1
|
Majumder T, Khot B, Suriyaarachchi H, Nathan A, Liu G. MYC regulation of the miR-92-Robo1 axis in Slit-mediated commissural axon guidance. Mol Biol Cell 2025; 36:ar50. [PMID: 40020181 PMCID: PMC12005101 DOI: 10.1091/mbc.e24-12-0534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 03/15/2025] Open
Abstract
In the developing spinal cord, translational repression of Robo1 expression by microRNA-92 (miR-92) in precrossing commissural axons (CAs) inhibits Slit/Robo1-mediated repulsion facilitating commissural axon projection and midline crossing; however, the regulatory mechanisms governing miR-92 expression in the developing commissural neurons are currently lacking. Here, we propose that the transcription factor MYC regulates miR-92 expression in the developing spinal cord (of either sex) to control Robo1 levels in precrossing CAs, modulating Slit/Robo1-mediated repulsion and midline crossing. MYC, miR-92, and Robo1 are differentially expressed in the developing chicken spinal cord. MYC binds to the promoter region upstream of the gga-miR-92 gene in vitro. MYC knockdown dramatically decreases miR-92 expression and increases chicken Robo1 (cRobo1) levels. In contrast, overexpression of MYC significantly induces miR-92 expression and reduces cRobo1 levels. MYC knockdown or overexpression results in significant inhibition or induction of miR-92 activity in the developing chicken spinal cord, respectively. Disruption of the MYC-dependent regulation of the miR-92-cRobo1 axis affects Slit2-mediated CA growth cone collapse in vitro and impairs CA projection and midline crossing in vivo. These results elucidate the role of the MYC-miR-92-cRobo1 axis in Slit2/Robo1-mediated CA repulsion and midline crossing.
Collapse
Affiliation(s)
- Tanushree Majumder
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606
| | - Bhakti Khot
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606
| | | | - Anagaa Nathan
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606
| | - Guofa Liu
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606
| |
Collapse
|
2
|
Zhu J, Xu B, Wu Z, Yu Z, Ji S, Lian J, Lu H. Integrative analysis of semaphorins family genes in colorectal cancer: implications for prognosis and immunotherapy. Front Immunol 2025; 16:1536545. [PMID: 40103807 PMCID: PMC11913869 DOI: 10.3389/fimmu.2025.1536545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 02/10/2025] [Indexed: 03/20/2025] Open
Abstract
Background Semaphorins (SEMAs), originally identified as axon guidance factors, have been found to play crucial roles in tumor growth, invasiveness, neoangiogenesis, and the modulation of immune responses. However, the prognostic value of SEMA-related genes in colorectal cancer (CRC) remains unclear. Methods We applied a novel machine learning framework that incorporated 10 machine learning algorithms and their 101 combinations to construct a SEMAs-related score (SRS). Multi-omics analysis was performed, including single-cell RNA sequencing (scRNA-seq), and spatial transcriptome (ST) to gain a more comprehensive understanding of the SRS. A series of cell experiments were conducted to prove the impact of key genes on CRC biological behavior. Result A consensus SRS was finally constructed based on a 101-combination machine learning computational framework, demonstrating outstanding performance in predicting overall survival. Moreover, distinct biological functions, mutation burden, immune cell infiltration, and immunotherapy response were observed between the high- and low-SRS groups. scRNA-seq and ST demonstrated unique cellular heterogeneity in CRC. We observed that SRS-high and SRS-low malignant epithelial cells exhibit different biological characteristics. High SRS malignant epithelial cells interact with myeloid and endothelial cells via SPP1 and COL4A2-ITGAV-ITGB8 pathways, respectively. Low SRS cells engage with myeloid and endothelial cells through MIF and JAG1-NOTCH4 pathways. Additionally, knocking down SEMA4C significantly inhibits the proliferation and invasion of CRC cells, while promoting apoptosis in vitro. Conclusion SRS could serve as an effective tool to predict survival and identify potential patients benefiting from immunotherapy in CRC. It also reveals tumor heterogeneity and provides valuable biological insights in CRC.
Collapse
Affiliation(s)
- Jiahao Zhu
- Department of Outpatient Chemotherapy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Benjie Xu
- Department of Outpatient Chemotherapy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Zhixing Wu
- Department of Computer Science, University of Liverpool, Liverpool, United Kingdom
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu, China
| | - Zhiwei Yu
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Shengjun Ji
- Department of Radiotherapy and Oncology, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Jie Lian
- Department of Outpatient Chemotherapy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Haibo Lu
- Department of Outpatient Chemotherapy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| |
Collapse
|
3
|
Li XL, Zhao YQ, Miao L, An YX, Wu F, Han JY, Han JY, Tay FR, Mu Z, Jiao Y, Wang J. Strategies for promoting neurovascularization in bone regeneration. Mil Med Res 2025; 12:9. [PMID: 40025573 PMCID: PMC11874146 DOI: 10.1186/s40779-025-00596-1] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 01/26/2025] [Indexed: 03/04/2025] Open
Abstract
Bone tissue relies on the intricate interplay between blood vessels and nerve fibers, both are essential for many physiological and pathological processes of the skeletal system. Blood vessels provide the necessary oxygen and nutrients to nerve and bone tissues, and remove metabolic waste. Concomitantly, nerve fibers precede blood vessels during growth, promote vascularization, and influence bone cells by secreting neurotransmitters to stimulate osteogenesis. Despite the critical roles of both components, current biomaterials generally focus on enhancing intraosseous blood vessel repair, while often neglecting the contribution of nerves. Understanding the distribution and main functions of blood vessels and nerve fibers in bone is crucial for developing effective biomaterials for bone tissue engineering. This review first explores the anatomy of intraosseous blood vessels and nerve fibers, highlighting their vital roles in bone embryonic development, metabolism, and repair. It covers innovative bone regeneration strategies directed at accelerating the intrabony neurovascular system over the past 10 years. The issues covered included material properties (stiffness, surface topography, pore structures, conductivity, and piezoelectricity) and acellular biological factors [neurotrophins, peptides, ribonucleic acids (RNAs), inorganic ions, and exosomes]. Major challenges encountered by neurovascularized materials during their clinical translation have also been highlighted. Furthermore, the review discusses future research directions and potential developments aimed at producing bone repair materials that more accurately mimic the natural healing processes of bone tissue. This review will serve as a valuable reference for researchers and clinicians in developing novel neurovascularized biomaterials and accelerating their translation into clinical practice. By bridging the gap between experimental research and practical application, these advancements have the potential to transform the treatment of bone defects and significantly improve the quality of life for patients with bone-related conditions.
Collapse
Affiliation(s)
- Xin-Ling Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Yu-Qing Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Li Miao
- Department of Stomatology, The Seventh Medical Center of PLA General Hospital, Beijing, 100700, China
| | - Yan-Xin An
- Department of General Surgery, The First Affiliated Hospital of Xi'an Medical University, Xi'an, 710077, China
| | - Fan Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Jin-Yu Han
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Jing-Yuan Han
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Franklin R Tay
- Graduate School of Augusta University, Augusta, GA, 30912, USA
| | - Zhao Mu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Yang Jiao
- Department of Stomatology, The Seventh Medical Center of PLA General Hospital, Beijing, 100700, China.
| | - Jing Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
4
|
Neuman K, Zhang X, Lejeune BT, Pizzarella D, Vázquez M, Lewis LH, Koppes AN, Koppes RA. Static Magnetic Stimulation and Magnetic Microwires Synergistically Enhance and Guide Neurite Outgrowth. Adv Healthc Mater 2025; 14:e2403956. [PMID: 39568232 PMCID: PMC11773108 DOI: 10.1002/adhm.202403956] [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: 10/11/2024] [Indexed: 11/22/2024]
Abstract
Axonal growth is heavily influenced by topography and biophysical stimuli including magnetic and electrical fields. Despite extensive investigation, the degree of influence and the underlying genetic mechanisms remain poorly understood. Here, a novel approach to guide neurite growth is undertaken using an innovative ferromagnetic composite material - glass-coated magnetic microwire - to furnish a synergistic combination of magnetic and topographical cues. Whole rat dorsal root ganglia (DRG) are cultured under five different conditions: control, static magnetic field, magnetic microwire, static magnetic field + glass fiber, and static magnetic field + magnetic microwire. DRG outgrowth responses under each condition, including total neurite outgrowth and directionality, are compared. The combination of both magnetic stimulation and topography significantly increases total neurite outgrowth compared to the controls. The combination of magnetic stimulation and magnetic microwire lead to a strong directional bias of growth along the microwire, double what is observed with the glass fiber. Next generation RNA sequencing of DRG exposed to static magnetic field + magnetic microwire reveals the downregulation of genes relating to the immune response, interleukin signaling, and signal transduction. These results set the stage for contemplating future biophysical stimulation for axonal guidance and improved understanding of material-tissue interactions.
