51
|
Binamé F, Pham-Van LD, Bagnard D. Manipulating oligodendrocyte intrinsic regeneration mechanism to promote remyelination. Cell Mol Life Sci 2021; 78:5257-5273. [PMID: 34019104 PMCID: PMC11073109 DOI: 10.1007/s00018-021-03852-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/14/2021] [Accepted: 05/08/2021] [Indexed: 02/06/2023]
Abstract
In demyelinated lesions, astrocytes, activated microglia and infiltrating macrophages secrete several factors regulating oligodendrocyte precursor cells' behaviour. What appears to be the initiation of an intrinsic mechanism of myelin repair is only leading to partial recovery and inefficient remyelination, a process worsening over the course of the disease. This failure is largely due to the concomitant accumulation of inhibitory cues in and around the lesion sites opposing to growth promoting factors. Here starts a complex game of interactions between the signalling pathways controlling oligodendrocytes migration or differentiation. Receptors of positive or negative cues are modulating Ras, PI3K or RhoGTPases pathways acting on oligodendrocyte cytoskeleton remodelling. From the description of this intricate signalling network, this review addresses the extent to which the modulation of the global response to inhibitory cues may pave the route towards novel therapeutic approaches for myelin repair.
Collapse
Affiliation(s)
- Fabien Binamé
- INSERM U1119, Biopathology of Myelin, Neuroprotection and Therapeutic Strategy (BMNST Lab), Labex Medalis, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Pôle API, Ecole Supérieure de Biotechnologie, 300 Boulevard Sébastien Brant, 67412, Illkirch, France
| | - Lucas D Pham-Van
- INSERM U1119, Biopathology of Myelin, Neuroprotection and Therapeutic Strategy (BMNST Lab), Labex Medalis, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Pôle API, Ecole Supérieure de Biotechnologie, 300 Boulevard Sébastien Brant, 67412, Illkirch, France
| | - Dominique Bagnard
- INSERM U1119, Biopathology of Myelin, Neuroprotection and Therapeutic Strategy (BMNST Lab), Labex Medalis, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Pôle API, Ecole Supérieure de Biotechnologie, 300 Boulevard Sébastien Brant, 67412, Illkirch, France.
| |
Collapse
|
52
|
Bianchi G, Czarnecki PG, Ho M, Roccaro AM, Sacco A, Kawano Y, Gullà A, Samur AA, Chen T, Wen K, Tai YT, Moscvin M, Wu X, Camci-Unal G, Da Vià MC, Bolli N, Sewastianik T, Carrasco RD, Ghobrial IM, Anderson KC. ROBO1 Promotes Homing, Dissemination, and Survival of Multiple Myeloma within the Bone Marrow Microenvironment. Blood Cancer Discov 2021; 2:338-353. [PMID: 34268498 PMCID: PMC8265993 DOI: 10.1158/2643-3230.bcd-20-0164] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 02/21/2021] [Accepted: 04/07/2021] [Indexed: 11/16/2022] Open
Abstract
The bone marrow (BM) microenvironment actively promotes multiple myeloma (MM) pathogenesis and therapies targeting both cancer cells and the niche are highly effective. We were interested in identifying novel signaling pathways supporting MM-BM crosstalk. Mutations in the transmembrane receptor Roundabout 1 (ROBO1) were recently identified in MM patients, however their functional consequences are uncertain. Through protein structure-function studies, we discovered that ROBO1 is necessary for MM adhesion to BM stromal and endothelial cells and ROBO1 knock out (KO) compromises BM homing and engraftment in a disseminated mouse model. ROBO1 KO significantly decreases MM proliferation in vitro and intra- and extramedullary tumor growth, in vivo. Mechanistically, ROBO1 C-terminus is cleaved in a ligand-independent fashion and is sufficient to promote MM proliferation. Viceversa, mutants lacking the cytoplasmic domain, including the human-derived G674* truncation, act dominantly negative. Interactomic and RNA sequencing studies suggest ROBO1 may be involved in RNA processing, supporting further studies.
Collapse
Affiliation(s)
- Giada Bianchi
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.
| | - Peter G Czarnecki
- Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Matthew Ho
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Aldo M Roccaro
- Clinical Research Development and Phase I Unit, CREA Laboratory, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Antonio Sacco
- Clinical Research Development and Phase I Unit, CREA Laboratory, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Yawara Kawano
- Departments of Hematology, Rheumatology, and Infectious Disease, Kumamoto University Hospital, Kumamoto, Japan
| | - Annamaria Gullà
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Anil Aktas Samur
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Tianzeng Chen
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Kenneth Wen
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Yu-Tzu Tai
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Maria Moscvin
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Xinchen Wu
- Department of Chemical Engineering, University of Massachusetts Lowell, Lowell, Massachusetts
| | - Gulden Camci-Unal
- Department of Chemical Engineering, University of Massachusetts Lowell, Lowell, Massachusetts
| | - Matteo C Da Vià
- Department of Oncology and Onco-Hematology, University of Milan, Milan, Italy
| | - Niccolo' Bolli
- Department of Oncology and Onco-Hematology, University of Milan, Milan, Italy
- Department of Clinical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Tomasz Sewastianik
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Ruben D Carrasco
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Irene M Ghobrial
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Kenneth C Anderson
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
53
|
Wurmser M, Muppavarapu M, Tait CM, Laumonnerie C, González-Castrillón LM, Wilson SI. Robo2 Receptor Gates the Anatomical Divergence of Neurons Derived From a Common Precursor Origin. Front Cell Dev Biol 2021; 9:668175. [PMID: 34249921 PMCID: PMC8263054 DOI: 10.3389/fcell.2021.668175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 05/03/2021] [Indexed: 12/25/2022] Open
Abstract
Sensory information relayed to the brain is dependent on complex, yet precise spatial organization of neurons. This anatomical complexity is generated during development from a surprisingly small number of neural stem cell domains. This raises the question of how neurons derived from a common precursor domain respond uniquely to their environment to elaborate correct spatial organization and connectivity. We addressed this question by exploiting genetically labeled mouse embryonic dorsal interneuron 1 (dI1) neurons that are derived from a common precursor domain and give rise to spinal projection neurons with distinct organization of cell bodies with axons projecting either commissurally (dI1c) or ipsilaterally (dI1i). In this study, we examined how the guidance receptor, Robo2, which is a canonical Robo receptor, influenced dI1 guidance during embryonic development. Robo2 was enriched in embryonic dI1i neurons, and loss of Robo2 resulted in misguidance of dI1i axons, whereas dI1c axons remained unperturbed within the mantle zone and ventral commissure. Further, Robo2 profoundly influenced dI1 cell body migration, a feature that was partly dependent on Slit2 signaling. These data suggest that dI1 neurons are dependent on Robo2 for their organization. This work integrated with the field support of a model whereby canonical Robo2 vs. non-canonical Robo3 receptor expression facilitates projection neurons derived from a common precursor domain to read out the tissue environment uniquely giving rise to correct anatomical organization.
Collapse
Affiliation(s)
- Maud Wurmser
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | | | | | | | | | - Sara Ivy Wilson
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| |
Collapse
|
54
|
Getmantseva L, Kolosova M, Bakoev F, Zimina A, Bakoev S. Genomic Regions and Candidate Genes Linked to Capped Hock in Pig. Life (Basel) 2021; 11:life11060510. [PMID: 34073088 PMCID: PMC8228005 DOI: 10.3390/life11060510] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/25/2021] [Accepted: 05/28/2021] [Indexed: 01/13/2023] Open
Abstract
Capped hock affects the exterior of pedigree pigs, making them unsalable and resulting in a negative impact on the efficiency of pig-breeding centers. The purpose of this paper was to carry out pilot studies aimed at finding genomic regions and genes linked to the capped hock in pigs. The studies were carried out on Landrace pigs (n = 75) and Duroc pigs (n = 70). To identify genomic regions linked to capped hock in pigs, we used smoothing FST statistics. Genotyping was performed with GeneSeek® GGP Porcine HD Genomic Profiler v1 (Illumina Inc, San Diego, CA, USA). The research results showed 70 SNPs linked to capped hock in Landrace (38 SNPs) and Duroc (32 SNPs). The identified regions overlapped with QTLs related with health traits (blood parameters) and meat and carcass traits (fatness). In total, 31 genes were identified (i.e., 17 genes in Landrace, 14 genes in Durocs). Three genes appeared in both the Landrace and Duroc groups, including A2ML1 (SSC5), ROBO2 (SSC13), and MSI1 (SSC14). We identified genomic regions directly or indirectly linked to capped hock, which thus might contribute to identifying genetic variants and using them as genetic markers in pig breeding.
Collapse
Affiliation(s)
- Lyubov Getmantseva
- Federal Research Center for Animal Husbandry Named after Academy Member L.K. Ernst, 142132 Dubrovitsy, Russia; (M.K.); (F.B.); (S.B.)
- Correspondence: (L.G.); (A.Z.); Tel.: +7-(4967)-65-11-01 (L.G. & A.Z.)
| | - Maria Kolosova
- Federal Research Center for Animal Husbandry Named after Academy Member L.K. Ernst, 142132 Dubrovitsy, Russia; (M.K.); (F.B.); (S.B.)
- Department of Biotechnology, Don State Agrarian University, 346493 Persianovski, Russia
| | - Faridun Bakoev
- Federal Research Center for Animal Husbandry Named after Academy Member L.K. Ernst, 142132 Dubrovitsy, Russia; (M.K.); (F.B.); (S.B.)
| | - Anna Zimina
- Federal Research Center for Animal Husbandry Named after Academy Member L.K. Ernst, 142132 Dubrovitsy, Russia; (M.K.); (F.B.); (S.B.)
- Correspondence: (L.G.); (A.Z.); Tel.: +7-(4967)-65-11-01 (L.G. & A.Z.)
| | - Siroj Bakoev
- Federal Research Center for Animal Husbandry Named after Academy Member L.K. Ernst, 142132 Dubrovitsy, Russia; (M.K.); (F.B.); (S.B.)
- Centre for Strategic Planning and Management of Biomedical Health Risks, 123182 Moscow, Russia
| |
Collapse
|
55
|
The Polygenic Nature and Complex Genetic Architecture of Specific Learning Disorder. Brain Sci 2021; 11:brainsci11050631. [PMID: 34068951 PMCID: PMC8156942 DOI: 10.3390/brainsci11050631] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 12/16/2022] Open
Abstract
Specific Learning Disorder (SLD) is a multifactorial, neurodevelopmental disorder which may involve persistent difficulties in reading (dyslexia), written expression and/or mathematics. Dyslexia is characterized by difficulties with speed and accuracy of word reading, deficient decoding abilities, and poor spelling. Several studies from different, but complementary, scientific disciplines have investigated possible causal/risk factors for SLD. Biological, neurological, hereditary, cognitive, linguistic-phonological, developmental and environmental factors have been incriminated. Despite worldwide agreement that SLD is highly heritable, its exact biological basis remains elusive. We herein present: (a) an update of studies that have shaped our current knowledge on the disorder’s genetic architecture; (b) a discussion on whether this genetic architecture is ‘unique’ to SLD or, alternatively, whether there is an underlying common genetic background with other neurodevelopmental disorders; and, (c) a brief discussion on whether we are at a position of generating meaningful correlations between genetic findings and anatomical data from neuroimaging studies or specific molecular/cellular pathways. We conclude with open research questions that could drive future research directions.
Collapse
|
56
|
Zalfa C, Paust S. Natural Killer Cell Interactions With Myeloid Derived Suppressor Cells in the Tumor Microenvironment and Implications for Cancer Immunotherapy. Front Immunol 2021; 12:633205. [PMID: 34025641 PMCID: PMC8133367 DOI: 10.3389/fimmu.2021.633205] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 02/12/2021] [Indexed: 12/17/2022] Open
Abstract
The tumor microenvironment (TME) is a complex and heterogeneous environment composed of cancer cells, tumor stroma, a mixture of tissue-resident and infiltrating immune cells, secreted factors, and extracellular matrix proteins. Natural killer (NK) cells play a vital role in fighting tumors, but chronic stimulation and immunosuppression in the TME lead to NK cell exhaustion and limited antitumor functions. Myeloid-derived suppressor cells (MDSCs) are a heterogeneous group of myeloid cells with potent immunosuppressive activity that gradually accumulate in tumor tissues. MDSCs interact with innate and adaptive immune cells and play a crucial role in negatively regulating the immune response to tumors. This review discusses MDSC-mediated NK cell regulation within the TME, focusing on critical cellular and molecular interactions. We review current strategies that target MDSC-mediated immunosuppression to enhance NK cell cytotoxic antitumor activity. We also speculate on how NK cell-based antitumor immunotherapy could be improved.
