1
|
Molnar N, Miskolci V. Imaging immunometabolism in situ in live animals. IMMUNOMETABOLISM (COBHAM, SURREY) 2024; 6:e00044. [PMID: 39296471 PMCID: PMC11406703 DOI: 10.1097/in9.0000000000000044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/21/2024]
Abstract
Immunometabolism is a rapidly developing field that holds great promise for diagnostic and therapeutic benefits to human diseases. The field has emerged based on seminal findings from in vitro and ex vivo studies that established the fundamental role of metabolism in immune cell effector functions. Currently, the field is acknowledging the necessity of investigating cellular metabolism within the natural context of biological processes. Examining cells in their native microenvironment is essential not only to reveal cell-intrinsic mechanisms but also to understand how cross-talk between neighboring cells regulates metabolism at the tissue level in a local niche. This necessity is driving innovation and advancement in multiple imaging-based technologies to enable analysis of dynamic intracellular metabolism at the single-cell level, with spatial and temporal resolution. In this review, we tally the currently available imaging-based technologies and explore the emerging methods of Raman and autofluorescence lifetime imaging microscopy, which hold significant potential and offer broad applications in the field of immunometabolism.
Collapse
Affiliation(s)
- Nicole Molnar
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers Health, Rutgers University, Newark, NJ, USA
- Center for Cell Signaling, Rutgers Health, Rutgers University, Newark, NJ, USA
- Center for Immunity and Inflammation, Rutgers Health, Rutgers University, Newark, NJ, USA
| | - Veronika Miskolci
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers Health, Rutgers University, Newark, NJ, USA
- Center for Cell Signaling, Rutgers Health, Rutgers University, Newark, NJ, USA
- Center for Immunity and Inflammation, Rutgers Health, Rutgers University, Newark, NJ, USA
| |
Collapse
|
2
|
Cassidy RM, Flores EM, Trinh Nguyen AK, Cheruvu SS, Uribe RA, Krachler AM, Odem MA. Systematic analysis of proximal midgut- and anorectal-originating contractions in larval zebrafish using event feature detection and supervised machine learning algorithms. Neurogastroenterol Motil 2023; 35:e14675. [PMID: 37743702 PMCID: PMC10841157 DOI: 10.1111/nmo.14675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 07/16/2023] [Accepted: 08/28/2023] [Indexed: 09/26/2023]
Abstract
BACKGROUND Zebrafish larvae are translucent, allowing in vivo analysis of gut development and physiology, including gut motility. While recent progress has been made in measuring gut motility in larvae, challenges remain which can influence results, such as how data are interpreted, opportunities for technical user error, and inconsistencies in methods. METHODS To overcome these challenges, we noninvasively introduced Nile Red fluorescent dye to fill the intraluminal gut space in zebrafish larvae and collected serial confocal microscopic images of gut fluorescence. We automated the detection of fluorescent-contrasted contraction events against the median-subtracted signal and compared it to manually annotated gut contraction events across anatomically defined gut regions. Supervised machine learning (multiple logistic regression) was then used to discriminate between true contraction events and noise. To demonstrate, we analyzed motility in larvae under control and reserpine-treated conditions. We also used automated event detection analysis to compare unfed and fed larvae. KEY RESULTS Automated analysis retained event features for proximal midgut-originating retrograde and anterograde contractions and anorectal-originating retrograde contractions. While manual annotation showed reserpine disrupted gut motility, machine learning only achieved equivalent contraction discrimination in controls and failed to accurately identify contractions after reserpine due to insufficient intraluminal fluorescence. Automated analysis also showed feeding had no effect on the frequency of anorectal-originating contractions. CONCLUSIONS & INFERENCES Automated event detection analysis rapidly and accurately annotated contraction events, including the previously neglected phenomenon of anorectal contractions. However, challenges remain to discriminate contraction events based on intraluminal fluorescence under treatment conditions that disrupt functional motility.
Collapse
Affiliation(s)
- Ryan M. Cassidy
- Brown Foundation Institute of Molecular Medicine, McGovern
Medical School at UTHealth, Houston, TX 77030, USA
| | - Erika M. Flores
- Department of Microbiology and Molecular Genetics, McGovern
Medical School at UTHealth, Houston, TX 77030, USA
| | - Anh K. Trinh Nguyen
- Department of Microbiology and Molecular Genetics, McGovern
Medical School at UTHealth, Houston, TX 77030, USA
| | - Sai S. Cheruvu
- Department of Integrative Biology and Pharmacology,
McGovern Medical School at UTHealth, Houston, TX 77030, USA
| | - Rosa A. Uribe
- Department of Biosciences, Rice University, Houston, TX
77005, USA
| | - Anne Marie Krachler
- Department of Microbiology and Molecular Genetics, McGovern
Medical School at UTHealth, Houston, TX 77030, USA
| | - Max A. Odem
- Department of Microbiology and Molecular Genetics, McGovern
Medical School at UTHealth, Houston, TX 77030, USA
| |
Collapse
|
3
|
Davies BM, Katayama JK, Monsivais JE, Adams JR, Dilts ME, Eberting AL, Hansen JM. Real-time analysis of dynamic compartmentalized GSH redox shifts and H 2O 2 availability in undifferentiated and differentiated cells. Biochim Biophys Acta Gen Subj 2023; 1867:130321. [PMID: 36870547 DOI: 10.1016/j.bbagen.2023.130321] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 01/26/2023] [Accepted: 01/29/2023] [Indexed: 03/06/2023]
Abstract
BACKGROUND Glutathione (GSH) is the most abundant, small biothiol antioxidant. GSH redox state (Eh) supports developmental processes, yet with disrupted GSH Eh, poor developmental outcomes may occur. The role of subcellular, compartmentalized redox environments in the context of redox regulation of differentiation is not well understood. Here, using the P19 neurogenesis model of cellular differentiation, kinetics of subcellular H2O2 availability and GSH Eh were evaluated following oxidant exposure. METHODS Stably transfected P19 cell lines expressing H2O2 availability or GSH Eh sensors, Orp1-roGFP or Grx1-roGFP, respectively, targeted to the cytosol, mitochondria, or nucleus were used. Dynamic, compartmentalized changes in H2O2 availability and GSH Eh were measured via spectrophotometric and confocal microscopy over 120 min following treatment with H2O2 (100 μM) in both differentiated and undifferentiated cells. RESULTS Generally, treated undifferentiated cells showed a greater degree and duration of both H2O2 availability and GSH Eh disruption than differentiated neurons. In treated undifferentiated cells, H2O2 availability was similar in all compartments. Interestingly, in treated undifferentiated cells, mitochondrial GSH Eh was most affected in both the initial oxidation and the rebound kinetics compared to other compartments. Pretreatment with an Nrf2 inducer prevented H2O2-induced effects in all compartments of undifferentiated cells. CONCLUSIONS Disruption of redox-sensitive developmental pathways is likely stage specific, where cells that are less differentiated and/or are actively differentiating are most affected. GENERAL SIGNIFICANCE Undifferentiated cells are more susceptible to oxidant-induced redox dysregulation but are protected by chemicals that induce Nrf2. This may preserve developmental programs and diminish the potential for poor developmental outcomes.
Collapse
Affiliation(s)
- Brandon M Davies
- Cell Biology and Physiology Department, Brigham Young University, Provo, UT 84602, USA
| | - Jenna K Katayama
- Cell Biology and Physiology Department, Brigham Young University, Provo, UT 84602, USA
| | - Joshua E Monsivais
- Cell Biology and Physiology Department, Brigham Young University, Provo, UT 84602, USA
| | - James R Adams
- Cell Biology and Physiology Department, Brigham Young University, Provo, UT 84602, USA
| | - Miriam E Dilts
- Cell Biology and Physiology Department, Brigham Young University, Provo, UT 84602, USA
| | - Arielle L Eberting
- Cell Biology and Physiology Department, Brigham Young University, Provo, UT 84602, USA
| | - Jason M Hansen
- Cell Biology and Physiology Department, Brigham Young University, Provo, UT 84602, USA.
| |
Collapse
|
4
|
Lebel M, Cliche DO, Charbonneau M, Adam D, Brochiero E, Dubois CM, Cantin AM. Invadosome Formation by Lung Fibroblasts in Idiopathic Pulmonary Fibrosis. Int J Mol Sci 2022; 24:ijms24010499. [PMID: 36613948 PMCID: PMC9820272 DOI: 10.3390/ijms24010499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/02/2022] [Accepted: 12/14/2022] [Indexed: 12/30/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is characterized by abnormal fibroblast accumulation in the lung leading to extracellular matrix deposition and remodeling that compromise lung function. However, the mechanisms of interstitial invasion and remodeling by lung fibroblasts remain poorly understood. The invadosomes, initially described in cancer cells, consist of actin-based adhesive structures that coordinate with numerous other proteins to form a membrane protrusion capable of degrading the extracellular matrix to promote their invasive phenotype. In this regard, we hypothesized that invadosome formation may be increased in lung fibroblasts from patients with IPF. Public RNAseq datasets from control and IPF lung tissues were used to identify differentially expressed genes associated with invadosomes. Lung fibroblasts isolated from bleomycin-exposed mice and IPF patients were seeded with and without the two approved drugs for treating IPF, nintedanib or pirfenidone on fluorescent gelatin-coated coverslips for invadosome assays. Several matrix and invadosome-associated genes were increased in IPF tissues and in IPF fibroblastic foci. Invadosome formation was significantly increased in lung fibroblasts isolated from bleomycin-exposed mice and IPF patients. The degree of lung fibrosis found in IPF tissues correlated strongly with invadosome production by neighboring cells. Nintedanib suppressed IPF and PDGF-activated lung fibroblast invadosome formation, an event associated with inhibition of the PDGFR/PI3K/Akt pathway and TKS5 expression. Fibroblasts derived from IPF lung tissues express a pro-invadosomal phenotype, which correlates with the severity of fibrosis and is responsive to antifibrotic treatment.
Collapse
Affiliation(s)
- Mégane Lebel
- Respiratory Division, Department of Medicine, Université de Sherbrooke, Sherbrooke, Québec, QC J1H 5N4, Canada
| | - Dominic O. Cliche
- Respiratory Division, Department of Medicine, Université de Sherbrooke, Sherbrooke, Québec, QC J1H 5N4, Canada
| | - Martine Charbonneau
- Department of Immunology and Cell Biology, Université de Sherbrooke, Sherbrooke, Québec, QC J1H 5N4, Canada
| | - Damien Adam
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada
| | - Emmanuelle Brochiero
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC H2X 0A9, Canada
- Department of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Claire M. Dubois
- Department of Immunology and Cell Biology, Université de Sherbrooke, Sherbrooke, Québec, QC J1H 5N4, Canada
| | - André M. Cantin
- Respiratory Division, Department of Medicine, Université de Sherbrooke, Sherbrooke, Québec, QC J1H 5N4, Canada
- Correspondence: ; Tel.: +819-346-1110 (ext. 14881)
| |
Collapse
|
5
|
Akinaga K, Azumi Y, Mogi K, Toyoizumi R. Stage-dependent sequential organization of nascent smooth muscle cells and its implications for the gut coiling morphogenesis in Xenopus larva. ZOOLOGY 2021; 146:125905. [PMID: 33631602 DOI: 10.1016/j.zool.2021.125905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 01/25/2021] [Accepted: 02/07/2021] [Indexed: 10/22/2022]
Abstract
In vertebrates, gut coiling proceeds left-right asymmetrically during expansion of the gastrointestinal tract with highly organized muscular structures facilitating peristalsis. In this report, we explored the mechanisms of larval gut coiling morphogenesis relevant to its nascent smooth muscle cells using highly transparent Xenopus early larvae. First, to visualize the dynamics of intestinal smooth muscle cells, whole-mount specimens were immunostained with anti-smooth muscle-specific actin (SM-actin) antibody. We found that the nascent gut of Xenopus early larvae gradually expands the SM-actin-positive region in a stage-dependent manner. Transverse orientation of smooth muscle cells was first established, and next, the cellular longitudinal orientation along the gut axis was followed to make a meshwork of the contractile cells. Finally, anisotropic torsion by the smooth muscle cells was generated in the center of gut coiling, suggesting that twisting force might be involved in the late phase of coiling morphogenesis of the gut. Administration of S-(-)-Blebbistatin to attenuate the actomyosin contraction in vivo resulted in cancellation of coiling of the gut. Development of decapitation embryos, trunk 'torso' explants, and gut-only explants revealed that initial coiling of the gut proceeds without interactions with the other parts of the body including the central nervous system. We newly developed an in vitro model to assess the gut coiling morphogenesis, indicating that coiling pattern of the nascent Xenopus gut is partially gut-autonomous. Using this gut explant culture technique, inhibition of actomyosin contraction was performed by administrating either actin polymerization inhibitor, myosin light chain kinase inhibitor, or calmodulin antagonist. All of these reagents decreased the extent of gut coiling morphogenesis in vitro. Taken together, these results suggest that the contraction force generated by actomyosin-rich intestinal smooth muscle cells during larval stages is essential for the normal coiling morphogenesis of this muscular tubular organ.