Collapse
Affiliation(s)
- Katelyn Neuman
- Dept. of Chemical EngineeringNortheastern UniversityBostonMA02115USA
| | - Xiaoyu Zhang
- Dept. of Mechanical and Industrial EngineeringNortheastern UniversityBostonMA02115USA
| | - Brian. T. Lejeune
- Dept. of Chemical EngineeringNortheastern UniversityBostonMA02115USA
| | | | - Manuel Vázquez
- Instituto de Ciencia de Materiales de MadridCSICMadrid28049Spain
| | - Laura H. Lewis
- Dept. of Chemical EngineeringNortheastern UniversityBostonMA02115USA
- Dept. of Mechanical and Industrial EngineeringNortheastern UniversityBostonMA02115USA
| | - Abigail N. Koppes
- Dept. of Chemical EngineeringNortheastern UniversityBostonMA02115USA
- Dept. of BioengineeringNortheastern UniversityBostonMA02115USA
- Dept. of BiologyNortheastern UniversityBostonMA02115USA
| | - Ryan A. Koppes
- Dept. of Chemical EngineeringNortheastern UniversityBostonMA02115USA
| |
Collapse
|
5
|
Wu T, Li X, Xue J, Xia Y. Rational Fabrication of Functionally-Graded Surfaces for Biological and Biomedical Applications. ACCOUNTS OF MATERIALS RESEARCH 2024; 5:1507-1519. [PMID: 39744454 PMCID: PMC11686510 DOI: 10.1021/accountsmr.4c00186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 09/07/2024] [Accepted: 09/14/2024] [Indexed: 03/11/2025]
Abstract
As a ubiquitous feature of the biological world, gradation, in either composition or structure, is essential to many functions and processes. Taking protein gradation as an example, it plays a pivotal role in the development and evolution of human bodies, including stimulation and direction of the outgrowth of peripheral nerves in a developing fetus. It is also critically involved in wound healing by attracting and guiding immune cells to the site of injury or infection. Another good example can be found in the tendon-to-bone enthesis that relies on gradations in composition, structure, and cell phenotype to create a gradual change in mechanical stiffness. It is these unique gradations that eliminate the high level of stress at the interface, enabling the effective transfer of mechanical load from tendon to bone. How to fabricate and utilize graded surfaces and materials has been a constant theme of research in the context of materials science, chemistry, cell biology, and biomedical engineering. In cell biology, for example, graded surfaces are employed to investigate the fundamental mechanisms related to embryo development and to elucidate cell behaviors under chemo-, hapto-, or mechano-taxis. Scaffolds based upon graded materials have also been widely explored to enhance tissue repair or regeneration by accelerating cell migration and/or controlling stem cell differentiation. In this Account, we review our efforts in the fabrication and utilization of functionally graded surfaces. The gradation typically occurs as gradual changes in terms of composition, structure (e.g., pore size or fiber alignment), and/or coverage density of molecular species or larger objects such as particles and cells. Specifically, we focus on two strategies for generating various types of gradations along the surface of a substrate. In the first strategy, the substrate is vertically placed in a container, followed by the addition of a solution containing the functional component at a constant rate. Owing to the variations in contact time, the amount of the component deposited on the substrate naturally takes a gradual change along the vertical direction. In the second strategy, a moving collector or mask is used to control the amount of the component deposited on a substrate during jet printing or electrospray. As for applications, we highlight the following examples: (i) promotion of neurite outgrowth for peripheral nerve repair; (ii) acceleration of cell migration for wound closure; and (iii) mimicking of the structure and/or force transition at the tendon-to-bone enthesis for interfacial tissue engineering. The surface gradation can be presented in a uniaxial or radial fashion, and further integrated with the structural features on the underlying substrate to suit a specific application. In addition to general issues such as diversity of the surface gradation and reproducibility of the fabrication method, we also offer perspectives on new directions for future development. The systems and strategies discussed in this Account are expected to open the door to a range of fundamental inquires while enabling various biological and biomedical applications.
Collapse
Affiliation(s)
- Tong Wu
- Medical
Research Center, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266000, P. R. China
| | - Xiaoran Li
- Innovation
Center for Textile Science and Technology, Donghua University, Shanghai 201620, P. R. China
| | - Jiajia Xue
- Beijing
Laboratory of Biomedical Materials, State Key Laboratory of Organic−Inorganic
Composites, Beijing University of Chemical
Technology, Beijing 100029, P. R. China
| | - Younan Xia
- The Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
- School
of Chemistry and Biochemistry, School of Chemical and Biomolecular
Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| |
Collapse
|
6
|
Onesto MM, Kim JI, Pasca SP. Assembloid models of cell-cell interaction to study tissue and disease biology. Cell Stem Cell 2024; 31:1563-1573. [PMID: 39454582 DOI: 10.1016/j.stem.2024.09.017] [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: 05/09/2024] [Revised: 07/26/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024]
Abstract
Neurodevelopment involves the migration, projection, and integration of various cell types across different regions of the nervous system. Assembloids are self-organizing systems formed by the integration of multiple organoids or cell types. Here, we outline the generation and application of assembloids. We illustrate how assembloids recapitulate critical neurodevelopmental steps, like migration, axon projection, and circuit formation, and how they are starting to provide biological insights into neuropsychiatric disorders. Additionally, we review how assembloids can be used to study properties emerging from cell-cell interactions within non-neural tissues. Overall, assembloid platforms represent a powerful tool for discovering human biology and developing therapeutics.
Collapse
Affiliation(s)
- Massimo M Onesto
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute and Bio-X, Stanford University, Stanford, CA, USA
| | - Ji-Il Kim
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute and Bio-X, Stanford University, Stanford, CA, USA
| | - Sergiu P Pasca
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute and Bio-X, Stanford University, Stanford, CA, USA.
| |
Collapse
|
7
|
Staii C. Nonlinear Growth Dynamics of Neuronal Cells Cultured on Directional Surfaces. Biomimetics (Basel) 2024; 9:203. [PMID: 38667214 PMCID: PMC11048115 DOI: 10.3390/biomimetics9040203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/20/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
During the development of the nervous system, neuronal cells extend axons and dendrites that form complex neuronal networks, which are essential for transmitting and processing information. Understanding the physical processes that underlie the formation of neuronal networks is essential for gaining a deeper insight into higher-order brain functions such as sensory processing, learning, and memory. In the process of creating networks, axons travel towards other recipient neurons, directed by a combination of internal and external cues that include genetic instructions, biochemical signals, as well as external mechanical and geometrical stimuli. Although there have been significant recent advances, the basic principles governing axonal growth, collective dynamics, and the development of neuronal networks remain poorly understood. In this paper, we present a detailed analysis of nonlinear dynamics for axonal growth on surfaces with periodic geometrical patterns. We show that axonal growth on these surfaces is described by nonlinear Langevin equations with speed-dependent deterministic terms and gaussian stochastic noise. This theoretical model yields a comprehensive description of axonal growth at both intermediate and long time scales (tens of hours after cell plating), and predicts key dynamical parameters, such as speed and angular correlation functions, axonal mean squared lengths, and diffusion (cell motility) coefficients. We use this model to perform simulations of axonal trajectories on the growth surfaces, in turn demonstrating very good agreement between simulated growth and the experimental results. These results provide important insights into the current understanding of the dynamical behavior of neurons, the self-wiring of the nervous system, as well as for designing innovative biomimetic neural network models.
Collapse
Affiliation(s)
- Cristian Staii
- Department of Physics and Astronomy, Tufts University, Medford, MA 02155, USA
| |
Collapse
|
8
|
Abushalbaq O, Baek J, Yaron A, Tran TS. Balancing act of small GTPases downstream of plexin-A4 signaling motifs promotes dendrite elaboration in mammalian cortical neurons. Sci Signal 2024; 17:eadh7673. [PMID: 38227686 DOI: 10.1126/scisignal.adh7673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 12/21/2023] [Indexed: 01/18/2024]
Abstract
The precise development of neuronal morphologies is crucial to the establishment of synaptic circuits and, ultimately, proper brain function. Signaling by the axon guidance cue semaphorin 3A (Sema3A) and its receptor complex of neuropilin-1 and plexin-A4 has multifunctional outcomes in neuronal morphogenesis. Downstream activation of the RhoGEF FARP2 through interaction with the lysine-arginine-lysine motif of plexin-A4 and consequent activation of the small GTPase Rac1 promotes dendrite arborization, but this pathway is dispensable for axon repulsion. Here, we investigated the interplay of small GTPase signaling mechanisms underlying Sema3A-mediated dendritic elaboration in mouse layer V cortical neurons in vitro and in vivo. Sema3A promoted the binding of the small GTPase Rnd1 to the amino acid motif lysine-valine-serine (LVS) in the cytoplasmic domain of plexin-A4. Rnd1 inhibited the activity of the small GTPase RhoA and the kinase ROCK, thus supporting the activity of the GTPase Rac1, which permitted the growth and branching of dendrites. Overexpression of a dominant-negative RhoA, a constitutively active Rac1, or the pharmacological inhibition of ROCK activity rescued defects in dendritic elaboration in neurons expressing a plexin-A4 mutant lacking the LVS motif. Our findings provide insights into the previously unappreciated balancing act between Rho and Rac signaling downstream of specific motifs in plexin-A4 to mediate Sema3A-dependent dendritic elaboration in mammalian cortical neuron development.
Collapse
Affiliation(s)
- Oday Abushalbaq
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | - Jiyeon Baek
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| | - Avraham Yaron
- Department of Biomolecular Sciences and Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Tracy S Tran
- Department of Biological Sciences, Rutgers University, Newark, NJ 07102, USA
| |
Collapse
|
9
|
Maeno T, Arimatsu R, Ojima K, Yamaya Y, Imakyure H, Watanabe N, Komiya Y, Kobayashi K, Nakamura M, Nishimura T, Tatsumi R, Suzuki T. Netrin-4 synthesized in satellite cell-derived myoblasts stimulates autonomous fusion. Exp Cell Res 2023; 430:113698. [PMID: 37437770 DOI: 10.1016/j.yexcr.2023.113698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 06/20/2023] [Accepted: 06/23/2023] [Indexed: 07/14/2023]
Abstract
Satellite cells are indispensable for skeletal muscle regeneration and hypertrophy by forming nascent myofibers (myotubes). They synthesize multi-potent modulator netrins (secreted subtypes: netrin-1, -3, and -4), originally found as classical neural axon guidance molecules. While netrin-1 and -3 have key roles in myogenic differentiation, the physiological significance of netrin-4 is still unclear. This study examined whether netrin-4 regulates myofiber type commitment and myotube formation. Initially, the expression profiles indicated that satellite cells isolated from the extensor digitorum longus muscle (EDL muscle: fast-twitch myofiber-abundant) expressed slightly more netrin-4 than the soleus muscle (slow-type abundant) cells. As netrin-4 knockdown inhibited both slow- and fast-type myotube formation, netrin-4 may not directly regulate myofiber type commitment. However, netrin-4 knockdown in satellite cell-derived myoblasts reduced the myotube fusion index, while exogenous netrin-4 promoted myotube formation, even though netrin-4 expression level was maximum during the initiation stage of myogenic differentiation. Furthermore, netrin-4 knockdown also inhibited MyoD (a master transcriptional factor of myogenesis) and Myomixer (a myoblast fusogenic molecule) expression. These data suggest that satellite cells synthesize netrin-4 during myogenic differentiation initiation to promote their own fusion, stimulating the MyoD-Myomixer signaling axis.