Collapse
Affiliation(s)
| | - Silke Paust
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, United States
| |
Collapse
|
57
|
Guzmán-Palma P, Contreras EG, Mora N, Smith M, González-Ramírez MC, Campusano JM, Sierralta J, Hassan BA, Oliva C. Slit/Robo Signaling Regulates Multiple Stages of the Development of the Drosophila Motion Detection System. Front Cell Dev Biol 2021; 9:612645. [PMID: 33968921 PMCID: PMC8097104 DOI: 10.3389/fcell.2021.612645] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 03/24/2021] [Indexed: 11/29/2022] Open
Abstract
Neurogenesis is achieved through a sequence of steps that include specification and differentiation of progenitors into mature neurons. Frequently, precursors migrate to distinct positions before terminal differentiation. The Slit-Robo pathway, formed by the secreted ligand Slit and its membrane bound receptor Robo, was first discovered as a regulator of axonal growth. However, today, it is accepted that this pathway can regulate different cellular processes even outside the nervous system. Since most of the studies performed in the nervous system have been focused on axonal and dendritic growth, it is less clear how versatile is this signaling pathway in the developing nervous system. Here we describe the participation of the Slit-Robo pathway in the development of motion sensitive neurons of the Drosophila visual system. We show that Slit and Robo receptors are expressed in different stages during the neurogenesis of motion sensitive neurons. Furthermore, we find that Slit and Robo regulate multiple aspects of their development including neuronal precursor migration, cell segregation between neural stem cells and daughter cells and formation of their connectivity pattern. Specifically, loss of function of slit or robo receptors in differentiated motion sensitive neurons impairs dendritic targeting, while knocking down robo receptors in migratory progenitors or neural stem cells leads to structural defects in the adult optic lobe neuropil, caused by migration and cell segregation defects during larval development. Thus, our work reveals the co-option of the Slit-Robo signaling pathway in distinct developmental stages of a neural lineage.
Collapse
Affiliation(s)
- Pablo Guzmán-Palma
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Esteban G Contreras
- Department of Neuroscience and Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Natalia Mora
- Institut du Cerveau-Paris Brain Institute (ICM), Inserm, CNRS, Hôpital Pitié-Salpêtrière, Sorbonne Université, Paris, France
| | - Macarena Smith
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - M Constanza González-Ramírez
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jorge M Campusano
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jimena Sierralta
- Department of Neuroscience and Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Bassem A Hassan
- Institut du Cerveau-Paris Brain Institute (ICM), Inserm, CNRS, Hôpital Pitié-Salpêtrière, Sorbonne Université, Paris, France
| | - Carlos Oliva
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
58
|
Chaudhari K, Gorla M, Chang C, Kania A, Bashaw GJ. Robo recruitment of the Wave regulatory complex plays an essential and conserved role in midline repulsion. eLife 2021; 10:e64474. [PMID: 33843588 PMCID: PMC8096436 DOI: 10.7554/elife.64474] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 04/06/2021] [Indexed: 12/23/2022] Open
Abstract
The Roundabout (Robo) guidance receptor family induces axon repulsion in response to its ligand Slit by inducing local cytoskeletal changes; however, the link to the cytoskeleton and the nature of these cytoskeletal changes are poorly understood. Here, we show that the heteropentameric Scar/Wave Regulatory Complex (WRC), which drives Arp2/3-induced branched actin polymerization, is a direct effector of Robo signaling. Biochemical evidence shows that Slit triggers WRC recruitment to the Robo receptor's WRC-interacting receptor sequence (WIRS) motif. In Drosophila embryos, mutants of the WRC enhance Robo1-dependent midline crossing defects. Additionally, mutating Robo1's WIRS motif significantly reduces receptor activity in rescue assays in vivo, and CRISPR-Cas9 mutagenesis shows that the WIRS motif is essential for endogenous Robo1 function. Finally, axon guidance assays in mouse dorsal spinal commissural axons and gain-of-function experiments in chick embryos demonstrate that the WIRS motif is also required for Robo1 repulsion in mammals. Together, our data support an essential conserved role for the WIRS-WRC interaction in Robo1-mediated axon repulsion.
Collapse
Affiliation(s)
- Karina Chaudhari
- Department of Neuroscience, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Madhavi Gorla
- Department of Neuroscience, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Chao Chang
- Institut de recherches cliniques de Montréal (IRCM)MontréalCanada
- Department of Anatomy and Cell Biology and Division of Experimental Medicine, McGill UniversityMontréalCanada
| | - Artur Kania
- Institut de recherches cliniques de Montréal (IRCM)MontréalCanada
- Department of Anatomy and Cell Biology and Division of Experimental Medicine, McGill UniversityMontréalCanada
| | - Greg J Bashaw
- Department of Neuroscience, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| |
Collapse
|
59
|
Doyle GA, Reiner BC, Crist RC, Rao AM, Ojeah NS, Arauco-Shapiro G, Levinson RN, Shah LD, Sperling MR, Ferraro TN, Buono RJ, Berrettini WH. Investigation of long interspersed element-1 retrotransposons as potential risk factors for idiopathic temporal lobe epilepsy. Epilepsia 2021; 62:1329-1342. [PMID: 33826137 DOI: 10.1111/epi.16897] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 03/19/2021] [Accepted: 03/19/2021] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To determine if long interspersed element-1 (L1) retrotransposons convey risk for idiopathic temporal lobe epilepsy (TLE). METHODS Surgically resected temporal cortex from individuals with TLE (N = 33) and postmortem temporal cortex from individuals with no known neurological disease (N = 33) were analyzed for L1 content by Restriction Enzyme Based Enriched L1Hs sequencing (REBELseq). Expression of three KCNIP4 splice variants was assessed by droplet digital PCR (ddPCR). Protein ANalysis THrough Evolutionary Relationships (PANTHER) was used to determine ontologies and pathways for lists of genes harboring L1 insertions. RESULTS We identified novel L1 insertions specific to individuals with TLE, and others specific to controls. Although there were no statistically significant differences between cases and controls in the numbers of known and novel L1 insertions, PANTHER analyses of intragenic L1 insertions showed statistically significant enrichments for epilepsy-relevant gene ontologies in both cases and controls. Gene ontologies "neuron projection development" and "calcium ion transmembrane transport" were among those found only in individuals with TLE. We confirmed novel L1 insertions in several genes associated with seizures/epilepsy, including a de novo somatic L1 retrotransposition in KCNIP4 that occurred after neural crest formation in one patient. However, ddPCR results suggest this de novo L1 did not alter KCNIP4 mRNA expression. SIGNIFICANCE Given current data from this small cohort, we conclude that L1 elements, either rare heritable germline insertions or de novo somatic retrotranspositions, may contribute only minimally to overall genetic risk for idiopathic TLE. We suggest that further studies in additional patients and additional brain regions are warranted.
Collapse
Affiliation(s)
- Glenn A Doyle
- Department of Psychiatry, Center for Neurobiology and Behavior, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Benjamin C Reiner
- Department of Psychiatry, Center for Neurobiology and Behavior, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Richard C Crist
- Department of Psychiatry, Center for Neurobiology and Behavior, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Aditya M Rao
- Department of Psychiatry, Center for Neurobiology and Behavior, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Nkechi S Ojeah
- Department of Psychiatry, Center for Neurobiology and Behavior, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,Lincoln University of School of Natural Sciences and Mathematics, Lincoln University, Lincoln, PA, USA
| | - Gabriella Arauco-Shapiro
- Department of Psychiatry, Center for Neurobiology and Behavior, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Rachel N Levinson
- Department of Psychiatry, Center for Neurobiology and Behavior, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Lokesh D Shah
- Department of Psychiatry, Center for Neurobiology and Behavior, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Michael R Sperling
- Department of Neurology, Comprehensive Epilepsy Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Thomas N Ferraro
- Department of Psychiatry, Center for Neurobiology and Behavior, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
| | - Russell J Buono
- Department of Neurology, Comprehensive Epilepsy Center, Thomas Jefferson University, Philadelphia, PA, USA.,Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
| | - Wade H Berrettini
- Department of Psychiatry, Center for Neurobiology and Behavior, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
60
|
Daiber T, VanderZwan-Butler CJ, Bashaw GJ, Evans TA. Conserved and divergent aspects of Robo receptor signaling and regulation between Drosophila Robo1 and C. elegans SAX-3. Genetics 2021; 217:iyab018. [PMID: 33789352 PMCID: PMC8045725 DOI: 10.1093/genetics/iyab018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 01/28/2021] [Indexed: 11/24/2022] Open
Abstract
The evolutionarily conserved Roundabout (Robo) family of axon guidance receptors control midline crossing of axons in response to the midline repellant ligand Slit in bilaterian animals including insects, nematodes, and vertebrates. Despite this strong evolutionary conservation, it is unclear whether the signaling mechanism(s) downstream of Robo receptors are similarly conserved. To directly compare midline repulsive signaling in Robo family members from different species, here we use a transgenic approach to express the Robo family receptor SAX-3 from the nematode Caenorhabditis elegans in neurons of the fruit fly, Drosophila melanogaster. We examine SAX-3's ability to repel Drosophila axons from the Slit-expressing midline in gain of function assays, and test SAX-3's ability to substitute for Drosophila Robo1 during fly embryonic development in genetic rescue experiments. We show that C. elegans SAX-3 is properly translated and localized to neuronal axons when expressed in the Drosophila embryonic CNS, and that SAX-3 can signal midline repulsion in Drosophila embryonic neurons, although not as efficiently as Drosophila Robo1. Using a series of Robo1/SAX-3 chimeras, we show that the SAX-3 cytoplasmic domain can signal midline repulsion to the same extent as Robo1 when combined with the Robo1 ectodomain. We show that SAX-3 is not subject to endosomal sorting by the negative regulator Commissureless (Comm) in Drosophila neurons in vivo, and that peri-membrane and ectodomain sequences are both required for Comm sorting of Drosophila Robo1.
Collapse
Affiliation(s)
- Trent Daiber
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR 72701, USA
| | | | - Greg J Bashaw
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Timothy A Evans
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR 72701, USA
| |
Collapse
|
61
|
Lysosomal Function and Axon Guidance: Is There a Meaningful Liaison? Biomolecules 2021; 11:biom11020191. [PMID: 33573025 PMCID: PMC7911486 DOI: 10.3390/biom11020191] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 01/26/2021] [Accepted: 01/26/2021] [Indexed: 01/25/2023] Open
Abstract
Axonal trajectories and neural circuit activities strongly rely on a complex system of molecular cues that finely orchestrate the patterning of neural commissures. Several of these axon guidance molecules undergo continuous recycling during brain development, according to incompletely understood intracellular mechanisms, that in part rely on endocytic and autophagic cascades. Based on their pivotal role in both pathways, lysosomes are emerging as a key hub in the sophisticated regulation of axonal guidance cue delivery, localization, and function. In this review, we will attempt to collect some of the most relevant research on the tight connection between lysosomal function and axon guidance regulation, providing some proof of concepts that may be helpful to understanding the relation between lysosomal storage disorders and neurodegenerative diseases.
Collapse
|
62
|
Abstract
Pituitary stalk interruption syndrome (PSIS) is a distinct developmental defect of the pituitary gland identified by magnetic resonance imaging and characterized by a thin, interrupted, attenuated or absent pituitary stalk, hypoplasia or aplasia of the adenohypophysis, and an ectopic posterior pituitary. The precise etiology of PSIS still remains elusive or incompletely confirmed in most cases. Adverse perinatal events, including breech delivery and hypoxia, were initially proposed as the underlying mechanism affecting the hypothalamic-pituitary axis. Nevertheless, recent findings have uncovered a wide variety of PSIS-associated molecular defects in genes involved in pituitary development, holoprosencephaly (HPE), neural development, and other important cellular processes such as cilia function. The application of whole exome sequencing (WES) in relatively large cohorts has identified an expanded pool of potential candidate genes, mostly related to the Wnt, Notch, and sonic hedgehog signaling pathways that regulate pituitary growth and development during embryogenesis. Importantly, WES has revealed coexisting pathogenic variants in a significant number of patients; therefore, pointing to a multigenic origin and inheritance pattern of PSIS. The disorder is characterized by inter- and intrafamilial variability and incomplete or variable penetrance. Overall, PSIS is currently viewed as a mild form of an expanded HPE spectrum. The wide and complex clinical manifestations include evolving pituitary hormone deficiencies (with variable timing of onset and progression) and extrapituitary malformations. Severe and life-threatening symptomatology is observed in a subset of patients with complete pituitary hormone deficiency during the neonatal period. Nevertheless, most patients are referred later in childhood for growth retardation. Prompt and appropriate hormone substitution therapy constitutes the cornerstone of treatment. Further studies are needed to uncover the etiopathogenesis of PSIS.