Collapse
Affiliation(s)
- Kaoru Akinaga
- Department of Biological Sciences, Faculty of Science, Kanagawa University, Tsuchiya 2946, Hiratsuka City, Kanagawa, 259-1293, Japan
| | - Yoshitaka Azumi
- Department of Biological Sciences, Faculty of Science, Kanagawa University, Tsuchiya 2946, Hiratsuka City, Kanagawa, 259-1293, Japan; Research Institute for Integrated Science, Kanagawa University, Japan
| | - Kazue Mogi
- Research Institute for Integrated Science, Kanagawa University, Japan
| | - Ryuji Toyoizumi
- Department of Biological Sciences, Faculty of Science, Kanagawa University, Tsuchiya 2946, Hiratsuka City, Kanagawa, 259-1293, Japan; Research Institute for Integrated Science, Kanagawa University, Japan.
| |
Collapse
|
6
|
Matthiesen CL, Hu L, Torslev AS, Poulsen ET, Larsen UG, Kjaer-Sorensen K, Thomsen JS, Brüel A, Enghild JJ, Oxvig C, Petersen SV. Superoxide dismutase 3 is expressed in bone tissue and required for normal bone homeostasis and mineralization. Free Radic Biol Med 2021; 164:399-409. [PMID: 33476796 DOI: 10.1016/j.freeradbiomed.2021.01.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/22/2020] [Accepted: 01/12/2021] [Indexed: 12/14/2022]
Abstract
Superoxide dismutase 3 (SOD3) is an extracellular protein with the capacity to convert superoxide into hydrogen peroxide, an important secondary messenger in redox regulation. To investigate the utility of zebrafish in functional studies of SOD3 and its relevance for redox regulation, we have characterized the zebrafish orthologues; Sod3a and Sod3b. Our analyses show that both recombinant Sod3a and Sod3b express SOD activity, however, only Sod3b is able to bind heparin. Furthermore, RT-PCR analyses reveal that sod3a and sod3b are expressed in zebrafish embryos and are present primarily in separate organs in adult zebrafish, suggesting distinct functions in vivo. Surprisingly, both RT-PCR and whole mount in situ hybridization showed specific expression of sod3b in skeletal tissue. To further investigate this observation, we compared femoral bone obtained from wild-type and SOD3-/- mice to determine whether a functional difference was apparent in healthy adult mice. Here we report, that bone from SOD3-/- mice is less mineralized and characterized by significant reduction of cortical and trabecular thickness in addition to reduced mechanical strength. These analyses show that SOD3 plays a hitherto unappreciated role in bone development and homeostasis.
Collapse
Affiliation(s)
| | - Lili Hu
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Ebbe T Poulsen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Ulrike G Larsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | | | - Annemarie Brüel
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Jan J Enghild
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | | |
Collapse
|
7
|
Naydenov NG, Lechuga S, Huang EH, Ivanov AI. Myosin Motors: Novel Regulators and Therapeutic Targets in Colorectal Cancer. Cancers (Basel) 2021; 13:741. [PMID: 33670106 PMCID: PMC7916823 DOI: 10.3390/cancers13040741] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 02/06/2021] [Accepted: 02/08/2021] [Indexed: 12/11/2022] Open
Abstract
Colorectal cancer (CRC) remains the third most common cause of cancer and the second most common cause of cancer deaths worldwide. Clinicians are largely faced with advanced and metastatic disease for which few interventions are available. One poorly understood aspect of CRC involves altered organization of the actin cytoskeleton, especially at the metastatic stage of the disease. Myosin motors are crucial regulators of actin cytoskeletal architecture and remodeling. They act as mechanosensors of the tumor environments and control key cellular processes linked to oncogenesis, including cell division, extracellular matrix adhesion and tissue invasion. Different myosins play either oncogenic or tumor suppressor roles in breast, lung and prostate cancer; however, little is known about their functions in CRC. This review focuses on the functional roles of myosins in colon cancer development. We discuss the most studied class of myosins, class II (conventional) myosins, as well as several classes (I, V, VI, X and XVIII) of unconventional myosins that have been linked to CRC development. Altered expression and mutations of these motors in clinical tumor samples and their roles in CRC growth and metastasis are described. We also evaluate the potential of using small molecular modulators of myosin activity to develop novel anticancer therapies.
Collapse
Affiliation(s)
- Nayden G. Naydenov
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA; (N.G.N.); (S.L.)
| | - Susana Lechuga
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA; (N.G.N.); (S.L.)
| | - Emina H. Huang
- Departments of Cancer Biology and Colorectal Surgery, Cleveland Clinic Foundation, Cleveland, OH 44195, USA;
| | - Andrei I. Ivanov
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA; (N.G.N.); (S.L.)
| |
Collapse
|
8
|
Nichols EL, Smith CJ. Functional Regeneration of the Sensory Root via Axonal Invasion. Cell Rep 2021; 30:9-17.e3. [PMID: 31914401 PMCID: PMC6996490 DOI: 10.1016/j.celrep.2019.12.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 10/15/2019] [Accepted: 12/03/2019] [Indexed: 12/11/2022] Open
Abstract
Regeneration following spinal root avulsion is broadly unsuccessful
despite the regenerative capacity of other PNS-located nerves. By combining
focal laser lesioning to model root avulsion in zebrafish, time-lapse imaging,
and transgenesis, we identify that regenerating DRG neurons fail to recapitulate
developmental paradigms of actin-based invasion after injury. We demonstrate
that inducing actin reorganization into invasive components via pharmacological
and genetic approaches in the regenerating axon can rescue sensory axon spinal
cord entry. Cell-autonomous induction of invasion components using
constitutively active Src induces DRG axon regeneration, suggesting an intrinsic
mechanism can be activated to drive regeneration. Furthermore, analyses of
neuronal activity and animal behavior show restoration of sensory circuit
activity and behavior upon stimulating axons to re-enter the spinal cord via
invasion. Altogether, our data identify induction of invasive components as
sufficient for functional sensory root regeneration after injury. Dorsal root ganglion (DRG) sensory axons are unable to regenerate into
the spinal cord after injury. Nichols and Smith demonstrate in zebrafish that
injured DRG axons do not initiate actin-based invasion components during
re-entry into the spinal cord. Pharmacological and cell-autonomous genetic
manipulations that promote actin-mediated cell invasion to restore sensory
behavior.
Collapse
Affiliation(s)
- Evan L Nichols
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA; Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, IN, USA
| | - Cody J Smith
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA; Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, IN, USA.
| |
Collapse
|
9
|
Kostyuk AI, Panova AS, Kokova AD, Kotova DA, Maltsev DI, Podgorny OV, Belousov VV, Bilan DS. In Vivo Imaging with Genetically Encoded Redox Biosensors. Int J Mol Sci 2020; 21:E8164. [PMID: 33142884 PMCID: PMC7662651 DOI: 10.3390/ijms21218164] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 12/13/2022] Open
Abstract
Redox reactions are of high fundamental and practical interest since they are involved in both normal physiology and the pathogenesis of various diseases. However, this area of research has always been a relatively problematic field in the context of analytical approaches, mostly because of the unstable nature of the compounds that are measured. Genetically encoded sensors allow for the registration of highly reactive molecules in real-time mode and, therefore, they began a new era in redox biology. Their strongest points manifest most brightly in in vivo experiments and pave the way for the non-invasive investigation of biochemical pathways that proceed in organisms from different systematic groups. In the first part of the review, we briefly describe the redox sensors that were used in vivo as well as summarize the model systems to which they were applied. Next, we thoroughly discuss the biological results obtained in these studies in regard to animals, plants, as well as unicellular eukaryotes and prokaryotes. We hope that this work reflects the amazing power of this technology and can serve as a useful guide for biologists and chemists who work in the field of redox processes.
Collapse
Affiliation(s)
- Alexander I. Kostyuk
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (A.I.K.); (A.S.P.); (A.D.K.); (D.A.K.); (D.I.M.); (O.V.P.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Anastasiya S. Panova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (A.I.K.); (A.S.P.); (A.D.K.); (D.A.K.); (D.I.M.); (O.V.P.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Aleksandra D. Kokova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (A.I.K.); (A.S.P.); (A.D.K.); (D.A.K.); (D.I.M.); (O.V.P.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Daria A. Kotova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (A.I.K.); (A.S.P.); (A.D.K.); (D.A.K.); (D.I.M.); (O.V.P.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Dmitry I. Maltsev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (A.I.K.); (A.S.P.); (A.D.K.); (D.A.K.); (D.I.M.); (O.V.P.); (V.V.B.)
- Federal Center for Cerebrovascular Pathology and Stroke, 117997 Moscow, Russia
| | - Oleg V. Podgorny
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (A.I.K.); (A.S.P.); (A.D.K.); (D.A.K.); (D.I.M.); (O.V.P.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Vsevolod V. Belousov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (A.I.K.); (A.S.P.); (A.D.K.); (D.A.K.); (D.I.M.); (O.V.P.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
- Federal Center for Cerebrovascular Pathology and Stroke, 117997 Moscow, Russia
- Institute for Cardiovascular Physiology, Georg August University Göttingen, D-37073 Göttingen, Germany
| | - Dmitry S. Bilan
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (A.I.K.); (A.S.P.); (A.D.K.); (D.A.K.); (D.I.M.); (O.V.P.); (V.V.B.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| |
Collapse
|
10
|
Breus O, Dickmeis T. Genetically encoded thiol redox-sensors in the zebrafish model: lessons for embryonic development and regeneration. Biol Chem 2020; 402:363-378. [PMID: 33021959 DOI: 10.1515/hsz-2020-0269] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 09/28/2020] [Indexed: 12/14/2022]
Abstract
Important roles for reactive oxygen species (ROS) and redox signaling in embryonic development and regenerative processes are increasingly recognized. However, it is difficult to obtain information on spatiotemporal dynamics of ROS production and signaling in vivo. The zebrafish is an excellent model for in vivo bioimaging and possesses a remarkable regenerative capacity upon tissue injury. Here, we review data obtained in this model system with genetically encoded redox-sensors targeting H2O2 and glutathione redox potential. We describe how such observations have prompted insight into regulation and downstream effects of redox alterations during tissue differentiation, morphogenesis and regeneration. We also discuss the properties of the different sensors and their consequences for the interpretation of in vivo imaging results. Finally, we highlight open questions and additional research fields that may benefit from further application of such sensor systems in zebrafish models of development, regeneration and disease.
