1
|
Nelson KA, Lenhart KF, Anllo L, DiNardo S. The Drosophila hematopoietic niche assembles through collective cell migration controlled by neighbor tissues and Slit-Robo signaling. eLife 2025; 13:RP100455. [PMID: 39750120 PMCID: PMC11698496 DOI: 10.7554/elife.100455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
Niches are often found in specific positions in tissues relative to the stem cells they support. Consistency of niche position suggests that placement is important for niche function. However, the complexity of most niches has precluded a thorough understanding of how their proper placement is established. To address this, we investigated the formation of a genetically tractable niche, the Drosophila Posterior Signaling Center (PSC), the assembly of which had not been previously explored. This niche controls hematopoietic progenitors of the lymph gland (LG). PSC cells were previously shown to be specified laterally in the embryo, but ultimately reside dorsally, at the LG posterior. Here, using live-imaging, we show that PSC cells migrate as a tight collective and associate with multiple tissues during their trajectory to the LG posterior. We find that Slit emanating from two extrinsic sources, visceral mesoderm and cardioblasts, is required for the PSC to remain a collective, and for its attachment to cardioblasts during migration. Without proper Slit-Robo signaling, PSC cells disperse, form aberrant contacts, and ultimately fail to reach their stereotypical position near progenitors. Our work characterizes a novel example of niche formation and identifies an extrinsic signaling relay that controls precise niche positioning.
Collapse
Affiliation(s)
- Kara A Nelson
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
- Institute for Regenerative Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| | - Kari F Lenhart
- Department of Biology, Drexel UniversityPhiladelphiaUnited States
| | - Lauren Anllo
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
- Institute for Regenerative Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| | - Stephen DiNardo
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
- Institute for Regenerative Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| |
Collapse
|
2
|
Nelson KA, Lenhart KF, Anllo L, DiNardo S. The Drosophila hematopoietic niche assembles through collective cell migration controlled by neighbor tissues and Slit-Robo signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.21.600069. [PMID: 38979182 PMCID: PMC11230208 DOI: 10.1101/2024.06.21.600069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Niches are often found in specific positions in tissues relative to the stem cells they support. Consistency of niche position suggests that placement is important for niche function. However, the complexity of most niches has precluded a thorough understanding of how their proper placement is established. To address this, we investigated the formation of a genetically tractable niche, the Drosophila Posterior Signaling Center (PSC), the assembly of which had not been previously explored. This niche controls hematopoietic progenitors of the lymph gland (LG). PSC cells were previously shown to be specified laterally in the embryo, but ultimately reside dorsally, at the LG posterior. Here, using live-imaging, we show that PSC cells migrate as a tight collective and associate with multiple tissues during their trajectory to the LG posterior. We find that Slit emanating from two extrinsic sources, visceral mesoderm and cardioblasts, is required for the PSC to remain a collective, and for its attachment to cardioblasts during migration. Without proper Slit-Robo signaling, PSC cells disperse, form aberrant contacts, and ultimately fail to reach their stereotypical position near progenitors. Our work characterizes a novel example of niche formation and identifies an extrinsic signaling relay that controls precise niche positioning.
Collapse
Affiliation(s)
- Kara A Nelson
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, 421 Curie Blvd. Philadelphia, PA 19104, United States
- Institute for Regenerative Medicine at the University of Pennsylvania, 3400 Civic Center Blvd. Philadelphia, PA 19104, United States
| | - Kari F Lenhart
- Department of Biology, Drexel University, 3245 Chestnut St. Philadelphia, PA 19104, United States
| | - Lauren Anllo
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, 421 Curie Blvd. Philadelphia, PA 19104, United States
- Institute for Regenerative Medicine at the University of Pennsylvania, 3400 Civic Center Blvd. Philadelphia, PA 19104, United States
- Present address: Department of Biology, East Carolina University, 458 Science & Tech Bldg. Greenville, NC 27858, United States
| | - Stephen DiNardo
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, 421 Curie Blvd. Philadelphia, PA 19104, United States
- Institute for Regenerative Medicine at the University of Pennsylvania, 3400 Civic Center Blvd. Philadelphia, PA 19104, United States
| |
Collapse
|
3
|
Johnson AN. Myotube Guidance: Shaping up the Musculoskeletal System. J Dev Biol 2024; 12:25. [PMID: 39311120 PMCID: PMC11417883 DOI: 10.3390/jdb12030025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/20/2024] [Accepted: 09/12/2024] [Indexed: 09/26/2024] Open
Abstract
Myofibers are highly specialized contractile cells of skeletal muscles, and dysregulation of myofiber morphogenesis is emerging as a contributing cause of myopathies and structural birth defects. Myotubes are the myofiber precursors and undergo a dramatic morphological transition into long bipolar myofibers that are attached to tendons on two ends. Similar to axon growth cones, myotube leading edges navigate toward target cells and form cell-cell connections. The process of myotube guidance connects myotubes with the correct tendons, orients myofiber morphology with the overall body plan, and generates a functional musculoskeletal system. Navigational signaling, addition of mass and volume, and identification of target cells are common events in myotube guidance and axon guidance, but surprisingly, the mechanisms regulating these events are not completely overlapping in myotubes and axons. This review summarizes the strategies that have evolved to direct myotube leading edges to predetermined tendon cells and highlights key differences between myotube guidance and axon guidance. The association of myotube guidance pathways with developmental disorders is also discussed.
Collapse
Affiliation(s)
- Aaron N Johnson
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| |
Collapse
|
4
|
Cazares O, Chen M, Menendez J, Molinuevo R, Thomas G, Cervantes J, Yee M, Cadell M, Durham M, Zhu Y, Strietzel C, Bubolz JW, Hinck L. SLIT Loss or Sequestration Increases Mammary Alveologenesis and Lactogenesis. MICROPUBLICATION BIOLOGY 2024; 2024:10.17912/micropub.biology.001264. [PMID: 39381643 PMCID: PMC11461027 DOI: 10.17912/micropub.biology.001264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/17/2024] [Accepted: 09/04/2024] [Indexed: 10/10/2024]
Abstract
SLITs comprise a family of secreted proteins that function as ligands for Roundabout (ROBO) receptors. Previous research showed that ROBO1 promotes the differentiation of milk-producing alveolar cells by inhibiting Notch signaling in mammary luminal cells. Here, we show enhanced alveolar development and increased milk production in Slit2-/-;Slit3-/- knockout mammary gland epithelia. This result can also be achieved by intraperitoneal delivery of recombinant ROBO1 extracellular domain fragment, ROBO1-5Ig-Fc, which sequesters SLITs. Together, our phenotypic studies suggest that SLITs restrict alveologenesis and lactogenesis by inhibiting ROBO1.
Collapse
Affiliation(s)
| | - Min Chen
- University of California, Santa Cruz, CA, USA
| | | | | | - Gwen Thomas
- University of California, Santa Cruz, CA, USA
| | | | - Michael Yee
- University of California, Santa Cruz, CA, USA
| | | | | | - Yaqi Zhu
- Zoetis (United States), Kalamazoo, MI, United States
| | | | | | | |
Collapse
|
5
|
Daponte V, Henke K, Drissi H. Current perspectives on the multiple roles of osteoclasts: Mechanisms of osteoclast-osteoblast communication and potential clinical implications. eLife 2024; 13:e95083. [PMID: 38591777 PMCID: PMC11003748 DOI: 10.7554/elife.95083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 03/29/2024] [Indexed: 04/10/2024] Open
Abstract
Bone remodeling is a complex process involving the coordinated actions of osteoblasts and osteoclasts to maintain bone homeostasis. While the influence of osteoblasts on osteoclast differentiation is well established, the reciprocal regulation of osteoblasts by osteoclasts has long remained enigmatic. In the past few years, a fascinating new role for osteoclasts has been unveiled in promoting bone formation and facilitating osteoblast migration to the remodeling sites through a number of different mechanisms, including the release of factors from the bone matrix following bone resorption and direct cell-cell interactions. Additionally, considerable evidence has shown that osteoclasts can secrete coupling factors known as clastokines, emphasizing the crucial role of these cells in maintaining bone homeostasis. Due to their osteoprotective function, clastokines hold great promise as potential therapeutic targets for bone diseases. However, despite long-standing work to uncover new clastokines and their effect in vivo, more substantial efforts are still required to decipher the mechanisms and pathways behind their activity in order to translate them into therapies. This comprehensive review provides insights into our evolving understanding of the osteoclast function, highlights the significance of clastokines in bone remodeling, and explores their potential as treatments for bone diseases suggesting future directions for the field.
Collapse
Affiliation(s)
- Valentina Daponte
- Department of Orthopaedics, Emory University School of MedicineAtlantaUnited States
- VA Medical CenterAtlantaUnited States
| | - Katrin Henke
- Department of Orthopaedics, Emory University School of MedicineAtlantaUnited States
| | - Hicham Drissi
- Department of Orthopaedics, Emory University School of MedicineAtlantaUnited States
- VA Medical CenterAtlantaUnited States
| |
Collapse
|
6
|
Zhou J, Zhang Z, Wu M, Liu H, Pang Y, Bartlett A, Peng Z, Ding W, Rivkin A, Lagos WN, Williams E, Lee CT, Miyazaki PA, Aldridge A, Zeng Q, Salinda JLA, Claffey N, Liem M, Fitzpatrick C, Boggeman L, Yao Z, Smith KA, Tasic B, Altshul J, Kenworthy MA, Valadon C, Nery JR, Castanon RG, Patne NS, Vu M, Rashid M, Jacobs M, Ito T, Osteen J, Emerson N, Lee J, Cho S, Rink J, Huang HH, Pinto-Duartec A, Dominguez B, Smith JB, O'Connor C, Zeng H, Chen S, Lee KF, Mukamel EA, Jin X, Margarita Behrens M, Ecker JR, Callaway EM. Brain-wide correspondence of neuronal epigenomics and distant projections. Nature 2023; 624:355-365. [PMID: 38092919 PMCID: PMC10719087 DOI: 10.1038/s41586-023-06823-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 11/01/2023] [Indexed: 12/17/2023]
Abstract
Single-cell analyses parse the brain's billions of neurons into thousands of 'cell-type' clusters residing in different brain structures1. Many cell types mediate their functions through targeted long-distance projections allowing interactions between specific cell types. Here we used epi-retro-seq2 to link single-cell epigenomes and cell types to long-distance projections for 33,034 neurons dissected from 32 different regions projecting to 24 different targets (225 source-to-target combinations) across the whole mouse brain. We highlight uses of these data for interrogating principles relating projection types to transcriptomics and epigenomics, and for addressing hypotheses about cell types and connections related to genetics. We provide an overall synthesis with 926 statistical comparisons of discriminability of neurons projecting to each target for every source. We integrate this dataset into the larger BRAIN Initiative Cell Census Network atlas, composed of millions of neurons, to link projection cell types to consensus clusters. Integration with spatial transcriptomics further assigns projection-enriched clusters to smaller source regions than the original dissections. We exemplify this by presenting in-depth analyses of projection neurons from the hypothalamus, thalamus, hindbrain, amygdala and midbrain to provide insights into properties of those cell types, including differentially expressed genes, their associated cis-regulatory elements and transcription-factor-binding motifs, and neurotransmitter use.