Collapse
Affiliation(s)
- Antonis Voutetakis
- Department of Pediatrics, School of Medicine, Democritus University of Thrace, Alexandroupolis, Thrace, Greece.
| |
Collapse
|
63
|
Gonda Y, Namba T, Hanashima C. Beyond Axon Guidance: Roles of Slit-Robo Signaling in Neocortical Formation. Front Cell Dev Biol 2020; 8:607415. [PMID: 33425915 PMCID: PMC7785817 DOI: 10.3389/fcell.2020.607415] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/07/2020] [Indexed: 12/11/2022] Open
Abstract
The formation of the neocortex relies on intracellular and extracellular signaling molecules that are involved in the sequential steps of corticogenesis, ranging from the proliferation and differentiation of neural progenitor cells to the migration and dendrite formation of neocortical neurons. Abnormalities in these steps lead to disruption of the cortical structure and circuit, and underly various neurodevelopmental diseases, including dyslexia and autism spectrum disorder (ASD). In this review, we focus on the axon guidance signaling Slit-Robo, and address the multifaceted roles of Slit-Robo signaling in neocortical development. Recent studies have clarified the roles of Slit-Robo signaling not only in axon guidance but also in progenitor cell proliferation and migration, and the maturation of neocortical neurons. We further discuss the etiology of neurodevelopmental diseases, which are caused by defects in Slit-Robo signaling during neocortical formation.
Collapse
Affiliation(s)
- Yuko Gonda
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| | - Takashi Namba
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- Neuroscience Center, HiLIFE – Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Carina Hanashima
- Faculty of Education and Integrated Arts and Sciences, Waseda University, Tokyo, Japan
- Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| |
Collapse
|
64
|
Mascheretti S, Riva V, Feng B, Trezzi V, Andreola C, Giorda R, Villa M, Dionne G, Gori S, Marino C, Facoetti A. The Mediation Role of Dynamic Multisensory Processing Using Molecular Genetic Data in Dyslexia. Brain Sci 2020; 10:brainsci10120993. [PMID: 33339203 PMCID: PMC7765588 DOI: 10.3390/brainsci10120993] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/04/2020] [Accepted: 12/11/2020] [Indexed: 12/21/2022] Open
Abstract
Although substantial heritability has been reported and candidate genes have been identified, we are far from understanding the etiopathogenetic pathways underlying developmental dyslexia (DD). Reading-related endophenotypes (EPs) have been established. Until now it was unknown whether they mediated the pathway from gene to reading (dis)ability. Thus, in a sample of 223 siblings from nuclear families with DD and 79 unrelated typical readers, we tested four EPs (i.e., rapid auditory processing, rapid automatized naming, multisensory nonspatial attention and visual motion processing) and 20 markers spanning five DD-candidate genes (i.e., DYX1C1, DCDC2, KIAA0319, ROBO1 and GRIN2B) using a multiple-predictor/multiple-mediator framework. Our results show that rapid auditory and visual motion processing are mediators in the pathway from ROBO1-rs9853895 to reading. Specifically, the T/T genotype group predicts impairments in rapid auditory and visual motion processing which, in turn, predict poorer reading skills. Our results suggest that ROBO1 is related to reading via multisensory temporal processing. These findings support the use of EPs as an effective approach to disentangling the complex pathways between candidate genes and behavior.
Collapse
Affiliation(s)
- Sara Mascheretti
- Child Psychopathology Unit, Scientific Institute, IRCCS E. Medea, 23842 Bosisio Parini, Italy; (S.M.); (V.R.); (V.T.); (C.A.)
| | - Valentina Riva
- Child Psychopathology Unit, Scientific Institute, IRCCS E. Medea, 23842 Bosisio Parini, Italy; (S.M.); (V.R.); (V.T.); (C.A.)
| | - Bei Feng
- École de Psychologie, Laval University, Québec, QC G1V 0A6, Canada; (B.F.); (G.D.)
| | - Vittoria Trezzi
- Child Psychopathology Unit, Scientific Institute, IRCCS E. Medea, 23842 Bosisio Parini, Italy; (S.M.); (V.R.); (V.T.); (C.A.)
| | - Chiara Andreola
- Child Psychopathology Unit, Scientific Institute, IRCCS E. Medea, 23842 Bosisio Parini, Italy; (S.M.); (V.R.); (V.T.); (C.A.)
- Laboratoire de Psychologie du Développement et de l’Éducation de l’Enfant (LaPsyDÉ), Universitè de Paris, 75005 Paris, France
| | - Roberto Giorda
- Molecular Biology Laboratory, Scientific Institute, IRCCS E. Medea, 23842 Bosisio Parini, Italy; (R.G.); (M.V.)
| | - Marco Villa
- Molecular Biology Laboratory, Scientific Institute, IRCCS E. Medea, 23842 Bosisio Parini, Italy; (R.G.); (M.V.)
| | - Ginette Dionne
- École de Psychologie, Laval University, Québec, QC G1V 0A6, Canada; (B.F.); (G.D.)
| | - Simone Gori
- Department of Human and Social Sciences, University of Bergamo, 24100 Bergamo, Italy;
| | - Cecilia Marino
- Child Psychopathology Unit, Scientific Institute, IRCCS E. Medea, 23842 Bosisio Parini, Italy; (S.M.); (V.R.); (V.T.); (C.A.)
- Department of Psychiatry, University of Toronto, Toronto, ON M5T 1R8, Canada
- The Division of Child and Youth Psychiatry, Centre for Addiction and Mental Health (CAMH), Toronto, ON M6J 1H4, Canada
- Correspondence: (C.M.); (A.F.)
| | - Andrea Facoetti
- Developmental Cognitive Neuroscience Lab, Department of General Psychology, University of Padua, 35131 Padua, Italy
- Correspondence: (C.M.); (A.F.)
| |
Collapse
|
65
|
Zhu J, Chen G. Protective effect of FOXP3-mediated miR-146b-5p/Robo1/NF-κB system on lipopolysaccharide-induced acute lung injury in mice. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1651. [PMID: 33490163 PMCID: PMC7812239 DOI: 10.21037/atm-20-7703] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background As a key transcription factor, forkhead box protein 3 (FOXP3) plays an important role in the development and function of natural cluster of differentiation 4 [CD4 (+)] regulatory T cells (Treg cells). However, the function of FOXP3 in Lipopolysaccharide (LPS)-induced acute lung injury (ALI) through regulating miR-146b-5p is unclear. This research aimed to disclose the regulatory effect of the FOXP3-mediated miR-146b-5p/Roundabout 1 (Robo1)/NF-κB system on LPS-induced ALI in mice. Methods The mice were subjected to 5 mg/kg of LPS via intratracheal instillation to induce ALI and generate the ALI model. Mice was divided into five group, including control group, ALI group, ALI + FOXP3 group, the ALI + miR antagomir group and ALI + miR antagomir+ FOXP3 group. Lung tissue injury were detected by hematoxylin and eosin (HE) staining. Lung wet/dry weight ratio, total cells in bronchoalveolar lavage fluid (BALF), total protein in BALF and the polymorphonuclear leukocyte (PMN) in BALF were detected. The levels of tumor necrosis factor-α (TNF-α), Interleukin 6 (IL-6) and IL-1β were detected by enzyme-linked immunosorbent assay (ELISA) kit. The dual-luciferase reporter assay were used to detect the target relationship between FOXP3 and Robo1. Mice was divided into five group, including control group, ALI group, ALI + FOXP3 group, ALI + Robo1 group and ALI + FOXP3+ Robo1 group. The protein levels of FOXP3, Robo1 and p-p65 were detected by western bolt. The mRNA levels of miR-146b-5p and Robo1 were detected by quantitative reverse transcription polymerase chain reaction (qRT-PCR). Results Although protein expression levels of FOXP3 were significantly down-regulated in the ALI model, the increased FOXP3 levels promoted an increase in miR-146b-5p. Compared with the control group, the ALI model group exhibited severe histopathologic injury, such as thickening of the alveolar wall, pulmonary congestion, and decreased alveolar numbers. By mediating the overexpression of miR-146b-5p, FOXP3 also increased alveolar clearance and inhibited inflammatory responses in the ALI model. Importantly, Robo1 is a potential target of miR-146b-5p. Conclusions FOXP3 could inhibit NF-κB activation, reduce lung pathological damage, and inhibit inflammatory responses by mediating the miR-146b-5p/Robo1/NF-κB system in the ALI model. These results may provide a new potential target for the treatment of ALI disease.
Collapse
Affiliation(s)
- Jiang Zhu
- Department of Respiratory and Critical Care Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University Hospital of Electronic Science & Technology of China, Chengdu, China
| | - Gaoli Chen
- Department of Blood Transfusion, Teaching Hospital of Chengdu University of TCM, Chengdu, China
| |
Collapse
|
66
|
Alvarez S, Varadarajan SG, Butler SJ. Dorsal commissural axon guidance in the developing spinal cord. Curr Top Dev Biol 2020; 142:197-231. [PMID: 33706918 DOI: 10.1016/bs.ctdb.2020.10.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Commissural axons have been a key model system for identifying axon guidance signals in vertebrates. This review summarizes the current thinking about the molecular and cellular mechanisms that establish a specific commissural neural circuit: the dI1 neurons in the developing spinal cord. We assess the contribution of long- and short-range signaling while sequentially following the developmental timeline from the birth of dI1 neurons, to the extension of commissural axons first circumferentially and then contralaterally into the ventral funiculus.
Collapse
Affiliation(s)
- Sandy Alvarez
- Department of Neurobiology, University of California, Los Angeles, CA, United States; Molecular Biology Interdepartmental Doctoral Program, University of California, Los Angeles, CA, United States
| | | | - Samantha J Butler
- Department of Neurobiology, University of California, Los Angeles, CA, United States; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, United States.
| |
Collapse
|
67
|
Kellermeyer R, Heydman LM, Gillis T, Mastick GS, Song M, Kidd T. Proteolytic cleavage of Slit by the Tolkin protease converts an axon repulsion cue to an axon growth cue in vivo. Development 2020; 147:dev.196055. [PMID: 32994163 PMCID: PMC7648596 DOI: 10.1242/dev.196055] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 09/18/2020] [Indexed: 12/27/2022]
Abstract
Slit is a secreted protein that has a canonical function of repelling growing axons from the CNS midline. The full-length Slit (Slit-FL) is cleaved into Slit-N and Slit-C fragments, which have potentially distinct functions via different receptors. Here, we report that the BMP-1/Tolloid family metalloprotease Tolkin (Tok) is responsible for Slit proteolysis in vivo and in vitro. In Drosophilatok mutants lacking Slit cleavage, midline repulsion of axons occurs normally, confirming that Slit-FL is sufficient to repel axons. However, longitudinal axon guidance is highly disrupted in tok mutants and can be rescued by midline expression of Slit-N, suggesting that Slit is the primary substrate for Tok in the embryonic CNS. Transgenic restoration of Slit-N or Slit-C does not repel axons in Slit-null flies. Slit-FL and Slit-N are both biologically active cues with distinct axon guidance functions in vivo Slit signaling is used in diverse biological processes; therefore, differentiating between Slit-FL and Slit fragments will be essential for evaluating Slit function in broader contexts.
Collapse
Affiliation(s)
| | | | | | | | | | - Thomas Kidd
- Department of Biology/MS 314, University of Nevada, 1664 North Virginia Street, Reno, NV 89557, USA
| |
Collapse
|
68
|
Brown HE, Evans TA. Minimal structural elements required for midline repulsive signaling and regulation of Drosophila Robo1. PLoS One 2020; 15:e0241150. [PMID: 33091076 PMCID: PMC7580999 DOI: 10.1371/journal.pone.0241150] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 10/08/2020] [Indexed: 02/07/2023] Open
Abstract
The Roundabout (Robo) family of axon guidance receptors has a conserved ectodomain arrangement of five immunoglobulin-like (Ig) domains plus three fibronectin type III (Fn) repeats. Based on the strong evolutionary conservation of this domain structure among Robo receptors, as well as in vitro structural and domain-domain interaction studies of Robo family members, this ectodomain arrangement is predicted to be important for Robo receptor signaling in response to Slit ligands. Here, we define the minimal ectodomain structure required for Slit binding and midline repulsive signaling in vivo by Drosophila Robo1. We find that the majority of the Robo1 ectodomain is dispensable for both Slit binding and repulsive signaling. We show that a significant level of midline repulsive signaling activity is retained when all Robo1 ectodomain elements apart from Ig1 are deleted, and that the combination of Ig1 plus one additional ectodomain element (Ig2, Ig5, or Fn3) is sufficient to restore midline repulsion to wild type levels. Further, we find that deleting four out of five Robo1 Ig domains (ΔIg2-5) does not affect negative regulation of Robo1 by Commissureless (Comm) or Robo2, while variants lacking all three fibronectin repeats (ΔFn1-3 and ΔIg2-Fn3) are insensitive to regulation by both Comm and Robo2, signifying a novel regulatory role for Robo1's Fn repeats. Our results provide an in vivo perspective on the importance of the conserved 5+3 ectodomain structure of Robo receptors, and suggest that specific biochemical properties and/or ectodomain structural conformations observed in vitro for domains other than Ig1 may have limited significance for in vivo signaling in the context of midline repulsion.