Collapse
Affiliation(s)
- Oksana Breus
- Institute of Biological and Chemical Systems - Biological Information Processing, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, D-76344Eggenstein-Leopoldshafen, Germany
| | - Thomas Dickmeis
- Institute of Biological and Chemical Systems - Biological Information Processing, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, D-76344Eggenstein-Leopoldshafen, Germany
| |
Collapse
|
11
|
Nichols EL, Smith CJ. Synaptic-like Vesicles Facilitate Pioneer Axon Invasion. Curr Biol 2019; 29:2652-2664.e4. [DOI: 10.1016/j.cub.2019.06.078] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 05/24/2019] [Accepted: 06/26/2019] [Indexed: 12/19/2022]
|
12
|
Peláez R, Pariente A, Pérez-Sala Á, Larrayoz IM. Integrins: Moonlighting Proteins in Invadosome Formation. Cancers (Basel) 2019; 11:cancers11050615. [PMID: 31052560 PMCID: PMC6562994 DOI: 10.3390/cancers11050615] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 04/26/2019] [Accepted: 04/28/2019] [Indexed: 12/24/2022] Open
Abstract
Invadopodia are actin-rich protrusions developed by transformed cells in 2D/3D environments that are implicated in extracellular matrix (ECM) remodeling and degradation. These structures have an undoubted association with cancer invasion and metastasis because invadopodium formation in vivo is a key step for intra/extravasation of tumor cells. Invadopodia are closely related to other actin-rich structures known as podosomes, which are typical structures of normal cells necessary for different physiological processes during development and organogenesis. Invadopodia and podosomes are included in the general term 'invadosomes,' as they both appear as actin puncta on plasma membranes next to extracellular matrix metalloproteinases, although organization, regulation, and function are slightly different. Integrins are transmembrane proteins implicated in cell-cell and cell-matrix interactions and other important processes such as molecular signaling, mechano-transduction, and cell functions, e.g., adhesion, migration, or invasion. It is noteworthy that integrin expression is altered in many tumors, and other pathologies such as cardiovascular or immune dysfunctions. Over the last few years, growing evidence has suggested a role of integrins in the formation of invadopodia. However, their implication in invadopodia formation and adhesion to the ECM is still not well known. This review focuses on the role of integrins in invadopodium formation and provides a general overview of the involvement of these proteins in the mechanisms of metastasis, taking into account classic research through to the latest and most advanced work in the field.
Collapse
Affiliation(s)
- Rafael Peláez
- Biomarkers and Molecular Signaling Group, Neurodegenerative Diseases Area Center for Biomedical Research of La Rioja, CIBIR, c.p., 26006. Logroño, Spain.
| | - Ana Pariente
- Biomarkers and Molecular Signaling Group, Neurodegenerative Diseases Area Center for Biomedical Research of La Rioja, CIBIR, c.p., 26006. Logroño, Spain.
| | - Álvaro Pérez-Sala
- Biomarkers and Molecular Signaling Group, Neurodegenerative Diseases Area Center for Biomedical Research of La Rioja, CIBIR, c.p., 26006. Logroño, Spain.
| | - Ignacio M Larrayoz
- Biomarkers and Molecular Signaling Group, Neurodegenerative Diseases Area Center for Biomedical Research of La Rioja, CIBIR, c.p., 26006. Logroño, Spain.
| |
Collapse
|
13
|
Li D, Wenger TL, Seiler C, March ME, Gutierrez-Uzquiza A, Kao C, Bhoj E, Tian L, Rosenbach M, Liu Y, Robinson N, Behr M, Chiavacci R, Hou C, Wang T, Bakay M, Pellegrino da Silva R, Perkins JA, Sleiman P, Levine MA, Hicks PJ, Itkin M, Dori Y, Hakonarson H. Pathogenic variant in EPHB4 results in central conducting lymphatic anomaly. Hum Mol Genet 2019; 27:3233-3245. [PMID: 29905864 DOI: 10.1093/hmg/ddy218] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 06/04/2018] [Indexed: 12/30/2022] Open
Abstract
Central conducting lymphatic anomaly (CCLA) is one of the complex lymphatic anomalies characterized by dilated lymphatic channels, lymphatic channel dysmotility and distal obstruction affecting lymphatic drainage. We performed whole exome sequencing (WES) of DNA from a four-generation pedigree and examined the consequences of the variant by transfection of mammalian cells and morpholino and rescue studies in zebrafish. WES revealed a heterozygous mutation in EPHB4 (RefSeq NM_004444.4; c.2334 + 1G>C) and RNA-Seq demonstrated that the EPHB4 mutation destroys the normal donor site, which leads to the use of a cryptic splice donor that results in retention of the intervening 12-bp intron sequence. Transient co-expression of the wild-type and mutant EPHB4 proteins showed reduced phosphorylation of tyrosine, consistent with a loss-of-function effect. Zebrafish ephb4a morpholino resulted in vessel misbranching and deformities in the lymphatic vessel development, indicative of possible differentiation defects in lymphatic vessels, mimicking the lymphatic presentations of the patients. Immunoblot analysis using zebrafish lysates demonstrated over-activation of mTORC1 as a consequence of reduced EPHB4 signaling. Strikingly, drugs that inhibit mTOR signaling or RAS-MAPK signaling effectively rescued the misbranching phenotype in a comparable manner. Moreover, knock-in of EPHB4 mutation in HEK293T cells also induced mTORC1 activity. Our data demonstrate the pathogenicity of the identified EPHB4 mutation as a novel cause of CCLA and suggesting that ERK inhibitors may have therapeutic benefits in such patients with complex lymphatic anomalies.
Collapse
Affiliation(s)
- Dong Li
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Tara L Wenger
- Division of Craniofacial Medicine, Seattle Children's Hospital, Seattle, WA, USA
| | - Christoph Seiler
- Zebrafish core, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Michael E March
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Charlly Kao
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Elizabeth Bhoj
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Lifeng Tian
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Misha Rosenbach
- Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Yichuan Liu
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Nora Robinson
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Mechenzie Behr
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Rosetta Chiavacci
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Cuiping Hou
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Tiancheng Wang
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Marina Bakay
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Jonathan A Perkins
- Division of Otolaryngology-Head and Neck Surgery, Seattle Children's Hospital, Seattle, WA, USA
| | - Patrick Sleiman
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Michael A Levine
- Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, PA, USA.,Division of Endocrinology and Diabetes
| | - Patricia J Hicks
- Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Maxim Itkin
- Center for Lymphatic Imaging and Interventions
| | - Yoav Dori
- Center for Lymphatic Imaging and Interventions
| | - Hakon Hakonarson
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, PA, USA.,Divisions of Human Genetics and Pulmonary Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
14
|
Zhang J, Xu S, Zhang Y, Zou S, Li X. Effects of equibiaxial mechanical stretch on extracellular matrix-related gene expression in human calvarial osteoblasts. Eur J Oral Sci 2018; 127:10-18. [PMID: 30474904 DOI: 10.1111/eos.12595] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mechanical stretch commonly promotes craniofacial suture remodeling during interceptive orthodontics. The mechanical responses of osteoblasts in craniofacial sutures play a role in suture remodeling. Moreover, the extracellular matrix (ECM) produced by osteoblasts is crucial for the transduction of mechanical signals that promote cell differentiation. Therefore, we aimed to investigate the effect of mechanical stretch on cell viability and ECM-related gene-expression changes in human osteoblasts. Human calvarial osteoblasts (HCObs) were subjected to 2% deformation. Caspase activity, MTT, and cell viability assays were used to estimate osteoblast apoptosis, proliferation, and viability, respectively. Real-time RT-PCR (RT2 -PCR) arrays were used to assess expression of cytoskeletal-, apoptosis-, osteogenesis-, and ECM-related genes. We found that mechanical stretch significantly increased osteoblast viability and cell proliferation, and decreased the activities of caspases 3 and 7. Moreover, the expression of 18 genes related to osteoblast differentiation, apoptosis, and ECM remodeling changed by more than two-fold in a time-dependent manner. Therefore, mechanical stretch promotes HCOb viability and alters expression of genes that are closely related to suture remodeling under mechanical stretch.
Collapse
Affiliation(s)
- Jiawei Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shuhao Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yanggen Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shujuan Zou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiaobing Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
15
|
Rampon C, Volovitch M, Joliot A, Vriz S. Hydrogen Peroxide and Redox Regulation of Developments. Antioxidants (Basel) 2018; 7:E159. [PMID: 30404180 PMCID: PMC6262372 DOI: 10.3390/antiox7110159] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 10/10/2018] [Accepted: 10/10/2018] [Indexed: 01/16/2023] Open
Abstract
Reactive oxygen species (ROS), which were originally classified as exclusively deleterious compounds, have gained increasing interest in the recent years given their action as bona fide signalling molecules. The main target of ROS action is the reversible oxidation of cysteines, leading to the formation of disulfide bonds, which modulate protein conformation and activity. ROS, endowed with signalling properties, are mainly produced by NADPH oxidases (NOXs) at the plasma membrane, but their action also involves a complex machinery of multiple redox-sensitive protein families that differ in their subcellular localization and their activity. Given that the levels and distribution of ROS are highly dynamic, in part due to their limited stability, the development of various fluorescent ROS sensors, some of which are quantitative (ratiometric), represents a clear breakthrough in the field and have been adapted to both ex vivo and in vivo applications. The physiological implication of ROS signalling will be presented mainly in the frame of morphogenetic processes, embryogenesis, regeneration, and stem cell differentiation. Gain and loss of function, as well as pharmacological strategies, have demonstrated the wide but specific requirement of ROS signalling at multiple stages of these processes and its intricate relationship with other well-known signalling pathways.
Collapse
Affiliation(s)
- Christine Rampon
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, 75231 Paris, France.
- Sorbonne Paris Cité, Univ Paris Diderot, Biology Department, 75205 Paris CEDEX 13, France.
| | - Michel Volovitch
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, 75231 Paris, France.
- École Normale Supérieure, Department of Biology, PSL Research University, 75005 Paris, France.
| | - Alain Joliot
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, 75231 Paris, France.
| | - Sophie Vriz
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, 75231 Paris, France.
- Sorbonne Paris Cité, Univ Paris Diderot, Biology Department, 75205 Paris CEDEX 13, France.
| |
Collapse
|
16
|
Swain L, Nanadikar MS, Borowik S, Zieseniss A, Katschinski DM. Transgenic Organisms Meet Redox Bioimaging: One Step Closer to Physiology. Antioxid Redox Signal 2018; 29:603-612. [PMID: 29320870 DOI: 10.1089/ars.2017.7469] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
SIGNIFICANCE Redox signaling is a common mechanism in the cellular response toward a variety of stimuli. For analyzing redox-dependent specific alterations in a cell, genetically encoded biosensors were highly instrumental in the past. To advance the knowledge about the importance of this signaling mechanism in vivo, models that are as close as possible to physiology are needed. Recent Advances: The development of transgenic (tg) redox biosensor animal models has enhanced the knowledge of redox signaling under patho(physio)logical conditions. So far, commonly used small animal models, that is, Caenorhabditis elegans, Drosophila melanogaster, and Danio rerio, and genetically modified mice were employed for redox biosensor transgenesis. However, especially the available mouse models are still limited. CRITICAL ISSUES The analysis of redox biosensor responses in vivo at the tissue level, especially for internal organs, is hampered by the detection limit of the available redox biosensors and microscopy techniques. Recent technical developments such as redox histology and the analysis of cell-type-specific biosensor responses need to be further refined and followed up in a systematic manner. FUTURE DIRECTIONS The usage of tg animal models in the field of redox signaling has helped to answer open questions. Application of the already established models and consequent development of more defined tg models will enable this research area to define the role of redox signaling in (patho)physiology in further depth. Antioxid. Redox Signal. 29, 603-612.