Collapse
Affiliation(s)
- Jingtian Zhou
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
- Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, CA, USA
| | - Zhuzhu Zhang
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
- Department of Human Genetics, The University of Chicago, Chicago, IL, USA
| | - May Wu
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
- Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Hanqing Liu
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Yan Pang
- Systems Neurobiology Laboratories, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Anna Bartlett
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Zihao Peng
- School of Mathematics and Computer Science, Nanchang University, Nanchang, China
- Henan Engineering Research Center of Intelligent Technology and Application, Henan University, Kaifeng, China
| | - Wubin Ding
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Angeline Rivkin
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Will N Lagos
- Systems Neurobiology Laboratories, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Elora Williams
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Cheng-Ta Lee
- Peptide Biology Laboratories, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Paula Assakura Miyazaki
- Systems Neurobiology Laboratories, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Andrew Aldridge
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Qiurui Zeng
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
- Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - J L Angelo Salinda
- Systems Neurobiology Laboratories, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Naomi Claffey
- Flow Cytometry Core Facility, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Michelle Liem
- Flow Cytometry Core Facility, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Conor Fitzpatrick
- Flow Cytometry Core Facility, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Lara Boggeman
- Flow Cytometry Core Facility, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Zizhen Yao
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Jordan Altshul
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Mia A Kenworthy
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Cynthia Valadon
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Joseph R Nery
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Rosa G Castanon
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Neelakshi S Patne
- Systems Neurobiology Laboratories, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Minh Vu
- Systems Neurobiology Laboratories, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Mohammad Rashid
- Systems Neurobiology Laboratories, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Matthew Jacobs
- Systems Neurobiology Laboratories, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Tony Ito
- Systems Neurobiology Laboratories, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Julia Osteen
- Computational Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Nora Emerson
- Computational Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Jasper Lee
- Computational Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Silvia Cho
- Computational Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Jon Rink
- Computational Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Hsiang-Hsuan Huang
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - António Pinto-Duartec
- Computational Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Bertha Dominguez
- Peptide Biology Laboratories, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Jared B Smith
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Carolyn O'Connor
- Flow Cytometry Core Facility, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Shengbo Chen
- Henan Engineering Research Center of Intelligent Technology and Application, Henan University, Kaifeng, China
- School of Computer and Information Engineering, Henan University, Kaifeng, China
| | - Kuo-Fen Lee
- Peptide Biology Laboratories, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Eran A Mukamel
- Department of Cognitive Science, University of California San Diego, La Jolla, CA, USA
| | - Xin Jin
- Center for Motor Control and Disease, Key Laboratory of Brain Functional Genomics, East China Normal University, Shanghai, China
- NYU-ECNU Institute of Brain and Cognitive Science, New York University Shanghai, Shanghai, China
| | - M Margarita Behrens
- Computational Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Joseph R Ecker
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA.
- Howard Hughes Medical Institute, The Salk Institute for Biological Studies, La Jolla, CA, USA.
| | - Edward M Callaway
- Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA.
- Systems Neurobiology Laboratories, The Salk Institute for Biological Studies, La Jolla, CA, USA.
| |
Collapse
|
7
|
Sarkany B, Kuthi L, Kovacs G. Novel concept of Wilms' tumor development: involvement of pluripotential cells of ureteric bud. Hum Pathol 2023; 138:34-40. [PMID: 37209922 DOI: 10.1016/j.humpath.2023.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 05/15/2023] [Indexed: 05/22/2023]
Abstract
It is acknowledged that nephron develops after bilateral induction of the metanephric mesenchyma and branching ureteric bud (UB), and that nephrogenic rest and Wilms' tumor (nephroblastoma) arises from impaired differentiation of metanephric blastema. The aim of this study was to obtain more information on the involvement of UB derivatives in nephrogenic rest and Wilms' tumor. We applied immunohistochemistry to analyze nephrogenic rests and Wilms' tumors with mixed histology, including regressive and blastemal types. We used antibodies recognizing UB tip cells (ROBO1, SLIT2, RET), principal cells (AQP2), α- and β-intercalated cells (SLC26A4, SLC4A1, ATP6V1B1, ATP6V0D2), and their precursors (CA2). Tubules surrounded by tumorous blastemal cells resembling UB tip were positive for RET, ROBO1, and SLIT2 in Wilms' tumor. Moreover, CA2-positive tubular structures and ATP6V1B1- and ATP6V0D2-positive immature non-α- and non-β-intercalated cells were detected in nephrogenic rest and Wilms' tumor. We suggest that Wilms' tumor is more than nephroblastoma and propose a definition that Wilms tumor is a malignant embryonal neoplasm derived from pluripotential cells of nephrogenic blastema and of ureteric bud tip.
Collapse
Affiliation(s)
- Beatrix Sarkany
- Department of Urology, Medical School, University of Pecs, 7621, Pecs, Hungary
| | - Levente Kuthi
- Department of Pathology, University of Szeged, 6725, Szeged, Hungary
| | - Gyula Kovacs
- Department of Urology, Medical School, University of Pecs, 7621, Pecs, Hungary; Medical Faculty, Ruprecht-Karls-University, 69120, Heidelberg, Germany.
| |
Collapse
|
8
|
Yang S, Johnson AN. The serine/threonine kinase Back seat driver prevents cell fusion to maintain cell identity. Dev Biol 2023; 495:35-41. [PMID: 36528051 PMCID: PMC11088746 DOI: 10.1016/j.ydbio.2022.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 11/23/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
Cell fate specification is essential for every major event of embryogenesis, and subsequent cell maturation ensures individual cell types acquire specialized functions. The mechanisms that regulate cell fate specification have been studied exhaustively, and each technological advance in developmental biology ushers in a new era of studies aimed at uncovering the most fundamental processes by which cells acquire unique identities. What is less appreciated is that mechanisms are in place to ensure cell identity is maintained throughout the life of the organism. The body wall musculature in the Drosophila embryo is a well-established model to study cell fate specification, as each hemisegment in the embryo generates and maintains thirty muscles with distinct identities. Once specified, the thirty body wall muscles fuse with mononucleate muscle precursors that lack a specific identity to form multinucleate striated muscles. Multinucleate body wall muscles do not fuse with each other, which maintains a diversification of muscle cell identities. Here we show the serine/threonine kinase Back seat driver (Bsd) prevents inappropriate muscle fusion to maintain cell identity. Thus, the regulation of cell fusion is one mechanism that maintains cell identity.
Collapse
Affiliation(s)
- Shuo Yang
- Department of Developmental Biology Washington University School of Medicine St. Louis, MO 63110
| | - Aaron N. Johnson
- Department of Developmental Biology Washington University School of Medicine St. Louis, MO 63110
| |
Collapse
|
9
|
Moucaud B, Prince E, Jagla K, Soler C. Developmental origin of tendon diversity in Drosophila melanogaster. Front Physiol 2023; 14:1176148. [PMID: 37143929 PMCID: PMC10151533 DOI: 10.3389/fphys.2023.1176148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 03/28/2023] [Indexed: 05/06/2023] Open
Abstract
Myogenesis is a developmental process that is largely conserved in both Drosophila and higher organisms. Consequently, the fruit fly is an excellent in vivo model for identifying the genes and mechanisms involved in muscle development. Moreover, there is growing evidence indicating that specific conserved genes and signaling pathways govern the formation of tissues that connect the muscles to the skeleton. In this review, we present an overview of the different stages of tendon development, from the specification of tendon progenitors to the assembly of a stable myotendinous junction across three different myogenic contexts in Drosophila: larval, flight and leg muscle development. We underline the different aspects of tendon cell specification and differentiation in embryo and during metamorphosis that result into tendon morphological and functional diversity.
Collapse
|
10
|
Hauptman G, Reichert MC, Abdal Rhida MA, Evans TA. Characterization of enhancer fragments in Drosophila robo2. Fly (Austin) 2022; 16:312-346. [PMID: 36217698 PMCID: PMC9559326 DOI: 10.1080/19336934.2022.2126259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 01/05/2023] Open
Abstract
Receptor proteins of the Roundabout (Robo) family regulate axon guidance decisions during nervous system development. Among the three Drosophila robo family genes (robo1, robo2 and robo3), robo2 displays a dynamic expression pattern and regulates multiple axon guidance outcomes, including preventing midline crossing in some axons, promoting midline crossing in others, forming lateral longitudinal axon pathways, and regulating motor axon guidance. The identity and location of enhancer elements regulating robo2's complex and dynamic expression pattern in different neural cell types are unknown. Here, we characterize a set of 17 transgenic lines expressing GAL4 under the control of DNA sequences derived from noncoding regions in and around robo2, to identify enhancers controlling specific aspects of robo2 expression in the embryonic ventral nerve cord. We identify individual fragments that confer expression in specific cell types where robo2 is known to function, including early pioneer neurons, midline glia and lateral longitudinal neurons. Our results indicate that robo2's dynamic expression pattern is specified by a combination of enhancer elements that are active in different subsets of cells. We show that robo2's expression in lateral longitudinal axons represents two genetically separable subsets of neurons, and compare their axon projections with each other and with Fasciclin II (FasII), a commonly used marker of longitudinal axon pathways. In addition, we provide a general description of each fragment's expression in embryonic tissues outside of the nervous system, to serve as a resource for other researchers interested in robo2 expression and its functional roles outside the central nervous system.
Collapse
Affiliation(s)
- Gina Hauptman
- Department of Biological Sciences, University of Arkansas, Fayetteville
| | - Marie C. Reichert
- Department of Biological Sciences, University of Arkansas, Fayetteville
| | - Muna A. Abdal Rhida
- Department of Biological Sciences, University of Arkansas, Fayetteville
- Department of Biology, Wasit University, Iraq
| | - Timothy A. Evans
- Department of Biological Sciences, University of Arkansas, Fayetteville
| |
Collapse
|
11
|
Chen Z, Chen P, Zheng M, Gao J, Liu D, Wang A, Zheng Q, Leys T, Tai A, Zheng M. Challenges and perspectives of tendon-derived cell therapy for tendinopathy: from bench to bedside. Stem Cell Res Ther 2022; 13:444. [PMID: 36056395 PMCID: PMC9438319 DOI: 10.1186/s13287-022-03113-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 08/03/2022] [Indexed: 11/18/2022] Open
Abstract
Tendon is composed of dense fibrous connective tissues, connecting muscle at the myotendinous junction (MTJ) to bone at the enthesis and allowing mechanical force to transmit from muscle to bone. Tendon diseases occur at different zones of the tendon, including enthesis, MTJ and midsubstance of the tendon, due to a variety of environmental and genetic factors which consequently result in different frequencies and recovery rates. Self-healing properties of tendons are limited, and cell therapeutic approaches in which injured tendon tissues are renewed by cell replenishment are highly sought after. Homologous use of individual’s tendon-derived cells, predominantly differentiated tenocytes and tendon-derived stem cells, is emerging as a treatment for tendinopathy through achieving minimal cell manipulation for clinical use. This is the first review summarizing the progress of tendon-derived cell therapy in clinical use and its challenges due to the structural complexity of tendons, heterogeneous composition of extracellular cell matrix and cells and unsuitable cell sources. Further to that, novel future perspectives to improve therapeutic effect in tendon-derived cell therapy based on current basic knowledge are discussed.
Collapse
Affiliation(s)
- Ziming Chen
- Division of Surgery, Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, WA, 6009, Australia
| | - Peilin Chen
- Division of Surgery, Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, WA, 6009, Australia
| | - Monica Zheng
- Department of Orthopaedic Surgery, Sir Charles Gairdner Hospital, Nedlands, WA, 6009, Australia
| | - Junjie Gao
- Perron Institute for Neurological and Translational Science, Nedlands, WA, 6009, Australia.,Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Shanghai Sixth People's Hospital, Shanghai, 200233, China
| | - Delin Liu
- Division of Surgery, Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, WA, 6009, Australia.,Perron Institute for Neurological and Translational Science, Nedlands, WA, 6009, Australia
| | - Allan Wang
- Division of Surgery, Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, WA, 6009, Australia
| | - Qiujian Zheng
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510000, Guangdong, China.,Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510000, Guangdong, China
| | - Toby Leys
- Department of Orthopaedic Surgery, Sir Charles Gairdner Hospital, Nedlands, WA, 6009, Australia
| | - Andrew Tai
- Perron Institute for Neurological and Translational Science, Nedlands, WA, 6009, Australia.
| | - Minghao Zheng
- Division of Surgery, Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, WA, 6009, Australia. .,Perron Institute for Neurological and Translational Science, Nedlands, WA, 6009, Australia.
| |
Collapse
|
12
|
Yang H, Zhou S, Lan D, Bin Y, Bao W, Wang M, Huang F, Peng Z. The expression of Slit2 and Robo1 increased during retinoic acid syndrome in acute promyelocytic leukemia and impacted differentiated cell migration. Transl Oncol 2022; 18:101370. [PMID: 35182953 PMCID: PMC8857660 DOI: 10.1016/j.tranon.2022.101370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/25/2022] [Accepted: 02/10/2022] [Indexed: 11/25/2022] Open
Abstract
The upregulation of Robo1 and Slit2 was first found in APL patients. The positive correlation between Robo1/Slit2 and retinoic acid syndrome was first demonstrated. It was demonstrated for the first time that Slit2 induces the migration of differentiated cells. Slit2 did not inhibit il8-induced differentiated cell migration.
