Cathepsin D Is Partly Endocytosed by the LRP1 Receptor and Inhibits LRP1-Regulated Intramembrane Proteolysis

Total Page:16

File Type:pdf, Size:1020Kb

Cathepsin D Is Partly Endocytosed by the LRP1 Receptor and Inhibits LRP1-Regulated Intramembrane Proteolysis Oncogene (2012) 31, 3202–3212 & 2012 Macmillan Publishers Limited All rights reserved 0950-9232/12 www.nature.com/onc SHORT COMMUNICATION Cathepsin D is partly endocytosed by the LRP1 receptor and inhibits LRP1-regulated intramembrane proteolysis D Derocq1,2,3,4, C Pre´bois1,2,3,4, M Beaujouin1,2,3,4, V Laurent-Matha1,2,3,4, S Pattingre1,2,3,4, GK Smith5 and E Liaudet-Coopman1,2,3,4 1IRCM, Institut de Recherche en Cance´rologie de Montpellier, Montpellier, France; 2INSERM U896, Montpellier, France; 3Universite´ Montpellier1, Montpellier, France; 4CRLC Val d’Aurelle Paul Lamarque, Montpellier, France and 5Screening and Compound Profiling, GlaxoSmithKline, Inc., Research Triangle Park, NC, USA The aspartic protease cathepsin-D (cath-D) is a marker of epithelial breast cancer cells (Vetvicka et al., 1994; poor prognosis in breast cancer that is overexpressed and Rochefort and Liaudet-Coopman, 1999; Liaudet-Coop- hypersecreted by human breast cancer cells. Secreted pro- man et al., 2006). This overproduction in breast cancer cath-D binds to the extracellular domain of the b-chain of is correlated with a poor prognosis (Ferrandina et al., the LDL receptor-related protein-1 (LRP1) in fibroblasts. 1997; Foekens et al., 1999; Rodriguez et al., 2005). The LRP1 receptor has an 85-kDa transmembrane b-chain Human cath-D is synthesized as a 52-kDa precursor and a noncovalently attached 515-kDa extracellular a-chain. that is rapidly converted in the endosomes to form an LRP1 acts by (1) internalizing many ligands via its a-chain, active, 48-kDa, single-chain intermediate, and then in (2) activating signaling pathways by phosphorylating the the lysosomes into the fully active mature protease, LRP1b-chain tyrosine and (3) modulating gene transcription composed of a 34-kDa heavy chain and a 14-kDa light by regulated intramembrane proteolysis (RIP) of its b-chain. chain (Gieselmann et al., 1985). The overexpression of LRP1 RIP involves two cleavages: the first liberates the cath-D in breast cancer cells leads to the hypersecretion LRP1 ectodomain to give a membrane-associated form, of the 52-kDa pro-cath-D into the extracellular envir- LRP1b-CTF, and the second generates the LRP1b-intra- onment (Vignon et al., 1986; Capony et al., 1989; Fusek cellular domain, LRP1b-ICD, that modulates gene tran- and Vetvicka, 1994). Cath-D affects both the cancer scription. Here, we investigated the endocytosis of pro-cath- cells and the stromal cells of the tumor microenviron- D by LRP1 and the effect of pro-cath-D/LRP1b interaction ment. Human pre-pro-cath-D cDNA transfected in on LRP1b tyrosine phosphorylation and/or LRP1b RIP. cancer cells promotes cancer cell proliferation, tumor Our results indicate that pro-cath-D was partially endocy- angiogenesis, and tumor growth and metastasis (Glondu tosedbyLRP1infibroblasts.However,pro-cath-Dand et al., 2001; Berchem et al., 2002). Human pre-pro-cath- ectopic cath-D did not stimulate phosphorylation of the D cDNA transfected in cath-DÀ/ÀMEF (mouse embryonic LRP1b-chain tyrosine. Interestingly, ectopic cath-D and its fibroblast) cells induces fibroblast outgrowth (Laurent- catalytically inactive D231Ncath-D, and pro-D231Ncath-D all Matha et al., 2005). Inhibition of cath-D expression significantly inhibited LRP1 RIP by preventing LRP1b- in breast cancer cells decreases tumor growth and CTF production. Thus, cath-D inhibits LRP1 RIP indepen- metastasis (Glondu et al., 2002; Ohri et al., 2007; dently of its catalytic activity by blocking the first cleavage. Vashishta et al., 2007). Human secreted pro-cath-D As cath-D triggers fibroblast outgrowth by LRP1, we stimulates breast cancer cell