Collapse
Affiliation(s)
- Lija Swain
- 1 Vascular Biology Unit, Boston University School of Medicine, Boston University , Boston, Massachusetts
| | - Maithily S Nanadikar
- 2 Institute of Cardiovascular Physiology, University Medical Center Göttingen, Georg-August University of Göttingen , Göttingen, Germany
| | - Sergej Borowik
- 2 Institute of Cardiovascular Physiology, University Medical Center Göttingen, Georg-August University of Göttingen , Göttingen, Germany
| | - Anke Zieseniss
- 2 Institute of Cardiovascular Physiology, University Medical Center Göttingen, Georg-August University of Göttingen , Göttingen, Germany
| | - Dörthe M Katschinski
- 2 Institute of Cardiovascular Physiology, University Medical Center Göttingen, Georg-August University of Göttingen , Göttingen, Germany
| |
Collapse
|
17
|
Byrnes J, Ganetzky R, Lightfoot R, Tzeng M, Nakamaru-Ogiso E, Seiler C, Falk MJ. Pharmacologic modeling of primary mitochondrial respiratory chain dysfunction in zebrafish. Neurochem Int 2018; 117:23-34. [PMID: 28732770 PMCID: PMC5773416 DOI: 10.1016/j.neuint.2017.07.008] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 07/11/2017] [Accepted: 07/16/2017] [Indexed: 02/07/2023]
Abstract
Mitochondrial respiratory chain (RC) disease is a heterogeneous and highly morbid group of energy deficiency disorders for which no proven effective therapies exist. Robust vertebrate animal models of primary RC dysfunction are needed to explore the effects of variation in RC disease subtypes, tissue-specific manifestations, and major pathogenic factors contributing to each disorder, as well as their pre-clinical response to therapeutic candidates. We have developed a series of zebrafish (Danio rerio) models that inhibit, to variable degrees, distinct aspects of RC function, and enable quantification of animal development, survival, behaviors, and organ-level treatment effects as well as effects on mitochondrial biochemistry and physiology. Here, we characterize four pharmacologic inhibitor models of mitochondrial RC dysfunction in early larval zebrafish, including rotenone (complex I inhibitor), azide (complex IV inhibitor), oligomycin (complex V inhibitor), and chloramphenicol (mitochondrial translation inhibitor that leads to multiple RC complex dysfunction). A range of concentrations and exposure times of each RC inhibitor were systematically evaluated on early larval development, animal survival, integrated behaviors (touch and startle responses), organ physiology (brain death, neurologic tone, heart rate), and fluorescence-based analyses of mitochondrial physiology in zebrafish skeletal muscle. Pharmacologic RC inhibitor effects were validated by spectrophotometric analysis of Complex I, II and IV enzyme activities, or relative quantitation of ATP levels in larvae. Outcomes were prioritized that utilize in vivo animal imaging and quantitative behavioral assessments, as may optimally inform the translational potential of pre-clinical drug screens for future clinical study in human mitochondrial disease subjects. The RC complex inhibitors each delayed early embryo development, with short-term exposures of these three agents or chloramphenicol from 5 to 7 days post fertilization also causing reduced larval survival and organ-specific defects ranging from brain death, behavioral and neurologic alterations, reduced mitochondrial membrane potential in skeletal muscle (rotenone), and/or cardiac edema with visible blood pooling (oligomycin). Remarkably, we demonstrate that treating animals with probucol, a nutrient-sensing signaling network modulating drug that has been shown to yield therapeutic effects in a range of other RC disease cellular and animal models, both prevented acute rotenone-induced brain death in zebrafish larvae, and significantly rescued early embryo developmental delay from either rotenone or oligomycin exposure. Overall, these zebrafish pharmacologic RC function inhibition models offer a unique opportunity to gain novel insights into diverse developmental, survival, organ-level, and behavioral defects of varying severity, as well as their individual response to candidate therapies, in a highly tractable and cost-effective vertebrate animal model system.
Collapse
Affiliation(s)
- James Byrnes
- Division of Human Genetics, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States
| | - Rebecca Ganetzky
- Division of Human Genetics, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States
| | - Richard Lightfoot
- Division of Human Genetics, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States
| | - Michael Tzeng
- Division of Human Genetics, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States
| | - Eiko Nakamaru-Ogiso
- Division of Human Genetics, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States
| | - Christoph Seiler
- Aquatics Core Facility, The Children's Hospital of Philadelphia Research Institute, Philadelphia, PA 19104, United States
| | - Marni J Falk
- Division of Human Genetics, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States; Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, United States.
| |
Collapse
|
18
|
Abstract
Although the zebrafish was initially developed as a model system to study embryonic development, it has gained increasing attention as an advantageous system to investigate human diseases, including intestinal disorders. Zebrafish embryos develop rapidly, and their digestive system is fully functional and visible by 5days post fertilization. There is a large degree of homology between the intestine of zebrafish and higher vertebrate organisms in terms of its cellular composition and function as both a digestive and immune organ. Furthermore, molecular pathways regulating injury and immune responses are highly conserved. In this chapter, we provide an overview of studies addressing developmental and physiological processes relevant to human intestinal disease. These studies include those related to congenital disorders, host-microbiota interactions, inflammatory diseases, motility disorders, and intestinal cancer. We also highlight the utility of zebrafish to functionally validate candidate genes identified through mutational analyses and genome-wide association studies, and discuss methodologies to investigate the intestinal biology that are unique to zebrafish.
Collapse
Affiliation(s)
- X Zhao
- University of Pennsylvania, Philadelphia, PA, United States
| | - M Pack
- University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
19
|
Kohrman AQ, Matus DQ. Divide or Conquer: Cell Cycle Regulation of Invasive Behavior. Trends Cell Biol 2017; 27:12-25. [PMID: 27634432 PMCID: PMC5186408 DOI: 10.1016/j.tcb.2016.08.003] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 07/30/2016] [Accepted: 08/10/2016] [Indexed: 12/18/2022]
Abstract
Cell invasion through the basement membrane (BM) occurs during normal embryonic development and is a fundamental feature of cancer metastasis. The underlying cellular and genetic machinery required for invasion has been difficult to identify, due to a lack of adequate in vivo models to accurately examine invasion in single cells at subcellular resolution. Recent evidence has documented a functional link between cell cycle arrest and invasive activity. While cancer progression is traditionally thought of as a disease of uncontrolled cell proliferation, cancer cell dissemination, a critical aspect of metastasis, may require a switch from a proliferative to an invasive state. In this work, we review evidence that BM invasion requires cell cycle arrest and discuss the implications of this concept with regard to limiting the lethality associated with cancer metastasis.
Collapse
Affiliation(s)
- Abraham Q Kohrman
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, USA
| | - David Q Matus
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, USA.
| |
Collapse
|
20
|
Zhao X, Lorent K, Wilkins B, Marchione DM, Gillespie K, Waisbourd-Zinman O, So J, Koo KA, Shin D, Porter JR, Wells RG, Blair I, Pack M. Glutathione antioxidant pathway activity and reserve determine toxicity and specificity of the biliary toxin biliatresone in zebrafish. Hepatology 2016; 64:894-907. [PMID: 27102575 PMCID: PMC5251204 DOI: 10.1002/hep.28603] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/15/2016] [Accepted: 04/12/2016] [Indexed: 12/13/2022]
Abstract
UNLABELLED Biliatresone is an electrophilic isoflavone isolated from Dysphania species plants that has been causatively linked to naturally occurring outbreaks of a biliary atresia (BA)-like disease in livestock. Biliatresone has selective toxicity for extrahepatic cholangiocytes (EHCs) in zebrafish larvae. To better understand its mechanism of toxicity, we performed transcriptional profiling of liver cells isolated from zebrafish larvae at the earliest stage of biliatresone-mediated biliary injury, with subsequent comparison of biliary and hepatocyte gene expression profiles. Transcripts encoded by genes involved in redox stress response, particularly those involved in glutathione (GSH) metabolism, were among the most prominently up-regulated in both cholangiocytes and hepatocytes of biliatresone-treated larvae. Consistent with these findings, hepatic GSH was depleted at the onset of biliary injury, and in situ mapping of the hepatic GSH redox potential using a redox-sensitive green fluorescent protein biosensor showed that it was significantly more oxidized in EHCs both before and after treatment with biliatresone. Pharmacological and genetic manipulation of GSH redox homeostasis confirmed the importance of GSH in modulating biliatresone-induced injury given that GSH depletion sensitized both EHCs and the otherwise resistant intrahepatic cholangiocytes to the toxin, whereas replenishing GSH level by N-acetylcysteine administration or activation of nuclear factor erythroid 2-like 2 (Nrf2), a transcriptional regulator of GSH synthesis, inhibited EHC injury. CONCLUSION These findings strongly support redox stress as a critical contributing factor in biliatresone-induced cholangiocyte injury, and suggest that variations in intrinsic stress responses underlie the susceptibility profile. Insufficient antioxidant capacity of EHCs may be critical to early pathogenesis of human BA. (Hepatology 2016;64:894-907).
Collapse
Affiliation(s)
- Xiao Zhao
- Division of Gastroenterology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kristin Lorent
- Division of Gastroenterology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Benjamin Wilkins
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Dylan M. Marchione
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kevin Gillespie
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Orith Waisbourd-Zinman
- Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Juhoon So
- Department of Developmental Biology, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Kyung Ah Koo
- Department of Biological Sciences, University of the Sciences, Philadelphia, PA 19104, USA
| | - Donghun Shin
- Department of Developmental Biology, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - John R. Porter
- Department of Biological Sciences, University of the Sciences, Philadelphia, PA 19104, USA
| | - Rebecca G. Wells
- Division of Gastroenterology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ian Blair
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael Pack
- Division of Gastroenterology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA., Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
21
|
Castro-Castro A, Marchesin V, Monteiro P, Lodillinsky C, Rossé C, Chavrier P. Cellular and Molecular Mechanisms of MT1-MMP-Dependent Cancer Cell Invasion. Annu Rev Cell Dev Biol 2016; 32:555-576. [PMID: 27501444 DOI: 10.1146/annurev-cellbio-111315-125227] [Citation(s) in RCA: 180] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Metastasis is responsible for most cancer-associated deaths. Accumulating evidence based on 3D migration models has revealed a diversity of invasive migratory schemes reflecting the plasticity of tumor cells to switch between proteolytic and nonproteolytic modes of invasion. Yet, initial stages of localized regional tumor dissemination require proteolytic remodeling of the extracellular matrix to overcome tissue barriers. Recent data indicate that surface-exposed membrane type 1-matrix metalloproteinase (MT1-MMP), belonging to a group of membrane-anchored MMPs, plays a central role in pericellular matrix degradation during basement membrane and interstitial tissue transmigration programs. In addition, a large body of work indicates that MT1-MMP is targeted to specialized actin-rich cell protrusions termed invadopodia, which are responsible for matrix degradation. This review describes the multistep assembly of actin-based invadopodia in molecular details. Mechanisms underlying MT1-MMP traffic to invadopodia through endocytosis/recycling cycles, which are key to the invasive program of carcinoma cells, are discussed.
Collapse
Affiliation(s)
| | | | - Pedro Monteiro
- Barts Cancer Institute, University of London John Vane Science Centre, London EC1M 6BQ, United Kingdom
| | - Catalina Lodillinsky
- Instituto de Oncologia Ángel H. Roffo, Research Area, Buenos Aires, C1417DTB, Argentina
| | - Carine Rossé
- Institut Curie, Paris, F-75248 France; .,PSL Research University, Paris, F-75005 France.,CNRS, UMR 144, Paris, F-75248 France
| | - Philippe Chavrier
- Institut Curie, Paris, F-75248 France; .,PSL Research University, Paris, F-75005 France.,CNRS, UMR 144, Paris, F-75248 France
| |
Collapse
|
22
|
Mrkonjic S, Destaing O, Albiges-Rizo C. Mechanotransduction pulls the strings of matrix degradation at invadosome. Matrix Biol 2016; 57-58:190-203. [PMID: 27392543 DOI: 10.1016/j.matbio.2016.06.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 06/16/2016] [Accepted: 06/28/2016] [Indexed: 02/07/2023]
Abstract
Degradation of the extracellular matrix is a critical step of tumor cell invasion. Both protease-dependent and -independent mechanisms have been described as alternate processes in cancer cell motility. Interestingly, some effectors of protease-dependent degradation are focalized at invadosomes and are directly coupled with contractile and adhesive machineries composed of multiple mechanosensitive proteins. This review presents recent findings in protease-dependent mechanisms elucidating the ways the force affects extracellular matrix degradation by targeting protease expression and activity at invadosome. The aim is to highlight mechanosensing and mechanotransduction processes to direct the degradative activity at invadosomes, with the focus on membrane tension, proteases and mechanosensitive ion channels.