Retinoic acid syndrome (RAS) is a serious complication developed during the induction therapy of acute promyelocytic leukemia (APL). Cytokines and differentiated cells migration play important roles in the development of RAS. Slit guidance ligand 2 (Slit2) and roundabout 1 (Robo1) involve in cell migration. Our study aimed to investigate the expression of Slit2 and Robo1 in APL and check whether they affected promyelocytes migration. 62 cases of newly diagnosed APL patients were involved and received all-trans retinoic acid (ATRA) and arsenic trioxide as induction therapy. Bone marrow cells (BMCs) were obtained on days 0 and 28, and promyelocytes and plasma were collected from day 1 to day 21. The expression of Robo1 in promyelocytes, and that of Slit2 and cytokines, including IL-8,IL-1β and others, in serum were monitored. 20 healthy individuals donated their cells as control. Of the 62 APL patients, 16 (25.81%) patients developed RAS. The expression of Robo1, Slit2 and IL-8 increased significantly with the development of RAS. In the 16 patients with RAS, levels of Slit2, Robo1 and IL-8 were higher during the development of RAS than before or after the RAS (P < 0.05). RhSlit2-N and rhIL-8 induced cells migration, and the migration induced by IL-8 was not inhibited by rhSlit2-N. Elevated Slit2 and Robo1 levels might be useful markers for the diagnosis and treatment of RAS. The levels of Slit2, Robo1 and IL-8 showed a positive correlation with the severity of RAS. Slit2 and IL-8 promoted the migration of differentiated cells.
Collapse
Affiliation(s)
- Haiyan Yang
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Shengsheng Zhou
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Dong Lan
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Yehong Bin
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Wenguang Bao
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Man Wang
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Fengxiang Huang
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Zhigang Peng
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China.
| |
Collapse
|
13
|
Yang S, McAdow J, Du Y, Trigg J, Taghert PH, Johnson AN. Spatiotemporal expression of regulatory kinases directs the transition from mitosis to cellular morphogenesis in Drosophila. Nat Commun 2022; 13:772. [PMID: 35140224 PMCID: PMC8828718 DOI: 10.1038/s41467-022-28322-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 01/17/2022] [Indexed: 02/01/2023] Open
Abstract
Embryogenesis depends on a tightly regulated balance between mitosis, differentiation, and morphogenesis. Understanding how the embryo uses a relatively small number of proteins to transition between growth and morphogenesis is a central question of developmental biology, but the mechanisms controlling mitosis and differentiation are considered to be fundamentally distinct. Here we show the mitotic kinase Polo, which regulates all steps of mitosis in Drosophila, also directs cellular morphogenesis after cell cycle exit. In mitotic cells, the Aurora kinases activate Polo to control a cytoskeletal regulatory module that directs cytokinesis. We show that in the post-mitotic mesoderm, the control of Polo activity transitions from the Aurora kinases to the uncharacterized kinase Back Seat Driver (Bsd), where Bsd and Polo cooperate to regulate muscle morphogenesis. Polo and its effectors therefore direct mitosis and cellular morphogenesis, but the transition from growth to morphogenesis is determined by the spatiotemporal expression of upstream activating kinases. The mechanisms regulating mitosis and differentiation during development are thought to be distinct. Here they show that in Drosophila the mitotic kinase Polo regulates cellular morphogenesis after cell cycle exit.
Collapse
Affiliation(s)
- Shuo Yang
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jennifer McAdow
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Yingqiu Du
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jennifer Trigg
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Paul H Taghert
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Aaron N Johnson
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
14
|
Maiorano AM, Cardoso DF, Carvalheiro R, Júnior GAF, de Albuquerque LG, de Oliveira HN. Signatures of selection in Nelore cattle revealed by whole-genome sequencing data. Genomics 2022; 114:110304. [DOI: 10.1016/j.ygeno.2022.110304] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 01/07/2022] [Accepted: 02/01/2022] [Indexed: 11/04/2022]
|
15
|
Pérez-Moreno JJ, Santa-Cruz Mateos C, Martín-Bermudo MD, Estrada B. LanB1 Cooperates With Kon-Tiki During Embryonic Muscle Migration in Drosophila. Front Cell Dev Biol 2022; 9:749723. [PMID: 35047493 PMCID: PMC8762229 DOI: 10.3389/fcell.2021.749723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 11/19/2021] [Indexed: 11/13/2022] Open
Abstract
Muscle development is a multistep process that involves cell specification, myoblast fusion, myotube migration, and attachment to the tendons. In spite of great efforts trying to understand the basis of these events, little is known about the molecular mechanisms underlying myotube migration. Knowledge of the few molecular cues that guide this migration comes mainly from studies in Drosophila. The migratory process of Drosophila embryonic muscles involves a first phase of migration, where muscle progenitors migrate relative to each other, and a second phase, where myotubes migrate searching for their future attachment sites. During this phase, myotubes form extensive filopodia at their ends oriented preferentially toward their attachment sites. This myotube migration and the subsequent muscle attachment establishment are regulated by cell adhesion receptors, such as the conserved proteoglycan Kon-tiki/Perdido. Laminins have been shown to regulate the migratory behavior of many cell populations, but their role in myotube migration remains largely unexplored. Here, we show that laminins, previously implicated in muscle attachment, are indeed required for muscle migration to tendon cells. Furthermore, we find that laminins genetically interact with kon-tiki/perdido to control both myotube migration and attachment. All together, our results uncover a new role for the interaction between laminins and Kon-tiki/Perdido during Drosophila myogenesis. The identification of new players and molecular interactions underlying myotube migration broadens our understanding of muscle development and disease.
Collapse
|
16
|
Junion G, Jagla K. Diversification of muscle types in Drosophila embryos. Exp Cell Res 2022; 410:112950. [PMID: 34838813 DOI: 10.1016/j.yexcr.2021.112950] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 11/22/2021] [Accepted: 11/24/2021] [Indexed: 12/31/2022]
Abstract
Drosophila embryonic somatic muscles represent a simple and tractable model system to study the gene regulatory networks that control diversification of cell types. Somatic myogenesis in Drosophila is initiated by intrinsic action of the mesodermal master gene twist, which activates a cascade of transcriptional outputs including myogenic differentiation factor Mef2, which triggers all aspects of the myogenic differentiation program. In parallel, the expression of a combinatorial code of identity transcription factors (iTFs) defines discrete particular features of each muscle fiber, such as number of fusion events, and specific attachment to tendon cells or innervation, thus ensuring diversification of muscle types. Here, we take the example of a subset of lateral transverse (LT) muscles and discuss how the iTF code and downstream effector genes progressively define individual LT properties such as fusion program, attachment and innervation. We discuss new challenges in the field including the contribution of posttranscriptional and epitranscriptomic regulation of gene expression in the diversification of cell types.
Collapse
Affiliation(s)
- Guillaume Junion
- Genetics Reproduction and Development Institute (iGReD), CNRS UMR6293, INSERM U1103, University of Clermont Auvergne, Clermont-Ferrand, France
| | - Krzysztof Jagla
- Genetics Reproduction and Development Institute (iGReD), CNRS UMR6293, INSERM U1103, University of Clermont Auvergne, Clermont-Ferrand, France.
| |
Collapse
|
17
|
Laurichesse Q, Moucaud B, Laddada L, Renaud Y, Jagla K, Soler C. Transcriptomic and Genetic Analyses Identify the Krüppel-Like Factor Dar1 as a New Regulator of Tube-Shaped Long Tendon Development. Front Cell Dev Biol 2021; 9:747563. [PMID: 34977007 PMCID: PMC8716952 DOI: 10.3389/fcell.2021.747563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 11/29/2021] [Indexed: 11/13/2022] Open
Abstract
To ensure locomotion and body stability, the active role of muscle contractions relies on a stereotyped muscle pattern set in place during development. This muscle patterning requires a precise assembly of the muscle fibers with the skeleton via a specialized connective tissue, the tendon. Like in vertebrate limbs, Drosophila leg muscles make connections with specific long tendons that extend through different segments. During the leg disc development, cell precursors of long tendons rearrange and collectively migrate to form a tube-shaped structure. A specific developmental program underlies this unique feature of tendon-like cells in the Drosophila model. We provide for the first time a transcriptomic profile of leg tendon precursors through fluorescence-based cell sorting. From promising candidates, we identified the Krüppel-like factor Dar1 as a critical actor of leg tendon development. Specifically expressed in the leg tendon precursors, loss of dar1 disrupts actin-rich filopodia formation and tendon elongation. Our findings show that Dar1 acts downstream of Stripe and is required to set up the correct number of tendon progenitors.
Collapse
|
18
|
Howard LJ, Reichert MC, Evans TA. The Slit-binding Ig1 domain is required for multiple axon guidance activities of Drosophila Robo2. Genesis 2021; 59:e23443. [PMID: 34411419 PMCID: PMC8446337 DOI: 10.1002/dvg.23443] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 07/28/2021] [Accepted: 08/01/2021] [Indexed: 12/02/2022]
Abstract
Drosophila Robo2 is a member of the evolutionarily conserved Roundabout (Robo) family of axon guidance receptors. Robo receptors signal midline repulsion in response to Slit ligands, which bind to the N‐terminal Ig1 domain in most family members. In the Drosophila embryonic ventral nerve cord, Robo1 and Robo2 signal Slit‐dependent midline repulsion, while Robo2 also regulates the medial‐lateral position of longitudinal axon pathways and acts non‐autonomously to promote midline crossing of commissural axons. While Robo2 signals midline repulsion in response to Slit, it is less clear whether Robo2's other activities are also Slit‐dependent. To determine which of Robo2's axon guidance roles depend on its Slit‐binding Ig1 domain, we used a clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9‐based strategy to replace the endogenous robo2 gene with a robo2 variant lacking the Ig1 domain (robo2∆Ig1). We compare the expression and localization of Robo2∆Ig1 protein with full‐length Robo2 in embryonic neurons in vivo and examine its ability to substitute for Robo2 to mediate midline repulsion and lateral axon pathway formation. We find that the removal of the Ig1 domain from Robo2∆Ig1 disrupts both of these axon guidance activities. In addition, we find that the Ig1 domain of Robo2 is required for its proper subcellular localization in embryonic neurons, a role that is not shared by the Ig1 domain of Robo1. Finally, we report that although FasII‐positive lateral axons are misguided in embryos expressing Robo2∆Ig1, the axons that normally express Robo2 are correctly guided to the lateral zone, suggesting that Robo2 may guide lateral longitudinal axons through a cell non‐autonomous mechanism.
Collapse
Affiliation(s)
- LaFreda J Howard
- Department of Biological Sciences, University of Arkansas, Fayetteville, Arkansas, USA.,City of Houston Health Department, University of Arkansas, Houston, Texas, USA
| | - Marie C Reichert
- Department of Biological Sciences, University of Arkansas, Fayetteville, Arkansas, USA
| | - Timothy A Evans
- Department of Biological Sciences, University of Arkansas, Fayetteville, Arkansas, USA
| |
Collapse
|
19
|
Ou T, Huang G, Wilson B, Gontarz P, Skeath JB, Johnson AN. A genetic screen for regulators of muscle morphogenesis in Drosophila. G3 (BETHESDA, MD.) 2021; 11:jkab172. [PMID: 33993253 PMCID: PMC8496313 DOI: 10.1093/g3journal/jkab172] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 05/03/2021] [Indexed: 11/14/2022]
Abstract
The mechanisms that determine the final topology of skeletal muscles remain largely unknown. We have been developing Drosophila body wall musculature as a model to identify and characterize the pathways that control muscle size, shape, and orientation during embryogenesis. Our working model argues muscle morphogenesis is regulated by (1) extracellular guidance cues that direct muscle cells toward muscle attachment sites, and (2) contact-dependent interactions between muscles and tendon cells. While we have identified several pathways that regulate muscle morphogenesis, our understanding is far from complete. Here, we report the results of a recent EMS-based forward genetic screen that identified a myriad of loci not previously associated with muscle morphogenesis. We recovered new alleles of known muscle morphogenesis genes, including back seat driver, kon-tiki, thisbe, and tumbleweed, arguing our screen had the depth and precision to uncover myogenic genes. We also identified new alleles of spalt-major, barren, and patched that presumably disrupt independent muscle morphogenesis pathways. Equally as important, our screen shows that at least 11 morphogenetic loci remain to be mapped and characterized. Our screen has developed exciting new tools to study muscle morphogenesis, which may provide future insights into the mechanisms that regulate skeletal muscle topology.
Collapse
Affiliation(s)
- Tiffany Ou
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Gary Huang
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Beth Wilson
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Paul Gontarz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - James B Skeath
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Aaron N Johnson
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
20
|
Yin C, Peterman E, Rasmussen JP, Parrish JZ. Transparent Touch: Insights From Model Systems on Epidermal Control of Somatosensory Innervation. Front Cell Neurosci 2021; 15:680345. [PMID: 34135734 PMCID: PMC8200473 DOI: 10.3389/fncel.2021.680345] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 04/28/2021] [Indexed: 12/28/2022] Open
Abstract
Somatosensory neurons (SSNs) densely innervate our largest organ, the skin, and shape our experience of the world, mediating responses to sensory stimuli including touch, pressure, and temperature. Historically, epidermal contributions to somatosensation, including roles in shaping innervation patterns and responses to sensory stimuli, have been understudied. However, recent work demonstrates that epidermal signals dictate patterns of SSN skin innervation through a variety of mechanisms including targeting afferents to the epidermis, providing instructive cues for branching morphogenesis, growth control and structural stability of neurites, and facilitating neurite-neurite interactions. Here, we focus onstudies conducted in worms (Caenorhabditis elegans), fruit flies (Drosophila melanogaster), and zebrafish (Danio rerio): prominent model systems in which anatomical and genetic analyses have defined fundamental principles by which epidermal cells govern SSN development.