Collapse
Affiliation(s)
- Haley E. Brown
- Department of Biological Sciences, University of Arkansas, Fayetteville, Arkansas, United States of America
| | - Timothy A. Evans
- Department of Biological Sciences, University of Arkansas, Fayetteville, Arkansas, United States of America
| |
Collapse
|
69
|
Sato K, Ito H, Yamamoto D. teiresias, a Fruitless target gene encoding an immunoglobulin-superfamily transmembrane protein, is required for neuronal feminization in Drosophila. Commun Biol 2020; 3:598. [PMID: 33087851 PMCID: PMC7578032 DOI: 10.1038/s42003-020-01327-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 09/25/2020] [Indexed: 01/02/2023] Open
Abstract
This study aims at identifying transcriptional targets of FruitlessBM (FruBM), which represents the major isoform of male-specific FruM transcription factors that induce neural sexual dimorphisms. A promoter of the axon-guidance factor gene robo1 carries the 16-bp palindrome motif Pal1, to which FruM binds. Our genome-wide search for Pal1-homologous sequences yielded ~200 candidate genes. Among these, CG17716 potentially encodes a transmembrane protein with extracellular immunoglobulin (Ig)-like domains similar to Robo1. Indeed, FruBM overexpression reduced CG17716 mRNA and protein expression. In the fru-expressing mAL neuron cluster exhibiting sexual dimorphism, we found that CG17716 knockdown in female neurons completely transformed all neurites to the male-type. Conversely, CG17716 overexpression suppressed male-specific midline crossing of fru-expressing sensory axons. We renamed CG17716 teiresias (tei) based on this feminizing function. We hypothesize that Tei interacts with other Ig superfamily transmembrane proteins, including Robo1, to feminize the neurite patterns in females, whereas FruBM represses tei transcription in males. Sato, Ito, and Yamamoto report the identification of a Fruitless target gene, teiresias (tei), required for neuronal feminization in Drosophila. They find that tei is responsible for specifying the sex-specific structure of neurites and propose that the Tei protein forms a complex with robo1, conferring the ligand specificity on the heteromeric receptor complex.
Collapse
Affiliation(s)
- Kosei Sato
- Neuro-Network Evolution Project, Advanced ICT Research Institute, National Institute of Information and Communications Technology, Kobe, Japan
| | - Hiroki Ito
- Division of Neurogenetics, Tohoku University Graduate School of Life Sciences, Sendai, Japan
| | - Daisuke Yamamoto
- Neuro-Network Evolution Project, Advanced ICT Research Institute, National Institute of Information and Communications Technology, Kobe, Japan.
| |
Collapse
|
70
|
Fujiwara K, Koyama K, Tsuji AB, Iwanari H, Kusano-Arai O, Higashi T, Momose T, Hamakubo T. Single-Dose Cisplatin Pre-Treatment Enhances Efficacy of ROBO1-Targeted Radioimmunotherapy. Int J Mol Sci 2020; 21:ijms21207728. [PMID: 33086574 PMCID: PMC7589062 DOI: 10.3390/ijms21207728] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/09/2020] [Accepted: 10/13/2020] [Indexed: 01/31/2023] Open
Abstract
We previously reported that radioimmunotherapy (RIT) using 90Y-labeled anti-ROBO1 IgG (90Y-B5209B) achieved significant anti-tumor effects against small-cell lung cancer (SCLC) xenografts. However, subsequent tumor regrowth suggested the necessity for more effective therapy. Here, we evaluated the efficacy of combination 90Y-B5209B and cisplatin therapy in NCI-H69 SCLC xenograft mice. Mice were divided into four therapeutic groups: saline, cisplatin only, RIT only, or combination therapy. Either saline or cisplatin was administered by injection one day prior to the administration of either saline or 90Y-B5209B. Tumor volume, body weight, and blood cell counts were monitored. The pathological analysis was performed on day seven post injection of 90Y-B5209B. The survival duration of the combination therapy group was significantly longer than that of the group treated with RIT alone. No significant survival benefit was observed following the isolated administration of cisplatin (relative to saline). Pathological changes following combination therapy were more significant than those following the isolated administration of RIT. Although combination therapy was associated with an increase of several adverse effects such as weight loss and pancytopenia, these were transient. Thus, cisplatin pre-treatment can potentially enhance the efficacy of 90Y-B5209B, making it a promising therapeutic strategy for SCLC.
Collapse
Affiliation(s)
- Kentaro Fujiwara
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology (QST-NIRS), Chiba 263-8555, Japan; (K.F.); (A.B.T.); (T.H.)
| | - Keitaro Koyama
- Department of Radiology, Faculty of Medicine, International University of Health and Welfare, Chiba 286-8686, Japan; (K.K.); (T.M.)
| | - Atsushi B. Tsuji
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology (QST-NIRS), Chiba 263-8555, Japan; (K.F.); (A.B.T.); (T.H.)
| | - Hiroko Iwanari
- Department of Quantitative Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo 153-8904, Japan; (H.I.); (O.K.-A.)
| | - Osamu Kusano-Arai
- Department of Quantitative Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo 153-8904, Japan; (H.I.); (O.K.-A.)
| | - Tatsuya Higashi
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology (QST-NIRS), Chiba 263-8555, Japan; (K.F.); (A.B.T.); (T.H.)
| | - Toshimitsu Momose
- Department of Radiology, Faculty of Medicine, International University of Health and Welfare, Chiba 286-8686, Japan; (K.K.); (T.M.)
| | - Takao Hamakubo
- Department of Quantitative Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo 153-8904, Japan; (H.I.); (O.K.-A.)
- Department of Protein-protein Interaction Research, Institute for Advanced Medical Sciences, Nippon Medical School, Kanagawa 211-8533, Japan
- Correspondence: ; Tel./Fax: +81-044-733-1825
| |
Collapse
|
71
|
Pasterkamp RJ, Burk K. Axon guidance receptors: Endocytosis, trafficking and downstream signaling from endosomes. Prog Neurobiol 2020; 198:101916. [PMID: 32991957 DOI: 10.1016/j.pneurobio.2020.101916] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/06/2020] [Accepted: 09/21/2020] [Indexed: 02/06/2023]
Abstract
During the development of the nervous system, axons extend through complex environments. Growth cones at the axon tip allow axons to find and innervate their appropriate targets and form functional synapses. Axon pathfinding requires axons to respond to guidance signals and these cues need to be detected by specialized receptors followed by intracellular signal integration and translation. Several downstream signaling pathways have been identified for axon guidance receptors and it has become evident that these pathways are often initiated from intracellular vesicles called endosomes. Endosomes allow receptors to traffic intracellularly, re-locating receptors from one cellular region to another. The localization of axon guidance receptors to endosomal compartments is crucial for their function, signaling output and expression levels. For example, active receptors within endosomes can recruit downstream proteins to the endosomal membrane and facilitate signaling. Also, endosomal trafficking can re-locate receptors back to the plasma membrane to allow re-activation or mediate downregulation of receptor signaling via degradation. Accumulating evidence suggests that axon guidance receptors do not follow a pre-set default trafficking route but may change their localization within endosomes. This re-routing appears to be spatially and temporally regulated, either by expression of adaptor proteins or co-receptors. These findings shed light on how signaling in axon guidance is regulated and diversified - a mechanism which explains how a limited set of guidance cues can help to establish billions of neuronal connections. In this review, we summarize and discuss our current knowledge of axon guidance receptor trafficking and provide directions for future research.
Collapse
Affiliation(s)
- R J Pasterkamp
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, the Netherlands.
| | - K Burk
- Department of Neurology, University Medical Center Göttingen, 37075 Göttingen, Germany; Center for Biostructural Imaging of Neurodegeneration, 37075 Göttingen, Germany.
| |
Collapse
|
72
|
Isumi Y, Hayashi S, Inoue T, Yoshigae Y, Sato T, Hasegawa J, Agatsuma T. DS-7080a, a Selective Anti-ROBO4 Antibody, Shows Anti-Angiogenic Efficacy with Distinctly Different Profiles from Anti-VEGF Agents. Transl Vis Sci Technol 2020; 9:7. [PMID: 32879763 PMCID: PMC7442859 DOI: 10.1167/tvst.9.9.7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 07/06/2020] [Indexed: 11/24/2022] Open
Abstract
Purpose Neovascular age-related macular degeneration (nAMD) results from choroidal neovascularization (CNV) and causes severe vision loss. Intravitreal anti-vascular endothelial growth factor (VEGF) therapies have significantly improved therapeutic outcomes; however, a substantial number of patients experience disease progression. Roundabout 4 (ROBO4) has been reported to be a vascular-specific protein that stabilizes vasculature in ocular pathological angiogenesis. To explore ROBO4 targeting as a novel treatment against neovascularization, we generated a humanized anti-human ROBO4 antibody, DS-7080a, and evaluated its efficacy. Methods ROBO4 mRNA in human whole eye cross-sections was examined by in situ hybridization. Human umbilical vein endothelial cell (HUVEC) migration was measured in the presence of VEGF, basic fibroblast growth factor (bFGF), hepatocyte growth factor (HGF), or conditioned medium of primary human retinal pigment epithelial (HRPE) cells. CNV was induced in cynomolgus monkeys by laser irradiation. Vascular leakage was measured by fluorescein angiography, and pathological changes were determined by histology. Results ROBO4 mRNA was detected in choroidal vessels of nAMD patients. DS-7080a suppressed HGF- or bFGF-induced HUVEC migration in addition to that induced by VEGF. Further, HUVEC migration induced by HRPE-conditioned medium was inhibited by either DS-7080a or ranibizumab in a similar manner, and the combination of these showed further inhibition. In a laser-induced CNV monkey model, single intravitreous administration of 1.1 mg/eye of DS-7080a reduced the incidence of grade 4 leakage from 44.45% in control eyes to 1.85% (P < 0.05 by Dunnett's test). Conclusions Anti-ROBO4 antibody DS-7080a suppressed HUVEC migration in a distinctly different fashion from anti-VEGF agents and improved laser-induced CNV in non-human primates. Translational Relevance DS-7080a may be a novel treatment option for nAMD.
Collapse
Affiliation(s)
- Yoshitaka Isumi
- Oncology Research Laboratories I, Oncology Function, R&D Division, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Shinko Hayashi
- Oncology Research Laboratories I, Oncology Function, R&D Division, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Tatsuya Inoue
- Specialty Medicine Research Laboratories I, Research Function, R&D Division, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Yasushi Yoshigae
- Research Planning Group, Research Function, R&D Division, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Toshiyuki Sato
- Specialty Medicine Research Laboratories II, Research Function, R&D Division, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Jun Hasegawa
- Modality Research Laboratories, Biologics Division, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Toshinori Agatsuma
- Oncology Research Laboratories I, Oncology Function, R&D Division, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| |
Collapse
|
73
|
Sherchan P, Travis ZD, Tang J, Zhang JH. The potential of Slit2 as a therapeutic target for central nervous system disorders. Expert Opin Ther Targets 2020; 24:805-818. [PMID: 32378435 PMCID: PMC7529836 DOI: 10.1080/14728222.2020.1766445] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 05/05/2020] [Indexed: 10/24/2022]
Abstract
Introduction: Slit2 is an extracellular matrix protein that regulates migration of developing axons during central nervous system (CNS) development. Roundabout (Robo) receptors expressed by various cell types in the CNS, mediate intracellular signal transduction pathways for Slit2. Recent studies indicate that Slit2 plays important protective roles in a myriad of processes such as cell migration, immune response, vascular permeability, and angiogenesis in CNS pathologies. Areas covered: This review provides an overview of the diverse functions of Slit2 in CNS disorders and discusses the potential of Slit2 as a therapeutic target. We reviewed preclinical studies reporting the role of Slit2 in various CNS disease models, transgenic animal research, and rodent models that utilized Slit2 as a therapy. Expert opinion: Slit2 exerts a wide array of beneficial effects ranging from anti-migration, blood-brain barrier (BBB) protection, inhibition of peripheral immune cell infiltration, and anti-apoptosis in various disease models. However, a dual role of Slit2 in endothelial permeability has been observed in transgenic animals. Further research on Slit2 will be crucial including key issues such as effects of transgenic overexpression versus exogenous Slit2, function of Slit2 dependent on cellular expression of Robo receptors and the underlying pathology for potential clinical translation.