Collapse
Affiliation(s)
- Sanela Mrkonjic
- INSERM U1209, Grenoble F-38042, France; Université Grenoble Alpes, Institut Albert Bonniot, F-38042 Grenoble, France; CNRS UMR 5309, F-38042 Grenoble, France
| | - Olivier Destaing
- INSERM U1209, Grenoble F-38042, France; Université Grenoble Alpes, Institut Albert Bonniot, F-38042 Grenoble, France; CNRS UMR 5309, F-38042 Grenoble, France.
| | - Corinne Albiges-Rizo
- INSERM U1209, Grenoble F-38042, France; Université Grenoble Alpes, Institut Albert Bonniot, F-38042 Grenoble, France; CNRS UMR 5309, F-38042 Grenoble, France.
| |
Collapse
|
23
|
Hastie EL, Sherwood DR. A new front in cell invasion: The invadopodial membrane. Eur J Cell Biol 2016; 95:441-448. [PMID: 27402208 DOI: 10.1016/j.ejcb.2016.06.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Revised: 06/18/2016] [Accepted: 06/20/2016] [Indexed: 01/16/2023] Open
Abstract
Invadopodia are F-actin-rich membrane protrusions that breach basement membrane barriers during cell invasion. Since their discovery more than 30 years ago, invadopodia have been extensively investigated in cancer cells in vitro, where great advances in understanding their composition, formation, cytoskeletal regulation, and control of the matrix metalloproteinase MT1-MMP trafficking have been made. In contrast, few studies examining invadopodia have been conducted in vivo, leaving their physiological regulation unclear. Recent live-cell imaging and gene perturbation studies in C. elegans have revealed that invadopodia are formed with a unique invadopodial membrane, defined by its specialized lipid and associated protein composition, which is rapidly recycled through the endolysosome. Here, we provide evidence that the invadopodial membrane is conserved and discuss its possible functions in traversing basement membrane barriers. Discovery and examination of the invadopodial membrane has important implications in understanding the regulation, assembly, and function of invadopodia in both normal and disease settings.
Collapse
Affiliation(s)
- Eric L Hastie
- Department of Biology, Duke University, 124 Science Drive, Box 90388, Durham, NC 27708, USA
| | - David R Sherwood
- Department of Biology, Duke University, 124 Science Drive, Box 90388, Durham, NC 27708, USA.
| |
Collapse
|
24
|
Schwarzländer M, Dick TP, Meyer AJ, Morgan B. Dissecting Redox Biology Using Fluorescent Protein Sensors. Antioxid Redox Signal 2016; 24:680-712. [PMID: 25867539 DOI: 10.1089/ars.2015.6266] [Citation(s) in RCA: 209] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
SIGNIFICANCE Fluorescent protein sensors have revitalized the field of redox biology by revolutionizing the study of redox processes in living cells and organisms. RECENT ADVANCES Within one decade, a set of fundamental new insights has been gained, driven by the rapid technical development of in vivo redox sensing. Redox-sensitive yellow and green fluorescent protein variants (rxYFP and roGFPs) have been the central players. CRITICAL ISSUES Although widely used as an established standard tool, important questions remain surrounding their meaningful use in vivo. We review the growing range of thiol redox sensor variants and their application in different cells, tissues, and organisms. We highlight five key findings where in vivo sensing has been instrumental in changing our understanding of redox biology, critically assess the interpretation of in vivo redox data, and discuss technical and biological limitations of current redox sensors and sensing approaches. FUTURE DIRECTIONS We explore how novel sensor variants may further add to the current momentum toward a novel mechanistic and integrated understanding of redox biology in vivo. Antioxid. Redox Signal. 24, 680-712.
Collapse
Affiliation(s)
- Markus Schwarzländer
- 1 Plant Energy Biology Lab, Department Chemical Signalling, Institute of Crop Science and Resource Conservation (INRES), University of Bonn , Bonn, Germany
| | - Tobias P Dick
- 2 Division of Redox Regulation, German Cancer Research Center (DKFZ) , DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Andreas J Meyer
- 3 Department Chemical Signalling, Institute of Crop Science and Resource Conservation (INRES), University of Bonn , Bonn, Germany
| | - Bruce Morgan
- 2 Division of Redox Regulation, German Cancer Research Center (DKFZ) , DKFZ-ZMBH Alliance, Heidelberg, Germany .,4 Cellular Biochemistry, Department of Biology, University of Kaiserslautern , Kaiserslautern, Germany
| |
Collapse
|
25
|
Di Martino J, Henriet E, Ezzoukhry Z, Goetz JG, Moreau V, Saltel F. The microenvironment controls invadosome plasticity. J Cell Sci 2016; 129:1759-68. [PMID: 27029343 DOI: 10.1242/jcs.182329] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Invadosomes are actin-based structures involved in extracellular matrix degradation. Invadosomes is a term that includes podosomes and invadopodia, which decorate normal and tumour cells, respectively. They are mainly organised into dots or rosettes, and podosomes and invadopodia are often compared and contrasted. Various internal or external stimuli have been shown to induce their formation and/or activity. In this Commentary, we address the impact of the microenvironment and the role of matrix receptors on the formation, and dynamic and degradative activities of invadosomes. In particular, we highlight recent findings regarding the role of type I collagen fibrils in inducing the formation of a new linear organisation of invadosomes. We will also discuss invadosome plasticity more generally and emphasise its physio-pathological relevance.
Collapse
Affiliation(s)
- Julie Di Martino
- Institut National de la Santé et de la Recherche Médicale, U1053, Bordeaux F-33076, France Université de Bordeaux, Bordeaux F-33076, France
| | - Elodie Henriet
- Institut National de la Santé et de la Recherche Médicale, U1053, Bordeaux F-33076, France Université de Bordeaux, Bordeaux F-33076, France
| | - Zakaria Ezzoukhry
- Institut National de la Santé et de la Recherche Médicale, U1053, Bordeaux F-33076, France Université de Bordeaux, Bordeaux F-33076, France
| | - Jacky G Goetz
- MN3T, Inserm U1109, Strasbourg 67200, France Université de Strasbourg, Strasbourg 67000, France LabEx Medalis, Université de Strasbourg, Strasbourg 67000, France Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg 67000, France
| | - Violaine Moreau
- Institut National de la Santé et de la Recherche Médicale, U1053, Bordeaux F-33076, France Université de Bordeaux, Bordeaux F-33076, France
| | - Frederic Saltel
- Institut National de la Santé et de la Recherche Médicale, U1053, Bordeaux F-33076, France Université de Bordeaux, Bordeaux F-33076, France
| |
Collapse
|
26
|
Kague E, Roy P, Asselin G, Hu G, Simonet J, Stanley A, Albertson C, Fisher S. Osterix/Sp7 limits cranial bone initiation sites and is required for formation of sutures. Dev Biol 2016; 413:160-72. [PMID: 26992365 DOI: 10.1016/j.ydbio.2016.03.011] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 03/11/2016] [Indexed: 12/16/2022]
Abstract
During growth, individual skull bones overlap at sutures, where osteoblast differentiation and bone deposition occur. Mutations causing skull malformations have revealed some required genes, but many aspects of suture regulation remain poorly understood. We describe a zebrafish mutation in osterix/sp7, which causes a generalized delay in osteoblast maturation. While most of the skeleton is patterned normally, mutants have specific defects in the anterior skull and upper jaw, and the top of the skull comprises a random mosaic of bones derived from individual initiation sites. Osteoblasts at the edges of the bones are highly proliferative and fail to differentiate, consistent with global changes in gene expression. We propose that signals from the bone itself are required for orderly recruitment of precursor cells and growth along the edges. The delay in bone maturation caused by loss of Sp7 leads to unregulated bone formation, revealing a new mechanism for patterning the skull and sutures.
Collapse
Affiliation(s)
- Erika Kague
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| | - Paula Roy
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Garrett Asselin
- Department of Biology, University of Massachusetts, Amherst, MA, USA
| | - Gui Hu
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Jacqueline Simonet
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Alexandra Stanley
- Cell and Molecular Biology Graduate Program, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Craig Albertson
- Department of Biology, University of Massachusetts, Amherst, MA, USA
| | - Shannon Fisher
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
27
|
Abrams J, Einhorn Z, Seiler C, Zong AB, Sweeney HL, Pack M. Graded effects of unregulated smooth muscle myosin on intestinal architecture, intestinal motility and vascular function in zebrafish. Dis Model Mech 2016; 9:529-40. [PMID: 26893369 PMCID: PMC4892660 DOI: 10.1242/dmm.023309] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 02/10/2016] [Indexed: 12/29/2022] Open
Abstract
Smooth muscle contraction is controlled by the regulated activity of the myosin heavy chain ATPase (Myh11). Myh11 mutations have diverse effects in the cardiovascular, digestive and genitourinary systems in humans and animal models. We previously reported a recessive missense mutation, meltdown (mlt), which converts a highly conserved tryptophan to arginine (W512R) in the rigid relay loop of zebrafish Myh11. The mlt mutation disrupts myosin regulation and non-autonomously induces invasive expansion of the intestinal epithelium. Here, we report two newly identified missense mutations in the switch-1 (S237Y) and coil-coiled (L1287M) domains of Myh11 that fail to complement mlt Cell invasion was not detected in either homozygous mutant but could be induced by oxidative stress and activation of oncogenic signaling pathways. The smooth muscle defect imparted by the mlt and S237Y mutations also delayed intestinal transit, and altered vascular function, as measured by blood flow in the dorsal aorta. The cell-invasion phenotype induced by the three myh11 mutants correlated with the degree of myosin deregulation. These findings suggest that the vertebrate intestinal epithelium is tuned to the physical state of the surrounding stroma, which, in turn, governs its response to physiologic and pathologic stimuli. Genetic variants that alter the regulation of smooth muscle myosin might be risk factors for diseases affecting the intestine, vasculature, and other tissues that contain smooth muscle or contractile cells that express smooth muscle proteins, particularly in the setting of redox stress.
Collapse
Affiliation(s)
- Joshua Abrams
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zev Einhorn
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christoph Seiler
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alan B Zong
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - H Lee Sweeney
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael Pack
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
28
|
Extracellular matrix stiffness dictates Wnt expression through integrin pathway. Sci Rep 2016; 6:20395. [PMID: 26854061 PMCID: PMC4745056 DOI: 10.1038/srep20395] [Citation(s) in RCA: 150] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 01/04/2016] [Indexed: 12/23/2022] Open
Abstract
It is well established that extracellular matrix (ECM) stiffness plays a significant role in regulating the phenotypes and behaviors of many cell types. However, the mechanism underlying the sensing of mechanical cues and subsequent elasticity-triggered pathways remains largely unknown. We observed that stiff ECM significantly enhanced the expression level of several members of the Wnt/β-catenin pathway in both bone marrow mesenchymal stem cells and primary chondrocytes. The activation of β-catenin by stiff ECM is not dependent on Wnt signals but is elevated by the activation of integrin/ focal adhesion kinase (FAK) pathway. The accumulated β-catenin then bound to the wnt1 promoter region to up-regulate the gene transcription, thus constituting a positive feedback of the Wnt/β-catenin pathway. With the amplifying effect of positive feedback, this integrin-activated β-catenin/Wnt pathway plays significant roles in mediating the enhancement of Wnt signal on stiff ECM and contributes to the regulation of mesenchymal stem cell differentiation and primary chondrocyte phenotype maintenance. The present integrin-regulated Wnt1 expression and signaling contributes to the understanding of the molecular mechanisms underlying the regulation of cell behaviors by ECM elasticity.
Collapse
|
29
|
Jiang L, Sun Z, Chen X, Li J, Xu Y, Zu Y, Hu J, Han D, Yang C. Cells Sensing Mechanical Cues: Stiffness Influences the Lifetime of Cell-Extracellular Matrix Interactions by Affecting the Loading Rate. ACS NANO 2016; 10:207-17. [PMID: 26701367 DOI: 10.1021/acsnano.5b03157] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
The question of how cells sense substrate mechanical cues has gained increasing attention among biologists. By introducing contour-based data analysis to single-cell force spectroscopy, we identified a loading-rate threshold for the integrin α2β1-DGEA bond beyond which a dramatic increase in bond lifetime was observed. On the basis of mechanical cues (elasticity or topography), the effective spring constant of substrates k is mapped to the loading rate r under actomyosin pulling speed v, which, in turn, affects the lifetime of the integrin-ligand bond. Additionally, downregulating v with a low-dose blebbistatin treatment promotes the neuronal lineage specification of mesenchymal stem cells on osteogenic stiff substrates. Thus, sensing of the loading rate is central to how cells sense mechanical cues that affect cell-extracellular matrix interactions and stem cell differentiation.