Collapse
Affiliation(s)
| | | | | | - Jay Z. Parrish
- Department of Biology, University of Washington, Seattle, WA, United States
| |
Collapse
|
21
|
Nie J, Shao J, Guo SW, Liu X. The relevance of plasma R-spondin 1 and Slit2 as predictive biomarkers in cervical cancer chemotherapy and radiotherapy. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:837. [PMID: 34164471 PMCID: PMC8184459 DOI: 10.21037/atm-21-87] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Background R-spondin 1 (Rspol) and Slit2 have been found to play a vital role in cancer development, and have the potential to act as therapeutic adjuvants to increase tolerance to aggressive chemotherapy and/or radiotherapy. This “proof of concept” study evaluates the role of Rspo1 and Slit2 expression in the clinical outcome of cervical cancer patients. Methods Using enzyme linked immunosorbent assays (ELISA), we analyzed Rspo1 and Slit2 levels from patients diagnosed with the International Federation of Gynecology and Obstetrics (FIGO) stage IB1–IIA2 cervical cancer (n=34) who received chemotherapy (CT) and/or radiotherapy (RT) and correlated the data with the acute radiation morbidity scoring criteria. Results Cervical cancer patients who underwent CT and/or RT showed that neither the level of Rspo1 nor the level of Slit2 changed significantly after the first round of CT (CT1), RT, or the second CT (CT2). However, neurological sensory scores and influence of infection scores were elevated following increasing rounds of therapies. Rspo1 levels correlated negatively with the morbidity score of neutrophils, hemoglobin, platelet, infection score, neurological sensory score, and performance status after CT1, RT, or CT2. We also found that Slit2 levels were negatively correlated with genitourinary, heart, and neurological sensory scores at RT and CT2. Conclusions The levels of Rspo1 and Slit2 correlate positively to the tolerance of the patients. In contrast, the levels of Rspo1 and Slit2 showed a negative correlation to the morbidity score of the patients undergoing CT and/or RT. Thus, Rspo1 and Slit2 may be potential predictive biomarkers for patients with cervical cancer receiving CT or RT postoperatively, which supports the current pursuit of the clinical significance of Rspo1 and Slit2.
Collapse
Affiliation(s)
- Jichan Nie
- Gynecology Department, Shanghai Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Jun Shao
- Gynecology Department, Shanghai Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Sun-Wei Guo
- Gynecology Department, Shanghai Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Xishi Liu
- Gynecology Department, Shanghai Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| |
Collapse
|
22
|
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
|
23
|
Marulanda K, Brokaw D, Gambarian M, Pareta R, McQuilling JP, Opara EC, McLean SE. Controlled Delivery of Slit3 Proteins from Alginate Microbeads Inhibits In Vitro Angiogenesis. J Surg Res 2021; 264:90-98. [PMID: 33794389 DOI: 10.1016/j.jss.2021.01.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 12/15/2020] [Accepted: 01/25/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND The Slit-Robo pathway is a key regulator of angiogenesis and cellular function in experimental models. Slit3 proteins exhibit both proangiogenic and antiangiogenic properties, but the exact mechanism remains unclear. It is theorized that Slit3 may be a potential treatment for vascular diseases and cancer. METHODS Slit3 labeled with I-125 was encapsulated in microbeads composed of low-viscosity alginate of high-glucuronic acid content, first coated with poly-L-ornithine for various durations and finally with low-viscosity high mannuronic acid. Gamma counter was used to measure microbead encapsulation efficiency and Slit3 release. Markers of angiogenesis were assessed with Boyden chamber, scratch wound, and Matrigel tube formation assays using human umbilical vein and mouse endothelial cells. RESULTS On incubation of Slit3-loaded microbeads, there was an initial burst phase release of Slit3 for the first 24 h followed by sustained release for 6 to 12 d. Microbead composition determined encapsulation efficiency and rate of release; Slit3 encapsulation was most efficient in microbeads with lower low-viscosity alginate of high-glucuronic acid content concentrations (1.5%) and no poly-L-ornithine coating. Compared with controls (media alone), Slit3 microbeads significantly inhibited in vitro cellular migration, endothelial cell migration for wound closure at 24 and 48 h and endothelial tube formation (P < 0.001, respectively). CONCLUSIONS Slit3 can be effectively encapsulated and delivered via a controlled release pattern using alginate microbeads. Microbead encapsulation reduces in vitro endothelial tube formation and inhibits cellular migration to impair angiogenesis. Thus, Slit3 microparticles may be explored as a therapeutic option to mitigate tumor proliferation.
Collapse
Affiliation(s)
- Kathleen Marulanda
- Department of Surgery, University of North Carolina, Chapel Hill, North Carolina
| | - Dylan Brokaw
- Department of Surgery, University of North Carolina, Chapel Hill, North Carolina
| | - Maria Gambarian
- Department of Surgery, University of North Carolina, Chapel Hill, North Carolina
| | - Rajesh Pareta
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston Salem, North Carolina
| | - John P McQuilling
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston Salem, North Carolina
| | - Emmanuel C Opara
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston Salem, North Carolina
| | - Sean E McLean
- Department of Surgery, University of North Carolina, Chapel Hill, North Carolina.
| |
Collapse
|
24
|
Rafipay A, Dun X, Parkinson DB, Erskine L, Vargesson N. Knockdown of slit signaling during limb development leads to a reduction in humerus length. Dev Dyn 2021; 250:1340-1357. [DOI: 10.1002/dvdy.284] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 12/16/2020] [Accepted: 12/16/2020] [Indexed: 12/18/2022] Open
Affiliation(s)
- Alexandra Rafipay
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition University of Aberdeen Aberdeen UK
| | - Xin‐Peng Dun
- Peninsula Medical School, Faculty of Health University of Plymouth Plymouth UK
| | - David B Parkinson
- Peninsula Medical School, Faculty of Health University of Plymouth Plymouth UK
| | - Lynda Erskine
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition University of Aberdeen Aberdeen UK
| | - Neil Vargesson
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition University of Aberdeen Aberdeen UK
| |
Collapse
|
25
|
Chen CP, Wang LK, Chen CY, Chen CY, Wu YH. Placental multipotent mesenchymal stromal cell-derived Slit2 may regulate macrophage motility during placental infection. Mol Hum Reprod 2020; 27:6020263. [PMID: 33275139 DOI: 10.1093/molehr/gaaa076] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 10/26/2020] [Indexed: 12/17/2022] Open
Abstract
Slit proteins have been reported to act as axonal repellents in Drosophila; however, their role in the placental microenvironment has not been explored. In this study, we found that human placental multipotent mesenchymal stromal cells (hPMSCs) constitutively express Slit2. Therefore, we hypothesized that Slit2 expressed by hPMSCs could be involved in macrophage migration during placental inflammation through membrane cognate Roundabout (Robo) receptor signaling. In order to develop a preclinical in vitro mouse model of hPMSCs in treatment of perinatal infection, RAW 264.7 cells were used in this study. Slit2 interacted with Robo4 that was highly expressed in RAW 264.7 macrophages: their interaction increased the adhesive ability of RAW 264.7 cells and inhibited migration. Lipopolysaccharide (LPS)-induced CD11bCD18 expression could be inhibited by Slit2 and by hPMSC-conditioned medium (CM). LPS-induced activation of p38 and Rap1 was also attenuated by Slit2 and by hPMSC-CM. Noticeably, these inhibitory effects of hPMSC-CM decreased after depletion of Slit2 from the CM. Furthermore, we found that p38 siRNA inhibited LPS-induced Rap1 expression in RAW 264.7 cells, indicating that Rap1 functions downstream of p38 signaling. p38 siRNA increased cell adhesion and inhibited migration through reducing LPS-stimulated CD11bCD18 expression in RAW 264.7 cells. Thus, hPMSC-derived Slit2 may inhibit LPS-induced CD11bCD18 expression to decrease cell migration and increase adhesion through modulating the activity and motility of inflammatory macrophages in placenta. This may represent a novel mechanism for LPS-induced placental infection.
Collapse
Affiliation(s)
- Chie-Pein Chen
- Division of High Risk Pregnancy, MacKay Memorial Hospital, Taipei, Taiwan.,Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
| | - Liang-Kai Wang
- Division of High Risk Pregnancy, MacKay Memorial Hospital, Taipei, Taiwan
| | - Chen-Yu Chen
- Division of High Risk Pregnancy, MacKay Memorial Hospital, Taipei, Taiwan
| | - Chia-Yu Chen
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
| | - Yi-Hsin Wu
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
| |
Collapse
|
26
|
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
|
27
|
Poovathumkadavil P, Jagla K. Genetic Control of Muscle Diversification and Homeostasis: Insights from Drosophila. Cells 2020; 9:cells9061543. [PMID: 32630420 PMCID: PMC7349286 DOI: 10.3390/cells9061543] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 06/19/2020] [Accepted: 06/23/2020] [Indexed: 12/13/2022] Open
Abstract
In the fruit fly, Drosophila melanogaster, the larval somatic muscles or the adult thoracic flight and leg muscles are the major voluntary locomotory organs. They share several developmental and structural similarities with vertebrate skeletal muscles. To ensure appropriate activity levels for their functions such as hatching in the embryo, crawling in the larva, and jumping and flying in adult flies all muscle components need to be maintained in a functionally stable or homeostatic state despite constant strain. This requires that the muscles develop in a coordinated manner with appropriate connections to other cell types they communicate with. Various signaling pathways as well as extrinsic and intrinsic factors are known to play a role during Drosophila muscle development, diversification, and homeostasis. In this review, we discuss genetic control mechanisms of muscle contraction, development, and homeostasis with particular emphasis on the contractile unit of the muscle, the sarcomere.
Collapse
|
28
|
Gong L, Wang S, Shen L, Liu C, Shenouda M, Li B, Liu X, Shaw JA, Wineman AL, Yang Y, Xiong D, Eichmann A, Evans SM, Weiss SJ, Si MS. SLIT3 deficiency attenuates pressure overload-induced cardiac fibrosis and remodeling. JCI Insight 2020; 5:136852. [PMID: 32644051 PMCID: PMC7406261 DOI: 10.1172/jci.insight.136852] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 05/06/2020] [Indexed: 01/28/2023] Open
Abstract
In pulmonary hypertension and certain forms of congenital heart disease, ventricular pressure overload manifests at birth and is an obligate hemodynamic abnormality that stimulates myocardial fibrosis, which leads to ventricular dysfunction and poor clinical outcomes. Thus, an attractive strategy is to attenuate the myocardial fibrosis to help preserve ventricular function. Here, by analyzing RNA-sequencing databases and comparing the transcript and protein levels of fibrillar collagen in WT and global-knockout mice, we found that slit guidance ligand 3 (SLIT3) was present predominantly in fibrillar collagen-producing cells and that SLIT3 deficiency attenuated collagen production in the heart and other nonneuronal tissues. We then performed transverse aortic constriction or pulmonary artery banding to induce left and right ventricular pressure overload, respectively, in WT and knockout mice. We discovered that SLIT3 deficiency abrogated fibrotic and hypertrophic changes and promoted long-term ventricular function and overall survival in both left and right ventricular pressure overload. Furthermore, we found that SLIT3 stimulated fibroblast activity and fibrillar collagen production, which coincided with the transcription and nuclear localization of the mechanotransducer yes-associated protein 1. These results indicate that SLIT3 is important for regulating fibroblast activity and fibrillar collagen synthesis in an autocrine manner, making it a potential therapeutic target for fibrotic diseases, especially myocardial fibrosis and adverse remodeling induced by persistent afterload elevation.