Collapse
Affiliation(s)
- Takahiro Maeno
- Laboratory of Muscle and Meat Science, Department of Animal and Marine Bioresource Sciences, Research Faculty of Agriculture, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Rio Arimatsu
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Graduate School of Agriculture, Hokkaido University, Sapporo, Japan
| | - Koichi Ojima
- Muscle Biology Research Unit, Division of Animal Products Research, Institute of Livestock and Grassland Science, NARO, Tsukuba, Japan
| | - Yuki Yamaya
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Graduate School of Agriculture, Hokkaido University, Sapporo, Japan
| | - Hikaru Imakyure
- Laboratory of Muscle and Meat Science, Department of Animal and Marine Bioresource Sciences, Research Faculty of Agriculture, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Naruha Watanabe
- Laboratory of Muscle and Meat Science, Department of Animal and Marine Bioresource Sciences, Research Faculty of Agriculture, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Yusuke Komiya
- Department of Animal Science, School of Veterinary Medicine, Kitasato University, Towada, Japan
| | - Ken Kobayashi
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Graduate School of Agriculture, Hokkaido University, Sapporo, Japan
| | - Mako Nakamura
- Laboratory of Muscle and Meat Science, Department of Animal and Marine Bioresource Sciences, Research Faculty of Agriculture, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Takanori Nishimura
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Graduate School of Agriculture, Hokkaido University, Sapporo, Japan
| | - Ryuichi Tatsumi
- Laboratory of Muscle and Meat Science, Department of Animal and Marine Bioresource Sciences, Research Faculty of Agriculture, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Takahiro Suzuki
- Laboratory of Muscle and Meat Science, Department of Animal and Marine Bioresource Sciences, Research Faculty of Agriculture, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
10
|
Staii C. Biased Random Walk Model of Neuronal Dynamics on Substrates with Periodic Geometrical Patterns. Biomimetics (Basel) 2023; 8:267. [PMID: 37366862 DOI: 10.3390/biomimetics8020267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/07/2023] [Accepted: 06/16/2023] [Indexed: 06/28/2023] Open
Abstract
Neuronal networks are complex systems of interconnected neurons responsible for transmitting and processing information throughout the nervous system. The building blocks of neuronal networks consist of individual neurons, specialized cells that receive, process, and transmit electrical and chemical signals throughout the body. The formation of neuronal networks in the developing nervous system is a process of fundamental importance for understanding brain activity, including perception, memory, and cognition. To form networks, neuronal cells extend long processes called axons, which navigate toward other target neurons guided by both intrinsic and extrinsic factors, including genetic programming, chemical signaling, intercellular interactions, and mechanical and geometrical cues. Despite important recent advances, the basic mechanisms underlying collective neuron behavior and the formation of functional neuronal networks are not entirely understood. In this paper, we present a combined experimental and theoretical analysis of neuronal growth on surfaces with micropatterned periodic geometrical features. We demonstrate that the extension of axons on these surfaces is described by a biased random walk model, in which the surface geometry imparts a constant drift term to the axon, and the stochastic cues produce a random walk around the average growth direction. We show that the model predicts key parameters that describe axonal dynamics: diffusion (cell motility) coefficient, average growth velocity, and axonal mean squared length, and we compare these parameters with the results of experimental measurements. Our findings indicate that neuronal growth is governed by a contact-guidance mechanism, in which the axons respond to external geometrical cues by aligning their motion along the surface micropatterns. These results have a significant impact on developing novel neural network models, as well as biomimetic substrates, to stimulate nerve regeneration and repair after injury.
Collapse
Affiliation(s)
- Cristian Staii
- Department of Physics and Astronomy, Tufts University, Medford, MA 02155, USA
| |
Collapse
|
11
|
Paşcalău R, Badea TC. Signaling - transcription interactions in mouse retinal ganglion cells early axon pathfinding -a literature review. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1180142. [PMID: 38983012 PMCID: PMC11182120 DOI: 10.3389/fopht.2023.1180142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 04/21/2023] [Indexed: 07/11/2024]
Abstract
Sending an axon out of the eye and into the target brain nuclei is the defining feature of retinal ganglion cells (RGCs). The literature on RGC axon pathfinding is vast, but it focuses mostly on decision making events such as midline crossing at the optic chiasm or retinotopic mapping at the target nuclei. In comparison, the exit of RGC axons out of the eye is much less explored. The first checkpoint on the RGC axons' path is the optic cup - optic stalk junction (OC-OS). OC-OS development and the exit of the RGC pioneer axons out of the eye are coordinated spatially and temporally. By the time the optic nerve head domain is specified, the optic fissure margins are in contact and the fusion process is ongoing, the first RGCs are born in its proximity and send pioneer axons in the optic stalk. RGC differentiation continues in centrifugal waves. Later born RGC axons fasciculate with the more mature axons. Growth cones at the end of the axons respond to guidance cues to adopt a centripetal direction, maintain nerve fiber layer restriction and to leave the optic cup. Although there is extensive information on OC-OS development, we still have important unanswered questions regarding its contribution to the exit of the RGC axons out of the eye. We are still to distinguish the morphogens of the OC-OS from the axon guidance molecules which are expressed in the same place at the same time. The early RGC transcription programs responsible for axon emergence and pathfinding are also unknown. This review summarizes the molecular mechanisms for early RGC axon guidance by contextualizing mouse knock-out studies on OC-OS development with the recent transcriptomic studies on developing RGCs in an attempt to contribute to the understanding of human optic nerve developmental anomalies. The published data summarized here suggests that the developing optic nerve head provides a physical channel (the closing optic fissure) as well as molecular guidance cues for the pioneer RGC axons to exit the eye.
Collapse
Affiliation(s)
- Raluca Paşcalău
- Research and Development Institute, Transilvania University of Braşov, Braşov, Romania
- Ophthalmology Clinic, Cluj County Emergency Hospital, Cluj-Napoca, Romania
| | - Tudor Constantin Badea
- Research and Development Institute, Transilvania University of Braşov, Braşov, Romania
- National Center for Brain Research, Institutul de Cercetări pentru Inteligență Artificială, Romanian Academy, Bucharest, Romania
| |
Collapse
|
12
|
Tomar M, Beros J, Meloni B, Rodger J. Interactions between Guidance Cues and Neuronal Activity: Therapeutic Insights from Mouse Models. Int J Mol Sci 2023; 24:ijms24086966. [PMID: 37108129 PMCID: PMC10138948 DOI: 10.3390/ijms24086966] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 03/31/2023] [Accepted: 04/03/2023] [Indexed: 04/29/2023] Open
Abstract
Topographic mapping of neural circuits is fundamental in shaping the structural and functional organization of brain regions. This developmentally important process is crucial not only for the representation of different sensory inputs but also for their integration. Disruption of topographic organization has been associated with several neurodevelopmental disorders. The aim of this review is to highlight the mechanisms involved in creating and refining such well-defined maps in the brain with a focus on the Eph and ephrin families of axon guidance cues. We first describe the transgenic models where ephrin-A expression has been manipulated to understand the role of these guidance cues in defining topography in various sensory systems. We further describe the behavioral consequences of lacking ephrin-A guidance cues in these animal models. These studies have given us unexpected insight into how neuronal activity is equally important in refining neural circuits in different brain regions. We conclude the review by discussing studies that have used treatments such as repetitive transcranial magnetic stimulation (rTMS) to manipulate activity in the brain to compensate for the lack of guidance cues in ephrin-knockout animal models. We describe how rTMS could have therapeutic relevance in neurodevelopmental disorders with disrupted brain organization.
Collapse
Affiliation(s)
- Maitri Tomar
- School of Biological Sciences, The University of Western Australia, Crawley, WA 6009, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| | - Jamie Beros
- School of Biological Sciences, The University of Western Australia, Crawley, WA 6009, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| | - Bruno Meloni
- Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
- Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Crawley, WA 6009, Australia
- Department of Neurosurgery, Sir Charles Gairdner Hospital, QEII Medical Centre, Nedlands, WA 6009, Australia
| | - Jennifer Rodger
- School of Biological Sciences, The University of Western Australia, Crawley, WA 6009, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| |
Collapse
|
13
|
Tsuchimochi R, Yamagami K, Kubo N, Amimoto N, Raudzus F, Samata B, Kikuchi T, Doi D, Yoshimoto K, Mihara A, Takahashi J. Viral delivery of L1CAM promotes axonal extensions by embryonic cerebral grafts in mouse brain. Stem Cell Reports 2023; 18:899-914. [PMID: 36963389 PMCID: PMC10147836 DOI: 10.1016/j.stemcr.2023.02.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 02/24/2023] [Accepted: 02/25/2023] [Indexed: 03/26/2023] Open
Abstract
Cell replacement therapy is expected as a new and more radical treatment against brain damage. We previously reported that transplanted human cerebral organoids extend their axons along the corticospinal tract in rodent brains. The axons reached the spinal cord but were still sparse. Therefore, this study optimized the host brain environment by the adeno-associated virus (AAV)-mediated expression of axon guidance proteins in mouse brain. Among netrin-1, SEMA3, and L1CAM, only L1CAM significantly promoted the axonal extension of mouse embryonic brain tissue-derived grafts. L1CAM was also expressed by donor neurons, and this promotion was exerted in a haptotactic manner by their homophilic binding. Primary cortical neurons cocultured on L1CAM-expressing HEK-293 cells supported this mechanism. These results suggest that optimizing the host environment by the AAV-mediated expression of axon guidance molecules enhances the effect of cell replacement therapy.