proliferation (Vignon et al., propose that cath-D modulates the growth of fibroblasts by 1986; Fusek and Vetvicka, 1994; Vetvicka et al., 1994; inhibiting LRP1 RIP in the breast tumor microenvironment. Ohri et al., 2008), fibroblast outgrowth (Laurent-Matha Oncogene (2012) 31, 3202–3212; doi:10.1038/onc.2011.501; et al., 2005) and angiogenesis (Hu et al., 2008). We have published online 14 November 2011 shown that a mutated catalytically inactive version of cath-D (D231N) is still mitogenic for tumor cells and Keywords: cancer; cathepsin D; LRP1; RIP; endocyto- fibroblasts (Glondu et al., 2001; Berchem et al., 2002; sis; tyrosine phosphorylation Laurent-Matha et al., 2005). We recently discovered that pro-cath-D is the first ligand that binds to the extracellular domain of the b-chain of the LDL receptor-related protein-1 (LRP1) in fibroblasts (Beau- Introduction jouin et al., 2010). We also showed that cath-D promotes LRP1-dependent fibroblast outgrowth by a The lysosomal aspartic protease cathepsin-D (cath-D) is mechanism that is independent of its catalytic activity overexpressed and abundantly secreted by human (Beaujouin et al., 2010). The LRP1 receptor consists of an 85-kDa transmem- Correspondence: Dr E Liaudet-Coopman, INSERM U896, IRCM Val brane b-chain and a noncovalently attached 515-kDa d’Aurelle-Paul Lamarque, 34298 Montpellier Cedex 5, France. E-mail: [email protected] extracellular a-chain (Strickland and Ranganathan, 2003; Received 24 July 2011; revised 21 September 2011; accepted 22 Lillis et al.,2005;Montelet al., 2007). The b-chain has an September 2011; published online 14 November 2011 extracellular domain, a transmembrane region and a Cathepsin D, endocytosis and LRP1 RIP D Derocq et al 3203 cytoplasmic tail. The extracellular a-chain contains bind- b-chain of the LRP1 receptor in fibroblasts (Beaujouin ing sites for numerous structurally diverse ligands, et al., 2010), and as LRP1 is an endocytic receptor, we including lipoprotein particles, proteases and protease- investigated the role of LRP1 in the internalization of inhibitor complexes, extracellular matrix proteins, cyto- secreted pro-cath-D in fibroblasts (Figure 1). We first kines and growth factors. LRP1 has a well-defined role as analyzed the endocytosis of S35-radiolabelled pro-cath-D a scavenger receptor mediating the endocytosis of 440 secreted by cancer cells in human mammary fibroblasts different extracellular ligands that bind to its a-chain. It (HMF) in which endogenous LRP1 synthesis had been delivers most, but not all, of these ligands to lysosomes for blocked by RNA interference (Figure 1A). Internalized degradation (Herz and Strickland, 2001; Gonias et al., labelled 52-kDa pro-cath-D was first transformed into a 2004; Emonard et al., 2005; May et al., 2007). It has also 48-kDa endosomal intermediate and then into the 34-kDa been shown that LRP1 is involvedinsignaltransduction lysosomal mature enzyme (Figure 1A, panel a, lane 1) by phosphorylation of the tyrosine in the cytoplasmic (Capony et al., 1987). Excess M6P prevented pro-cath-D NPXY motifs of its b-chain and modulation of signaling from binding to its M6P receptors and inhibited the pathways such as the MAP kinase pathway (Barnes et al., internalization of pro-cath-D by 82% (Figure 1A, panel a, 2001, 2003; Boucher et al., 2002; Boucher and Gotthardt, compare lanes 1 and 3). Thus, 18% of pro-cath-D was 2004; Loukinova et al., 2002; Yang et al., 2004; Newton taken up by HMF fibroblasts in a manner that was et al., 2005; Hu et al., 2006). More recent studies have independent of the M6P receptors (Figure 1A, panel a, shown that LRP1 influences gene transcription by lane 3), as shown for mouse fibroblasts (Laurent-Matha regulated intramembrane proteolysis (RIP) of its b-chain et al., 2005). Inhibiting LRP1 synthesis with siRNA (May et al., 2002; Kinoshita et al., 2003; von Arnim et al., (Figure 1A, panel b) led to a 50% decrease