Collapse
Affiliation(s)
- Lianghui Gong
- Section of Pediatric Cardiovascular Surgery, Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA.,Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Shuyun Wang
- Section of Pediatric Cardiovascular Surgery, Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Li Shen
- Section of Pediatric Cardiovascular Surgery, Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Catherine Liu
- Section of Pediatric Cardiovascular Surgery, Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Mena Shenouda
- Section of Pediatric Cardiovascular Surgery, Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Baolei Li
- Section of Pediatric Cardiovascular Surgery, Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Xiaoxiao Liu
- Section of Pediatric Cardiovascular Surgery, Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Alan L. Wineman
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Yifeng Yang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Dingding Xiong
- Section of Pediatric Cardiovascular Surgery, Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Anne Eichmann
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.,Paris Cardiovascular Research Center, INSERM U970, Paris, France.,Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sylvia M. Evans
- Skaggs School of Pharmacy and Pharmaceutical Sciences,,Department of Medicine, and,Department of Pharmacology, UCSD, La Jolla, California, USA
| | - Stephen J. Weiss
- Division of Genetic Medicine,,Department of Internal Medicine,,Life Sciences Institute,,Cellular and Molecular Biology Graduate Program, and,Rogel Cancer Center, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Ming-Sing Si
- Section of Pediatric Cardiovascular Surgery, Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
29
|
Strenzke M, Alberton P, Aszodi A, Docheva D, Haas E, Kammerlander C, Böcker W, Saller MM. Tenogenic Contribution to Skeletal Muscle Regeneration: The Secretome of Scleraxis Overexpressing Mesenchymal Stem Cells Enhances Myogenic Differentiation In Vitro. Int J Mol Sci 2020; 21:E1965. [PMID: 32183051 PMCID: PMC7139530 DOI: 10.3390/ijms21061965] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/09/2020] [Accepted: 03/12/2020] [Indexed: 01/07/2023] Open
Abstract
Integrity of the musculoskeletal system is essential for the transfer of muscular contraction force to the associated bones. Tendons and skeletal muscles intertwine, but on a cellular level, the myotendinous junctions (MTJs) display a sharp transition zone with a highly specific molecular adaption. The function of MTJs could go beyond a mere structural role and might include homeostasis of this musculoskeletal tissue compound, thus also being involved in skeletal muscle regeneration. Repair processes recapitulate several developmental mechanisms, and as myotendinous interaction does occur already during development, MTJs could likewise contribute to muscle regeneration. Recent studies identified tendon-related, scleraxis-expressing cells that reside in close proximity to the MTJs and the muscle belly. As the muscle-specific function of these scleraxis positive cells is unknown, we compared the influence of two immortalized mesenchymal stem cell (MSC) lines-differing only by the overexpression of scleraxis-on myoblasts morphology, metabolism, migration, fusion, and alignment. Our results revealed a significant increase in myoblast fusion and metabolic activity when exposed to the secretome derived from scleraxis-overexpressing MSCs. However, we found no significant changes in myoblast migration and myofiber alignment. Further analysis of differentially expressed genes between native MSCs and scleraxis-overexpressing MSCs by RNA sequencing unraveled potential candidate genes, i.e., extracellular matrix (ECM) proteins, transmembrane receptors, or proteases that might enhance myoblast fusion. Our results suggest that musculotendinous interaction is essential for the development and healing of skeletal muscles.
Collapse
Affiliation(s)
- Maximilian Strenzke
- Experimental Surgery and Regenerative Medicine (ExperiMed), Department of General Trauma and Reconstructive Surgery, Ludwig-Maximilians-University (LMU), Fraunhoferstraße 20, 82152 Planegg-Martinsried, Germany; (M.S.); (P.A.); (A.A.); (E.H.); (C.K.); (W.B.)
| | - Paolo Alberton
- Experimental Surgery and Regenerative Medicine (ExperiMed), Department of General Trauma and Reconstructive Surgery, Ludwig-Maximilians-University (LMU), Fraunhoferstraße 20, 82152 Planegg-Martinsried, Germany; (M.S.); (P.A.); (A.A.); (E.H.); (C.K.); (W.B.)
| | - Attila Aszodi
- Experimental Surgery and Regenerative Medicine (ExperiMed), Department of General Trauma and Reconstructive Surgery, Ludwig-Maximilians-University (LMU), Fraunhoferstraße 20, 82152 Planegg-Martinsried, Germany; (M.S.); (P.A.); (A.A.); (E.H.); (C.K.); (W.B.)
| | - Denitsa Docheva
- Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, 93053 Regensburg, Germany;
| | - Elisabeth Haas
- Experimental Surgery and Regenerative Medicine (ExperiMed), Department of General Trauma and Reconstructive Surgery, Ludwig-Maximilians-University (LMU), Fraunhoferstraße 20, 82152 Planegg-Martinsried, Germany; (M.S.); (P.A.); (A.A.); (E.H.); (C.K.); (W.B.)
- Division of Hand, Plastic and Aesthetic Surgery, Ludwig-Maximilians-University (LMU), Pettenkoferstraße 8a, 80336 Munich, Germany
| | - Christian Kammerlander
- Experimental Surgery and Regenerative Medicine (ExperiMed), Department of General Trauma and Reconstructive Surgery, Ludwig-Maximilians-University (LMU), Fraunhoferstraße 20, 82152 Planegg-Martinsried, Germany; (M.S.); (P.A.); (A.A.); (E.H.); (C.K.); (W.B.)
| | - Wolfgang Böcker
- Experimental Surgery and Regenerative Medicine (ExperiMed), Department of General Trauma and Reconstructive Surgery, Ludwig-Maximilians-University (LMU), Fraunhoferstraße 20, 82152 Planegg-Martinsried, Germany; (M.S.); (P.A.); (A.A.); (E.H.); (C.K.); (W.B.)
| | - Maximilian Michael Saller
- Experimental Surgery and Regenerative Medicine (ExperiMed), Department of General Trauma and Reconstructive Surgery, Ludwig-Maximilians-University (LMU), Fraunhoferstraße 20, 82152 Planegg-Martinsried, Germany; (M.S.); (P.A.); (A.A.); (E.H.); (C.K.); (W.B.)
| |
Collapse
|
30
|
Yang S, Weske A, Du Y, Valera JM, Jones KL, Johnson AN. FGF signaling directs myotube guidance by regulating Rac activity. Development 2020; 147:dev.183624. [PMID: 31932350 DOI: 10.1242/dev.183624] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 12/27/2019] [Indexed: 01/25/2023]
Abstract
Nascent myotubes undergo a dramatic morphological transformation during myogenesis, in which the myotubes elongate over several cell diameters and are directed to the correct muscle attachment sites. Although this process of myotube guidance is essential to pattern the musculoskeletal system, the mechanisms that control myotube guidance remain poorly understood. Using transcriptomics, we found that components of the Fibroblast Growth Factor (FGF) signaling pathway were enriched in nascent myotubes in Drosophila embryos. Null mutations in the FGF receptor heartless (htl), or its ligands, caused significant myotube guidance defects. The FGF ligand Pyramus is expressed broadly in the ectoderm, and ectopic Pyramus expression disrupted muscle patterning. Mechanistically, Htl regulates the activity of Rho/Rac GTPases in nascent myotubes and effects changes in the actin cytoskeleton. FGF signals are thus essential regulators of myotube guidance that act through cytoskeletal regulatory proteins to pattern the musculoskeletal system.
Collapse
Affiliation(s)
- Shuo Yang
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Allison Weske
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Yingqiu Du
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Juliana M Valera
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Kenneth L Jones
- Hematology, Oncology, and Bone Marrow Transplant, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Aaron N Johnson
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| |
Collapse
|
31
|
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
|
32
|
Richier B, Inoue Y, Dobramysl U, Friedlander J, Brown NH, Gallop JL. Integrin signaling downregulates filopodia during muscle-tendon attachment. J Cell Sci 2018; 131:jcs.217133. [PMID: 30054384 PMCID: PMC6127725 DOI: 10.1242/jcs.217133] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 07/12/2018] [Indexed: 11/25/2022] Open
Abstract
Cells need to sense their environment to ensure accurate targeting to specific destinations. This occurs in developing muscles, which need to attach to tendon cells before muscle contractions can begin. Elongating myotube tips form filopodia, which are presumed to have sensory roles, and are later suppressed upon building the attachment site. Here, we use live imaging and quantitative image analysis of lateral transverse (LT) myotubes in Drosophila to show that filopodia suppression occurs as a result of integrin signaling. Loss of the integrin subunits αPS2 and βPS (also known as If and Mys, respectively, in flies) increased filopodia number and length at stages when they are normally suppressed. Conversely, inducing integrin signaling, achieved by the expression of constitutively dimerised βPS cytoplasmic domain (diβ), prematurely suppressed filopodia. We discovered that the integrin signal is transmitted through the protein G protein-coupled receptor kinase interacting ArfGAP (Git) and its downstream kinase p21-activated kinase (Pak). Absence of these proteins causes profuse filopodia and prevents the filopodial inhibition mediated by diβ. Thus, integrin signaling terminates the exploratory behavior of myotubes seeking tendons, enabling the actin machinery to focus on forming a strong attachment and assembling the contractile apparatus. Summary: Integrins signal through Git and Pak to downregulate filopodia when muscles reach their target attachment site in Drosophila.
Collapse
Affiliation(s)
- Benjamin Richier
- The Gurdon Institute, Tennis Court Rd, Cambridge CB2 1QN, UK.,Dept. of Biochemistry, University of Cambridge, Cambridge CB2 1GA, UK
| | - Yoshiko Inoue
- The Gurdon Institute, Tennis Court Rd, Cambridge CB2 1QN, UK.,Dept. of Biochemistry, University of Cambridge, Cambridge CB2 1GA, UK.,Dept. of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK
| | - Ulrich Dobramysl
- The Gurdon Institute, Tennis Court Rd, Cambridge CB2 1QN, UK.,Dept. of Biochemistry, University of Cambridge, Cambridge CB2 1GA, UK
| | - Jonathan Friedlander
- The Gurdon Institute, Tennis Court Rd, Cambridge CB2 1QN, UK.,Dept. of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK
| | - Nicholas H Brown
- The Gurdon Institute, Tennis Court Rd, Cambridge CB2 1QN, UK.,Dept. of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK
| | - Jennifer L Gallop
- The Gurdon Institute, Tennis Court Rd, Cambridge CB2 1QN, UK .,Dept. of Biochemistry, University of Cambridge, Cambridge CB2 1GA, UK
| |
Collapse
|
33
|
Liu KY, Sengillo JD, Velez G, Jauregui R, Sakai LY, Maumenee IH, Bassuk AG, Mahajan VB, Tsang SH. Missense mutation in SLIT2 associated with congenital myopia, anisometropia, connective tissue abnormalities, and obesity. Orphanet J Rare Dis 2018; 13:138. [PMID: 30111362 PMCID: PMC6094464 DOI: 10.1186/s13023-018-0885-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 07/31/2018] [Indexed: 12/26/2022] Open
Abstract
Background SLIT2 is a protein ligand for the Roundabout (ROBO) receptor and was found to play a major role in repulsive midline axon guidance in central nervous system development. Based on studies utilizing knockout models, it has been postulated that SLIT2 is important for preventing inappropriate axonal routing during mammalian optic chiasm development. Methods Case report. Results Here, we report a case of congenital myopia, anisometropia, and obesity in a patient with a SLIT2 point mutation. Examination of the patient’s skin biopsy revealed abnormalities in elastin and collagen fibrils that suggest an underlying connective tissue disorder. Structural modeling placed the novel mutation (p.D1407G) in the EGF-like domain 8 and was predicted to affect interactions with SLIT2 binding partners. Conclusions To the authors’ knowledge, this is the first report of a SLIT2 variant in the context of these ocular findings. Electronic supplementary material The online version of this article (10.1186/s13023-018-0885-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Katherine Y Liu
- Stony Brook University School of Medicine, Stony Brook, NY, USA
| | - Jesse D Sengillo
- Department of Ophthalmology, Columbia University, New York, NY, USA.,Department of Medicine, Reading Hospital, West Reading, PA, USA
| | - Gabriel Velez
- Omics Laboratory, Stanford University, Palo Alto, CA, USA.,Department of Ophthalmology, Byers Eye Institute, Stanford University, Palo Alto, CA, USA.,Medical Scientist Training Program, University of Iowa, Iowa City, IA, USA
| | - Ruben Jauregui
- Department of Ophthalmology, Columbia University, New York, NY, USA.,Weill Cornell Medical College, New York, NY, USA
| | - Lynn Y Sakai
- Departments of Molecular and Medical Genetics and Biochemistry and Molecular Biology, Oregon Health and Science University and Shriners Hospital for Children, Portland, USA
| | - Irene H Maumenee
- Department of Ophthalmology, Columbia University, New York, NY, USA
| | | | - Vinit B Mahajan
- Omics Laboratory, Stanford University, Palo Alto, CA, USA.,Department of Ophthalmology, Byers Eye Institute, Stanford University, Palo Alto, CA, USA.,Palo Alto Veterans Administration, Palo Alto, CA, USA
| | - Stephen H Tsang
- Jonas Children's Vision Care, and Bernard and Shirlee Brown Glaucoma Laboratory, New York, USA. .,Department of Ophthalmology, Columbia University, New York, NY, USA. .,Department of Pathology and Cell Biology, Stem Cell Initiative (CSCI), Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, NY, USA. .,Harkness Eye Institute, Columbia University Medical Center, 635 West 165th Street, Box 212, New York, NY, 10032, USA.