Collapse
Affiliation(s)
- Ryosuke Tsuchimochi
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan; Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Keitaro Yamagami
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan; Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Naoko Kubo
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Naoya Amimoto
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Fabian Raudzus
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Bumpei Samata
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Tetsuhiro Kikuchi
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Daisuke Doi
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Koji Yoshimoto
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Aya Mihara
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Jun Takahashi
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan; Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan.
| |
Collapse
|
14
|
García-Bonilla M, Ojeda-Pérez B, Shumilov K, Rodríguez-Pérez LM, Domínguez-Pinos D, Vitorica J, Jiménez S, Ramírez-Lorca R, Echevarría M, Cárdenas-García C, Iglesias T, Gutiérrez A, McAllister JP, Limbrick DD, Páez-González P, Jiménez AJ. Generation of Periventricular Reactive Astrocytes Overexpressing Aquaporin 4 Is Stimulated by Mesenchymal Stem Cell Therapy. Int J Mol Sci 2023; 24:5640. [PMID: 36982724 PMCID: PMC10057840 DOI: 10.3390/ijms24065640] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/09/2023] [Accepted: 03/11/2023] [Indexed: 03/18/2023] Open
Abstract
Aquaporin-4 (AQP4) plays a crucial role in brain water circulation and is considered a therapeutic target in hydrocephalus. Congenital hydrocephalus is associated with a reaction of astrocytes in the periventricular white matter both in experimental models and human cases. A previous report showed that bone marrow-derived mesenchymal stem cells (BM-MSCs) transplanted into the lateral ventricles of hyh mice exhibiting severe congenital hydrocephalus are attracted by the periventricular astrocyte reaction, and the cerebral tissue displays recovery. The present investigation aimed to test the effect of BM-MSC treatment on astrocyte reaction formation. BM-MSCs were injected into the lateral ventricles of four-day-old hyh mice, and the periventricular reaction was detected two weeks later. A protein expression analysis of the cerebral tissue differentiated the BM-MSC-treated mice from the controls and revealed effects on neural development. In in vivo and in vitro experiments, BM-MSCs stimulated the generation of periventricular reactive astrocytes overexpressing AQP4 and its regulatory protein kinase D-interacting substrate of 220 kDa (Kidins220). In the cerebral tissue, mRNA overexpression of nerve growth factor (NGF), vascular endothelial growth factor (VEGF), hypoxia-inducible factor-1 (HIF1α), and transforming growth factor beta 1 (TGFβ1) could be related to the regulation of the astrocyte reaction and AQP4 expression. In conclusion, BM-MSC treatment in hydrocephalus can stimulate a key developmental process such as the periventricular astrocyte reaction, where AQP4 overexpression could be implicated in tissue recovery.
Collapse
Affiliation(s)
- María García-Bonilla
- Department of Cell Biology, Genetics and Physiology, University of Malaga, 29010 Malaga, Spain
- Instituto de Investigación Biomédica de Málaga (IBIMA), 29010 Malaga, Spain
| | - Betsaida Ojeda-Pérez
- Department of Cell Biology, Genetics and Physiology, University of Malaga, 29010 Malaga, Spain
- Instituto de Investigación Biomédica de Málaga (IBIMA), 29010 Malaga, Spain
| | - Kirill Shumilov
- Department of Cell Biology, Genetics and Physiology, University of Malaga, 29010 Malaga, Spain
- Instituto de Investigación Biomédica de Málaga (IBIMA), 29010 Malaga, Spain
- Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Luis-Manuel Rodríguez-Pérez
- Instituto de Investigación Biomédica de Málaga (IBIMA), 29010 Malaga, Spain
- Departamento de Fisiología Humana, Histología Humana, Anatomía Patológica y Educación Física y Deportiva, University of Malaga, 29010 Malaga, Spain
| | | | - Javier Vitorica
- Department of Molecular Biology and Biochemistry, University of Seville, 41013 Sevilla, Spain
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital, (HUVR)/Spanish National Research Council (CSIC)/University of Seville, 41013 Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos II, 28029 Madrid, Spain
| | - Sebastián Jiménez
- Department of Molecular Biology and Biochemistry, University of Seville, 41013 Sevilla, Spain
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital, (HUVR)/Spanish National Research Council (CSIC)/University of Seville, 41013 Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos II, 28029 Madrid, Spain
| | - Reposo Ramírez-Lorca
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos II, 28029 Madrid, Spain
- Department of Physiology and Biophysics, University of Seville, 41009 Seville, Spain
| | - Miriam Echevarría
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos II, 28029 Madrid, Spain
- Department of Physiology and Biophysics, University of Seville, 41009 Seville, Spain
| | - Casimiro Cárdenas-García
- Servicios Centrales de Apoyo a la Investigación (SCAI), University of Malaga, 29010 Malaga, Spain
| | - Teresa Iglesias
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos II, 28029 Madrid, Spain
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28029 Madrid, Spain
| | - Antonia Gutiérrez
- Department of Cell Biology, Genetics and Physiology, University of Malaga, 29010 Malaga, Spain
- Instituto de Investigación Biomédica de Málaga (IBIMA), 29010 Malaga, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos II, 28029 Madrid, Spain
| | - James P. McAllister
- Department of Neurosurgery, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - David D. Limbrick
- Department of Neurosurgery, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Patricia Páez-González
- Department of Cell Biology, Genetics and Physiology, University of Malaga, 29010 Malaga, Spain
- Instituto de Investigación Biomédica de Málaga (IBIMA), 29010 Malaga, Spain
| | - Antonio J. Jiménez
- Department of Cell Biology, Genetics and Physiology, University of Malaga, 29010 Malaga, Spain
- Instituto de Investigación Biomédica de Málaga (IBIMA), 29010 Malaga, Spain
| |
Collapse
|
15
|
Wood KL, Fonseca MIA, Gunderson KA, Nkana ZH, Israel JS, Poore SO, Dingle AM. Local Environment Induces Differential Gene Expression in Regenerating Nerves. J Surg Res 2022; 278:418-432. [PMID: 35618492 DOI: 10.1016/j.jss.2022.04.017] [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: 05/18/2021] [Revised: 03/18/2022] [Accepted: 04/07/2022] [Indexed: 11/24/2022]
Abstract
INTRODUCTION Approximately 80% of amputations are complicated by neuromas. Methods for neuroma management include nerve translocation into bone and implantation into skeletal muscle grafts, which have also facilitated the development of regenerative neural interfaces to enable fixation of prosthetics with motor and sensory feedback. However, molecular-level differences between nerves in these environments have not been investigated. This study aimed to elucidate the physiology of regenerating nerves in different settings by assessing gene expression. MATERIALS AND METHODS New Zealand white rabbits underwent transfemoral amputation with sciatic nerve transposition into the femur or tacked to skeletal muscle. At 5 wk, ribonucleic acid (RNA) sequencing of samples of distal nerve terminating in bone or muscle and nerve of the contralateral limb (control) identified differentially expressed genes (DEGs) and biochemical pathways (α = 0.05). RESULTS Three samples of nerve housed in bone, four of nerve tacked to muscle, and seven naïve controls were analyzed. Relative to controls, nerve housed in bone had little within-group variation and 13,028 DEGs, and nerve tacked to muscle had dramatic within-group variation and 12,811 DEGs. These samples upregulated the following pathways: lysosome, phagosome, antigen processing/presentation, and cell adhesion molecule. Relative to nerve housed in bone, nerve tacked to muscle had 12,526 DEGs, demonstrating upregulation of pathways of B-cell receptor signaling, focal adhesion, natural killer-cell mediated cytotoxicity, leukocyte transendothelial migration, and extracellular matrix-receptor interactions. CONCLUSIONS Nerve housed in bone has a more predictable molecular profile than does nerve tacked to muscle. Thus, the intramedullary canal may provide a more reliable setting for neuroma prevention and neural interfacing.
Collapse
Affiliation(s)
- Kasey Leigh Wood
- Division of Plastic Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Marina I Adrianzen Fonseca
- Division of Plastic Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Kirsten A Gunderson
- Division of Plastic Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Zeeda H Nkana
- Division of Plastic Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Jacqueline S Israel
- Division of Plastic Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Samuel O Poore
- Division of Plastic Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Aaron M Dingle
- Division of Plastic Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin.
| |
Collapse
|
16
|
Chandrasekaran A, Clarke A, McQueen P, Fang HY, Papoian GA, Giniger E. Computational simulations reveal that Abl activity controls cohesiveness of actin networks in growth cones. Mol Biol Cell 2022; 33:ar92. [PMID: 35857718 PMCID: PMC9582807 DOI: 10.1091/mbc.e21-11-0535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 06/30/2022] [Accepted: 07/12/2022] [Indexed: 11/11/2022] Open
Abstract
Extensive studies of growing axons have revealed many individual components and protein interactions that guide neuronal morphogenesis. Despite this, however, we lack any clear picture of the emergent mechanism by which this nanometer-scale biochemistry generates the multimicron-scale morphology and cell biology of axon growth and guidance in vivo. To address this, we studied the downstream effects of the Abl signaling pathway using a computer simulation software (MEDYAN) that accounts for mechanochemical dynamics of active polymers. Previous studies implicate two Abl effectors, Arp2/3 and Enabled, in Abl-dependent axon guidance decisions. We now find that Abl alters actin architecture primarily by activating Arp2/3, while Enabled plays a more limited role. Our simulations show that simulations mimicking modest levels of Abl activity bear striking similarity to actin profiles obtained experimentally from live imaging of actin in wild-type axons in vivo. Using a graph theoretical filament-filament contact analysis, moreover, we find that networks mimicking hyperactivity of Abl (enhanced Arp2/3) are fragmented into smaller domains of actin that interact weakly with each other, consistent with the pattern of actin fragmentation observed upon Abl overexpression in vivo. Two perturbative simulations further confirm that high-Arp2/3 actin networks are mechanically disconnected and fail to mount a cohesive response to perturbation. Taken together, these data provide a molecular-level picture of how the large-scale organization of the axonal cytoskeleton arises from the biophysics of actin networks.