Collapse
Affiliation(s)
- Prativa Sherchan
- Center for Neuroscience Research, Department of Physiology and Pharmacology, Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| | - Zachary D. Travis
- Department of Earth and Biological Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, 92354, USA and Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA
- Department of Physiology and Pharmacology, Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| | - John H. Zhang
- Center for Neuroscience Research, Department of Physiology and Pharmacology, Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
- Departments of Anesthesiology, Neurosurgery and Neurology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| |
Collapse
|
74
|
Manhart A, Azevedo M, Baylies M, Mogilner A. Reverse-engineering forces responsible for dynamic clustering and spreading of multiple nuclei in developing muscle cells. Mol Biol Cell 2020; 31:1802-1814. [PMID: 32129712 PMCID: PMC7521854 DOI: 10.1091/mbc.e19-12-0711] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
How cells position their organelles is a fundamental biological question. During Drosophila embryonic muscle development, multiple nuclei transition from being clustered together to splitting into two smaller clusters to spreading along the myotube’s length. Perturbations of microtubules and motor proteins disrupt this sequence of events. These perturbations do not allow intuiting which molecular forces govern the nuclear positioning; we therefore used computational screening to reverse-engineer and identify these forces. The screen reveals three models. Two suggest that the initial clustering is due to nuclear repulsion from the cell poles, while the third, most robust, model poses that this clustering is due to a short-ranged internuclear attraction. All three models suggest that the nuclear spreading is due to long-ranged internuclear repulsion. We test the robust model quantitatively by comparing it with data from perturbed muscle cells. We also test the model using agent-based simulations with elastic dynamic microtubules and molecular motors. The model predicts that, in longer mammalian myotubes with a large number of nuclei, the spreading stage would be preceded by segregation of the nuclei into a large number of clusters, proportional to the myotube length, with a small average number of nuclei per cluster.
Collapse
Affiliation(s)
- Angelika Manhart
- Mathematics Department, University College London, London WC1H 0AY, UK
| | - Mafalda Azevedo
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065.,Graduate Program in Areas of Basic and Applied Biology (GABBA), Abel Salazar Biomedical Sciences Institute, University of Porto, 4050 Porto, Portugal
| | - Mary Baylies
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Alex Mogilner
- Courant Institute for Mathematical Sciences and Department of Biology, New York University, New York, NY 10012
| |
Collapse
|
75
|
Kulus M, Kranc W, Jeseta M, Sujka-Kordowska P, Konwerska A, Ciesiółka S, Celichowski P, Moncrieff L, Kocherova I, Józkowiak M, Kulus J, Wieczorkiewicz M, Piotrowska-Kempisty H, Skowroński MT, Bukowska D, Machatkova M, Hanulakova S, Mozdziak P, Jaśkowski JM, Kempisty B, Antosik P. Cortical Granule Distribution and Expression Pattern of Genes Regulating Cellular Component Size, Morphogenesis, and Potential to Differentiation are Related to Oocyte Developmental Competence and Maturational Capacity In Vivo and In Vitro. Genes (Basel) 2020; 11:genes11070815. [PMID: 32708880 PMCID: PMC7397037 DOI: 10.3390/genes11070815] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 07/10/2020] [Accepted: 07/13/2020] [Indexed: 12/13/2022] Open
Abstract
Polyspermia is an adverse phenomenon during mammalian fertilization when more than one sperm fuses with a single oocyte. The egg cell is prepared to prevent polyspermia by, among other ways, producing cortical granules (CGs), which are specialized intracellular structures containing enzymes that aim to harden the zona pellucida and block the fusion of subsequent sperm. This work focused on exploring the expression profile of genes that may be associated with cortical reactions, and evaluated the distribution of CGs in immature oocytes and the peripheral density of CGs in mature oocytes. Oocytes were isolated and then processed for in vitro maturation (IVM). Transcriptomic analysis of genes belonging to five ontological groups has been conducted. Six genes showed increased expression after IVM (ARHGEF2, MAP1B, CXCL12, FN1, DAB2, and SOX9), while the majority of genes decreased expression after IVM. Using CG distribution analysis in immature oocytes, movement towards the cortical zone of the oocyte during meiotic competence acquisition was observed. CGs peripheral density decreased with the rise in meiotic competence during the IVM process. The current results reveal important new insights into the in vitro maturation of oocytes. Our results may serve as a basis for further studies to investigate the cortical reaction of oocytes.
Collapse
Affiliation(s)
- Magdalena Kulus
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (M.K.); (P.A.)
| | - Wiesława Kranc
- Department of Anatomy, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (W.K.); (I.K.)
| | - Michal Jeseta
- Department of Obstetrics and Gynecology, University Hospital and Masaryk University, 602 00 Brno, Czech Republic;
- Department of Veterinary Sciences, Czech University of Life Sciences in Prague, 165 00 Prague, Czech Republic
| | - Patrycja Sujka-Kordowska
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (P.S.-K.); (A.K.); (S.C.); (P.C.); (L.M.)
- Department of Anatomy and Histology, University of Zielona Gora, 65-046 Zielona Gora, Poland
| | - Aneta Konwerska
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (P.S.-K.); (A.K.); (S.C.); (P.C.); (L.M.)
| | - Sylwia Ciesiółka
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (P.S.-K.); (A.K.); (S.C.); (P.C.); (L.M.)
| | - Piotr Celichowski
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (P.S.-K.); (A.K.); (S.C.); (P.C.); (L.M.)
| | - Lisa Moncrieff
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (P.S.-K.); (A.K.); (S.C.); (P.C.); (L.M.)
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Ievgeniia Kocherova
- Department of Anatomy, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (W.K.); (I.K.)
| | - Małgorzata Józkowiak
- Department of Toxicology, Poznan University of Medical Sciences, 60-631 Poznan, Poland; (M.J.); (H.P.-K.)
| | - Jakub Kulus
- Department of Diagnostics and Clinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (J.K.); (D.B.); (J.M.J.)
| | - Maria Wieczorkiewicz
- Department of Basic and Preclinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (M.W.); (M.T.S.)
| | - Hanna Piotrowska-Kempisty
- Department of Toxicology, Poznan University of Medical Sciences, 60-631 Poznan, Poland; (M.J.); (H.P.-K.)
| | - Mariusz T. Skowroński
- Department of Basic and Preclinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (M.W.); (M.T.S.)
| | - Dorota Bukowska
- Department of Diagnostics and Clinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (J.K.); (D.B.); (J.M.J.)
| | - Marie Machatkova
- Veterinary Research Institute, 621 00 Brno, Czech Republic; (M.M.); (S.H.)
| | - Sarka Hanulakova
- Veterinary Research Institute, 621 00 Brno, Czech Republic; (M.M.); (S.H.)
| | - Paul Mozdziak
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC 27695, USA;
| | - Jędrzej M. Jaśkowski
- Department of Diagnostics and Clinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (J.K.); (D.B.); (J.M.J.)
| | - Bartosz Kempisty
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (M.K.); (P.A.)
- Department of Anatomy, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (W.K.); (I.K.)
- Department of Obstetrics and Gynecology, University Hospital and Masaryk University, 602 00 Brno, Czech Republic;
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (P.S.-K.); (A.K.); (S.C.); (P.C.); (L.M.)
- Correspondence: ; Tel.: +48-61-854-6418
| | - Paweł Antosik
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (M.K.); (P.A.)
| |
Collapse
|
76
|
Vaid S, Huttner WB. Transcriptional Regulators and Human-Specific/Primate-Specific Genes in Neocortical Neurogenesis. Int J Mol Sci 2020; 21:ijms21134614. [PMID: 32610533 PMCID: PMC7369782 DOI: 10.3390/ijms21134614] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/09/2020] [Accepted: 06/26/2020] [Indexed: 12/15/2022] Open
Abstract
During development, starting from a pool of pluripotent stem cells, tissue-specific genetic programs help to shape and develop functional organs. To understand the development of an organ and its disorders, it is important to understand the spatio-temporal dynamics of the gene expression profiles that occur during its development. Modifications in existing genes, the de-novo appearance of new genes, or, occasionally, even the loss of genes, can greatly affect the gene expression profile of any given tissue and contribute to the evolution of organs or of parts of organs. The neocortex is evolutionarily the most recent part of the brain, it is unique to mammals, and is the seat of our higher cognitive abilities. Progenitors that give rise to this tissue undergo sequential waves of differentiation to produce the complete sets of neurons and glial cells that make up a functional neocortex. We will review herein our understanding of the transcriptional regulators that control the neural precursor cells (NPCs) during the generation of the most abundant class of neocortical neurons, the glutametergic neurons. In addition, we will discuss the roles of recently-identified human- and primate-specific genes in promoting neurogenesis, leading to neocortical expansion.
Collapse
|
77
|
Ding C, Li Y, Xing C, Zhang H, Wang S, Dai M. Research Progress on Slit/Robo Pathway in Pancreatic Cancer: Emerging and Promising. JOURNAL OF ONCOLOGY 2020; 2020:2845906. [PMID: 32670371 PMCID: PMC7341381 DOI: 10.1155/2020/2845906] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 12/12/2022]
Abstract
Pancreatic cancer is a highly malignant digestive system tumor which is the leading cause of cancer-related deaths. The basic and clinical research of pancreatic cancer has made great progress in recent years, and kinds of signaling pathways have been found in the tumorigenesis and progression in pancreatic cancer. The Slit glycoprotein (Slit) and Roundabout receptor (Robo) signaling pathway acts as a neural targeting factor with the axonal remnant, axon guidance, and inhibition of neuronal migration in the nervous system. In recent years, it has been found that the Slit/Robo signaling pathway has different degrees of expression changes in various tumor cells. In different tumor cells, the signaling pathway gene expression is different and regulates tumor angiogenesis, cell invasion, metastasis, and nerve infiltration. Herein, we summarize the mechanisms of the Slit/Robo pathway in the development and progression of pancreatic cancer, in order to have more understanding of the role of Slit/Robo in pancreatic cancer.
Collapse
Affiliation(s)
- Cheng Ding
- Department of General Surgery, Peking Union Medical College Hospital (PUMCH), Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730, China
- National Translational Medicine of China, Beijing 100730, China
| | - Yatong Li
- National Translational Medicine of China, Beijing 100730, China
| | - Cheng Xing
- National Translational Medicine of China, Beijing 100730, China
| | - Hanyu Zhang
- National Translational Medicine of China, Beijing 100730, China
| | - Shunda Wang
- National Translational Medicine of China, Beijing 100730, China
| | - Menghua Dai
- Department of General Surgery, Peking Union Medical College Hospital (PUMCH), Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730, China
| |
Collapse
|
78
|
Sasaki T, Komatsu Y, Yamamori T. Expression patterns of SLIT/ROBO mRNAs reveal a characteristic feature in the entorhinal-hippocampal area of macaque monkeys. BMC Res Notes 2020; 13:262. [PMID: 32460877 PMCID: PMC7251749 DOI: 10.1186/s13104-020-05100-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 05/19/2020] [Indexed: 01/06/2023] Open
Abstract
OBJECTIVE SLITs are secreted glycoproteins that bind to Roundabouts (ROBOs) which are a family member of transmembrane receptors. SLIT signaling has well-conserved roles in mediating axon repulsion in a developing nervous system. We previously reported that SLIT1 mRNA is enriched in middle layers of the prefrontal cortex of macaque monkeys in a developmentally regulated manner. Other SLIT (SLIT2 and SLIT3) mRNAs showed preferential expressions in the prefrontal cortex with a distinct laminar pattern. To obtain further clues to the role of SLIT signaling in the organization of the primate brain, we performed ISH analysis of SLIT and ROBO mRNAs using adult macaque brain tissues. RESULTS In this study, we examined the expression patterns of SLITs and ROBOs (ROBO1 and ROBO2) in other brain regions, and found intense and characteristic expression patterns of these genes in the entorhinal-hippocampal area. In situ hybridization analysis revealed that SLIT1 and SLIT2 mRNAs showed marked complementary distribution in the entorhinal cortex. SLIT and ROBO mRNAs were widely expressed in the hippocampus with modest regional preference. These findings suggest that each SLIT gene has a specialized role that is particularly important for prefrontal as well as hippocampal connectivity in the primate cortex.
Collapse
Affiliation(s)
- Tetsuya Sasaki
- Division of Brain Biology, National Institute for Basic Biology, 38 Nishigonaka Myodaiji, Okazaki, 444-8585, Japan.
- Department of Anatomy and Neuroscience, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan.
- Ph.D Program of Neurosciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan.
| | - Yusuke Komatsu
- Division of Brain Biology, National Institute for Basic Biology, 38 Nishigonaka Myodaiji, Okazaki, 444-8585, Japan
- Laboratory of Veterinary Hygiene, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, 060-0816, Japan
| | - Tetsuo Yamamori
- Division of Brain Biology, National Institute for Basic Biology, 38 Nishigonaka Myodaiji, Okazaki, 444-8585, Japan.