Collapse
Affiliation(s)
- Li Jiang
- Institute of Biomechanics and Medical Engineering, School of Aerospace, Tsinghua University , Beijing 100084, People's Republic of China
| | - Zhenglong Sun
- Institute of Biomechanics and Medical Engineering, School of Aerospace, Tsinghua University , Beijing 100084, People's Republic of China
- Suzhou Institute of Biomedical Engineering and Technology, China Academy of Science , Suzhou 215163, People's Republic of China
| | - Xiaofei Chen
- Institute of Biomechanics and Medical Engineering, School of Aerospace, Tsinghua University , Beijing 100084, People's Republic of China
- Beijing Institute of Aerospace Systems Engineering , Beijing 100076, People's Republic of China
| | - Jing Li
- Institute of Biomechanics and Medical Engineering, School of Aerospace, Tsinghua University , Beijing 100084, People's Republic of China
| | - Yue Xu
- Institute of Biomechanics and Medical Engineering, School of Aerospace, Tsinghua University , Beijing 100084, People's Republic of China
| | - Yan Zu
- Institute of Biomechanics and Medical Engineering, School of Aerospace, Tsinghua University , Beijing 100084, People's Republic of China
| | - Jiliang Hu
- Institute of Biomechanics and Medical Engineering, School of Aerospace, Tsinghua University , Beijing 100084, People's Republic of China
| | - Dong Han
- National Center for Nanoscience and Technology , Beijing 100190, People's Republic of China
| | - Chun Yang
- Institute of Biomechanics and Medical Engineering, School of Aerospace, Tsinghua University , Beijing 100084, People's Republic of China
| |
Collapse
|
30
|
|
31
|
Lohmer LL, Clay MR, Naegeli KM, Chi Q, Ziel JW, Hagedorn EJ, Park JE, Jayadev R, Sherwood DR. A Sensitized Screen for Genes Promoting Invadopodia Function In Vivo: CDC-42 and Rab GDI-1 Direct Distinct Aspects of Invadopodia Formation. PLoS Genet 2016; 12:e1005786. [PMID: 26765257 PMCID: PMC4713207 DOI: 10.1371/journal.pgen.1005786] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 12/12/2015] [Indexed: 12/12/2022] Open
Abstract
Invadopodia are specialized membrane protrusions composed of F-actin, actin regulators, signaling proteins, and a dynamically trafficked invadopodial membrane that drive cell invasion through basement membrane (BM) barriers in development and cancer. Due to the challenges of studying invasion in vivo, mechanisms controlling invadopodia formation in their native environments remain poorly understood. We performed a sensitized genome-wide RNAi screen and identified 13 potential regulators of invadopodia during anchor cell (AC) invasion into the vulval epithelium in C. elegans. Confirming the specificity of this screen, we identified the Rho GTPase cdc-42, which mediates invadopodia formation in many cancer cell lines. Using live-cell imaging, we show that CDC-42 localizes to the AC-BM interface and is activated by an unidentified vulval signal(s) that induces invasion. CDC-42 is required for the invasive membrane localization of WSP-1 (N-WASP), a CDC-42 effector that promotes polymerization of F-actin. Loss of CDC-42 or WSP-1 resulted in fewer invadopodia and delayed BM breaching. We also characterized a novel invadopodia regulator, gdi-1 (Rab GDP dissociation inhibitor), which mediates membrane trafficking. We show that GDI-1 functions in the AC to promote invadopodia formation. In the absence of GDI-1, the specialized invadopodial membrane was no longer trafficked normally to the invasive membrane, and instead was distributed to plasma membrane throughout the cell. Surprisingly, the pro-invasive signal(s) from the vulval cells also controls GDI-1 activity and invadopodial membrane trafficking. These studies represent the first in vivo screen for genes regulating invadopodia and demonstrate that invadopodia formation requires the integration of distinct cellular processes that are coordinated by an extracellular cue. During animal development specialized cells acquire the ability move and invade into other tissues to form complex organs and structures. Understanding this cellular behavior is important medically, as cancer cells can hijack the developmental program of invasion to metastasize throughout the body. One of the most formidable barriers invasive cells face is basement membrane–-a thin, dense, sheet-like assembly of proteins and carbohydrates that surrounds most tissues. Cells deploy small, protrusive, membrane associated structures called invadopodia (invasive feet) to breach basement membranes. How invadopodia are formed and controlled during invasion has been challenging to understand, as it is difficult to examine these dynamic structures in live animals. Using the nematode worm Caenorhabditis elegans, we have conducted the first large-scale screen to isolate genes that control invadopodia in live animals. Our screen isolated 13 genes and we confirmed two are key invadopodia regulators: the Rho GTPase CDC-42 that promotes F-actin polymerization at invadopodia to generate the force to breach basement membranes, and the Rab GDI-1 that promotes membrane addition at invadopodia that may allow invadopodia to extend through basement membranes. This work provides new insights into invadopodia construction and identifies potential novel targets for anti-metastasis therapies.
Collapse
Affiliation(s)
- Lauren L. Lohmer
- Department of Biology, Duke University, Durham, North Carolina, United States of America
| | - Matthew R. Clay
- Department of Biology, Duke University, Durham, North Carolina, United States of America
| | - Kaleb M. Naegeli
- Department of Biology, Duke University, Durham, North Carolina, United States of America
| | - Qiuyi Chi
- Department of Biology, Duke University, Durham, North Carolina, United States of America
| | - Joshua W. Ziel
- Department of Biology, Duke University, Durham, North Carolina, United States of America
| | - Elliott J. Hagedorn
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jieun E. Park
- Department of Biology, Duke University, Durham, North Carolina, United States of America
| | - Ranjay Jayadev
- Department of Biology, Duke University, Durham, North Carolina, United States of America
| | - David R. Sherwood
- Department of Biology, Duke University, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
32
|
Lobert VH, Mouradov D, Heath JK. Focusing the Spotlight on the Zebrafish Intestine to Illuminate Mechanisms of Colorectal Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 916:411-37. [PMID: 27165364 DOI: 10.1007/978-3-319-30654-4_18] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Colorectal cancer, encompassing colon and rectal cancer, arises from the epithelial lining of the large bowel. It is most prevalent in Westernised societies and is increasing in frequency as the world becomes more industrialised. Unfortunately, metastatic colorectal cancer is not cured by chemotherapy and the annual number of deaths caused by colorectal cancer, currently 700,000, is expected to rise. Our understanding of the contribution that genetic mutations make to colorectal cancer, although incomplete, is reasonably well advanced. However, it has only recently become widely appreciated that in addition to the ongoing accumulation of genetic mutations, chronic inflammation also plays a critical role in the initiation and progression of this disease. While a robust and tractable genetic model of colorectal cancer in zebrafish, suitable for pre-clinical studies, is not yet available, the identification of genes required for the rapid proliferation of zebrafish intestinal epithelial cells during development has highlighted a number of essential genes that could be targeted to disable colorectal cancer cells. Moreover, appreciation of the utility of zebrafish to study intestinal inflammation is on the rise. In particular, zebrafish provide unique opportunities to investigate the impact of genetic and environmental factors on the integrity of intestinal epithelial barrier function. With currently available tools, the interplay between epigenetic regulators, intestinal injury, microbiota composition and innate immune cell mobilisation can be analysed in exquisite detail. This provides excellent opportunities to define critical events that could potentially be targeted therapeutically. Further into the future, the use of zebrafish larvae as hosts for xenografts of human colorectal cancer tissue, while still in its infancy, holds great promise that zebrafish could one day provide a practical, preclinical personalized medicine platform for the rapid assessment of the metastatic potential and drug-sensitivity of patient-derived cancers.
Collapse
Affiliation(s)
- Viola H Lobert
- Development and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia.,Department of Biochemistry, Institute for Cancer Research, Oslo University Hospital, Montebello, N-0379, Oslo, Norway
| | - Dmitri Mouradov
- Systems Biology and Personalised Medicine Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
| | - Joan K Heath
- Development and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia. .,Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia.
| |
Collapse
|
33
|
Li S, Xue H, Wu J, Rao MS, Kim DH, Deng W, Liu Y. Human Induced Pluripotent Stem Cell NEUROG2 Dual Knockin Reporter Lines Generated by the CRISPR/Cas9 System. Stem Cells Dev 2015; 24:2925-42. [PMID: 26414932 DOI: 10.1089/scd.2015.0131] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Human induced pluripotent stem cell (hiPSC) technologies are powerful tools for modeling development and disease, drug screening, and regenerative medicine. Faithful gene targeting in hiPSCs greatly facilitates these applications. We have developed a fast and precise clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR associated protein 9 (Cas9) technology-based method and obtained fluorescent protein and antibiotic resistance dual knockin reporters in hiPSC lines for neurogenin2 (NEUROG2), an important proneural transcription factor. Gene targeting efficiency was greatly improved in CRISPR/Cas9-mediated homology directed recombination (∼ 33% correctly targeted clones) compared to conventional targeting protocol (∼ 3%) at the same locus. No off-target events were detected. In addition, taking the advantage of the versatile applications of the CRISPR/Cas9 system, we designed transactivation components to transiently induce NEUROG2 expression, which helps identify transcription factor binding sites and trans-regulation regions of human NEUROG2. The strategy of using CRISPR/Cas9 genome editing coupled with fluorescence-activated cell sorting of neural progenitor cells in a knockin lineage hiPSC reporter platform might be broadly applicable in other stem cell derivatives and subpopulations.
Collapse
Affiliation(s)
- Shenglan Li
- 1 Department of Neurosurgery, Medical School, the Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston , Houston, Texas.,2 Center for Stem Cell and Regenerative Medicine, the Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston , Houston, Texas
| | - Haipeng Xue
- 1 Department of Neurosurgery, Medical School, the Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston , Houston, Texas.,2 Center for Stem Cell and Regenerative Medicine, the Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston , Houston, Texas
| | - Jianbo Wu
- 1 Department of Neurosurgery, Medical School, the Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston , Houston, Texas.,2 Center for Stem Cell and Regenerative Medicine, the Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston , Houston, Texas
| | - Mahendra S Rao
- 3 The New York Stem Cell Foundation , New York, New York
| | - Dong H Kim
- 1 Department of Neurosurgery, Medical School, the Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston , Houston, Texas.,2 Center for Stem Cell and Regenerative Medicine, the Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston , Houston, Texas
| | - Wenbin Deng
- 4 Department of Biochemistry and Molecular Medicine, School of Medicine, University of California , Davis, California.,5 Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children , Sacramento, California
| | - Ying Liu
- 1 Department of Neurosurgery, Medical School, the Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston , Houston, Texas.,2 Center for Stem Cell and Regenerative Medicine, the Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston , Houston, Texas.,6 The Senator Lloyd and B.A. Bentsen Center for Stroke Research, the Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston , Houston, Texas
| |
Collapse
|
34
|
Glentis A, Gurchenkov V, Matic Vignjevic D. Assembly, heterogeneity, and breaching of the basement membranes. Cell Adh Migr 2015; 8:236-45. [PMID: 24727304 DOI: 10.4161/cam.28733] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Basement membranes are thin sheets of self-assembled extracellular matrices that are essential for embryonic development and for the homeostasis of adult tissues. They play a role in structuring, protecting, polarizing, and compartmentalizing cells, as well as in supplying them with growth factors. All basement membranes are built from laminin and collagen IV networks stabilized by nidogen/perlecan bridges. The precise composition of basement membranes, however, varies between different tissues. Even though basement membranes represent physical barriers that delimit different tissues, they are breached in many physiological or pathological processes, including development, the immune response, and tumor invasion. Here, we provide a brief overview of the molecular composition of basement membranes and the process of their assembly. We will then illustrate the heterogeneity of basement membranes using two examples, the epithelial basement membrane in the gut and the vascular basement membrane. Finally, we examine the different strategies cells use to breach the basement membrane.