| |
Collapse
|
34
|
Xu R, Qin N, Xu X, Sun X, Chen X, Zhao J. Inhibitory effect of SLIT2 on granulosa cell proliferation mediated by the CDC42-PAKs-ERK1/2 MAPK pathway in the prehierarchical follicles of the chicken ovary. Sci Rep 2018; 8:9168. [PMID: 29907785 PMCID: PMC6003946 DOI: 10.1038/s41598-018-27601-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 06/06/2018] [Indexed: 01/09/2023] Open
Abstract
The SLIT2 ligand and ROBO receptors of the SLIT/ROBO pathway are expressed in hen ovarian follicles and have been shown to play critical roles in ovary development, cell proliferation and apoptosis in mammals. However, the exact roles of SLIT2 and the molecular mechanisms of chicken follicle development remain poorly understood. Here, we discovered that high levels of SLIT2 suppress FSHR, GDF9, STAR and CYP11A1 mRNA and protein expression in granulosa cells (GCs) and cell proliferation (p < 0.01). However, these inhibitory effects can be abolished by the siRNA-mediated knockdown of the ROBO1 and ROBO2 receptors. Furthermore, the activity of CDC42, which is a key Rho GTPase in the SLIT/ROBO pathway, is regulated by the ligand SLIT2 because the intrinsic GTPase activation activity of CDC42 is activated or repressed by regulating SRGAP1 expression (p < 0.01). The effects of the SLIT2 overexpression on GC proliferation and phosphorylation of the B-RAF, RAF1 and ERK1/2 kinases were completely abrogated by knocking down endogenous PAK1 and partially abrogated by the knockdown of PAK2 and PAK3 in the GCs. Collectively, our findings indicate that SLIT2 suppresses GC proliferation, differentiation and follicle selection mainly by a mechanism involving ROBO1 and ROBO2 and that this suppression is mediated by the CDC42-PAKs-ERK1/2 MAPK signaling cascade in the prehierarchical follicles of the chicken ovary.
Collapse
Affiliation(s)
- Rifu Xu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, People's Republic of China. .,Key Laboratory of Animal Production and Product Quality Safety of the Ministry of Education, Changchun, 130118, People's Republic of China.
| | - Ning Qin
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, People's Republic of China.,Key Laboratory of Animal Production and Product Quality Safety of the Ministry of Education, Changchun, 130118, People's Republic of China
| | - Xiaoxing Xu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, People's Republic of China
| | - Xue Sun
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, People's Republic of China
| | - Xiaoxia Chen
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, People's Republic of China
| | - Jinghua Zhao
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, People's Republic of China
| |
Collapse
|
35
|
Kim BJ, Lee YS, Lee SY, Baek WY, Choi YJ, Moon SA, Lee SH, Kim JE, Chang EJ, Kim EY, Yoon J, Kim SW, Ryu SH, Lee SK, Lorenzo JA, Ahn SH, Kim H, Lee KU, Kim GS, Koh JM. Osteoclast-secreted SLIT3 coordinates bone resorption and formation. J Clin Invest 2018; 128:1429-1441. [PMID: 29504949 PMCID: PMC5873876 DOI: 10.1172/jci91086] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 12/28/2017] [Indexed: 01/15/2023] Open
Abstract
Coupling is the process that links bone resorption to bone formation in a temporally and spatially coordinated manner within the remodeling cycle. Several lines of evidence point to the critical roles of osteoclast-derived coupling factors in the regulation of osteoblast performance. Here, we used a fractionated secretomic approach and identified the axon-guidance molecule SLIT3 as a clastokine that stimulated osteoblast migration and proliferation by activating β-catenin. SLIT3 also inhibited bone resorption by suppressing osteoclast differentiation in an autocrine manner. Mice deficient in Slit3 or its receptor, Robo1, exhibited osteopenic phenotypes due to a decrease in bone formation and increase in bone resorption. Mice lacking Slit3 specifically in osteoclasts had low bone mass, whereas mice with either neuron-specific Slit3 deletion or osteoblast-specific Slit3 deletion had normal bone mass, thereby indicating the importance of SLIT3 as a local determinant of bone metabolism. In postmenopausal women, higher circulating SLIT3 levels were associated with increased bone mass. Notably, injection of a truncated recombinant SLIT3 markedly rescued bone loss after an ovariectomy. Thus, these results indicate that SLIT3 plays an osteoprotective role by synchronously stimulating bone formation and inhibiting bone resorption, making it a potential therapeutic target for metabolic bone diseases.
Collapse
Affiliation(s)
- Beom-Jun Kim
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Young-Sun Lee
- Asan Institute for Life Sciences, Seoul, South Korea
| | - Sun-Young Lee
- Asan Institute for Life Sciences, Seoul, South Korea
| | | | | | - Sung Ah Moon
- Asan Institute for Life Sciences, Seoul, South Korea
| | - Seung Hun Lee
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jung-Eun Kim
- Department of Molecular Medicine, Cell and Matrix Research Institute, Kyungpook National University School of Medicine, Daegu, South Korea
| | | | | | - Jin Yoon
- Department of Pharmacology, University of Ulsan College of Medicine, Seoul, South Korea
| | - Seung-Whan Kim
- Department of Pharmacology, University of Ulsan College of Medicine, Seoul, South Korea
| | - Sung Ho Ryu
- Department of Life Science, Pohang University of Science and Technology, Pohang, Kyungbook, South Korea
| | | | - Joseph A. Lorenzo
- Departments of Medicine and Orthopaedics, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Seong Hee Ahn
- Department of Internal Medicine, Inha University School of Medicine, Incheon, South Korea
| | - Hyeonmok Kim
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Ki-Up Lee
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Ghi Su Kim
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jung-Min Koh
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| |
Collapse
|
36
|
Brown HE, Reichert MC, Evans TA. In Vivo Functional Analysis of Drosophila Robo1 Fibronectin Type-III Repeats. G3 (BETHESDA, MD.) 2018; 8:621-630. [PMID: 29217730 PMCID: PMC5919748 DOI: 10.1534/g3.117.300418] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 12/05/2017] [Indexed: 12/02/2022]
Abstract
The repellant ligand Slit and its Roundabout (Robo) family receptors regulate midline crossing of axons during development of the embryonic central nervous system (CNS). Slit proteins are produced at the midline and signal through Robo receptors to repel axons from the midline. Disruption of Slit-Robo signaling causes ectopic midline-crossing phenotypes in the CNS of a broad range of animals, including insects and vertebrates. While previous studies have investigated the roles of Drosophila melanogaster Robo1's five Immunoglobulin-like (Ig) domains, little is known about the importance of the three evolutionarily conserved Fibronectin (Fn) type-III repeats. We have individually deleted each of Drosophila Robo1's three Fn repeats, and then tested these Robo1 variants in vitro to determine their ability to bind Slit in cultured Drosophila cells and in vivo to investigate the requirement for each domain in regulating Robo1's embryonic expression pattern, axonal localization, midline repulsive function, and sensitivity to Commissureless (Comm) downregulation. We demonstrate that the Fn repeats are not required for Robo1 to bind Slit or for proper expression of Robo1 in Drosophila embryonic neurons. When expressed in a robo1 mutant background, these variants are able to restore midline repulsion to an extent equivalent to full-length Robo1. We identify a novel requirement for Fn3 in the exclusion of Robo1 from commissures and downregulation of Robo1 by Comm. Our results indicate that each of the Drosophila Robo1 Fn repeats are individually dispensable for the protein's role in midline repulsion, despite the evolutionarily conserved "5 + 3" protein structure.
Collapse
Affiliation(s)
- Haley E Brown
- Department of Biological Sciences, University of Arkansas, Fayetteville, Arkansas 72701
| | - Marie C Reichert
- Department of Biological Sciences, University of Arkansas, Fayetteville, Arkansas 72701
| | - Timothy A Evans
- Department of Biological Sciences, University of Arkansas, Fayetteville, Arkansas 72701
| |
Collapse
|
37
|
Lee JK, Hallock PT, Burden SJ. Abelson tyrosine-protein kinase 2 regulates myoblast proliferation and controls muscle fiber length. eLife 2017; 6:29905. [PMID: 29231808 PMCID: PMC5752197 DOI: 10.7554/elife.29905] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Accepted: 12/02/2017] [Indexed: 01/15/2023] Open
Abstract
Muscle fiber length is nearly uniform within a muscle but widely different among different muscles. We show that Abelson tyrosine-protein kinase 2 (Abl2) has a key role in regulating myofiber length, as a loss of Abl2 leads to excessively long myofibers in the diaphragm, intercostal and levator auris muscles but not limb muscles. Increased myofiber length is caused by enhanced myoblast proliferation, expanding the pool of myoblasts and leading to increased myoblast fusion. Abl2 acts in myoblasts, but as a consequence of expansion of the diaphragm muscle, the diaphragm central tendon is reduced in size, likely contributing to reduced stamina of Abl2 mutant mice. Ectopic muscle islands, each composed of myofibers of uniform length and orientation, form within the central tendon of Abl2+/− mice. Specialized tendon cells, resembling tendon cells at myotendinous junctions, form at the ends of these muscle islands, suggesting that myofibers induce differentiation of tendon cells, which reciprocally regulate myofiber length and orientation.
Collapse
Affiliation(s)
- Jennifer K Lee
- Helen L and Martin S Kimmel Center for Biology and Medicine at the Skirball Institute of Biomolecular Medicine, NYU Medical School, New York, United States
| | - Peter T Hallock
- Helen L and Martin S Kimmel Center for Biology and Medicine at the Skirball Institute of Biomolecular Medicine, NYU Medical School, New York, United States
| | - Steven J Burden
- Helen L and Martin S Kimmel Center for Biology and Medicine at the Skirball Institute of Biomolecular Medicine, NYU Medical School, New York, United States
| |
Collapse
|
38
|
Pons M, Soulard C, Soustelle L, Parmentier ML, Grau Y, Layalle S. A New Behavioral Test and Associated Genetic Tools Highlight the Function of Ventral Abdominal Muscles in Adult Drosophila. Front Cell Neurosci 2017; 11:371. [PMID: 29209177 PMCID: PMC5702315 DOI: 10.3389/fncel.2017.00371] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 11/08/2017] [Indexed: 11/23/2022] Open
Abstract
The function of the nervous system in complex animals is reflected by the achievement of specific behaviors. For years in Drosophila, both simple and complex behaviors have been studied and their genetic bases have emerged. The neuromuscular junction is maybe one of the prototypal simplest examples. A motor neuron establishes synaptic connections on its muscle cell target and elicits behavior: the muscle contraction. Different muscles in adult fly are related to specific behaviors. For example, the thoracic muscles are associated with flight and the leg muscles are associated with locomotion. However, specific tools are still lacking for the study of cellular physiology in distinct motor neuron subpopulations. Here we decided to use the abdominal muscles and in particular the ventral abdominal muscles (VAMs) in adult Drosophila as new model to link a precise behavior to specific motor neurons. Hence, we developed a new behavioral test based on the folding movement of the adult abdomen. Further, we performed a genetic screen and identify two specific Gal4 lines with restricted expression patterns to the adult motor neurons innervating the VAMs or their precursor cells. Using these genetic tools, we showed that the lack of the VAMs or the loss of the synaptic transmission in their innervating motor neurons lead to a significant impairment of the abdomen folding behavior. Altogether, our results allow establishing a direct link between specific motor neurons and muscles for the realization of particular behavior: the folding behavior of the abdomen in Drosophila.