- Laboratory for Molecular Analysis of Higher Brain Function, RIKEN Center for Brain Science, Wako, Saitama, 351-0198, Japan.
| |
Collapse
|
79
|
Price KM, Wigg KG, Feng Y, Blokland K, Wilkinson M, He G, Kerr EN, Carter TC, Guger SL, Lovett MW, Strug LJ, Barr CL. Genome-wide association study of word reading: Overlap with risk genes for neurodevelopmental disorders. GENES BRAIN AND BEHAVIOR 2020; 19:e12648. [PMID: 32108986 DOI: 10.1111/gbb.12648] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 01/28/2020] [Accepted: 02/24/2020] [Indexed: 12/12/2022]
Abstract
Reading disabilities (RD) are the most common neurocognitive disorder, affecting 5% to 17% of children in North America. These children often have comorbid neurodevelopmental/psychiatric disorders, such as attention deficit/hyperactivity disorder (ADHD). The genetics of RD and their overlap with other disorders is incompletely understood. To contribute to this, we performed a genome-wide association study (GWAS) for word reading. Then, using summary statistics from neurodevelopmental/psychiatric disorders, we computed polygenic risk scores (PRS) and used them to predict reading ability in our samples. This enabled us to test the shared aetiology between RD and other disorders. The GWAS consisted of 5.3 million single nucleotide polymorphisms (SNPs) and two samples; a family-based sample recruited for reading difficulties in Toronto (n = 624) and a population-based sample recruited in Philadelphia [Philadelphia Neurodevelopmental Cohort (PNC)] (n = 4430). The Toronto sample SNP-based analysis identified suggestive SNPs (P ~ 5 × 10-7 ) in the ARHGAP23 gene, which is implicated in neuronal migration/axon pathfinding. The PNC gene-based analysis identified significant associations (P < 2.72 × 10-6 ) for LINC00935 and CCNT1, located in the region of the KANSL2/CCNT1/LINC00935/SNORA2B/SNORA34/MIR4701/ADCY6 genes on chromosome 12q, with near significant SNP-based analysis. PRS identified significant overlap between word reading and intelligence (R2 = 0.18, P = 7.25 × 10-181 ), word reading and educational attainment (R2 = 0.07, P = 4.91 × 10-48 ) and word reading and ADHD (R2 = 0.02, P = 8.70 × 10-6 ; threshold for significance = 7.14 × 10-3 ). Overlap was also found between RD and autism spectrum disorder (ASD) as top-ranked genes were previously implicated in autism by rare and copy number variant analyses. These findings support shared risk between word reading, cognitive measures, educational outcomes and neurodevelopmental disorders, including ASD.
Collapse
Affiliation(s)
- Kaitlyn M Price
- Genetics and Development Division, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Program in Neuroscience and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Karen G Wigg
- Genetics and Development Division, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Yu Feng
- Genetics and Development Division, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Kirsten Blokland
- Program in Neuroscience and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Margaret Wilkinson
- Program in Neuroscience and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Gengming He
- Genetics and Genome Biology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Elizabeth N Kerr
- Department of Psychology, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Tasha-Cate Carter
- Program in Neuroscience and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada.,Holland Bloorview Rehabilitation Hospital, Toronto, Ontario, Canada
| | - Sharon L Guger
- Department of Psychology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Maureen W Lovett
- Program in Neuroscience and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Lisa J Strug
- Genetics and Genome Biology, Hospital for Sick Children, Toronto, Ontario, Canada.,Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Cathy L Barr
- Genetics and Development Division, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Program in Neuroscience and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
80
|
Saponin Facilitates Anti-Robo1 Immunotoxin Cytotoxic Effects on Maxillary Sinus Squamous Cell Carcinoma. JOURNAL OF ONCOLOGY 2020; 2020:9593516. [PMID: 32256588 PMCID: PMC7086449 DOI: 10.1155/2020/9593516] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 12/08/2019] [Accepted: 01/18/2020] [Indexed: 01/06/2023]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is one of the most common cancers worldwide. The standard treatment of surgery, chemotherapy, and radiotherapy can result in long-term complications which lower the patient's quality of life, such as eating disorders, speech problems, and disfiguring or otherwise untoward cosmetic issues. Antibody therapy against cancer-specific antigens is advantageous in terms of its lesser side effects achieved by its greater specificity, though the antitumor activity is still usually not enough to obtain a complete cure. Robo1, an axon guidance receptor, has received considerable attention as a possible drug target in various cancers. We have shown previously the enhanced cytotoxic effects of saporin-conjugated anti-Robo1 immunotoxin (IT-Robo1) on the HNSCC cell line HSQ-89 in combination with a photochemical internalization technique. Considering the light source, which has only limited tissue penetrance, we examined the drug internalization effect of saponin. Treatment with saponin facilitated significant cytotoxic effects of IT-Robo1 on HSQ-89 cells. Saponin exerts its own nonspecific cytotoxicity, which may cover the actual extent of the internalization effect. We thus examined whether a flashed treatment with saponin exerted a significant specific cytotoxic effect on cancer cells. The combination of an immunotoxin with saponin also exhibited a significant tumor-suppressive effect on mice HSQ-19 xenografts. These results suggest the utility of saponin treatment as an enhancer of immunotoxin treatment in cancer.
Collapse
|
81
|
Abstract
The spinal cord receives, relays and processes sensory information from the periphery and integrates this information with descending inputs from supraspinal centres to elicit precise and appropriate behavioural responses and orchestrate body movements. Understanding how the spinal cord circuits that achieve this integration are wired during development is the focus of much research interest. Several families of proteins have well-established roles in guiding developing spinal cord axons, and recent findings have identified new axon guidance molecules. Nevertheless, an integrated view of spinal cord network development is lacking, and many current models have neglected the cellular and functional diversity of spinal cord circuits. Recent advances challenge the existing spinal cord axon guidance dogmas and have provided a more complex, but more faithful, picture of the ontogenesis of vertebrate spinal cord circuits.
Collapse
|
82
|
Niimi T. Roles of Slit Ligands and Their Roundabout (Robo) Family of Receptors in Bone Remodeling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 21:143-154. [PMID: 32986130 DOI: 10.1007/5584_2020_586] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Slit guidance ligands (Slits) and their roundabout (Robo) family of receptors are well-known axon guidance molecules that were originally identified in Drosophila mutants with commissural axon pathfinding defects. However, Slit-Robo signaling has been shown to be involved in not only neurogenesis, but also the development of other organs such as the kidney and heart. Recently, it was also revealed that Slit-Robo signaling plays an important role in bone metabolism. For example, osteoclast-derived Slit3 plays an osteoprotective role by synchronously stimulating bone formation by osteoblasts and suppressing bone resorption by osteoclasts through Robo receptors expressed on osteoblastic and osteoclastic cell lineages, making it a potential therapeutic target for metabolic bone disorders. Furthermore, osteoblast-derived Slit3 promotes bone formation indirectly as a proangiogenic factor. This review summarizes the recent progress on defining the roles of the Slit-Robo signaling in bone metabolism, and discusses the possible roles of the interaction between Robo and neural epidermal growth factor-like (NEL)-like (NELL) proteins that are novel ligands for Robo receptors.
Collapse
Affiliation(s)
- Tomoaki Niimi
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan.
| |
Collapse
|
83
|
Sato K, Goto J, Yamamoto D. Sex Mysteries of the Fly Courtship Master Regulator Fruitless. Front Behav Neurosci 2019; 13:245. [PMID: 31680899 PMCID: PMC6813181 DOI: 10.3389/fnbeh.2019.00245] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 10/07/2019] [Indexed: 01/21/2023] Open
Abstract
The fruitless (fru) gene of Drosophila melanogaster generates two groups of protein products, the male-specific FruM proteins and non-sex-specific FruCOM proteins. The FruM proteins have a 101 amino acids (a.a.)-long extension at the N-terminus which is absent from FruCOM. We suggest that this N-terminal extension might confer male-specific roles on FruM interaction partner proteins such as Lola, which otherwise operates as a transcription factor common to both sexes. FruM-expressing neurons are known to connect with other neurons to form a sexually dimorphic circuit for male mating behavior. We propose that FruM proteins expressed in two synaptic partners specify, at the transcriptional level, signaling pathways through which select pre- and post-synaptic partners communicate, and thereby pleiotropic ligand-receptor pairs for cell-cell interactions acquire the high specificity for mutual connections between two FruM-positive cells. We further discuss the possibility that synaptic connections made by FruM-positive neurons are regulated by neural activities, which in turn upregulate Fru expression in active cells, resulting in feedforward enhancement of courtship activities of the male fly.
Collapse
Affiliation(s)
- Kosei Sato
- Neuro-Network Evolution Project, Advanced ICT Research Institute, National Institute of Information and Communications Technology, Kobe, Japan
| | - Junpei Goto
- Division of Neurogenetics, Tohoku University Graduate School of Life Sciences, Sendai, Japan
| | - Daisuke Yamamoto
- Neuro-Network Evolution Project, Advanced ICT Research Institute, National Institute of Information and Communications Technology, Kobe, Japan
| |
Collapse
|
84
|
Gonsior M, Ismat A. sli is required for proper morphology and migration of sensory neurons in the Drosophila PNS. Neural Dev 2019; 14:10. [PMID: 31651354 PMCID: PMC6813078 DOI: 10.1186/s13064-019-0135-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 10/17/2019] [Indexed: 11/12/2022] Open
Abstract
Neurons and glial cells coordinate with each other in many different aspects of nervous system development. Both types of cells are receiving multiple guidance cues to guide the neurons and glial cells to their proper final position. The lateral chordotonal organs (lch5) of the Drosophila peripheral nervous system (PNS) are composed of five sensory neurons surrounded by four different glial cells, scolopale cells, cap cells, attachment cells and ligament cells. During embryogenesis, the lch5 neurons go through a rotation and ventral migration to reach their final position in the lateral region of the abdomen. We show here that the extracellular ligand sli is required for the proper ventral migration and morphology of the lch5 neurons. We further show that mutations in the Sli receptors Robo and Robo2 also display similar defects as loss of sli, suggesting a role for Slit-Robo signaling in lch5 migration and positioning. Additionally, we demonstrate that the scolopale, cap and attachment cells follow the mis-migrated lch5 neurons in sli mutants, while the ventral stretching of the ligament cells seems to be independent of the lch5 neurons. This study sheds light on the role of Slit-Robo signaling in sensory neuron development.
Collapse
Affiliation(s)
- Madison Gonsior
- Department of Biology, University of St. Thomas, Saint Paul, MN, 55104, USA
| | - Afshan Ismat
- Department of Biology, University of St. Thomas, Saint Paul, MN, 55104, USA.
| |
Collapse
|
85
|
Koohini Z, Koohini Z, Teimourian S. Slit/Robo Signaling Pathway in Cancer; a New Stand Point for Cancer Treatment. Pathol Oncol Res 2019; 25:1285-1293. [PMID: 30610466 DOI: 10.1007/s12253-018-00568-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 12/17/2018] [Indexed: 10/27/2022]
Abstract
Angiogenesis and metastasis are two critical steps for cancer cells survival and migration. The microenvironment of tumor sphere induces new blood vessels formation for enhancing tumor mass. Preexisting capillaries and postcapillary venules in tumors bring about new blood vessels. ROBO1-ROBO4 are transmembrane receptors family which act as guidance molecules of the nervous system. The SLITs family is secreted glycoproteins that bind to these receptors. SLIT-ROBO signaling pathway plays an important role in neurogenesis and immune response. Linkage between ROBOs and their ligands (SLITs) induce chemorepllent signal for regulation of axon guidance and leukocyte cell migration, recent finding shows that it is also involved in endothelial cell migration and angiogenesis in various type of cancers. In this article we review recent finding of SLIT-ROBO pathway in angiogenesis and metastasis.
Collapse
Affiliation(s)
- Zahra Koohini
- Department of Medical Genetics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zohreh Koohini
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shahram Teimourian
- Department of Medical Genetics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
86
|
Nisha, Aggarwal P, Sarkar S. Adequate expression of Globin1 is required for development and maintenance of nervous system in Drosophila. Mol Cell Neurosci 2019; 100:103398. [DOI: 10.1016/j.mcn.2019.103398] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 08/07/2019] [Accepted: 08/25/2019] [Indexed: 10/26/2022] Open
|
87
|
Comer JD, Alvarez S, Butler SJ, Kaltschmidt JA. Commissural axon guidance in the developing spinal cord: from Cajal to the present day. Neural Dev 2019; 14:9. [PMID: 31514748 PMCID: PMC6739980 DOI: 10.1186/s13064-019-0133-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Accepted: 08/23/2019] [Indexed: 12/11/2022] Open
Abstract
During neuronal development, the formation of neural circuits requires developing axons to traverse a diverse cellular and molecular environment to establish synaptic contacts with the appropriate postsynaptic partners. Essential to this process is the ability of developing axons to navigate guidance molecules presented by specialized populations of cells. These cells partition the distance traveled by growing axons into shorter intervals by serving as intermediate targets, orchestrating the arrival and departure of axons by providing attractive and repulsive guidance cues. The floor plate in the central nervous system (CNS) is a critical intermediate target during neuronal development, required for the extension of commissural axons across the ventral midline. In this review, we begin by giving a historical overview of the ventral commissure and the evolutionary purpose of decussation. We then review the axon guidance studies that have revealed a diverse assortment of midline guidance cues, as well as genetic and molecular regulatory mechanisms required for coordinating the commissural axon response to these cues. Finally, we examine the contribution of dysfunctional axon guidance to neurological diseases.