Collapse
|
35
|
Lohmer LL, Kelley LC, Hagedorn EJ, Sherwood DR. Invadopodia and basement membrane invasion in vivo. Cell Adh Migr 2015; 8:246-55. [PMID: 24717190 DOI: 10.4161/cam.28406] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Over 20 years ago, protrusive, F-actin-based membrane structures, termed invadopodia, were identified in highly metastatic cancer cell lines. Invadopodia penetrate artificial or explanted extracellular matrices in 2D culture conditions and have been hypothesized to facilitate the migration of cancer cells through basement membrane, a thin, dense, barrier-like matrix surrounding most tissues. Despite intensive study, the identification of invadopodia in vivo has remained elusive and until now their possible roles during invasion or even existence have remained unclear. Studies in remarkably different cellular contexts-mouse tumor models, zebrafish intestinal epithelia, and C. elegans organogenesis-have recently identified invadopodia structures associated with basement membrane invasion. These studies are providing the first in vivo insight into the regulation, function, and role of these fascinating subcellular devices with critical importance to both development and human disease.
Collapse
|
36
|
Di Martino J, Paysan L, Gest C, Lagrée V, Juin A, Saltel F, Moreau V. Cdc42 and Tks5: a minimal and universal molecular signature for functional invadosomes. Cell Adh Migr 2015; 8:280-92. [PMID: 24840388 DOI: 10.4161/cam.28833] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Invadosomes are actin-based structures involved in extracellular-matrix degradation. Invadosomes, either known as podosomes or invadopodia, are found in an increasing number of cell types. Moreover, their overall organization and molecular composition may vary from one cell type to the other. Some are constitutive such as podosomes in hematopoietic cells whereas others are inducible. However, they share the same feature, their ability to interact and to degrade the extracellular matrix. Based on the literature and our own experiments, the aim of this study was to establish a minimal molecular definition of active invadosomes. We first highlighted that Cdc42 is the key RhoGTPase involved in invadosome formation in all described models. Using different cellular models, such as NIH-3T3, HeLa, and endothelial cells, we demonstrated that overexpression of an active form of Cdc42 is sufficient to form invadosome actin cores. Therefore, active Cdc42 must be considered not only as an inducer of filopodia, but also as an inducer of invadosomes. Depending on the expression level of Tks5, these Cdc42-dependent actin cores were endowed or not with a proteolytic activity. In fact, Tks5 overexpression rescued this activity in Tks5 low expressing cells. We thus described the adaptor protein Tks5 as a major actor of the invadosome degradation function. Surprisingly, we found that Src kinases are not always required for invadosome formation and function. These data suggest that even if Src family members are the principal kinases involved in the majority of invadosomes, it cannot be considered as a common element for all invadosome structures. We thus define a minimal and universal molecular signature of invadosome that includes Cdc42 activity and Tks5 presence in order to drive the actin machinery and the proteolytic activity of these invasive structures.
Collapse
Affiliation(s)
- Julie Di Martino
- INSERM; Physiopathologie du cancer du foie; U1053; Bordeaux, France; Univ. Bordeaux; Physiopathologie du cancer du foie; U1053; Bordeaux, France
| | - Lisa Paysan
- INSERM; Physiopathologie du cancer du foie; U1053; Bordeaux, France; Univ. Bordeaux; Physiopathologie du cancer du foie; U1053; Bordeaux, France
| | - Caroline Gest
- INSERM; Physiopathologie du cancer du foie; U1053; Bordeaux, France; Univ. Bordeaux; Physiopathologie du cancer du foie; U1053; Bordeaux, France
| | - Valérie Lagrée
- INSERM; Physiopathologie du cancer du foie; U1053; Bordeaux, France; Univ. Bordeaux; Physiopathologie du cancer du foie; U1053; Bordeaux, France
| | - Amélie Juin
- INSERM; Physiopathologie du cancer du foie; U1053; Bordeaux, France; Univ. Bordeaux; Physiopathologie du cancer du foie; U1053; Bordeaux, France
| | - Frédéric Saltel
- INSERM; Physiopathologie du cancer du foie; U1053; Bordeaux, France; Univ. Bordeaux; Physiopathologie du cancer du foie; U1053; Bordeaux, France
| | - Violaine Moreau
- INSERM; Physiopathologie du cancer du foie; U1053; Bordeaux, France; Univ. Bordeaux; Physiopathologie du cancer du foie; U1053; Bordeaux, France
| |
Collapse
|
37
|
Seiler C, Gebhart N, Zhang Y, Shinton SA, Li YS, Ross NL, Liu X, Li Q, Bilbee AN, Varshney GK, LaFave MC, Burgess SM, Balciuniene J, Balciunas D, Hardy RR, Kappes DJ, Wiest DL, Rhodes J. Mutagenesis Screen Identifies agtpbp1 and eps15L1 as Essential for T lymphocyte Development in Zebrafish. PLoS One 2015; 10:e0131908. [PMID: 26161877 PMCID: PMC4498767 DOI: 10.1371/journal.pone.0131908] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 06/08/2015] [Indexed: 11/19/2022] Open
Abstract
Genetic screens are a powerful tool to discover genes that are important in immune cell development and function. The evolutionarily conserved development of lymphoid cells paired with the genetic tractability of zebrafish make this a powerful model system for this purpose. We used a Tol2-based gene-breaking transposon to induce mutations in the zebrafish (Danio rerio, AB strain) genome, which served the dual purpose of fluorescently tagging cells and tissues that express the disrupted gene and provided a means of identifying the disrupted gene. We identified 12 lines in which hematopoietic tissues expressed green fluorescent protein (GFP) during embryonic development, as detected by microscopy. Subsequent analysis of young adult fish, using a novel approach in which single cell suspensions of whole fish were analyzed by flow cytometry, revealed that 8 of these lines also exhibited GFP expression in young adult cells. An additional 15 lines that did not have embryonic GFP+ hematopoietic tissue by microscopy, nevertheless exhibited GFP+ cells in young adults. RT-PCR analysis of purified GFP+ populations for expression of T and B cell-specific markers identified 18 lines in which T and/or B cells were fluorescently tagged at 6 weeks of age. As transposon insertion is expected to cause gene disruption, these lines can be used to assess the requirement for the disrupted genes in immune cell development. Focusing on the lines with embryonic GFP+ hematopoietic tissue, we identified three lines in which homozygous mutants exhibited impaired T cell development at 6 days of age. In two of the lines we identified the disrupted genes, agtpbp1 and eps15L1. Morpholino-mediated knockdown of these genes mimicked the T cell defects in the corresponding mutant embryos, demonstrating the previously unrecognized, essential roles of agtpbp1 and eps15L1 in T cell development.
Collapse
Affiliation(s)
- Christoph Seiler
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania, United States of America
| | - Nichole Gebhart
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania, United States of America
| | - Yong Zhang
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania, United States of America
| | - Susan A. Shinton
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania, United States of America
| | - Yue-sheng Li
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania, United States of America
| | - Nicola L. Ross
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania, United States of America
| | - Xingjun Liu
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania, United States of America
| | - Qin Li
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania, United States of America
| | - Alison N. Bilbee
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania, United States of America
| | - Gaurav K. Varshney
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Matthew C. LaFave
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Shawn M. Burgess
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jorune Balciuniene
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania, United States of America
| | - Darius Balciunas
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania, United States of America
| | - Richard R. Hardy
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania, United States of America
| | - Dietmar J. Kappes
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania, United States of America
| | - David L. Wiest
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania, United States of America
| | - Jennifer Rhodes
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
38
|
Jordi J, Guggiana-Nilo D, Soucy E, Song EY, Lei Wee C, Engert F. A high-throughput assay for quantifying appetite and digestive dynamics. Am J Physiol Regul Integr Comp Physiol 2015; 309:R345-57. [PMID: 26108871 DOI: 10.1152/ajpregu.00225.2015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 06/16/2015] [Indexed: 01/23/2023]
Abstract
Food intake and digestion are vital functions, and their dysregulation is fundamental for many human diseases. Current methods do not support their dynamic quantification on large scales in unrestrained vertebrates. Here, we combine an infrared macroscope with fluorescently labeled food to quantify feeding behavior and intestinal nutrient metabolism with high temporal resolution, sensitivity, and throughput in naturally behaving zebrafish larvae. Using this method and rate-based modeling, we demonstrate that zebrafish larvae match nutrient intake to their bodily demand and that larvae adjust their digestion rate, according to the ingested meal size. Such adaptive feedback mechanisms make this model system amenable to identify potential chemical modulators. As proof of concept, we demonstrate that nicotine, l-lysine, ghrelin, and insulin have analogous impact on food intake as in mammals. Consequently, the method presented here will promote large-scale translational research of food intake and digestive function in a naturally behaving vertebrate.
Collapse
Affiliation(s)
- Josua Jordi
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts; and
| | - Drago Guggiana-Nilo
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts; and Committee for Higher Degrees in Biophysics, Harvard University, Cambridge, Massachusetts
| | - Edward Soucy
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts; and
| | - Erin Yue Song
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts; and
| | - Caroline Lei Wee
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts; and
| | - Florian Engert
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts; and
| |
Collapse
|
39
|
Jiang F, Chen J, Ma X, Huang C, Zhu S, Wang F, Li L, Luo L, Ruan H, Huang H. Analysis of mutants from a genetic screening reveals the control of intestine and liver development by many common genes in zebrafish. Biochem Biophys Res Commun 2015; 460:838-44. [DOI: 10.1016/j.bbrc.2015.03.119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 03/21/2015] [Indexed: 01/10/2023]
|
40
|
Jacob A, Prekeris R. The regulation of MMP targeting to invadopodia during cancer metastasis. Front Cell Dev Biol 2015; 3:4. [PMID: 25699257 PMCID: PMC4313772 DOI: 10.3389/fcell.2015.00004] [Citation(s) in RCA: 217] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 01/09/2015] [Indexed: 01/07/2023] Open
Abstract
The dissemination of cancer cells from the primary tumor to a distant site, known as metastasis, is the main cause of mortality in cancer patients. Metastasis is a very complex cellular process that involves many steps, including the breaching of the basement membrane (BM) to allow the movement of cells through tissues. The BM breach occurs via highly regulated and localized remodeling of the extracellular matrix (ECM), which is mediated by formation of structures, known as invadopodia, and targeted secretion of matrix metalloproteinases (MMPs). Recently, invadopodia have emerged as key cellular structures that regulate the metastasis of many cancers. Furthermore, targeting of various cytoskeletal modulators and MMPs has been shown to play a major role in regulating invadopodia function. Here, we highlight recent findings regarding the regulation of protein targeting during invadopodia formation and function.
Collapse
Affiliation(s)
- Abitha Jacob
- Department of Cell and Developmental Biology, School of Medicine, Anschutz Medical Campus, University of Colorado Denver Aurora, CO, USA
| | - Rytis Prekeris
- Department of Cell and Developmental Biology, School of Medicine, Anschutz Medical Campus, University of Colorado Denver Aurora, CO, USA
| |
Collapse
|
41
|
Génot E, Gligorijevic B. Invadosomes in their natural habitat. Eur J Cell Biol 2014; 93:367-79. [PMID: 25457677 DOI: 10.1016/j.ejcb.2014.10.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 08/15/2014] [Accepted: 10/14/2014] [Indexed: 01/01/2023] Open
Abstract
Podosomes and invadopodia (collectively known as invadosomes) are small, F-actin-rich protrusions that are located at points of cell-ECM contacts and endow cells with invasive capabilities. So far, they have been identified in human or murine immune (myelomonocytic), vascular and cancer cells. The overarching reason for studying invadosomes is their connection to human disease. For example, macrophages and osteoclasts lacking Wiskott-Aldrich syndrome protein (WASp) are not able to form podosomes, and this leads to altered macrophage chemotaxis and defective bone resorption by osteoclasts. In contrast, the ability of cancer cells to form invadopodia is associated with high invasive and metastatic potentials. While invadosome composition, dynamics and signaling cascades leading to their assembly can be followed easily in in vitro assays, studying their contribution to pathophysiological processes in situ remains challenging. A number of recent papers have started to address this issue and describe invadosomes in situ in mouse models of cancer, cardiovascular disease and angiogenesis. In addition, in vivo invadosome homologs have been reported in developmental model systems such as C. elegans, zebrafish and sea squirt. Comparative analyses among different invasion mechanisms as they happen in their natural habitats, i.e., in situ, may provide an outline of the invadosome evolutionary history, and guide our understanding of the roles of the invasion process in pathophysiology versus development.