in reminiscent 2005; Zurhove et al., 2008). RIP is a process that involves pro-cath-D internalization (Figure 1A, panel a, compare two cleavages, as described for Notch (De Strooper et al., lanes 3 and 4). There was also a decrease in pro-cath-D 1999), APP (amyloid precursor protein) (De Strooper endocytosis in LRP1-silenced fibroblasts (Figure 1A, et al., 1998) and LRP6 (Mi and Johnson, 2007). The first panel d) when uptake was measured over 18 h cleavage, performed by metalloproteinases (Quinn et al., (Figure 1A, panel c, compare lanes 3 and 4, and panel e 1999; Rozanov et al., 2004; Liu et al., 2009; Selvais et al., for quantification). We then investigated pro-cath-D 2009, 2011) and by the membrane-associated b-secretase, internalization by LRP1 using MEF that lacked M6P BACE1 (von Arnim et al., 2005), occurs in the extra- receptors (Figure 1B). Silencing LRP1 in RM6PÀ/ÀMEF cellular region, close to the plasma membrane, and leads to cells (Figure 1B, panel b) partly decreased pro-cath-D shedding of the LRP1b ectodomain. This produces the endocytosis over 3 h (Figure 1B, panel a, left) and 18 h membrane-associated carboxyl-terminal fragment, (Figure 1B, panel a, right, and panel c for quantification). LRP1b-CTF,whichisthencleavedbyg-secretases within Re-expression of the LRP1b chain alone in LRP1- its transmembrane domain (Hass et al., 2009). The silenced RM6PÀ/ÀMEF cells caused accumulation of 52- LRP1b-intracellular domain, LRP1b-ICD, is released into kDa pro-cath-D (Figure 1B, panel a, right and left). We the cytosol, where it may interact with signaling proteins, obtained further evidence by examining pro-cath-D translocate to the nucleus and control gene transcription endocytosis in LRP1À/ÀMEF cells re-transfected with full- (May et al., 2002; Kinoshita et al., 2003; von Arnim et al., length LRP1 (clone B-41) or with the LRP1b chaininthe 2005; Zurhove et al., 2008). We have now investigated the presence of M6P (Figure 1C). Re-expressing full-length mechanisms by which cath-D affects the behavior of LRP1 in LRP1À/ÀMEF cells (Figure 1C, panel b) stimulated LRP1. We studied the endocytosis of pro-cath-D by LRP1 pro-cath-D endocytosis (Figure 1C, panel a).
Recommended publications
  • LRP1 Shedding in Human Brain: Roles of ADAM10 and ADAM17 Qiang Liu Washington University School of Medicine in St
    Washington University School of Medicine Digital Commons@Becker ICTS Faculty Publications Institute of Clinical and Translational Sciences 2009 LRP1 shedding in human brain: roles of ADAM10 and ADAM17 Qiang Liu Washington University School of Medicine in St. Louis Juan Zhang Washington University School of Medicine in St. Louis Hien Tran Washington University School of Medicine in St. Louis Marcel M. Verbeek Radboud University Nijmegen Medical Centre Karina Reiss Christian-Albrecht University Kiel See next page for additional authors Follow this and additional works at: https://digitalcommons.wustl.edu/icts_facpubs Part of the Medicine and Health Sciences Commons Recommended Citation Liu, Qiang; Zhang, Juan; Tran, Hien; Verbeek, Marcel M.; Reiss, Karina; Estus, Steven; and Bu, Guojun, "LRP1 shedding in human brain: roles of ADAM10 and ADAM17". Molecular Neurodegeneration, 17. 2009. Paper 85. https://digitalcommons.wustl.edu/icts_facpubs/85 This Article is brought to you for free and open access by the Institute of Clinical and Translational Sciences at Digital Commons@Becker. It has been accepted for inclusion in ICTS Faculty Publications by an authorized administrator of Digital Commons@Becker. For more information, please contact [email protected]. Authors Qiang Liu, Juan Zhang, Hien Tran, Marcel M. Verbeek, Karina Reiss, Steven Estus, and Guojun Bu This article is available at Digital Commons@Becker: https://digitalcommons.wustl.edu/icts_facpubs/85 Molecular Neurodegeneration BioMed Central Research article Open Access LRP1 shedding
    [Show full text]