Collapse
Affiliation(s)
- J D Comer
- Neuroscience Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA.,Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA.,Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY, USA
| | - S Alvarez
- Department of Neurobiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA.,Molecular Biology Interdepartmental Graduate Program, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - S J Butler
- Department of Neurobiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - J A Kaltschmidt
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
88
|
Sato K, Yamamoto D. The mode of action of Fruitless: Is it an easy matter to switch the sex? GENES BRAIN AND BEHAVIOR 2019; 19:e12606. [PMID: 31420927 PMCID: PMC7027472 DOI: 10.1111/gbb.12606] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/13/2019] [Accepted: 08/14/2019] [Indexed: 11/28/2022]
Abstract
The fruitless (fru) locus was originally defined by a male sterile mutation that promotes male-to-male courtship while suppressing male-to-female courtship in Drosophila melanogaster. The fru promoter-1 pre-RNA generates a set of BTB-zinc finger family FruM proteins expressed exclusively in the male neurons, leading to the formation of sexual dimorphisms in neurons via male-specific neuroblast proliferation, male-specific neural survival, male-specific neuritegenesis or male-specific arbor patterning. Such a wide spectrum of phenotypic effects seems to result from chromatin modifications, in which FruBM recruits Bonus, Histone deacetylase 1 (HDAC1) and/or Heterochromatin protein 1a (HP1a) to ~130 target sites. One established FruBM transcriptional target is the axon guidance protein gene robo1. Multiple transcriptional regulator-binding sites are nested around the FruBM-binding site, and mediate sophisticated modulation of the repressor activity of FruBM. FruBM also binds to the Lola-Q transcriptional repressor to protect it from proteasome-dependent degradation in male but not female neurons as FruBM exists only in male neurons, leading to the formation of sexually dimorphic neural structures. These findings shed light on the multilayered network of transcription regulation orchestrated by the master regulator FruBM.
Collapse
Affiliation(s)
- Kosei Sato
- Neuro-Network Evolution Project, Advanced ICT Research Institute, National Institute of Information and Communications Technology, Kobe, Japan
| | - Daisuke Yamamoto
- Neuro-Network Evolution Project, Advanced ICT Research Institute, National Institute of Information and Communications Technology, Kobe, Japan
| |
Collapse
|
89
|
Lee YJ, Ch'ng TH. RIP at the Synapse and the Role of Intracellular Domains in Neurons. Neuromolecular Med 2019; 22:1-24. [PMID: 31346933 DOI: 10.1007/s12017-019-08556-4] [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] [Received: 05/30/2019] [Accepted: 07/12/2019] [Indexed: 12/18/2022]
Abstract
Regulated intramembrane proteolysis (RIP) occurs in a cell when transmembrane proteins are cleaved by intramembrane proteases such as secretases to generate soluble protein fragments in the extracellular environment and the cytosol. In the cytosol, these soluble intracellular domains (ICDs) have local functions near the site of cleavage or in many cases, translocate to the nucleus to modulate gene expression. While the mechanism of RIP is relatively well studied, the fate and function of ICDs for most substrate proteins remain poorly characterized. In neurons, RIP occurs in various subcellular compartments including at the synapse. In this review, we summarize current research on RIP in neurons, focusing specifically on synaptic proteins where the presence and function of the ICDs have been reported. We also briefly discuss activity-driven processing of RIP substrates at the synapse and the cellular machinery that support long-distance transport of ICDs from the synapse to the nucleus. Finally, we describe future challenges in this field of research in the context of understanding the contribution of ICDs in neuronal function.
Collapse
Affiliation(s)
- Yan Jun Lee
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Science Building, 11 Mandalay Road, 10-01-01 M, Singapore, 308232, Singapore.,Interdisciplinary Graduate School (IGS), Nanyang Technological University, Singapore, Singapore
| | - Toh Hean Ch'ng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Science Building, 11 Mandalay Road, 10-01-01 M, Singapore, 308232, Singapore. .,School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
90
|
Regulatory mechanisms of Robo4 and their effects on angiogenesis. Biosci Rep 2019; 39:BSR20190513. [PMID: 31160487 PMCID: PMC6620384 DOI: 10.1042/bsr20190513] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 05/30/2019] [Accepted: 05/31/2019] [Indexed: 12/13/2022] Open
Abstract
Roundabout4 (Robo4) is a transmembrane receptor that belongs to the Roundabout (Robo) family of axon guidance molecules. Robo4 is an endothelial-specific receptor that participates in endothelial cell migration, proliferation, and angiogenesis and the maintenance of vasculature homeostasis. The purpose of this review is to summarize and analyze three main mechanisms related to the expression and function of Robo4 during developmental and pathological angiogenesis. In this review, static shear stress and the binding of transcription factors such as E26 transformation-specific variant 2 (ETV2) and Slit3 induce Robo4 expression and activate Robo4 during tissue and organ development. Robo4 interacts with Slit2 or UNC5B to maintain vascular integrity, while a disturbed flow and the expression of transcription factors in inflammatory or neoplastic environments alter Robo4 expression levels, although these changes have uncertain functions. Based on the mechanisms described above, we discuss the aberrant expression of Robo4 in angiogenesis-related diseases and propose antiangiogenic therapies targeting the Robo4 signaling pathway for the treatment of ocular neovascularization lesions and tumors. Finally, although many problems related to Robo4 signaling pathways remain to be resolved, Robo4 is a promising and potentially valuable therapeutic target for treating pathological angiogenesis and developmental defects in angiogenesis.
Collapse
|
91
|
Inactivation of Lgi1 in murine neuronal precursor cells leads to dysregulation of axon guidance pathways. Genomics 2019; 112:1167-1172. [PMID: 31276752 DOI: 10.1016/j.ygeno.2019.07.001] [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: 01/09/2019] [Revised: 05/22/2019] [Accepted: 07/01/2019] [Indexed: 11/20/2022]
Abstract
LGI1 mutations predispose to a rare epilepsy syndrome and when inactivated in mice leads to early onset seizures and premature death. Histopathology of the mature brain soon after birth shows cortical dysplasia in Lgi1 null mice with hypercellularity in the outer cortical layers. Here we show extensive gene expression changes in neuronal precursor cells from Lgi1 null mice compared with wild type mice. The most significantly dysregulated pathway involves canonical axon guidance signaling with multiple networks involved in cell movement, adhesion and invasion related to actin cytoskeleton reorganization. The Lgi1 null NPCs show increased cell motility in vitro compared with normal counterparts. Dysregulation of genes critical to cell movement/migration and critical transcription factors involved in early neuronal development is a prominent feature. These studies provide a critical mechanistic link to the observation of increased cellularity in the outer layers of the developing cortex in Lgi1 null mice.
Collapse
|
92
|
Coelho DS, Moreno E. Emerging links between cell competition and Alzheimer's disease. J Cell Sci 2019; 132:132/13/jcs231258. [PMID: 31263078 DOI: 10.1242/jcs.231258] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Alzheimer's disease (AD) causes a progressive loss of memory and other cognitive functions, which inexorably debilitates patients. There is still no cure for AD and effective treatments to delay or revert AD are urgently needed. On a molecular level, the excessive accumulation of amyloid-β (Aβ) peptides triggers a complex cascade of pathological events underlying neuronal death, whose details are not yet completely understood. Our laboratory recently discovered that cell competition may play a protective role against AD by eliminating less fit neurons from the brain of Aβ-transgenic flies. Loss of Aβ-damaged neurons through fitness comparison with healthy counterparts is beneficial for the organism, delaying cognitive decline and motor disability. In this Review, we introduce the molecular mechanisms of cell competition, including seminal works on the field and latest advances regarding genetic triggers and effectors of cell elimination. We then describe the biological relevance of competition in the nervous system and discuss how competitive interactions between neurons may arise and be exacerbated in the context of AD. Selection of neurons through fitness comparison is a promising, but still emerging, research field that may open new avenues for the treatment of neurological disorders.
Collapse
Affiliation(s)
- Dina S Coelho
- Cell Fitness Laboratory, Champalimaud Centre for the Unknown, Av. Brasília., 1400-038 Lisbon, Portugal
| | - Eduardo Moreno
- Cell Fitness Laboratory, Champalimaud Centre for the Unknown, Av. Brasília., 1400-038 Lisbon, Portugal
| |
Collapse
|
93
|
Rutledge EA, Parvez RK, Short KM, Smyth IM, McMahon AP. Morphogenesis of the kidney and lung requires branch-tip directed activity of the Adamts18 metalloprotease. Dev Biol 2019; 454:156-169. [PMID: 31242448 DOI: 10.1016/j.ydbio.2019.06.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 06/18/2019] [Accepted: 06/18/2019] [Indexed: 12/22/2022]
Abstract
Adamts18 encodes a secreted metalloprotease restricted to branch-tip progenitor pools directing the morphogenesis of multiple mammalian organs. Adamts18 was targeted to explore a potential role in branching morphogenesis. In the kidney, an arborized collecting system develops through extensive branching morphogenesis of an initial epithelial outgrowth of the mesonephric duct, the ureteric bud. Adamts18 mutants displayed a weakly penetrant phenotype: duplicated ureteric outgrowths forming enlarged, bi-lobed kidneys with an increased nephron endowment. In contrast, Adamts18 mutants showed a fully penetrant lung phenotype: epithelial growth was markedly reduced and early secondary branching scaled to the reduced length of the primary airways. Furthermore, there was a pronounced delay in the appearance of differentiated cell types in both proximal and distally positions of the developing airways. Adamts18 is closely related to Adamts16. In the kidney but not the lung, broad epithelial Adamts16 expression overlaps Adamts18 in branch tips. However, compound Adamts16/18 mutants displayed a comparable low penetrance duplicated ureteric phenotype, ruling out a possible role for Adamts16 as a functional modifier of the Adamts18 kidney phenotype. Given the predicted action of secreted Adamts18 metalloprotease, and broad expression of Adamts18 in branching organ systems, these findings suggest distinct requirements for matrix modelling in the morphogenesis of epithelial networks.
Collapse
Affiliation(s)
- Elisabeth A Rutledge
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, W.M. Keck School of Medicine of the University of Southern California, CA, 90089, USA
| | - Riana K Parvez
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, W.M. Keck School of Medicine of the University of Southern California, CA, 90089, USA
| | - Kieran M Short
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 3800, Australia; Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Ian M Smyth
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 3800, Australia; Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia; Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, W.M. Keck School of Medicine of the University of Southern California, CA, 90089, USA.
| |
Collapse
|
94
|
Anbalagan S, Blechman J, Gliksberg M, Gordon L, Rotkopf R, Dadosh T, Shimoni E, Levkowitz G. Robo2 regulates synaptic oxytocin content by affecting actin dynamics. eLife 2019; 8:45650. [PMID: 31180321 PMCID: PMC6590984 DOI: 10.7554/elife.45650] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 06/08/2019] [Indexed: 12/28/2022] Open
Abstract
The regulation of neuropeptide level at the site of release is essential for proper neurophysiological functions. We focused on a prominent neuropeptide, oxytocin (OXT) in the zebrafish as an in vivo model to visualize and quantify OXT content at the resolution of a single synapse. We found that OXT-loaded synapses were enriched with polymerized actin. Perturbation of actin filaments by either cytochalasin-D or conditional Cofilin expression resulted in decreased synaptic OXT levels. Genetic loss of robo2 or slit3 displayed decreased synaptic OXT content and robo2 mutants displayed reduced mobility of the actin probe Lifeact-EGFP in OXT synapses. Using a novel transgenic reporter allowing real-time monitoring of OXT-loaded vesicles, we show that robo2 mutants display slower rate of vesicles accumulation. OXT-specific expression of dominant-negative Cdc42, which is a key regulator of actin dynamics and a downstream effector of Robo2, led to a dose-dependent increase in OXT content in WT, and a dampened effect in robo2 mutants. Our results link Slit3-Robo2-Cdc42, which controls local actin dynamics, with the maintenance of synaptic neuropeptide levels.
Collapse
Affiliation(s)
- Savani Anbalagan
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Janna Blechman
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Michael Gliksberg
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ludmila Gordon
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ron Rotkopf
- Bioinformatics Unit, LSCF, Weizmann Institute of Science, Rehovot, Israel.,Electron Microscopy Unit, Weizmann Institute of Science, Rehovot, Israel
| | - Tali Dadosh
- Department of Chemical Research Support, Weizmann Institute of Science, Rehovot, Israel
| | - Eyal Shimoni
- Department of Chemical Research Support, Weizmann Institute of Science, Rehovot, Israel
| | - Gil Levkowitz
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
95
|
Jiang Z, Liang G, Xiao Y, Qin T, Chen X, Wu E, Ma Q, Wang Z. Targeting the SLIT/ROBO pathway in tumor progression: molecular mechanisms and therapeutic perspectives. Ther Adv Med Oncol 2019; 11:1758835919855238. [PMID: 31217826 PMCID: PMC6557020 DOI: 10.1177/1758835919855238] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 05/07/2019] [Indexed: 01/14/2023] Open
Abstract
The SLITs (SLIT1, SLIT2, and SLIT3) are a family of secreted proteins that mediate positional interactions between cells and their environment during development by signaling through ROBO receptors (ROBO1, ROBO2, ROBO3, and ROBO4). The SLIT/ROBO signaling pathway has been shown to participate in axonal repulsion, axon guidance, and neuronal migration in the nervous system and the formation of the vascular system. However, the role of the SLIT/ROBO pathway has not been thoroughly clarified in tumor development. The SLIT/ROBO pathway can produce both beneficial and detrimental effects in the growth of malignant cells. It has been confirmed that SLIT/ROBO play contradictory roles in tumorigenesis. Here, we discuss the tumor promotion and tumor suppression roles of the SLIT/ROBO pathway in tumor growth, angiogenesis, migration, and the tumor microenvironment. Understanding these roles will help us develop more effective cancer therapies.