Collapse
Affiliation(s)
- Elisabeth Génot
- Université de Bordeaux, F-33000 Bordeaux, France; INSERM U1045, F-33000 Bordeaux, France; European Institute of Chemistry and Biology, 2 rue Robert Escarpit, 33 600 Pessac, France.
| | - Bojana Gligorijevic
- Department of Systems & Computational Biology and Albert Einstein College of Medicine, Price Center, 1301 Morris Park Avenue, 10461 Bronx, NY, USA.
| |
Collapse
|
42
|
Kelley LC, Lohmer LL, Hagedorn EJ, Sherwood DR. Traversing the basement membrane in vivo: a diversity of strategies. ACTA ACUST UNITED AC 2014; 204:291-302. [PMID: 24493586 PMCID: PMC3912525 DOI: 10.1083/jcb.201311112] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The basement membrane is a dense, highly cross-linked, sheet-like extracellular matrix that underlies all epithelia and endothelia in multicellular animals. During development, leukocyte trafficking, and metastatic disease, cells cross the basement membrane to disperse and enter new tissues. Based largely on in vitro studies, cells have been thought to use proteases to dissolve and traverse this formidable obstacle. Surprisingly, recent in vivo studies have uncovered a remarkably diverse range of cellular- and tissue-level strategies beyond proteolysis that cells use to navigate through the basement membrane. These fascinating and unexpected mechanisms have increased our understanding of how cells cross this matrix barrier in physiological and disease settings.
Collapse
Affiliation(s)
- Laura C Kelley
- Department of Biology, Duke University, Durham, NC 27708
| | | | | | | |
Collapse
|
43
|
Chun HS, Shin SH, Ahn S, Shin DS, Choi SS, Ahn JH, Bae MA. KR-62980 Suppresses Lipid Metabolism Through Inhibition of Cytosolic NADP Isocitrate Dehydrogenase in Zebrafish. Zebrafish 2014; 11:122-8. [DOI: 10.1089/zeb.2013.0919] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Affiliation(s)
- Hang-Suk Chun
- Drug Discovery Platform Technology Team, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Sun Hye Shin
- Drug Discovery Platform Technology Team, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Sunjoo Ahn
- Drug Discovery Platform Technology Team, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Dae-Seop Shin
- Drug Discovery Platform Technology Team, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Sun-Sil Choi
- Drug Discovery Platform Technology Team, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Jin Hee Ahn
- Drug Discovery Platform Technology Team, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Myung Ae Bae
- Drug Discovery Platform Technology Team, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| |
Collapse
|
44
|
Wansleeben C, Bowie E, Hotten DF, Yu YRA, Hogan BLM. Age-related changes in the cellular composition and epithelial organization of the mouse trachea. PLoS One 2014; 9:e93496. [PMID: 24675804 PMCID: PMC3968161 DOI: 10.1371/journal.pone.0093496] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 03/05/2014] [Indexed: 01/25/2023] Open
Abstract
We report here senescent changes in the structure and organization of the mucociliary pseudostratified epithelium of the mouse trachea and main stem bronchi. We confirm previous reports of the gradual appearance of age-related, gland-like structures (ARGLS) in the submucosa, especially in the intercartilage regions and carina. Immunohistochemistry shows these structures contain ciliated and secretory cells and Krt5+ basal cells, but not the myoepithelial cells or ciliated ducts typical of normal submucosal glands. Data suggest they arise de novo by budding from the surface epithelium rather than by delayed growth of rudimentary or cryptic submucosal glands. In old mice the surface epithelium contains fewer cells per unit length than in young mice and the proportion of Krt5+, p63+ basal cells is reduced in both males and females. However, there appears to be no significant difference in the ability of basal stem cells isolated from individual young and old mice to form clonal tracheospheres in culture or in the ability of the epithelium to repair after damage by inhaled sulfur dioxide. Gene expression analysis by Affymetrix microarray and quantitative PCR, as well as immunohistochemistry and flow sorting studies, are consistent with low-grade chronic inflammation in the tracheas of old versus young mice and an increase in the number of immune cells. The significance of these changes for ARGL formation are not clear since several treatments that induce acute inflammation in young mice did not result in budding of the surface epithelium.
Collapse
Affiliation(s)
- Carolien Wansleeben
- Department of Cell Biology, Duke University Medical Centre, Durham, North Carolina, United States of America
| | - Emily Bowie
- Department of Cell Biology, Duke University Medical Centre, Durham, North Carolina, United States of America
| | - Danielle F. Hotten
- Department of Medicine, Duke University Medical Centre, Durham, North Carolina, United States of America
| | - Yen-Rei A. Yu
- Department of Medicine, Duke University Medical Centre, Durham, North Carolina, United States of America
| | - Brigid L. M. Hogan
- Department of Cell Biology, Duke University Medical Centre, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
45
|
Bergman A, Condeelis JS, Gligorijevic B. Invadopodia in context. Cell Adh Migr 2014; 8:273-9. [PMID: 24713806 PMCID: PMC4198352 DOI: 10.4161/cam.28349] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 02/21/2014] [Accepted: 02/25/2014] [Indexed: 12/16/2022] Open
Abstract
Invadopodia are dynamic protrusions in motile tumor cells whose function is to degrade extracellular matrix so that cells can enter into new environments. Invadopodia are specifically identified by microscopy as proteolytic invasive protrusions containing TKS5 and cortactin. The increasing complexity in models for the study of invadopodia, including engineered 3D environments, explants, or animal models in vivo, entails a higher level of microenvironment complexity as well as cancer cell heterogeneity. Such experimental setups are rich in information and offer the possibility of contextualizing invadopodia and other motility-related structures. That is, they hold the promise of revealing more realistic microenvironmental conditions under which the invadopodium assembles and functions or in which tumor cells switch to a different cellular phenotype (focal adhesion, lamellipodia, proliferation, and apoptosis). For such an effort, we need a systemic approach to microscopy, which will integrate information from multiple modalities. While the individual technologies needed to achieve this are mostly available, data integration and standardization is not a trivial process. In a systems microscopy approach, microscopy is used to extract information on cell phenotypes and the microenvironment while -omics technologies assess profiles of cancer cell and microenvironment genetic, transcription, translation, and protein makeups. Data are classified and linked via in silico modeling (including statistical and mathematical models and bioinformatics). Computational considerations create predictions to be validated experimentally by perturbing the system through use of genetic manipulations and molecular biology. With such a holistic approach, a deeper understanding of function of invadopodia in vivo will be reached, opening the potential for personalized diagnostics and therapies.
Collapse
Affiliation(s)
- Aviv Bergman
- Department of Systems and Computational Biology; Albert Einstein College of Medicine; Price Center; Bronx, NY USA
| | - John S Condeelis
- Gruss-Lipper Biophotonic Center; Albert Einstein College of Medicine; Price Center, Bronx, NY USA
- Department of Anatomy and Structural Biology; Albert Einstein College of Medicine; Price Center; Bronx, NY USA
| | - Bojana Gligorijevic
- Department of Systems and Computational Biology; Albert Einstein College of Medicine; Price Center; Bronx, NY USA
- Gruss-Lipper Biophotonic Center; Albert Einstein College of Medicine; Price Center, Bronx, NY USA
| |
Collapse
|
46
|
Opitz R, Antonica F, Costagliola S. New model systems to illuminate thyroid organogenesis. Part I: an update on the zebrafish toolbox. Eur Thyroid J 2013; 2:229-42. [PMID: 24783054 PMCID: PMC3923603 DOI: 10.1159/000357079] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 11/07/2013] [Indexed: 12/16/2022] Open
Abstract
Thyroid dysgenesis (TD) resulting from defects during embryonic thyroid development represents a major cause of congenital hypothyroidism. The pathogenetic mechanisms of TD in human newborns, however, are still poorly understood and disease-causing genetic variants have been identified in only a small percentage of TD cases. This limited understanding of the pathogenesis of TD is partly due to a lack of knowledge on how intrinsic factors and extrinsic signalling cues orchestrate the differentiation of thyroid follicular cells and the morphogenesis of thyroid tissue. Recently, embryonic stem cells and zebrafish embryos emerged as novel model systems that allow for innovative experimental approaches in order to decipher cellular and molecular mechanisms of thyroid development and to unravel pathogenic mechanisms of TD. Zebrafish embryos offer several salient properties for studies on thyroid organogenesis including rapid and external development, optical transparency, ease of breeding, relative short generation time and amenability for genome editing. In this review, we will highlight recent advances in the zebrafish toolkit to visualize cellular dynamics of organ development and discuss specific prospects of the zebrafish model for studies on vertebrate thyroid development and human congenital thyroid diseases.
Collapse
Affiliation(s)
- Robert Opitz
- Institute of Interdisciplinary Research in Molecular Human Biology, Université Libre de Bruxelles, Brussels, Belgium
| | - Francesco Antonica
- Institute of Interdisciplinary Research in Molecular Human Biology, Université Libre de Bruxelles, Brussels, Belgium
| | - Sabine Costagliola
- Institute of Interdisciplinary Research in Molecular Human Biology, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
47
|
Morrissey MA, Hagedorn EJ, Sherwood DR. Cell invasion through basement membrane: The netrin receptor DCC guides the way. WORM 2013; 2:e26169. [PMID: 24778942 PMCID: PMC3875654 DOI: 10.4161/worm.26169] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 08/14/2013] [Accepted: 08/15/2013] [Indexed: 01/23/2023]
Abstract
Cell invasion through basement membrane is an essential part of normal development and physiology, and occurs during the pathological progression of human inflammatory diseases and cancer. F-actin-rich membrane protrusions, called invadopodia, have been hypothesized to be the “drill bits” of invasive cells, mediating invasion through the dense, highly cross-linked basement membrane matrix. Though studied in vitro for over 30 y, invadopodia function in vivo has remained elusive. We have recently discovered that invadopodia breach basement membrane during anchor cell invasion in C. elegans, a genetically and visually tractable in vivo invasion event. Further, we found that the netrin receptor DCC localizes to the initial site of basement membrane breach and directs invasion through a single gap in the matrix. In this commentary, we examine how the dynamics and structure of AC-invadopodia compare with in vitro invadopodia and how the netrin receptor guides invasion through a single basement membrane breach. We end with a discussion of our surprising result that the anchor cell pushes the basement membrane aside, instead of completely dissolving it through proteolysis, and provide some ideas for how proteases and physical displacement may work together to ensure efficient and robust invasion.
Collapse
|
48
|
Hagedorn EJ, Ziel JW, Morrissey MA, Linden LM, Wang Z, Chi Q, Johnson SA, Sherwood DR. The netrin receptor DCC focuses invadopodia-driven basement membrane transmigration in vivo. ACTA ACUST UNITED AC 2013; 201:903-13. [PMID: 23751497 PMCID: PMC3678161 DOI: 10.1083/jcb.201301091] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Localized activation of netrin signaling induces focused F-actin formation and the protrusive force necessary for physical displacement of basement membrane during cell transmigration. Though critical to normal development and cancer metastasis, how cells traverse basement membranes is poorly understood. A central impediment has been the challenge of visualizing invasive cell interactions with basement membrane in vivo. By developing live-cell imaging methods to follow anchor cell (AC) invasion in Caenorhabditis elegans, we identify F-actin–based invadopodia that breach basement membrane. When an invadopodium penetrates basement membrane, it rapidly transitions into a stable invasive process that expands the breach and crosses into the vulval tissue. We find that the netrin receptor UNC-40 (DCC) specifically enriches at the site of basement membrane breach and that activation by UNC-6 (netrin) directs focused F-actin formation, generating the invasive protrusion and the cessation of invadopodia. Using optical highlighting of basement membrane components, we further demonstrate that rather than relying solely on proteolytic dissolution, the AC’s protrusion physically displaces basement membrane. These studies reveal an UNC-40–mediated morphogenetic transition at the cell–basement membrane interface that directs invading cells across basement membrane barriers.
Collapse
|