Collapse
Affiliation(s)
- Aravind Chandrasekaran
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742
- National Institute of Neurological Diseases and Stroke, Bethesda, MD 20892
| | - Akanni Clarke
- National Institute of Neurological Diseases and Stroke, Bethesda, MD 20892
- Department of Biochemistry and Molecular Medicine, George Washington University School of Medicine/National Institutes of Health Graduate Partnerships Program, Washington, DC 20037
| | - Philip McQueen
- Center for Information Technology, National Institutes of Health, Bethesda, MD 20892
| | - Hsiao Yu Fang
- National Institute of Neurological Diseases and Stroke, Bethesda, MD 20892
| | - Garegin A. Papoian
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742
- Institute for Physical Science and Technology, University of Maryland, College Park, MD 20742
| | - Edward Giniger
- National Institute of Neurological Diseases and Stroke, Bethesda, MD 20892
| |
Collapse
|
17
|
Chen Z, Cao T, Zhong X, Wu Y, Fu W, Fan C, Jiang Y, Zhou Q, Peng J, Liao J, You Z, Yi X, Tan J. Association between serum netrin-1 levels and early neurological deterioration after acute ischemic stroke. Front Neurol 2022; 13:953557. [PMID: 36090888 PMCID: PMC9449874 DOI: 10.3389/fneur.2022.953557] [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: 05/26/2022] [Accepted: 08/03/2022] [Indexed: 11/15/2022] Open
Abstract
Background and purposes Experimental studies demonstrated that netrin-1 (NT-1) has anti-inflammatory, tissue regeneration, and immune modulation properties. We aimed to discern the utility of NT-1 as a biomarker for assessing the risk of early neurological deterioration (END) after ischemic stroke. Methods This was a prospective study enrolling ischemic stroke patients with symptoms onset <24 h. Serum NT-1 concentrations were measured at admission. The National Institutes of Health Stroke Scale increased by ≥2 points and ≥4 points during the first 72 h after admission and was defined as END2 and END4, respectively. Results The study included 268 patients (146 men and 122 women) with a mean age of 63.0 ± 9.6 years. The median NT-1 concentrations were 466.4 pg/ml (interquartile range, 341.4–589.2 pg/ml). During the initial 72 h after admission, END2 was found in 83 (31.0%) patients, and END4 was observed in 48 (17.9%) subjects. After adjusted for potential confounders, multivariate analysis indicated that decreased NT-1 levels is an independent predictor for END2 [odds ratio (OR) 0.62, 95% confidence interval (CI) 0.46–0.84, p < 0.001) and END4 (OR 0.53, 95% CI 0.36–0.76, p < 0.001). Similar results were found when the NT-1 levels were analyzed as a categorical variable. Furthermore, restricted cubic spline analysis showed a linear association between NT-1 concentrations and the risk of END (END2, p = 0.006 for linearity; END4, p < 0.001 for linearity). Conclusions Our results suggest that decreased NT-1 levels were significantly associated with a higher risk of END after ischemic stroke.
Collapse
Affiliation(s)
- Zhuo Chen
- Department of Neurology, Mianzhu People's Hospital, Mianzhu, China
| | - Tianli Cao
- Department of Neurology, Mianzhu People's Hospital, Mianzhu, China
| | - Xingju Zhong
- Department of Neurology, Mianzhu People's Hospital, Mianzhu, China
| | - Yong Wu
- Department of Neurology, Mianzhu People's Hospital, Mianzhu, China
| | - Wei Fu
- Department of Neurology, Mianzhu People's Hospital, Mianzhu, China
| | - Chaoli Fan
- Department of Neurology, Mianzhu People's Hospital, Mianzhu, China
| | - Yu Jiang
- Department of Neurology, Mianzhu People's Hospital, Mianzhu, China
| | - Qi Zhou
- Department of Neurology, Mianzhu People's Hospital, Mianzhu, China
| | - Jie Peng
- Department of Neurology, Mianzhu People's Hospital, Mianzhu, China
| | - Jieyu Liao
- Department of Neurology, Mianzhu People's Hospital, Mianzhu, China
| | - Zhike You
- Department of Neurology, Mianzhu People's Hospital, Mianzhu, China
| | - Xin Yi
- Department of Neurology, Mianzhu People's Hospital, Mianzhu, China
| | - Jingyu Tan
- Department of Endocrinology, Mianzhu People's Hospital, Mianzhu, China
- *Correspondence: Jingyu Tan
| |
Collapse
|
18
|
Ruiz Tejada Segura ML, Abou Moussa E, Garabello E, Nakahara TS, Makhlouf M, Mathew LS, Wang L, Valle F, Huang SSY, Mainland JD, Caselle M, Osella M, Lorenz S, Reisert J, Logan DW, Malnic B, Scialdone A, Saraiva LR. A 3D transcriptomics atlas of the mouse nose sheds light on the anatomical logic of smell. Cell Rep 2022; 38:110547. [PMID: 35320714 PMCID: PMC8995392 DOI: 10.1016/j.celrep.2022.110547] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 01/26/2022] [Accepted: 03/01/2022] [Indexed: 12/26/2022] Open
Abstract
The sense of smell helps us navigate the environment, but its molecular architecture and underlying logic remain understudied. The spatial location of odorant receptor genes (Olfrs) in the nose is thought to be independent of the structural diversity of the odorants they detect. Using spatial transcriptomics, we create a genome-wide 3D atlas of the mouse olfactory mucosa (OM). Topographic maps of genes differentially expressed in space reveal that both Olfrs and non-Olfrs are distributed in a continuous and overlapping fashion over at least five broad zones in the OM. The spatial locations of Olfrs correlate with the mucus solubility of the odorants they recognize, providing direct evidence for the chromatographic theory of olfaction. This resource resolves the molecular architecture of the mouse OM and will inform future studies on mechanisms underlying Olfr gene choice, axonal pathfinding, patterning of the nervous system, and basic logic for the peripheral representation of smell.
Collapse
Affiliation(s)
- Mayra L Ruiz Tejada Segura
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, Feodor-Lynen-Strasse 21, 81377 München, Germany; Institute of Functional Epigenetics, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany; Institute of Computational Biology, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | | | - Elisa Garabello
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, Feodor-Lynen-Strasse 21, 81377 München, Germany; Physics Department, University of Turin and INFN, Via P. Giuria 1, 10125 Turin, Italy; Department of Civil and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Thiago S Nakahara
- Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | | | | | - Li Wang
- Sidra Medicine, P.O. Box 26999, Doha, Qatar
| | - Filippo Valle
- Physics Department, University of Turin and INFN, Via P. Giuria 1, 10125 Turin, Italy
| | | | - Joel D Mainland
- Monell Chemical Senses Center, 3500 Market Street, Philadelphia, PA 19104, USA; Department of Neuroscience, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michele Caselle
- Physics Department, University of Turin and INFN, Via P. Giuria 1, 10125 Turin, Italy
| | - Matteo Osella
- Physics Department, University of Turin and INFN, Via P. Giuria 1, 10125 Turin, Italy
| | - Stephan Lorenz
- Sidra Medicine, P.O. Box 26999, Doha, Qatar; Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Johannes Reisert
- Monell Chemical Senses Center, 3500 Market Street, Philadelphia, PA 19104, USA
| | - Darren W Logan
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Bettina Malnic
- Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Antonio Scialdone
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, Feodor-Lynen-Strasse 21, 81377 München, Germany; Institute of Functional Epigenetics, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany; Institute of Computational Biology, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany.
| | - Luis R Saraiva
- Sidra Medicine, P.O. Box 26999, Doha, Qatar; Monell Chemical Senses Center, 3500 Market Street, Philadelphia, PA 19104, USA; College of Health and Life Sciences, Hamad Bin Khalifa University, P.O. Box 34110, Doha, Qatar.
| |
Collapse
|
19
|
Kumar VB, Porat Z, Gedanken A. Synthesis of Doped/Hybrid Carbon Dots and Their Biomedical Application. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:898. [PMID: 35335711 PMCID: PMC8951121 DOI: 10.3390/nano12060898] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/04/2022] [Accepted: 03/05/2022] [Indexed: 02/07/2023]
Abstract
Carbon dots (CDs) are a novel type of carbon-based nanomaterial that has gained considerable attention for their unique optical properties, including tunable fluorescence, stability against photobleaching and photoblinking, and strong fluorescence, which is attributed to a large number of organic functional groups (amino groups, hydroxyl, ketonic, ester, and carboxyl groups, etc.). In addition, they also demonstrate high stability and electron mobility. This article reviews the topic of doped CDs with organic and inorganic atoms and molecules. Such doping leads to their functionalization to obtain desired physical and chemical properties for biomedical applications. We have mainly highlighted modification techniques, including doping, polymer capping, surface functionalization, nanocomposite and core-shell structures, which are aimed at their applications to the biomedical field, such as bioimaging, bio-sensor applications, neuron tissue engineering, drug delivery and cancer therapy. Finally, we discuss the key challenges to be addressed, the future directions of research, and the possibilities of a complete hybrid format of CD-based materials.
Collapse
Affiliation(s)
- Vijay Bhooshan Kumar
- Bar-Ilan Institute for Nanotechnology and Advanced Materials, Department of Chemistry, Bar-Ilan University, Ramat-Gan 5290002, Israel
- Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ze’ev Porat
- Division of Chemistry, Nuclear Research Center-Negev, Beer-Sheva 8419001, Israel
- Unit of Environmental Engineering, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Aharon Gedanken
- Bar-Ilan Institute for Nanotechnology and Advanced Materials, Department of Chemistry, Bar-Ilan University, Ramat-Gan 5290002, Israel
| |
Collapse
|
20
|
Feedback-controlled dynamics of neuronal cells on directional surfaces. Biophys J 2022; 121:769-781. [PMID: 35101418 PMCID: PMC8943704 DOI: 10.1016/j.bpj.2022.01.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 12/16/2021] [Accepted: 01/25/2022] [Indexed: 11/21/2022] Open
Abstract
The formation of neuronal networks is a complex phenomenon of fundamental importance for understanding the development of the nervous system. The basic process underlying the network formation is axonal growth, a process involving the extension of axons from the cell body and axonal navigation toward target neurons. Axonal growth is guided by the interactions between the tip of the axon (growth cone) and its extracellular environmental cues, which include intercellular interactions, the biochemical landscape around the neuron, and the mechanical and geometrical features of the growth substrate. Here, we present a comprehensive experimental and theoretical analysis of axonal growth for neurons cultured on micropatterned polydimethylsiloxane (PDMS) surfaces. We demonstrate that closed-loop feedback is an essential component of axonal dynamics on these surfaces: the growth cone continuously measures environmental cues and adjusts its motion in response to external geometrical features. We show that this model captures all the characteristics of axonal dynamics on PDMS surfaces for both untreated and chemically modified neurons. We combine experimental data with theoretical analysis to measure key parameters that describe axonal dynamics: diffusion (cell motility) coefficients, speed and angular distributions, and cell-substrate interactions. The experiments performed on neurons treated with Taxol (inhibitor of microtubule dynamics) and Y-27632 (disruptor of actin filaments) indicate that the internal dynamics of microtubules and actin filaments plays a critical role for the proper function of the feedback mechanism. Our results demonstrate that axons follow geometrical patterns through a contact-guidance mechanism, in which high-curvature geometrical features impart high traction forces to the growth cone. These results have important implications for our fundamental understanding of axonal growth as well as for bioengineering novel substrate to guide neuronal growth and promote nerve repair.