Collapse
Affiliation(s)
- Zhengdong Jiang
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Gang Liang
- Department of Hepatobiliary Surgery, No. 215 Hospital of Shaanxi Nuclear Industry, Xianyang, Shaanxi, China
| | - Ying Xiao
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Tao Qin
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xin Chen
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Erxi Wu
- Department of Neurosurgery, Neuroscience Institute, Baylor Scott and White Health, Temple, TX, USA
| | - Qingyong Ma
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Zheng Wang
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| |
Collapse
|
96
|
Ye X, Qiu Y, Gao Y, Wan D, Zhu H. A Subtle Network Mediating Axon Guidance: Intrinsic Dynamic Structure of Growth Cone, Attractive and Repulsive Molecular Cues, and the Intermediate Role of Signaling Pathways. Neural Plast 2019; 2019:1719829. [PMID: 31097955 PMCID: PMC6487106 DOI: 10.1155/2019/1719829] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/25/2019] [Accepted: 03/06/2019] [Indexed: 01/01/2023] Open
Abstract
A fundamental feature of both early nervous system development and axon regeneration is the guidance of axonal projections to their targets in order to assemble neural circuits that control behavior. In the navigation process where the nerves grow toward their targets, the growth cones, which locate at the tips of axons, sense the environment surrounding them, including varies of attractive or repulsive molecular cues, then make directional decisions to adjust their navigation journey. The turning ability of a growth cone largely depends on its highly dynamic skeleton, where actin filaments and microtubules play a very important role in its motility. In this review, we summarize some possible mechanisms underlying growth cone motility, relevant molecular cues, and signaling pathways in axon guidance of previous studies and discuss some questions regarding directions for further studies.
Collapse
Affiliation(s)
- Xiyue Ye
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China
- Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China
- Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China
| | - Yan Qiu
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China
- Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China
- Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China
| | - Yuqing Gao
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China
- Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China
- Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China
| | - Dong Wan
- Department of Emergency, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Huifeng Zhu
- College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing 400715, China
- Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China
- Engineering Research Center for Chongqing Pharmaceutical Process and Quality Control, Chongqing 400715, China
| |
Collapse
|
97
|
Phuong TTT, Walker AE, Henson GD, Machin DR, Li DY, Donato AJ, Lesniewski LA. Deletion of Robo4 prevents high-fat diet-induced adipose artery and systemic metabolic dysfunction. Microcirculation 2019; 26:e12540. [PMID: 30825241 DOI: 10.1111/micc.12540] [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] [Received: 09/21/2018] [Revised: 01/22/2019] [Accepted: 02/27/2019] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Accumulating evidence suggests the vascular endothelium plays a fundamental role in the pathophysiology of obesity by regulating the functional status of white adipose and systemic metabolism. Robo4 is expressed specifically in endothelial cells and increases vascular stability and inhibits angiogenesis. We sought to determine the role of Robo4 in modulating cardiometabolic function in response to high-fat feeding. METHODS We examined exercise capacity, glucose tolerance, and white adipose tissue artery gene expression, endothelium-dependent dilation (EDD), and angiogenesis in wild type and Robo4 knockout (KO) mice fed normal chow (NC) or a high-fat diet (HFD). RESULTS We found Robo4 deletion enhances exercise capacity in NC-fed mice and HFD markedly increased the expression of the Robo4 ligand, Slit2, in white adipose tissue. Deletion of Robo4 increased angiogenesis in white adipose tissue and protected against HFD-induced impairments in white adipose artery vasodilation and glucose intolerance. CONCLUSIONS We demonstrate a novel functional role for Robo4 in endothelial cell function and metabolic homeostasis in white adipose tissue, with Robo4 deletion protecting against endothelial and metabolic dysfunction associated with a HFD. Our findings suggest that Robo4-dependent signaling pathways may be a novel target in anti-obesity therapy.
Collapse
Affiliation(s)
- Tam T T Phuong
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah
| | - Ashley E Walker
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah
| | - Grant D Henson
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah
| | - Daniel R Machin
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah
| | - Dean Y Li
- Department of Medicine, Program in Molecular Medicine, University of Utah, Salt Lake City, Utah.,Division of Cardiovascular Medicine Department of Medicine, University of Utah, Salt Lake City, Utah.,Department of Human Genetics, University of Utah, Salt Lake City, Utah
| | - Anthony J Donato
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah.,Salt Lake City Veteran's Affair Medical Center, Geriatrics Research Education and Clinic Center, Salt Lake City, Utah.,Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah
| | - Lisa A Lesniewski
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah.,Salt Lake City Veteran's Affair Medical Center, Geriatrics Research Education and Clinic Center, Salt Lake City, Utah.,Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah
| |
Collapse
|
98
|
Kumar SR, Gajagowni S, Bryan JN, Bodenhausen HM. Molecular targets for tivantinib (ARQ 197) and vasculogenic mimicry in human melanoma cells. Eur J Pharmacol 2019; 853:316-324. [PMID: 30954563 DOI: 10.1016/j.ejphar.2019.04.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 03/27/2019] [Accepted: 04/02/2019] [Indexed: 11/25/2022]
Abstract
Tivantinib (TivB) was reported previously to target MET and microtubule assembly in different cells resulting in cytotoxicity. However, its other cellular targets remain unknown, especially the proteins involved in focal adhesion and cytoskeletal organization. We studied the effect of TivB on vinculin a focal adhesion protein, and RhoC, a GTPase which promote the reorganization of cytoskeleton. Biomolecules involved in vasculogenic mimicry (VM) previously not reported in melanoma, and their susceptibility to TivB was also evaluated. TivB affects the viability and apoptosis of human melanoma cells depending on the cell type. Vinculin and RhoC were increased in the presence of TivB and affected the integrity of actin filaments and altered the cellular morphology. TivB disrupts the VM exhibited by melanoma cells in 3D matrix. Roundabout Guidance Receptor 4 (Robo4), a receptor protein implicated in axonal guidance and angiogenesis and its ligand Slit2 are expressed in human C8161 and WM793 melanoma cells, but absent in other melanoma cells including normal melanocytes. VM is more prominent in C8161 cells and could be blocked by siRNA mediated silencing of Robo4 mRNA, but TivB does not affect Robo4 in C8161 cells. Immunoblot analysis indicated no changes in Robo4 and Slit2 protein expression, however, both vinculin and RhoC protein increased in TivB treated melanoma cells. These results suggest that TivB affects cell cytoskeleton and morphology by altering proteins such as vinculin and RhoC. Our studies indicate TivB could target molecules other than MET in melanoma cells, which may provide insight into its alternate mechanism of action.
Collapse
Affiliation(s)
- Senthil R Kumar
- Comparative Oncology, Radiobiology and Epigenetics Laboratory, College of Veterinary Medicine, University of Missouri, Columbia, MO, 65211, USA; Department of Surgery, Ellis Fischel Cancer Centre, School of Medicine, University of Missouri, Columbia, MO, 65201, USA; Harry S. Truman Veterans Medical Center, Columbia, MO, 65201, USA.
| | - Saivaroon Gajagowni
- Comparative Oncology, Radiobiology and Epigenetics Laboratory, College of Veterinary Medicine, University of Missouri, Columbia, MO, 65211, USA
| | - Jeffrey N Bryan
- Comparative Oncology, Radiobiology and Epigenetics Laboratory, College of Veterinary Medicine, University of Missouri, Columbia, MO, 65211, USA
| | - Hannah M Bodenhausen
- Comparative Oncology, Radiobiology and Epigenetics Laboratory, College of Veterinary Medicine, University of Missouri, Columbia, MO, 65211, USA
| |
Collapse
|
99
|
Barak R, Yom-Tov G, Guez-Haddad J, Gasri-Plotnitsky L, Maimon R, Cohen-Berkman M, McCarthy AA, Perlson E, Henis-Korenblit S, Isupov MN, Opatowsky Y. Structural Principles in Robo Activation and Auto-inhibition. Cell 2019; 177:272-285.e16. [PMID: 30853216 DOI: 10.1016/j.cell.2019.02.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 12/06/2018] [Accepted: 02/06/2019] [Indexed: 01/28/2023]
Abstract
Proper brain function requires high-precision neuronal expansion and wiring, processes controlled by the transmembrane Roundabout (Robo) receptor family and their Slit ligands. Despite their great importance, the molecular mechanism by which Robos' switch from "off" to "on" states remains unclear. Here, we report a 3.6 Å crystal structure of the intact human Robo2 ectodomain (domains D1-8). We demonstrate that Robo cis dimerization via D4 is conserved through hRobo1, 2, and 3 and the C. elegans homolog SAX-3 and is essential for SAX-3 function in vivo. The structure reveals two levels of auto-inhibition that prevent premature activation: (1) cis blocking of the D4 dimerization interface and (2) trans interactions between opposing Robo receptors that fasten the D4-blocked conformation. Complementary experiments in mouse primary neurons and C. elegans support the auto-inhibition model. These results suggest that Slit stimulation primarily drives the release of Robo auto-inhibition required for dimerization and activation.
Collapse
Affiliation(s)
- Reut Barak
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Israel
| | - Galit Yom-Tov
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Israel
| | - Julia Guez-Haddad
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Israel
| | | | - Roy Maimon
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Israel
| | - Moran Cohen-Berkman
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Israel
| | | | - Eran Perlson
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Israel
| | | | | | - Yarden Opatowsky
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Israel.
| |
Collapse
|
100
|
Gołos A, Jesionek-Kupnicka D, Gil L, Braun M, Komarnicki M, Robak T, Wierzbowska A. The Expression of the SLIT-ROBO Family in Adult Patients with Acute Myeloid Leukemia. Arch Immunol Ther Exp (Warsz) 2019; 67:109-123. [PMID: 30820596 PMCID: PMC6420492 DOI: 10.1007/s00005-019-00535-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Accepted: 11/23/2018] [Indexed: 12/13/2022]
Abstract
INTRODUCTION SLIT-ROBO is a ligand-receptor family of neuronal guidance cues that has been involved in pathological and physiological angiogenesis. SLIT-ROBO expression is altered in many tumours. However, no data exist about the role of the whole family in acute myelogenous myeloid leukemia (AML). PURPOSE Herein, we assessed the expression of all SLIT-ROBO family in bone marrow (BM) biopsy of AML patients and control group on both protein and RNA levels. METHODS The paraffin-embedded tissue blocks were subjected to immunohistochemistry for SLIT1, SLIT2, SLIT3, ROBO1, ROBO2, ROBO3, and ROBO4. Microvessel density (MVD) was evaluated by CD34 immunohistochemistry. An in silico analysis using The Cancer Genome Atlas data repository was conducted for assessment of RNA level. RESULTS Acute myeloid leukemia patients were generally high expressers of ROBO1 and ROBO2 compared to the controls (p < 0.0001, p < 0.001, respectively). In contrast, low expression of SLIT1, SLIT2, and SLIT3 ligands has been noted more commonly in AML than in control BM samples (p < 0.0001, p = 0.003, and p = 0.001, respectively). ROBO4 expression correlated with MVD. The in silico analysis showed a poor prognostic value of high ROBO3 and low SLIT2 RNA levels (p = 0.0003 and p = 0.0008, respectively), as well as high ROBO3 and ROBO4 RNA levels in cytogenetic poor risk groups of patients (p = 0.0029 and p = 0.0003, respectively). CONCLUSIONS These data indicate that SLIT-ROBO family members play a role in the biology of AML. Low expression of SLIT in BM of AML patients may suggest its expression alterations in AML. Increased expression of ROBO1 and ROBO2 in AML patients suggests their participation in AML pathogenesis.
Collapse
Affiliation(s)
- Aleksandra Gołos
- Department of Hematology, Medical University, Lodz, Poland.
- Department of Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland.
| | | | - Lidia Gil
- Department of Hematology, University of Medical Sciences, Poznan, Poland
| | - Marcin Braun
- Department of Pathology, Medical University, Lodz, Poland
- Postgraduate School of Medicine, Medical University of Warsaw, Warsaw, Poland
| | | | - Tadeusz Robak
- Department of Hematology, Medical University, Lodz, Poland
| | | |
Collapse
|