  • Reconfiguring Nature's Cholesterol Accepting Lipoproteins As
    nanomaterials Review Reconfiguring Nature’s Cholesterol Accepting Lipoproteins as Nanoparticle Platforms for Transport and Delivery of Therapeutic and Imaging Agents , Skylar T. Chuang y z, Siobanth Cruz y and Vasanthy Narayanaswami * Department of Chemistry and Biochemistry, California State University, Long Beach, 1250 Bellflower Blvd, Long Beach, CA 90840, USA; [email protected] (S.T.C.); [email protected] (S.C.) * Correspondence: [email protected]; Tel.: +1-562-985-4953; Fax: +1-562-985-8557 These authors contributed equally to this work. y Current address: Department of Chemistry and Chemical Biology, Rutgers University, 123 Bevier Road, z Piscataway, NJ 08854, USA. Received: 2 March 2020; Accepted: 29 April 2020; Published: 8 May 2020 Abstract: Apolipoproteins are critical structural and functional components of lipoproteins, which are large supramolecular assemblies composed predominantly of lipids and proteins, and other biomolecules such as nucleic acids. A signature feature of apolipoproteins is the preponderance of amphipathic α-helical motifs that dictate their ability to make extensive non-covalent inter- or intra-molecular helix–helix interactions in lipid-free states or helix–lipid interactions with hydrophobic biomolecules in lipid-associated states. This review focuses on the latter ability of apolipoproteins, which has been capitalized on to reconstitute synthetic nanoscale binary/ternary lipoprotein complexes composed of apolipoproteins/peptides and lipids that mimic native high-density lipoproteins (HDLs) with the goal to transport drugs. It traces the historical development of our understanding of these nanostructures and how the cholesterol accepting property of HDL has been reconfigured to develop them as drug-loading platforms. The review provides the structural perspective of these platforms with different types of apolipoproteins and an overview of their synthesis.
    [Show full text]
  • Management of Brain and Leptomeningeal Metastases from Breast Cancer
    International Journal of Molecular Sciences Review Management of Brain and Leptomeningeal Metastases from Breast Cancer Alessia Pellerino 1,* , Valeria Internò 2 , Francesca Mo 1, Federica Franchino 1, Riccardo Soffietti 1 and Roberta Rudà 1,3 1 Department of Neuro-Oncology, University and City of Health and Science Hospital, 10126 Turin, Italy; [email protected] (F.M.); [email protected] (F.F.); riccardo.soffi[email protected] (R.S.); [email protected] (R.R.) 2 Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, 70121 Bari, Italy; [email protected] 3 Department of Neurology, Castelfranco Veneto and Treviso Hospital, 31100 Treviso, Italy * Correspondence: [email protected]; Tel.: +39-011-6334904 Received: 11 September 2020; Accepted: 10 November 2020; Published: 12 November 2020 Abstract: The management of breast cancer (BC) has rapidly evolved in the last 20 years. The improvement of systemic therapy allows a remarkable control of extracranial disease. However, brain (BM) and leptomeningeal metastases (LM) are frequent complications of advanced BC and represent a challenging issue for clinicians. Some prognostic scales designed for metastatic BC have been employed to select fit patients for adequate therapy and enrollment in clinical trials. Different systemic drugs, such as targeted therapies with either monoclonal antibodies or small tyrosine kinase molecules, or modified chemotherapeutic agents are under investigation. Major aims are to improve the penetration of active drugs through the blood–brain barrier (BBB) or brain–tumor barrier (BTB), and establish the best sequence and timing of radiotherapy and systemic therapy to avoid neurocognitive impairment. Moreover, pharmacologic prevention is a new concept driven by the efficacy of targeted agents on macrometastases from specific molecular subgroups.