Collapse
|
21
|
Maternal exercise and high-fat diet affect hypothalamic neural projections in rat offspring in a sex-specific manner. J Nutr Biochem 2022; 103:108958. [DOI: 10.1016/j.jnutbio.2022.108958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 10/20/2021] [Accepted: 01/05/2022] [Indexed: 11/23/2022]
|
22
|
Jiang J, Zhang F, Wan Y, Fang K, Yan ZD, Ren XL, Zhang R. Semaphorins as Potential Immune Therapeutic Targets for Cancer. Front Oncol 2022; 12:793805. [PMID: 35155237 PMCID: PMC8830438 DOI: 10.3389/fonc.2022.793805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 01/04/2022] [Indexed: 11/28/2022] Open
Abstract
Semaphorins are a large class of secreted or membrane-bound molecules. It has been reported that semaphorins play important roles in regulating several hallmarks of cancer, including angiogenesis, metastasis, and immune evasion. Semaphorins and their receptors are widely expressed on tumor cells and immune cells. However, the biological role of semaphorins in tumor immune microenvironment is intricate. The dysregulation of semaphorins influences the recruitment and infiltration of immune cells, leading to abnormal anti-tumor effect. Although the underlying mechanisms of semaphorins on regulating tumor-infiltrating immune cell activation and functions are not fully understood, semaphorins can notably be promising immunotherapy targets for cancer.
Collapse
Affiliation(s)
- Jun Jiang
- Department of Health Service, Fourth Military Medical University, Xi’an, China
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi’an, China
| | - Fang Zhang
- Department of Respiratory Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yi Wan
- Department of Health Service, Fourth Military Medical University, Xi’an, China
| | - Ke Fang
- Department of Health Service, Fourth Military Medical University, Xi’an, China
| | - Ze-dong Yan
- Department of Biomedical Engineering, Fourth Military Medical University, Xi’an, China
| | - Xin-ling Ren
- Department of Respiratory Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, China
- Department of Pulmonary Medicine, Shenzhen General Hospital, Shenzhen University, Shenzhen, China
| | - Rui Zhang
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
23
|
Li Y, Fraser D, Mereness J, Van Hove A, Basu S, Newman M, Benoit DSW. Tissue Engineered Neurovascularization Strategies for Craniofacial Tissue Regeneration. ACS APPLIED BIO MATERIALS 2022; 5:20-39. [PMID: 35014834 PMCID: PMC9016342 DOI: 10.1021/acsabm.1c00979] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Craniofacial tissue injuries, diseases, and defects, including those within bone, dental, and periodontal tissues and salivary glands, impact an estimated 1 billion patients globally. Craniofacial tissue dysfunction significantly reduces quality of life, and successful repair of damaged tissues remains a significant challenge. Blood vessels and nerves are colocalized within craniofacial tissues and act synergistically during tissue regeneration. Therefore, the success of craniofacial regenerative approaches is predicated on successful recruitment, regeneration, or integration of both vascularization and innervation. Tissue engineering strategies have been widely used to encourage vascularization and, more recently, to improve innervation through host tissue recruitment or prevascularization/innervation of engineered tissues. However, current scaffold designs and cell or growth factor delivery approaches often fail to synergistically coordinate both vascularization and innervation to orchestrate successful tissue regeneration. Additionally, tissue engineering approaches are typically investigated separately for vascularization and innervation. Since both tissues act in concert to improve craniofacial tissue regeneration outcomes, a revised approach for development of engineered materials is required. This review aims to provide an overview of neurovascularization in craniofacial tissues and strategies to target either process thus far. Finally, key design principles are described for engineering approaches that will support both vascularization and innervation for successful craniofacial tissue regeneration.
Collapse
Affiliation(s)
- Yiming Li
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - David Fraser
- Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States.,Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, New York 14620, United States.,Translational Biomedical Sciences Program, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Jared Mereness
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States.,Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Amy Van Hove
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Sayantani Basu
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Maureen Newman
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Danielle S W Benoit
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States.,Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, New York 14620, United States.,Translational Biomedical Sciences Program, University of Rochester Medical Center, Rochester, New York 14642, United States.,Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York 14642, United States.,Materials Science Program, University of Rochester, Rochester, New York 14627, United States.,Department of Chemical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Biomedical Genetics and Center for Oral Biology, University of Rochester Medical Center, Rochester, New York 14642, United States
| |
Collapse
|
24
|
Taylor L, Wankell M, Saxena P, McFarlane C, Hebbard L. Cell adhesion an important determinant of myogenesis and satellite cell activity. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1869:119170. [PMID: 34763027 DOI: 10.1016/j.bbamcr.2021.119170] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 10/18/2021] [Accepted: 11/01/2021] [Indexed: 10/19/2022]
Abstract
Skeletal muscles represent a complex and highly organised tissue responsible for all voluntary body movements. Developed through an intricate and tightly controlled process known as myogenesis, muscles form early in development and are maintained throughout life. Due to the constant stresses that muscles are subjected to, skeletal muscles maintain a complex course of regeneration to both replace and repair damaged myofibers and to form new functional myofibers. This process, made possible by a pool of resident muscle stem cells, termed satellite cells, and controlled by an array of transcription factors, is additionally reliant on a diverse range of cell adhesion molecules and the numerous signaling cascades that they initiate. This article will review the literature surrounding adhesion molecules and their roles in skeletal muscle myogenesis and repair.
Collapse
Affiliation(s)
- Lauren Taylor
- Department of Molecular and Cell Biology, College of Public Health, Medical and Veterinary Sciences, Centre for Molecular Therapeutics, Centre for Tropical Bioinformatics and Molecular Biology, Australian Institute of Tropical Medicine and Health, James Cook University, Townsville, Queensland, Australia
| | - Miriam Wankell
- Department of Molecular and Cell Biology, College of Public Health, Medical and Veterinary Sciences, Centre for Molecular Therapeutics, Centre for Tropical Bioinformatics and Molecular Biology, Australian Institute of Tropical Medicine and Health, James Cook University, Townsville, Queensland, Australia
| | - Pankaj Saxena
- Department of Cardiothoracic Surgery, The Townsville University Hospital, Townsville, Queensland, Australia; College of Medicine, Dentistry, James Cook University, Townsville, Queensland, Australia
| | - Craig McFarlane
- Department of Molecular and Cell Biology, College of Public Health, Medical and Veterinary Sciences, Centre for Molecular Therapeutics, Centre for Tropical Bioinformatics and Molecular Biology, Australian Institute of Tropical Medicine and Health, James Cook University, Townsville, Queensland, Australia.
| | - Lionel Hebbard
- Department of Molecular and Cell Biology, College of Public Health, Medical and Veterinary Sciences, Centre for Molecular Therapeutics, Centre for Tropical Bioinformatics and Molecular Biology, Australian Institute of Tropical Medicine and Health, James Cook University, Townsville, Queensland, Australia; Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
25
|
Abstract
Two of the most prevalent human viruses worldwide, herpes simplex virus type 1 and type 2 (HSV-1 and HSV-2, respectively), cause a variety of diseases, including cold sores, genital herpes, herpes stromal keratitis, meningitis and encephalitis. The intrinsic, innate and adaptive immune responses are key to control HSV, and the virus has developed mechanisms to evade them. The immune response can also contribute to pathogenesis, as observed in stromal keratitis and encephalitis. The fact that certain individuals are more prone than others to suffer severe disease upon HSV infection can be partially explained by the existence of genetic polymorphisms in humans. Like all herpesviruses, HSV has two replication cycles: lytic and latent. During lytic replication HSV produces infectious viral particles to infect other cells and organisms, while during latency there is limited gene expression and lack of infectious virus particles. HSV establishes latency in neurons and can cause disease both during primary infection and upon reactivation. The mechanisms leading to latency and reactivation and which are the viral and host factors controlling these processes are not completely understood. Here we review the HSV life cycle, the interaction of HSV with the immune system and three of the best-studied pathologies: Herpes stromal keratitis, herpes simplex encephalitis and genital herpes. We also discuss the potential association between HSV-1 infection and Alzheimer's disease.
Collapse
Affiliation(s)
- Shuyong Zhu
- Institute of Virology, Hannover Medical School, Cluster of Excellence RESIST (Exc 2155), Hannover Medical School, Hannover, Germany
| | - Abel Viejo-Borbolla
- Institute of Virology, Hannover Medical School, Cluster of Excellence RESIST (Exc 2155), Hannover Medical School, Hannover, Germany
| |
Collapse
|
26
|
Reduced hippocampal inhibition and enhanced autism-epilepsy comorbidity in mice lacking neuropilin 2. Transl Psychiatry 2021; 11:537. [PMID: 34663783 PMCID: PMC8523694 DOI: 10.1038/s41398-021-01655-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 08/18/2021] [Accepted: 09/17/2021] [Indexed: 12/19/2022] Open
Abstract
The neuropilin receptors and their secreted semaphorin ligands play key roles in brain circuit development by regulating numerous crucial neuronal processes, including the maturation of synapses and migration of GABAergic interneurons. Consistent with its developmental roles, the neuropilin 2 (Nrp2) locus contains polymorphisms in patients with autism spectrum disorder (ASD). Nrp2-deficient mice show autism-like behavioral deficits and propensity to develop seizures. In order to determine the pathophysiology in Nrp2 deficiency, we examined the hippocampal numbers of interneuron subtypes and inhibitory regulation of hippocampal CA1 pyramidal neurons in mice lacking one or both copies of Nrp2. Immunostaining for interneuron subtypes revealed that Nrp2-/- mice have a reduced number of parvalbumin, somatostatin, and neuropeptide Y cells, mainly in CA1. Whole-cell recordings identified reduced firing and hyperpolarized shift in resting membrane potential in CA1 pyramidal neurons from Nrp2+/- and Nrp2-/- mice compared to age-matched wild-type controls indicating decrease in intrinsic excitability. Simultaneously, the frequency and amplitude of spontaneous inhibitory postsynaptic currents (sIPSCs) are reduced in Nrp2-deficient mice. A convulsive dose of kainic acid evoked electrographic and behavioral seizures with significantly shorter latency, longer duration, and higher severity in Nrp2-/- compared to Nrp2+/+ animals. Finally, Nrp2+/- and Nrp2-/- but not Nrp2+/+, mice have impaired cognitive flexibility demonstrated by reward-based reversal learning, a task associated with hippocampal circuit function. Together these data demonstrate a broad reduction in interneuron subtypes and compromised inhibition in CA1 of Nrp2-/- mice, which could contribute to the heightened seizure susceptibility and behavioral deficits consistent with an ASD/epilepsy phenotype.
