of exocytic vesicles near FAs. We propose that CLASPs couple microtubule organization, vesicle transport and cell interactions with the ECM, establishing a local secretion pathway that facilitates FA turnover by severing cell-matrix connections.


Cell Adhesion , Cell Movement , Exocytosis , Focal Adhesions/metabolism , Keratinocytes/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Transport Vesicles/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Adhesion/drug effects , Cell Movement/drug effects , Exocytosis/drug effects , Extracellular Matrix/metabolism , Focal Adhesions/drug effects , HEK293 Cells , Humans , Keratinocytes/drug effects , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Microtubule-Associated Proteins/genetics , RNA Interference , Recombinant Fusion Proteins/metabolism , Time Factors , Transfection , Transport Vesicles/drug effects
16.
Curr Biol ; 22(9): 753-62, 2012 May 08.
Article En | MEDLINE | ID: mdl-22483942

BACKGROUND: Epithelial remodeling, in which apical-basal polarized cells switch to a migratory phenotype, plays a central role in development and disease of multicellular organisms. Although dynamic microtubules (MTs) are required for directed migration on flat surfaces, how MT dynamics are controlled or contribute to epithelial remodeling in a more physiological three-dimensional (3D) environment is not understood. We use confocal live-cell imaging to analyze MT function and dynamics during 3D epithelial morphogenesis and remodeling of polarized Madin-Darby canine kidney epithelial cells that undergo partial epithelial-to-mesenchymal transition in response to hepatocyte growth factor (HGF). RESULTS: We find that HGF treatment increases MT growth rate before morphological changes are evident and that large numbers of MTs grow into HGF-induced cell extensions independent of centrosome reorientation. Using lentivirus-mediated small hairpin RNA, we demonstrate that EB1, an adaptor protein that mediates recruitment of numerous other +TIP proteins to growing MT plus ends, is required for this HGF-induced MT reorganization. We further show that protrusion and adhesion dynamics are disorganized and that vesicular trafficking to the tip of HGF-induced cell extensions is disrupted in EB1-depleted cells. CONCLUSIONS: We conclude that EB1-mediated interactions with growing MTs are important to coordinate cell shape changes and directed migration into the surrounding extracellular matrix during epithelial remodeling in a physiological 3D environment. In contrast, EB1 is not required for the establishment or maintenance of apical-basal cell polarity, suggesting different functions of +TIPs and MTs in different types of cell polarity.


Epithelium/chemistry , Microtubule-Associated Proteins/physiology , Microscopy, Confocal
17.
Methods Cell Biol ; 97: 15-33, 2010.
Article En | MEDLINE | ID: mdl-20719263

The spatiotemporal regulation of intracellular microtubule polymerization dynamics, by numerous microtubule-associated proteins and other mechanisms, is central to many cell processes. Here, we give an overview and practical guide on how to acquire and analyze time-lapse sequences of dynamic microtubules in live cells by either fluorescently labeling entire microtubules or by utilizing proteins that specifically associate only with growing microtubule ends and summarize the strengths and weaknesses of different approaches. We give practical recommendations for imaging conditions, and discuss important limitations of such analysis that are dictated by the maximum achievable spatial and temporal sampling frequencies.


Cell Physiological Phenomena , Clinical Laboratory Techniques , Microtubules/metabolism , Protein Multimerization/physiology , Adenoviridae/physiology , Animals , Cell Proliferation , Humans , Microscopy, Fluorescence/methods , Microtubules/chemistry , Models, Biological
18.
J Cell Biol ; 184(6): 895-908, 2009 Mar 23.
Article En | MEDLINE | ID: mdl-19289791

Polarity of the microtubule (MT) cytoskeleton is essential for many cell functions. Cytoplasmic linker-associated proteins (CLASPs) are MT-associated proteins thought to organize intracellular MTs and display a unique spatiotemporal regulation. In migrating epithelial cells, CLASPs track MT plus ends in the cell body but bind along MTs in the lamella. In this study, we demonstrate that glycogen synthase kinase 3beta (GSK3beta) directly phosphorylates CLASPs at multiple sites in the domain required for MT plus end tracking. Although complete phosphorylation disrupts both plus end tracking and association along lamella MTs, we show that partial phosphorylation of the identified GSK3beta motifs determines whether CLASPs track plus ends or associate along MTs. In addition, we find that expression of constitutively active GSK3beta destabilizes lamella MTs by disrupting lateral MT interactions with the cell cortex. GSK3beta-induced lamella MT destabilization was partially rescued by expression of CLASP2 with mutated phosphorylation sites. This indicates that CLASP-mediated stabilization of peripheral MTs, which likely occurs in the vicinity of focal adhesions, may be regulated by local GSK3beta inactivation.


Cell Adhesion , Cell Movement , Epithelial Cells/enzymology , Glycogen Synthase Kinase 3/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/enzymology , Amino Acid Motifs , Amino Acid Sequence , Cell Polarity , Focal Adhesions/metabolism , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , HeLa Cells , Humans , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/genetics , Molecular Sequence Data , Phosphorylation , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Time Factors , Transfection
19.
Cell ; 128(5): 901-13, 2007 Mar 09.
Article En | MEDLINE | ID: mdl-17350575

Actin filament networks exert protrusive and attachment forces on membranes and thereby drive membrane deformation and movement. Here, we show that N-WASP WH2 domains play a previously unanticipated role in vesicle movement by transiently attaching actin filament barbed ends to the membrane. To dissect the attachment mechanism, we reconstituted the propulsive motility of lipid-coated glass beads, using purified soluble proteins. N-WASP WH2 mutants assembled actin comet tails and initiated movement, but the comet tails catastrophically detached from the membrane. When presented on the surface of a lipid-coated bead, WH2 domains were sufficient to maintain comet tail attachment. In v-Src-transformed fibroblasts, N-WASP WH2 mutants were severely defective in the formation of circular podosome arrays. In addition to creating an attachment force, interactions between WH2 domains and barbed ends may locally amplify signals for dendritic actin nucleation.


Actin Cytoskeleton/metabolism , Actins/metabolism , Cell Membrane/physiology , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism , Actin-Related Protein 2-3 Complex/metabolism , Animals , Cell Membrane/metabolism , Cell Transformation, Viral , Fibroblasts , Genes, src , Membrane Lipids , Microfilament Proteins/metabolism , Microspheres , Movement , Mutation , Protein Structure, Tertiary , Rabbits , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Wiskott-Aldrich Syndrome Protein, Neuronal/chemistry , Wiskott-Aldrich Syndrome Protein, Neuronal/genetics
...