    [Show full text]
  • Endothelial LRP1 Transports Amyloid-Β1–42 Across the Blood- Brain Barrier
    Endothelial LRP1 transports amyloid-β1–42 across the blood- brain barrier Steffen E. Storck, … , Thomas A. Bayer, Claus U. Pietrzik J Clin Invest. 2016;126(1):123-136. https://doi.org/10.1172/JCI81108. Research Article Neuroscience According to the neurovascular hypothesis, impairment of low-density lipoprotein receptor–related protein-1 (LRP1) in brain capillaries of the blood-brain barrier (BBB) contributes to neurotoxic amyloid-β (Aβ) brain accumulation and drives Alzheimer’s disease (AD) pathology. However, due to conflicting reports on the involvement of LRP1 in Aβ transport and the expression of LRP1 in brain endothelium, the role of LRP1 at the BBB is uncertain. As global Lrp1 deletion in mice is lethal, appropriate models to study the function of LRP1 are lacking. Moreover, the relevance of systemic Aβ clearance to AD pathology remains unclear, as no BBB-specific knockout models have been available. Here, we developed transgenic mouse strains that allow for tamoxifen-inducible deletion of Lrp1 specifically within brain endothelial cells (Slco1c1- CreERT2 Lrp1fl/fl mice) and used these mice to accurately evaluate LRP1-mediated Aβ BBB clearance in vivo. Selective 125 deletion of Lrp1 in the brain endothelium of C57BL/6 mice strongly reduced brain efflux of injected [ I] Aβ1–42. Additionally, in the 5xFAD mouse model of AD, brain endothelial–specific Lrp1 deletion reduced plasma Aβ levels and elevated soluble brain Aβ, leading to aggravated spatial learning and memory deficits, thus emphasizing the importance of systemic Aβ elimination via the BBB. Together, our results suggest that receptor-mediated Aβ BBB clearance may be a potential target for treatment and prevention of Aβ brain accumulation in AD.
    [Show full text]
  • Over-Expression of Low-Density Lipoprotein Receptor-Related Protein-1 Is Associated with Poor Prognosis and Invasion in Pancreatic Ductal Adenocarcinoma
    Pancreatology 19 (2019) 429e435 Contents lists available at ScienceDirect Pancreatology journal homepage: www.elsevier.com/locate/pan Over-expression of low-density lipoprotein receptor-related Protein-1 is associated with poor prognosis and invasion in pancreatic ductal adenocarcinoma Ali Gheysarzadeh a, Amir Ansari a, Mohammad Hassan Emami b, * Amirnader Emami Razavi c, Mohammad Reza Mofid a, a Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran b Gastrointestinal and Hepatobiliary Diseases Research Center, Poursina Hakim Research Institute for Health Care Development, Isfahan, Iran c Iran National Tumor Bank, Cancer Biology Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran article info abstract Article history: Background: Low-density lipoprotein receptor-Related Protein-1 (LRP-1) has been reported to involve in Received 1 December 2018 tumor development. However, its role in pancreatic cancer has not been elucidated. The present study Received in revised form was designed to evaluate the expression of LRP-1 in Pancreatic Ductal Adenocarcinoma Cancer (PDAC) as 16 February 2019 well as its association with prognosis. Accepted 23 February 2019 Methods: Here, 478 pancreatic cancers were screened for suitable primary PDAC tumors. The samples Available online 14 March 2019 were analyzed using qRT-PCR, western blotting, and Immunohistochemistry (IHC) staining as well as LRP-1 expression in association with clinicopathological features. Keywords: PDAC Results: The relative LRP-1 mRNA expression was up-regulated in 82.3% (42/51) of the PDAC tumors and ± fi ± LRP-1 its expression (3.72 1.25) was signi cantly higher than that in pancreatic normal margins (1.0 0.23, Prognosis P < 0.05).