Collapse
|
27
|
Sunnerberg JP, Descoteaux M, Kaplan DL, Staii C. Axonal growth on surfaces with periodic geometrical patterns. PLoS One 2021; 16:e0257659. [PMID: 34555083 PMCID: PMC8459970 DOI: 10.1371/journal.pone.0257659] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 09/08/2021] [Indexed: 11/18/2022] Open
Abstract
The formation of neuron networks is a complex phenomenon of fundamental importance for understanding the development of the nervous system, and for creating novel bioinspired materials for tissue engineering and neuronal repair. The basic process underlying the network formation is axonal growth, a process involving the extension of axons from the cell body towards target neurons. Axonal growth is guided by environmental stimuli that include intercellular interactions, biochemical cues, and the mechanical and geometrical features of the growth substrate. The dynamics of the growing axon and its biomechanical interactions with the growing substrate remains poorly understood. In this paper, we develop a model of axonal motility which incorporates mechanical interactions between the axon and the growth substrate. We combine experimental data with theoretical analysis to measure the parameters that describe axonal growth on surfaces with micropatterned periodic geometrical features: diffusion (cell motility) coefficients, speed and angular distributions, and axon bending rigidities. Experiments performed on neurons treated Taxol (inhibitor of microtubule dynamics) and Blebbistatin (disruptor of actin filaments) show that the dynamics of the cytoskeleton plays a critical role in the axon steering mechanism. Our results demonstrate that axons follow geometrical patterns through a contact-guidance mechanism, in which high-curvature geometrical features impart high traction forces to the growth cone. These results have important implications for our fundamental understanding of axonal growth as well as for bioengineering novel substrates that promote neuronal growth and nerve repair.
Collapse
Affiliation(s)
- Jacob P. Sunnerberg
- Department of Physics and Astronomy, Tufts University, Medford, Massachusetts, United States of America
| | - Marc Descoteaux
- Department of Physics and Astronomy, Tufts University, Medford, Massachusetts, United States of America
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, United States of America
| | - Cristian Staii
- Department of Physics and Astronomy, Tufts University, Medford, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
28
|
Functional Analysis of Actin-Binding Proteins in the Central Nervous System of Drosophila. Methods Mol Biol 2021. [PMID: 34542862 DOI: 10.1007/978-1-0716-1661-1_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Using Drosophila actin-binding protein Dunc-115 as an example, this chapter describes a MARCM (mosaic analysis with a repressible cell marker)-based method for analyzing cytoskeletal components for their functions in the nervous system. Following a concise description about the principle, a step-by-step protocol is provided for generating the needed stocks and for histological analysis. Additional details and explanations have been given in the accompanying notes. Together, this should form a practical and sufficient recipe for performing at the single cell level loss-of-function and gain-of-function analyses of proteins associated with the cytoskeleton.
Collapse
|
29
|
Malheiro A, Wieringa P, Moroni L. Peripheral neurovascular link: an overview of interactions and in vitro models. Trends Endocrinol Metab 2021; 32:623-638. [PMID: 34127366 DOI: 10.1016/j.tem.2021.05.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 04/23/2021] [Accepted: 05/10/2021] [Indexed: 12/26/2022]
Abstract
Nerves and blood vessels (BVs) establish extensive arborized networks to innervate tissues and deliver oxygen/metabolic support. Developmental cues direct the formation of these intricate and often overlapping patterns, which reflect close interactions within the peripheral neurovascular system. Besides the mutual dependence to survive and function, nerves and BVs share several receptors and ligands, as well as principles of differentiation, growth and pathfinding. Neurovascular (NV) interactions are maintained in adult life and are essential for certain regenerative mechanisms, such as wound healing. In pathological situations (e.g., type 2 diabetes mellitus), the NV system can be severely perturbed and become dysfunctional. Unwanted neural growth and vascularization are also associated with the progression of some pathologies, such as cancer and endometriosis. In this review, we describe the fundamental NV interactions in development, highlighting the similarities between both networks and wiring mechanisms. We also describe the NV contribution to regenerative processes and potential pathological dysfunctions. Finally, we provide an overview of current in vitro models used to replicate and investigate the NV ecosystem, addressing present limitations and future perspectives.
Collapse
Affiliation(s)
- Afonso Malheiro
- Complex Tissue Regeneration Department, MERLN Institute for Technology-Inspired Regenerative Medicine, Universiteitssingel 40, 6229ER Maastricht, The Netherlands
| | - Paul Wieringa
- Complex Tissue Regeneration Department, MERLN Institute for Technology-Inspired Regenerative Medicine, Universiteitssingel 40, 6229ER Maastricht, The Netherlands
| | - Lorenzo Moroni
- Complex Tissue Regeneration Department, MERLN Institute for Technology-Inspired Regenerative Medicine, Universiteitssingel 40, 6229ER Maastricht, The Netherlands.
| |
Collapse
|
30
|
Vinton J, Aninweze A, Birgbauer E. Ibuprofen does not inhibit RhoA-mediated growth cone collapse of embryonic chicken retinal axons by LPA. Exp Brain Res 2021; 239:2969-2977. [PMID: 34322723 DOI: 10.1007/s00221-021-06172-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 05/15/2021] [Indexed: 11/28/2022]
Abstract
Lysophosphatidic acid (LPA) is a bioactive lysophospholipid that causes neuronal growth cones to collapse and neurites to retract through a RhoA-ROCK mediated pathway. It has been reported that the NSAID ibuprofen improves regeneration after spinal cord injury through a mechanism of inhibiting RhoA. This leads to the hypothesis that ibuprofen should block LPA-mediated growth cone collapse. We tested this hypothesis by treating embryonic chick retinal neurons with ibuprofen followed by LPA. Retinal growth cones collapsed with LPA in the presence of ibuprofen similar to control; however, growth cone collapse was effectively blocked by a ROCK inhibitor. Thus, our results do not support the designation of ibuprofen as a direct RhoA inhibitor.
Collapse
Affiliation(s)
- James Vinton
- Department of Biology, Winthrop University, Rock Hill, SC, 29733, USA
| | - Adaeze Aninweze
- Department of Biology, Winthrop University, Rock Hill, SC, 29733, USA
| | - Eric Birgbauer
- Department of Biology, Winthrop University, Rock Hill, SC, 29733, USA.
| |
Collapse
|
31
|
Yurchenko I, Farwell M, Brady DD, Staii C. Neuronal Growth and Formation of Neuron Networks on Directional Surfaces. Biomimetics (Basel) 2021; 6:biomimetics6020041. [PMID: 34208649 PMCID: PMC8293217 DOI: 10.3390/biomimetics6020041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/26/2021] [Accepted: 06/10/2021] [Indexed: 11/26/2022] Open
Abstract
The formation of neuron networks is a process of fundamental importance for understanding the development of the nervous system and for creating biomimetic devices for tissue engineering and neural repair. The basic process that controls the network formation is the growth of an axon from the cell body and its extension towards target neurons. Axonal growth is directed by environmental stimuli that include intercellular interactions, biochemical cues, and the mechanical and geometrical properties of the growth substrate. Despite significant recent progress, the steering of the growing axon remains poorly understood. In this paper, we develop a model of axonal motility, which incorporates substrate-geometry sensing. We combine experimental data with theoretical analysis to measure the parameters that describe axonal growth on micropatterned surfaces: diffusion (cell motility) coefficients, speed and angular distributions, and cell-substrate interactions. Experiments performed on neurons treated with inhibitors for microtubules (Taxol) and actin filaments (Y-27632) indicate that cytoskeletal dynamics play a critical role in the steering mechanism. Our results demonstrate that axons follow geometrical patterns through a contact-guidance mechanism, in which geometrical patterns impart high traction forces to the growth cone. These results have important implications for bioengineering novel substrates to guide neuronal growth and promote nerve repair.
Collapse
|
32
|
Abstract
Abnormalities in cranial motor nerve development cause paralytic strabismus syndromes, collectively referred to as congenital cranial dysinnervation disorders, in which patients cannot fully move their eyes. These disorders can arise through one of two mechanisms: (a) defective motor neuron specification, usually by loss of a transcription factor necessary for brainstem patterning, or (b) axon growth and guidance abnormalities of the oculomotor, trochlear, and abducens nerves. This review focuses on our current understanding of axon guidance mechanisms in the cranial motor nerves and how disease-causing mutations disrupt axon targeting. Abnormalities of axon growth and guidance are often limited to a single nerve or subdivision, even when the causative gene is ubiquitously expressed. Additionally, when one nerve is absent, its normal target muscles attract other motor neurons. Study of these disorders highlights the complexities of axon guidance and how each population of neurons uses a unique but overlapping set of axon guidance pathways. Expected final online publication date for the Annual Review of Vision Science, Volume 7 is September 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Mary C Whitman
- Department of Ophthalmology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA;
| |
Collapse
|
33
|
Crawley O, Grill B. Autophagy in axonal and presynaptic development. Curr Opin Neurobiol 2021; 69:139-148. [PMID: 33940492 DOI: 10.1016/j.conb.2021.03.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/17/2021] [Accepted: 03/21/2021] [Indexed: 10/21/2022]
Abstract
The study of autophagy in the nervous system has predominantly centered on degeneration. Evidence is now cementing crucial roles for autophagy in neuronal development and growth, especially in axonal and presynaptic compartments. A picture is emerging that autophagy typically promotes the growth of axons and reduces presynaptic stability. Nonetheless, these are not rigid principles, and it remains unclear why autophagy does not always display these relationships during axonal and presynaptic development. Recent progress has identified mechanisms underlying spatiotemporal control of autophagy in neurons and begun to unravel how autophagy is integrated with other cellular processes, such as proteasomal degradation and axon guidance. Ultimately, understanding how autophagy is regulated and its role in the developing nervous system is key to comprehending how the nervous system assembles its stereotyped yet plastic configuration. It is also likely to inform how we think about neurodevelopmental disorders and neurodegenerative diseases.