Collapse
Affiliation(s)
- Marine Pons
- Institut de Génomique Fonctionnelle, CNRS, INSERM, Université de Montpellier, Montpellier, France
| | - Claire Soulard
- Institut de Génomique Fonctionnelle, CNRS, INSERM, Université de Montpellier, Montpellier, France
| | - Laurent Soustelle
- Institut de Génomique Fonctionnelle, CNRS, INSERM, Université de Montpellier, Montpellier, France
| | - Marie-Laure Parmentier
- Institut de Génomique Fonctionnelle, CNRS, INSERM, Université de Montpellier, Montpellier, France
| | - Yves Grau
- Institut de Génomique Fonctionnelle, CNRS, INSERM, Université de Montpellier, Montpellier, France
| | - Sophie Layalle
- Institut de Génomique Fonctionnelle, CNRS, INSERM, Université de Montpellier, Montpellier, France
| |
Collapse
|
39
|
Ahmad SM. Conserved signaling mechanisms in Drosophila heart development. Dev Dyn 2017; 246:641-656. [PMID: 28598558 PMCID: PMC11546222 DOI: 10.1002/dvdy.24530] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 04/06/2017] [Accepted: 05/08/2017] [Indexed: 12/24/2022] Open
Abstract
Signal transduction through multiple distinct pathways regulates and orchestrates the numerous biological processes comprising heart development. This review outlines the roles of the FGFR, EGFR, Wnt, BMP, Notch, Hedgehog, Slit/Robo, and other signaling pathways during four sequential phases of Drosophila cardiogenesis-mesoderm migration, cardiac mesoderm establishment, differentiation of the cardiac mesoderm into distinct cardiac cell types, and morphogenesis of the heart and its lumen based on the proper positioning and cell shape changes of these differentiated cardiac cells-and illustrates how these same cardiogenic roles are conserved in vertebrates. Mechanisms bringing about the regulation and combinatorial integration of these diverse signaling pathways in Drosophila are also described. This synopsis of our present state of knowledge of conserved signaling pathways in Drosophila cardiogenesis and the means by which it was acquired should facilitate our understanding of and investigations into related processes in vertebrates. Developmental Dynamics 246:641-656, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Shaad M. Ahmad
- Department of Biology, Indiana State University, Terre Haute, IN, USA
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN, USA
| |
Collapse
|
40
|
Kang YE, Choung S, Lee JH, Kim HJ, Ku BJ. The Role of Circulating Slit2, the One of the Newly Batokines, in Human Diabetes Mellitus. Endocrinol Metab (Seoul) 2017; 32:383-388. [PMID: 28956369 PMCID: PMC5620036 DOI: 10.3803/enm.2017.32.3.383] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 07/25/2017] [Accepted: 08/04/2017] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Slit2 is a new secreted protein from adipose tissue that improves glucose hemostasis in mice; however, there is no study about the serum levels and precise role of Slit2 in human. The aim of this study is to explore the serum level of Slit2 in human, and to identify the role of Slit2 in diabetes mellitus (DM). METHODS The participants of this study consist of 38 subjects with newly diagnosed DM, and 75 healthy subjects as a control group. Serum Slit2 levels were measured using an enzyme-linked immunosorbent assay. Relationship between circulating Slit2 and diabetic related factors was investigated in diabetic group compared with non-diabetic group. Additionally, the correlations between the serum level of Slit2 and diverse metabolic parameters were analyzed. RESULTS Circulating Slit2 level was more decreased in diabetic group than in control group, but there was no significant difference statistically. Interestingly, serum levels of Slit2 were significantly negatively correlated to the serum concentrations of fasting glucose (coefficient r=-0.246, P=0.008), the serum concentrations of postprandial glucose (coefficient r=-0.233, P=0.017), and glycosylated hemoglobin (HbA1c; coefficient r=-0.357, P<0.001). CONCLUSION From our study, the first report of circulating Slit2 levels in human, circulating Slit2 level significantly negatively correlated with serum glucose and HbA1c. Our results suggest that the circulating Slit2 may play a role in maintainence of glucose homeostasis in human, even though exact contribution and mechanism are not yet known.
Collapse
Affiliation(s)
- Yea Eun Kang
- Department of Endocrinology and Metabolism, Chungnam National University School of Medicine, Daejeon, Korea
| | - Sorim Choung
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Korea
| | - Ju Hee Lee
- Department of Endocrinology and Metabolism, Chungnam National University School of Medicine, Daejeon, Korea
| | - Hyun Jin Kim
- Department of Endocrinology and Metabolism, Chungnam National University School of Medicine, Daejeon, Korea
| | - Bon Jeong Ku
- Department of Endocrinology and Metabolism, Chungnam National University School of Medicine, Daejeon, Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Korea.
| |
Collapse
|
41
|
Bashamboo A, Bignon-Topalovic J, Moussi N, McElreavey K, Brauner R. Mutations in the Human ROBO1 Gene in Pituitary Stalk Interruption Syndrome. J Clin Endocrinol Metab 2017; 102:2401-2406. [PMID: 28402530 DOI: 10.1210/jc.2016-1095] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 04/06/2017] [Indexed: 12/21/2022]
Abstract
Context Pituitary stalk interruption syndrome (PSIS) is characterized by a thin or absent pituitary stalk usually in association with an ectopic posterior pituitary and hypoplasia/aplasia of the anterior pituitary. Associated phenotypes include varied ocular anomalies, hypoglycemia, micropenis/cryptorchidism, growth failure, or combined pituitary hormone deficiencies. Although genetic causes have been identified, they explain only around 5% of PSIS cases. Objective To identify genetic causes of PSIS by exome sequencing. Design Exon enrichment was performed using the Agilent SureSelect Human All Exon V4. Paired-end sequencing was performed on the Illumina HiSeq2000 platform with an average sequencing coverage of ×50. Patients Patients with unexplained PSIS were included in the study. Results In five cases of unexplained PSIS including two familial cases, we identified a novel heterozygous frameshift and nonsense and missense mutations in the ROBO1 gene (p.Ala977Glnfs*40, two affected sibs; p.Tyr1114Ter, sporadic case, and p.Cys240Ser, affected child and paternal aunt) that controls embryonic axon guidance, and branching in the nervous system. Interestingly, four of the five cases of PSIS also presented with ocular anomalies, including hypermetropia with strabismus as well as ptosis. Conclusions These data suggest that mutations in ROBO1 contribute to PSIS and associated ocular anomalies.
Collapse
Affiliation(s)
- Anu Bashamboo
- Human Developmental Genetics, Institut Pasteur, 75015 Paris, France
| | | | - Nasser Moussi
- Human Developmental Genetics, Institut Pasteur, 75015 Paris, France
| | - Ken McElreavey
- Human Developmental Genetics, Institut Pasteur, 75015 Paris, France
| | - Raja Brauner
- Université Paris Descartes and Pediatric Endocrinology Unit, Fondation Ophtalmologique Adolphe de Rothschild, 75019 Paris, France
| |
Collapse
|
42
|
Sasse S, Klämbt C. Repulsive Epithelial Cues Direct Glial Migration along the Nerve. Dev Cell 2017; 39:696-707. [PMID: 27997826 DOI: 10.1016/j.devcel.2016.11.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 10/14/2016] [Accepted: 11/19/2016] [Indexed: 11/29/2022]
Abstract
Most glial cells show pronounced migratory abilities and generally follow axonal trajectories to reach their final destination. However, the molecular cues controlling their directional migration are largely unknown. To address this, we established glial migration onto the developing Drosophila leg imaginal disc as a model. Here, CNS-derived glial cells move along nerves containing motoaxons and sensory axons. Along their path, glial cells encounter at least three choice points where directional decisions are needed. Subsequent genetic analyses allowed uncovering mechanisms that escaped previous studies. Most strikingly, we found that glial cells require the expression of the repulsive guidance receptors PlexinA/B and Robo2 to prevent breaking away from the nerve. Interestingly, the repulsive ligands are presented by the underlying leg imaginal disc epithelium, which appears to push glial cells toward the axon fascicle. In conclusion, nerve formation not only requires neuron-glia interaction but also depends on glial-epithelial communication.
Collapse
Affiliation(s)
- Sofia Sasse
- Institut für Neuro- und Verhaltensbiologie, Badestraße 9, 48149 Münster, Germany
| | - Christian Klämbt
- Institut für Neuro- und Verhaltensbiologie, Badestraße 9, 48149 Münster, Germany.
| |
Collapse
|
43
|
Nassari S, Duprez D, Fournier-Thibault C. Non-myogenic Contribution to Muscle Development and Homeostasis: The Role of Connective Tissues. Front Cell Dev Biol 2017; 5:22. [PMID: 28386539 PMCID: PMC5362625 DOI: 10.3389/fcell.2017.00022] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 03/07/2017] [Indexed: 12/22/2022] Open
Abstract
Skeletal muscles belong to the musculoskeletal system, which is composed of bone, tendon, ligament and irregular connective tissue, and closely associated with motor nerves and blood vessels. The intrinsic molecular signals regulating myogenesis have been extensively investigated. However, muscle development, homeostasis and regeneration require interactions with surrounding tissues and the cellular and molecular aspects of this dialogue have not been completely elucidated. During development and adult life, myogenic cells are closely associated with the different types of connective tissue. Connective tissues are defined as specialized (bone and cartilage), dense regular (tendon and ligament) and dense irregular connective tissue. The role of connective tissue in muscle morphogenesis has been investigated, thanks to the identification of transcription factors that characterize the different types of connective tissues. Here, we review the development of the various connective tissues in the context of the musculoskeletal system and highlight their important role in delivering information necessary for correct muscle morphogenesis, from the early step of myoblast differentiation to the late stage of muscle maturation. Interactions between muscle and connective tissue are also critical in the adult during muscle regeneration, as impairment of the regenerative potential after injury or in neuromuscular diseases results in the progressive replacement of the muscle mass by fibrotic tissue. We conclude that bi-directional communication between muscle and connective tissue is critical for a correct assembly of the musculoskeletal system during development as well as to maintain its homeostasis in the adult.
Collapse
Affiliation(s)
- Sonya Nassari
- Developmental Biology Laboratory, IBPS, Centre National de la Recherche Scientifique UMR7622, Institut National de la Santé Et de la Recherche Médicale U1156, Université Pierre et Marie Curie, Sorbonne Universités Paris, France
| | - Delphine Duprez
- Developmental Biology Laboratory, IBPS, Centre National de la Recherche Scientifique UMR7622, Institut National de la Santé Et de la Recherche Médicale U1156, Université Pierre et Marie Curie, Sorbonne Universités Paris, France
| | - Claire Fournier-Thibault
- Developmental Biology Laboratory, IBPS, Centre National de la Recherche Scientifique UMR7622, Institut National de la Santé Et de la Recherche Médicale U1156, Université Pierre et Marie Curie, Sorbonne Universités Paris, France
| |
Collapse
|
44
|
Valdivia M, Vega-Macaya F, Olguín P. Mechanical Control of Myotendinous Junction Formation and Tendon Differentiation during Development. Front Cell Dev Biol 2017; 5:26. [PMID: 28386542 PMCID: PMC5362613 DOI: 10.3389/fcell.2017.00026] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 03/07/2017] [Indexed: 01/01/2023] Open
Abstract
The development of the musculoskeletal system is a great model to study the interplay between chemical and mechanical inter-tissue signaling in cell adhesion, tissue morphogenesis and differentiation. In both vertebrates and invertebrates (e.g., Drosophila melanogaster) the formation of muscle-tendon interaction generates mechanical forces which are required for myotendinous junction maturation and tissue differentiation. In addition, these forces must be withstood by muscles and tendons in order to prevent detachment from each other, deformation or even losing their integrity. Extracellular matrix remodeling at the myotendinous junction is key to resist mechanical load generated by muscle contraction. Recent evidences in vertebrates indicate that mechanical forces generated during junction formation regulate chemical signaling leading to extracellular matrix remodeling, however, the mechanotransduction mechanisms associated to this response remains elusive. In addition to extracellular matrix remodeling, the ability of Drosophila tendon-cells to bear mechanical load depends on rearrangement of tendon cell cytoskeleton, thus studying the molecular mechanisms involved in this process is critical to understand the contribution of mechanical forces to the development of the musculoskeletal system. Here, we review recent findings regarding the role of chemical and mechanical signaling in myotendinous junction formation and tendon differentiation, and discuss molecular mechanisms of mechanotransduction that may allow tendon cells to withstand mechanical load during development of the musculoskeletal system.