    [Show full text]
  • Lrp1 Modulators
    Last updated on February 14, 2021 Cognitive Vitality Reports® are reports written by neuroscientists at the Alzheimer’s Drug Discovery Foundation (ADDF). These scientific reports include analysis of drugs, drugs-in- development, drug targets, supplements, nutraceuticals, food/drink, non-pharmacologic interventions, and risk factors. Neuroscientists evaluate the potential benefit (or harm) for brain health, as well as for age-related health concerns that can affect brain health (e.g., cardiovascular diseases, cancers, diabetes/metabolic syndrome). In addition, these reports include evaluation of safety data, from clinical trials if available, and from preclinical models. Lrp1 Modulators Evidence Summary Lrp1 has a variety of essential functions, mediated by a diverse array of ligands. Therapeutics will need to target specific interactions. Neuroprotective Benefit: Lrp1-mediated interactions promote Aβ clearance, Aβ generation, tau propagation, brain glucose utilization, and brain lipid homeostasis. The therapeutic effect will depend on the interaction targeted. Aging and related health concerns: Lrp1 plays mixed roles in cardiovascular diseases and cancer, dependent on context. Lrp1 is dysregulated in metabolic disease, which may contribute to insulin resistance. Safety: Broad-spectrum Lrp1 modulators are untenable therapeutics due to the high potential for extensive side effects. Therapies that target a specific Lrp1-ligand interaction are expected to have a better therapeutic profile. 1 Last updated on February 14, 2021 Availability: Research use Dose: N/A Chemical formula: N/A S16 is in clinical trials MW: N/A Half life: N/A BBB: Angiopep is a peptide that facilitates BBB penetrance by interacting with Lrp1 Clinical trials: S16, an Lrp1 Observational studies: sLrp1 levels are agonist was tested in healthy altered in Alzheimer’s disease, volunteers (n=10) in a Phase 1 cardiovascular disease, and metabolic study.
    [Show full text]
  • LRP1 Influences Trafficking of N-Type Calcium Channels Via Interaction
    www.nature.com/scientificreports OPEN LRP1 influences trafficking of N-type calcium channels via interaction with the auxiliary α2δ-1 Received: 17 November 2016 Accepted: 30 January 2017 subunit Published: 03 March 2017 Ivan Kadurin, Simon W. Rothwell, Beatrice Lana, Manuela Nieto-Rostro & Annette C. Dolphin 2+ Voltage-gated Ca (CaV) channels consist of a pore-forming α1 subunit, which determines the main functional and pharmacological attributes of the channel. The CaV1 and CaV2 channels are associated with auxiliary β- and α2δ-subunits. The molecular mechanisms involved in α2δ subunit trafficking, and the effect ofα 2δ subunits on trafficking calcium channel complexes remain poorly understood. Here we show that α2δ-1 is a ligand for the Low Density Lipoprotein (LDL) Receptor-related Protein-1 (LRP1), a multifunctional receptor which mediates trafficking of cargoes. This interaction with LRP1 is direct, and is modulated by the LRP chaperone, Receptor-Associated Protein (RAP). LRP1 regulates α2δ binding to gabapentin, and influences calcium channel trafficking and function. Whereas LRP1 alone reducesα 2δ-1 trafficking to the cell-surface, the LRP1/RAP combination enhances mature glycosylation, proteolytic processing and cell-surface expression of α2δ-1, and also increase plasma-membrane expression and function of CaV2.2 when co-expressed with α2δ-1. Furthermore RAP alone produced a small increase in cell-surface expression of CaV2.2, α2δ-1 and the associated calcium currents. It is likely to be interacting with an endogenous member of the LDL receptor family to have these effects. Our findings now provide a key insight and new tools to investigate the trafficking of calcium channelα 2δ subunits.
    [Show full text]
  • Leptomeningeal Metastases from Solid Tumors: Recent Advances in Diagnosis and Molecular Approaches
    cancers Review Leptomeningeal Metastases from Solid Tumors: Recent Advances in Diagnosis and Molecular Approaches Alessia Pellerino 1,* , Priscilla K. Brastianos 2, Roberta Rudà 1,3 and Riccardo Soffietti 1 1 Department of Neuro-Oncology, University and City of Health and Science Hospital, 10126 Turin, Italy; [email protected] (R.R.); riccardo.soffi[email protected] (R.S.) 2 Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02115, USA; [email protected] 3 Department of Neurology, Castelfranco Veneto and Brain Tumor Board Treviso Hospital, 31100 Treviso, Italy * Correspondence: [email protected]; Tel.: +39-011-633-4904 Simple Summary: Leptomeningeal metastases are a devastating complication of solid tumors with poor survival, regardless of the type of treatments. The limited efficacy of targeted agents is due to the molecular divergence between leptomeningeal recurrences and primary site, as well as the presence of a heterogeneous blood-brain barrier and blood-tumor barrier that interfere with the penetration of drugs into the brain. The diagnosis of leptomeningeal metastases is achieved by neurological examination, and/or brain and spinal magnetic resonance, and/or a positive cerebrospinal fluid cytology. The presence of neoplastic cells in the cerebrospinal fluid examination is the gold-standard for the diagnosis of leptomeningeal metastases; however, novel techniques known as “liquid biopsy” aim to improve the sensitivity and specificity in detecting circulating neoplastic cells or DNA in the cerebrospinal fluid. Targeted therapies and immunotherapies have changed the natural history Citation: Pellerino, A.; Brastianos, of metastatic solid tumors, including lung, breast cancer, and melanoma. Targeting actionable P.K.; Rudà, R.; Soffietti, R.