Collapse
Affiliation(s)
- Mauricio Valdivia
- Program in Human Genetics, Faculty of Medicine, Institute of Biomedical Sciences, Biomedical Neurosciences Institute, University of Chile Santiago, Chile
| | - Franco Vega-Macaya
- Program in Human Genetics, Faculty of Medicine, Institute of Biomedical Sciences, Biomedical Neurosciences Institute, University of Chile Santiago, Chile
| | - Patricio Olguín
- Program in Human Genetics, Faculty of Medicine, Institute of Biomedical Sciences, Biomedical Neurosciences Institute, University of Chile Santiago, Chile
| |
Collapse
|
45
|
Li GJ, Yang Y, Yang GK, Wan J, Cui DL, Ma ZH, Du LJ, Zhang GM. Slit2 suppresses endothelial cell proliferation and migration by inhibiting the VEGF-Notch signaling pathway. Mol Med Rep 2017; 15:1981-1988. [PMID: 28260032 PMCID: PMC5364956 DOI: 10.3892/mmr.2017.6240] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 11/10/2016] [Indexed: 11/11/2022] Open
Abstract
Slit homolog 2 (Slit2) is distributed in various tissues and participates in numerous cellular processes; however, the role of Slit2 in the regulation of angiogenesis remains controversial, since it has previously been reported to exert proangiogenic and antiangiogenic activities. The present study aimed to investigate the effects of Slit2 on vascular endothelial cell proliferation and migration in vitro, and to reveal the possible underlying signaling pathway. Aortic endothelial cells were isolated from Sprague Dawley rats and cultured. Cell proliferation assay, cell migration assay, immunocytochemistry and small interfering RNA transfection were subsequently performed. The results demonstrated that exogenous Slit2 administration markedly suppressed TNF-α-induced endothelial cell proliferation and migration in vitro. In addition, TNF-α application upregulated the protein expression levels of vascular endothelial growth factor (VEGF) and Notch in RAECs, whereas Slit2 administration downregulated VEGF and Notch expression in RAECs cultured in TNF-α conditioned medium. Further studies indicated that knockdown of VEGF suppressed the effects of TNF-α on the induction of RAEC proliferation and migration. VEGF knockdown-induced inhibition of RAEC proliferation and migration in TNF-α conditioned medium was also achieved without Slit2 administration. Furthermore, VEGF knockdown markedly decreased Notch1 and Notch2 expression. These results indicated that Slit2 suppresses TNF-α-induced vascular endothelial cell proliferation and migration in vitro by inhibiting the VEGF-Notch signaling pathway. Therefore, Slit2 may inhibit the proliferation and migration of endothelial cells during vascular development.
Collapse
Affiliation(s)
- Guo-Jian Li
- Department of Vascular Surgery, The Fourth Affiliated Hospital, Kunming Medical University, Kunming, Yunnan 650021, P.R. China
| | - Yong Yang
- Department of Vascular Surgery, The Fourth Affiliated Hospital, Kunming Medical University, Kunming, Yunnan 650021, P.R. China
| | - Guo-Kai Yang
- Department of Vascular Surgery, The Fourth Affiliated Hospital, Kunming Medical University, Kunming, Yunnan 650021, P.R. China
| | - Jia Wan
- Department of Vascular Surgery, The Fourth Affiliated Hospital, Kunming Medical University, Kunming, Yunnan 650021, P.R. China
| | - Dao-Lei Cui
- Department of Vascular Surgery, The Fourth Affiliated Hospital, Kunming Medical University, Kunming, Yunnan 650021, P.R. China
| | - Zhen-Huan Ma
- Department of Vascular Surgery, The Fourth Affiliated Hospital, Kunming Medical University, Kunming, Yunnan 650021, P.R. China
| | - Ling-Juan Du
- Department of Vascular Surgery, The Fourth Affiliated Hospital, Kunming Medical University, Kunming, Yunnan 650021, P.R. China
| | - Gui-Min Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| |
Collapse
|
46
|
Ordan E, Volk T. Amontillado is required for Drosophila Slit processing and for tendon-mediated muscle patterning. Biol Open 2016; 5:1530-1534. [PMID: 27628033 PMCID: PMC5087687 DOI: 10.1242/bio.020636] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Slit cleavage into N-terminal and C-terminal polypeptides is essential for restricting the range of Slit activity. Although the Slit cleavage site has been characterized previously and is evolutionally conserved, the identity of the protease that cleaves Slit remains elusive. Our previous analysis indicated that Slit cleavage is essential to immobilize the active Slit-N at the tendon cell surfaces, mediating the arrest of muscle elongation. In an attempt to identify the protease required for Slit cleavage we performed an RNAi-based assay in the ectoderm and followed the process of elongation of the lateral transverse muscles toward tendon cells. The screen led to the identification of the Drosophila homolog of pheromone convertase 2 (PC2), Amontillado (Amon), as an essential protease for Slit cleavage. Further analysis indicated that Slit mobility on SDS polyacrylamide gel electrophoresis (SDS-PAGE) is slightly up-shifted in amon mutants, and its conventional cleavage into the Slit-N and Slit-C polypeptides is attenuated. Consistent with the requirement for amon to promote Slit cleavage and membrane immobilization of Slit-N, the muscle phenotype of amon mutant embryos was rescued by co-expressing a membrane-bound form of full-length Slit lacking the cleavage site and knocked into the slit locus. The identification of a novel protease component essential for Slit processing may represent an additional regulatory step in the Slit signaling pathway. Summary: The Drosophila homolog of pheromone convertase 2 (PC2), amontillado (Amon), is shown to contribute to Slit processing and further cleavage into an N-Slit, essential for Slit activity in directing muscle patterning.
Collapse
Affiliation(s)
- Elly Ordan
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Talila Volk
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
47
|
Abstract
Intensive investigations on angiogenesis and vasculogenesis have increased our understanding of molecular mechanisms of blood vessel formation during pathologic and developmental conditions. However, endothelial cells (ECs), the main component of vasculature, are heterogeneous, as revealed by our phenotypic and molecular biological studies in the laboratory, and it is still hard to adequately understand the molecular mechanisms of angiogenesis and vasculogenesis. Indeed, there are several major ligand/receptor signal pathways: VEGF/VEGFR, Jagged-1/Notch, Wnt ligand/frizzled receptor, and ephrin/Eph; each of which having distinct and independent roles during vascular formation. In this review, we focus on the angiogenic effect of the Slit and Robo signal pathway that was formally known as neuronal axon guidance. Among the existing vascular signals, this pathway is the most recently found ligand/receptor vascular signal, and may play important physiological roles as other major receptor/ligand signals do. Here, we briefly address: (1) the background of Slit and Robo families; (2) expression patterns of Slit and Robo; (3) functional roles of the Slit/Robo pathway in vascular formation; and (4) confronting tasks of this novel vascular pathway in the near future. Together, a summary of these data suggest the essential role of the Slit/Robo pathway in angiogenesis, and may explain why multiple vascular signals exist in heterogenic endothelial cells.
Collapse
Affiliation(s)
- Masakazu Fujiwara
- Department of Molecular Pathology, Nippon Medical School, Graduate School of Medicine, Institute of Gerontology, Kawasaki, Kanagawa, Japan
| | | | | |
Collapse
|
48
|
Alavi M, Song M, King GLA, Gillis T, Propst R, Lamanuzzi M, Bousum A, Miller A, Allen R, Kidd T. Dscam1 Forms a Complex with Robo1 and the N-Terminal Fragment of Slit to Promote the Growth of Longitudinal Axons. PLoS Biol 2016; 14:e1002560. [PMID: 27654876 PMCID: PMC5031454 DOI: 10.1371/journal.pbio.1002560] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 08/23/2016] [Indexed: 11/19/2022] Open
Abstract
The Slit protein is a major midline repellent for central nervous system (CNS) axons. In vivo, Slit is proteolytically cleaved into N- and C-terminal fragments, but the biological significance of this is unknown. Analysis in the Drosophila ventral nerve cord of a slit allele (slit-UC) that cannot be cleaved revealed that midline repulsion is still present but longitudinal axon guidance is disrupted, particularly across segment boundaries. Double mutants for the Slit receptors Dscam1 and robo1 strongly resemble the slit-UC phenotype, suggesting they cooperate in longitudinal axon guidance, and through biochemical approaches, we found that Dscam1 and Robo1 form a complex dependent on Slit-N. In contrast, Robo1 binding alone shows a preference for full-length Slit, whereas Dscam1 only binds Slit-N. Using a variety of transgenes, we demonstrated that Dscam1 appears to modify the output of Robo/Slit complexes so that signaling is no longer repulsive. Our data suggest that the complex is promoting longitudinal axon growth across the segment boundary. The ability of Dscam1 to modify the output of other receptors in a ligand-dependent fashion may be a general principle for Dscam proteins.
Collapse
Affiliation(s)
- Maryam Alavi
- Department of Biology, University of Nevada, Reno, Nevada, United States of America
| | - Minmin Song
- Department of Biology, University of Nevada, Reno, Nevada, United States of America
| | | | - Taylor Gillis
- Department of Biology, University of Nevada, Reno, Nevada, United States of America
| | - Robert Propst
- Department of Biology, University of Nevada, Reno, Nevada, United States of America
| | - Matthew Lamanuzzi
- Department of Biology, University of Nevada, Reno, Nevada, United States of America
| | - Adam Bousum
- Department of Biology, University of Nevada, Reno, Nevada, United States of America
| | - Amanda Miller
- Department of Biology, University of Nevada, Reno, Nevada, United States of America
| | - Ryan Allen
- Department of Biology, University of Nevada, Reno, Nevada, United States of America
| | - Thomas Kidd
- Department of Biology, University of Nevada, Reno, Nevada, United States of America
| |
Collapse
|
49
|
Transgenic over-expression of slit2 enhances disruption of blood-brain barrier and increases cell death after traumatic brain injury in mice. Neurosci Lett 2016; 631:85-90. [DOI: 10.1016/j.neulet.2016.08.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 07/24/2016] [Accepted: 08/09/2016] [Indexed: 11/22/2022]
|
50
|
Saller MM, Huettl RE, Hanuschick P, Amend AL, Alberton P, Aszodi A, Huber AB. The role of Sema3-Npn-1 signaling during diaphragm innervation and muscle development. J Cell Sci 2016; 129:3295-308. [PMID: 27466379 PMCID: PMC5047703 DOI: 10.1242/jcs.186015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 07/20/2016] [Indexed: 11/20/2022] Open
Abstract
Correct innervation of the main respiratory muscle in mammals, namely the thoracic diaphragm, is a crucial pre-requisite for the functionality of this muscle and the viability of the entire organism. Systemic impairment of Sema3A-Npn-1 (Npn-1 is also known as NRP1) signaling causes excessive branching of phrenic nerves in the diaphragm and into the central tendon region, where the majority of misguided axons innervate ectopic musculature. To elucidate whether these ectopic muscles are a result of misguidance of myoblast precursors due to the loss of Sema3A-Npn-1 signaling, we conditionally ablated Npn-1 in somatic motor neurons, which led to a similar phenotype of phrenic nerve defasciculation and, intriguingly, also formation of innervated ectopic muscles. We therefore hypothesize that ectopic myocyte fusion is caused by additional factors released by misprojecting growth cones. Slit2 and its Robo receptors are expressed by phrenic motor axons and migrating myoblasts, respectively, during innervation of the diaphragm. In vitro analyses revealed a chemoattractant effect of Slit2 on primary diaphragm myoblasts. Thus, we postulate that factors released by motor neuron growth cones have an influence on the migration properties of myoblasts during establishment of the diaphragm.
Collapse
Affiliation(s)
- Maximilian Michael Saller
- Experimental Surgery and Regenerative Medicine, Department of Surgery, Ludwig-Maximilians-University (LMU), Nußbaumstraße 20, Munich 80336, Germany Institute of Developmental Genetics, Helmholtz Zentrum München - German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, Neuherberg 85764, Germany
| | - Rosa-Eva Huettl
- Institute of Developmental Genetics, Helmholtz Zentrum München - German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, Neuherberg 85764, Germany Institute of Physiology, Department of Physiological Genomics, Ludwig-Maximilians-University (LMU), Schillerstraße 46, Munich 80336, Germany
| | - Philipp Hanuschick
- Institute of Developmental Genetics, Helmholtz Zentrum München - German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, Neuherberg 85764, Germany
| | - Anna-Lena Amend
- Institute of Developmental Genetics, Helmholtz Zentrum München - German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, Neuherberg 85764, Germany
| | - Paolo Alberton
- Experimental Surgery and Regenerative Medicine, Department of Surgery, Ludwig-Maximilians-University (LMU), Nußbaumstraße 20, Munich 80336, Germany
| | - Attila Aszodi
- Experimental Surgery and Regenerative Medicine, Department of Surgery, Ludwig-Maximilians-University (LMU), Nußbaumstraße 20, Munich 80336, Germany
| | - Andrea B Huber
- Institute of Developmental Genetics, Helmholtz Zentrum München - German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, Neuherberg 85764, Germany Bernstein Network for Computational Neuroscience, Albert-Ludwigs-University, Freiburg, Germany
| |
Collapse
|