    [Show full text]
  • Sleeping Beauty Transposon Mutagenesis Identifies Genes That
    Sleeping Beauty transposon mutagenesis identifies PNAS PLUS genes that cooperate with mutant Smad4 in gastric cancer development Haruna Takedaa,b, Alistair G. Rustc,d, Jerrold M. Warda, Christopher Chin Kuan Yewa, Nancy A. Jenkinsa,e, and Neal G. Copelanda,e,1 aDivision of Genomics and Genetics, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore 138673; bDepartment of Pathology, School of Medicine, Kanazawa Medical University, Ishikawa 920-0293, Japan; cExperimental Cancer Genetics, Wellcome Trust Sanger Institute, Cambridge CB10 1HH, United Kingdom; dTumour Profiling Unit, The Institute of Cancer Research, Chester Beatty Laboratories, London SW3 6JB, United Kingdom; and eCancer Research Program, Houston Methodist Research Institute, Houston, TX 77030 Contributed by Neal G. Copeland, February 27, 2016 (sent for review October 15, 2015; reviewed by Yoshiaki Ito and David A. Largaespada) Mutations in SMAD4 predispose to the development of gastroin- animal models that mimic human GC, researchers have infected testinal cancer, which is the third leading cause of cancer-related mice with H. pylori and then, treated them with carcinogens. They deaths. To identify genes driving gastric cancer (GC) development, have also used genetic engineering to develop a variety of trans- we performed a Sleeping Beauty (SB) transposon mutagenesis genic and KO mouse models of GC (10). Smad4 KO mice are one + − screen in the stomach of Smad4 / mutant mice. This screen iden- GC model that has been of particular interest to us (11, 12). tified 59 candidate GC trunk drivers and a much larger number of Heterozygous Smad4 KO mice develop polyps in the pyloric re- candidate GC progression genes.
    [Show full text]
  • LRP1B Mutations Are Associated with Favorable Outcomes to Immune Checkpoint Inhibitors Across Multiple Cancer Types
    Open access Original research J Immunother Cancer: first published as 10.1136/jitc-2020-001792 on 2 March 2021. Downloaded from LRP1B mutations are associated with favorable outcomes to immune checkpoint inhibitors across multiple cancer types 1 2 3 4 Landon C Brown , Matthew D Tucker , Ramy Sedhom, Eric B Schwartz, 5 1 1 1 Jason Zhu, Chester Kao, Matthew K Labriola , Rajan T Gupta, 1 6 1 1 1 Daniele Marin, Yuan Wu, Santosh Gupta, Tian Zhang , Michael R Harrison, Daniel J George,1 Ajjai Alva,4 Emmanuel S Antonarakis,3 Andrew J Armstrong1 To cite: Brown LC, Tucker MD, ABSTRACT lung cancer (NSCLC) and renal cell carci- Sedhom R, et al. LRP1B Background Low- density lipoprotein receptor- related noma.1–3 However, not all patients respond mutations are associated with protein 1b (encoded by LRP1B) is a putative tumor favorable outcomes to immune to treatment with ICIs. The use of predictive suppressor, and preliminary evidence suggests LRP1B- checkpoint inhibitors across biomarkers for response has been explored mutated cancers may have improved outcomes with multiple cancer types. Journal in many tumor types with varying degrees immune checkpoint inhibitors (ICI). for ImmunoTherapy of Cancer of success. Programmed-death ligand-1 (PD- Methods We conducted a multicenter, retrospective pan- 2021;9:e001792. doi:10.1136/ L1) expression,1 microsatellite instability jitc-2020-001792 cancer analysis of patients with LRP1B alterations treated 4 with ICI at Duke University, Johns Hopkins University (JHU) (MSI)/mismatch repair deficiency and 5 6 These data were presented at and University of Michigan (UM). The primary objective tumor mutational burden (TMB) are the the ASCO annual meeting 2020, was to assess the association between overall response only FDA- approved predictive biomarkers for Abstract # 3007.
    [Show full text]
  • Identification of LRP1B-Interacting Proteins and Inhibition of Protein
    FEBS Letters 583 (2009) 43–48 journal homepage: www.FEBSLetters.org Identification of LRP1B-interacting proteins and inhibition of protein kinase Ca-phosphorylation of LRP1B by association with PICK1 Tomoko Shiroshima, Chio Oka, Masashi Kawaichi * Division of Gene Function in Animals, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara 630-0192, Japan article info abstract Article history: Recent studies show LDL receptor-related protein 1B, LRP1B as a transducer of extracellular signals. Received 17 October 2008 Here, we identify six interacting partners of the LRP1B cytoplasmic region by yeast two-hybrid Revised 9 November 2008 screen and confirmed their in vivo binding by immunoprecipitation. One of the partners, PICK1 rec- Accepted 18 November 2008 ognizes the C-terminus of LRP1B and LRP1. The cytoplasmic domains of LRP1B are phosphorylated Available online 9 December 2008 by PKCa about 100 times more efficiently than LRP1. Binding of PICK1 inhibits phosphorylation of Edited by Gianni Cesareni LRP1B, but does not affect LRP1 phosphorylation. This study presents the possibility that LRP1B participates in signal transduction which PICK1 may regulate by inhibiting PKC phosphorylation of LRP1B. Keywords: a LDL receptor family LRP1B Structured summary: LRP1 MINT-6801075: Lrp1b (uniprotkb:Q9JI18) physically interacts (MI:0218) with SNTG2 (uniprotkb:Q925E0) PICK1 by two hybrid (MI:0018) JIP MINT-6801030, MINT-6801468: Lrp1b (uniprotkb:Q9JI18) physically interacts (MI:0218) with Pick1 (uni- PDZ domain protkb:Q80VC8) by two
    [Show full text]
  • Activated Α2-Macroglobulin Regulates LRP1 Levels at the Plasma
    www.nature.com/scientificreports OPEN Activated α2-Macroglobulin Regulates LRP1 Levels at the Plasma Membrane through the Received: 3 May 2019 Accepted: 19 August 2019 Activation of a Rab10-dependent Published: xx xx xxxx Exocytic Pathway in Retinal Müller Glial Cells Javier R. Jaldín-Fincati 1,2,3, Virginia Actis Dato1,2, Nicolás M. Díaz1,2, María C. Sánchez1,2, Pablo F. Barcelona1,2 & Gustavo A. Chiabrando 1,2 Activated α2-macroglobulin (α2M*) and its receptor, low-density lipoprotein receptor-related protein 1 (LRP1), have been linked to proliferative retinal diseases. In Müller glial cells (MGCs), the α2M*/LRP1 interaction induces cell signaling, cell migration, and extracellular matrix remodeling, processes closely associated with proliferative disorders. However, the mechanism whereby α2M* and LRP1 participate in the aforementioned pathologies remains incompletely elucidated. Here, we investigate whether α2M* regulates both the intracellular distribution and sorting of LRP1 to the plasma membrane (PM) and how this regulation is involved in the cell migration of MGCs. Using a human Müller glial-derived cell line, MIO-M1, we demonstrate that the α2M*/LRP1 complex is internalized and rapidly reaches early endosomes. Afterward, α2M* is routed to degradative compartments, while LRP1 is accumulated at the PM through a Rab10-dependent exocytic pathway regulated by PI3K/Akt. Interestingly, Rab10 knockdown reduces both LRP1 accumulation at the PM and cell migration of MIO-M1 cells induced by α2M*. Given the importance of MGCs in the maintenance of retinal homeostasis, unravelling this molecular mechanism can potentially provide new therapeutic targets for the treatment of proliferative retinopathies. Te low-density lipoprotein (LDL) receptor-related protein 1 (LRP1) is a member of the LDL receptor gene family.
    [Show full text]