Collapse
Affiliation(s)
- Oliver Crawley
- Unidad de Neurobiología Celular y de Sistemas, Instituto de Neurociencias (CSIC-UMH), San Juan de Alicante, 03550, Spain.
| | - Brock Grill
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, 98199, USA; Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA; Department of Pharmacology, University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
34
|
Abundant Synthesis of Netrin-1 in Satellite Cell-Derived Myoblasts Isolated from EDL Rather Than Soleus Muscle Regulates Fast-Type Myotube Formation. Int J Mol Sci 2021; 22:ijms22094499. [PMID: 33925862 PMCID: PMC8123454 DOI: 10.3390/ijms22094499] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 04/23/2021] [Indexed: 01/05/2023] Open
Abstract
Resident myogenic stem cells (satellite cells) are attracting attention for their novel roles in myofiber type regulation. In the myogenic differentiation phase, satellite cells from soleus muscle (slow fiber-abundant) synthesize and secrete higher levels of semaphorin 3A (Sema3A, a multifunctional modulator) than those derived from extensor digitorum longus (EDL; fast fiber-abundant), suggesting the role of Sema3A in forming slow-twitch myofibers. However, the regulatory mechanisms underlying fast-twitch myotube commitment remain unclear. Herein, we focused on netrin family members (netrin-1, -3, and -4) that compete with Sema3A in neurogenesis and osteogenesis. We examined whether netrins affect fast-twitch myotube generation by evaluating their expression in primary satellite cell cultures. Initially, netrins are upregulated during myogenic differentiation. Next, we compared the expression levels of netrins and their cell membrane receptors between soleus- and EDL-derived satellite cells; only netrin-1 showed higher expression in EDL-derived satellite cells than in soleus-derived satellite cells. We also performed netrin-1 knockdown experiments and additional experiments with recombinant netrin-1 in differentiated satellite cell-derived myoblasts. Netrin-1 knockdown in myoblasts substantially reduced fast-type myosin heavy chain (MyHC) expression; exogenous netrin-1 upregulated fast-type MyHC in satellite cells. Thus, netrin-1 synthesized in EDL-derived satellite cells may promote myofiber type commitment of fast muscles.
Collapse
|
35
|
Hisano K, Kawase S, Mimura T, Yoshida H, Yamada H, Haniu H, Tsukahara T, Kurihara T, Matsuda Y, Saito N, Uemura T. Structurally different lysophosphatidylethanolamine species stimulate neurite outgrowth in cultured cortical neurons via distinct G-protein-coupled receptors and signaling cascades. Biochem Biophys Res Commun 2021; 534:179-185. [PMID: 33298313 DOI: 10.1016/j.bbrc.2020.11.119] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 11/29/2020] [Indexed: 12/31/2022]
Abstract
Neurite outgrowth is important in neuronal circuit formation and functions, and for regeneration of neuronal networks following trauma and disease in the brain. Thus, identification and characterization of the molecules that regulate neurite outgrowth are essential for understanding how brain circuits form and function and for the development of treatment of neurological disorders. In this study, we found that structurally different lysophosphatidylethanolamine (LPE) species, palmitoyl-LPE (16:0 LPE) and stearoyl-LPE (18:0 LPE), stimulate neurite growth in cultured cortical neurons. Interestingly, YM-254890, an inhibitor of Gq/11 protein, inhibited 16:0 LPE-stimulated neurite outgrowth but not 18:0 LPE-stimulated neurite outgrowth. In contrast, pertussis toxin, an inhibitor of Gi/Go proteins, inhibited 18:0 LPE-stimulated neurite outgrowth but not 16:0 LPE-stimulated neurite outgrowth. The effects of protein kinase C inhibitors on neurite outgrowth were also different. In addition, both 16:0 LPE and 18:0 LPE activate mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase 1/2, but the effect of the MAPK inhibitor differed between the 16:0 LPE- and 18:0 LPE-treated cultures. Collectively, the results suggest that the structurally different LPE species, 16:0 LPE and 18:0 LPE stimulate neurite outgrowth through distinct signaling cascades in cultured cortical neurons and that distinct G protein-coupled receptors are involved in these processes.
Collapse
Affiliation(s)
- Kazutoshi Hisano
- Graduate School of Medicine, Science and Technology, Department of Biomedical Engineering, Shinshu University, Nagano, 390-8621, Japan; Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano, 390-8621, Japan
| | - Shiori Kawase
- Division of Gene Research, Research Center for Supports to Advanced Science, Shinshu University, Nagano, 390-8621, Japan
| | - Tetsuhiko Mimura
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, Nagano, 390-8621, Japan
| | - Hironori Yoshida
- Graduate School of Medicine, Science and Technology, Department of Biomedical Engineering, Shinshu University, Nagano, 390-8621, Japan; Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano, 390-8621, Japan
| | - Hiroki Yamada
- Shinshu University School of Medicine, Nagano, 390-8621, Japan
| | - Hisao Haniu
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano, 390-8621, Japan
| | - Tamotsu Tsukahara
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8521, Japan
| | - Taiga Kurihara
- Division of Microbiology and Molecular Cell Biology, Nihon Pharmaceutical University, Saitama, 362-0806, Japan
| | - Yoshikazu Matsuda
- Division of Clinical Pharmacology and Pharmaceutics, Nihon Pharmaceutical University, Saitama, 362-0806, Japan
| | - Naoto Saito
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano, 390-8621, Japan
| | - Takeshi Uemura
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Nagano, 390-8621, Japan; Division of Gene Research, Research Center for Supports to Advanced Science, Shinshu University, Nagano, 390-8621, Japan.
| |
Collapse
|
36
|
Halakos EG, Connell AJ, Glazewski L, Wei S, Mason RW. Bottom up proteomics identifies neuronal differentiation pathway networks activated by cathepsin inhibition treatment in neuroblastoma cells that are enhanced by concurrent 13-cis retinoic acid treatment. J Proteomics 2020; 232:104068. [PMID: 33278663 DOI: 10.1016/j.jprot.2020.104068] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 10/16/2020] [Accepted: 11/29/2020] [Indexed: 12/19/2022]
Abstract
Neuroblastoma is the second most common pediatric cancer involving the peripheral nervous system in which stage IVS metastatic tumors regress due to spontaneous differentiation. 13-cis retinoic acid (13-cis RA) is currently used in the clinic for its differentiation effects and although it improves outcomes, relapse is seen in half of high-risk patients. Combinatorial therapies have been shown to be more effective in oncotherapy and since cathepsin inhibition reduces tumor growth, we explored the potential of coupling 13-cis RA with a cathepsin inhibitor (K777) to enhance therapeutic efficacy against neuroblastoma. Shotgun proteomics was used to identify proteins affected by K777 and dual (13-cis RA/K777) treatment in neuroblastoma SK-N-SH cells. Cathepsin inhibition was more effective in increasing proteins involved in neuronal differentiation and neurite outgrowth than 13-cis RA alone, but the combination of both treatments enhanced the neuronal differentiation effect. SIGNIFICANCE: As neuroblastoma can spontaneously differentiate, determining which proteins are involved in differentiation can guide development of more accurate diagnostic markers and more effective treatments. In this study, we established a differentiation proteomic map of SK-N-SH cells treated with a cathepsin inhibitor (K777) and K777/13-cis RA (dual). Bioinformatic analysis revealed these treatments enhanced neuronal differentiation and axonogenesis pathways. The most affected proteins in these pathways may become valuable biomarkers of efficacy of drugs designed to enhance differentiation of neuroblastoma [1].
Collapse
Affiliation(s)
- Effie G Halakos
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA; Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Andrew J Connell
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Lisa Glazewski
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA
| | - Shuo Wei
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Robert W Mason
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA; Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA.
| |
Collapse
|
37
|
Ahammad I. A comprehensive review of tumor proliferative and suppressive role of semaphorins and therapeutic approaches. Biophys Rev 2020; 12:1233-1247. [PMID: 32577918 PMCID: PMC7575654 DOI: 10.1007/s12551-020-00709-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 06/15/2020] [Indexed: 01/05/2023] Open
Abstract
Semaphorins have been traditionally known as axon guidance proteins that negatively regulate axonal growth. However, in the past couple of decades, their versatile role in so many other biological processes has come to prominence as well. One such example is their role in cancer. In this review article, the focus was on the tumor proliferative and tumor suppressive role of all 20 semaphorin family members under the 7 semaphorin classes found in vertebrates and invertebrates as well as the ongoing and emerging therapeutic approaches to combat semaphorin-mediated cancers. Except sema6C, 19 of the 20 non-viral semaphorin family members have been discovered to be associated with cancer in one way or another. Eleven semaphorin family members have been discovered to be tumor proliferative and 8 to be tumor suppressive. Six therapeutic avenues and their safety profiles have been discussed which are currently at use or at the various stages of development. Finally, perspectives on which approach is the best for treating cancers associated with semaphorins have been given.
Collapse
Affiliation(s)
- Ishtiaque Ahammad
- Department of Biochemistry and Microbiology, North South University, Dhaka, 1229, Bangladesh.
| |
Collapse
|
38
|
Herpes Simplex Virus 2 Counteracts Neurite Outgrowth Repulsion during Infection in a Nerve Growth Factor-Dependent Manner. J Virol 2020; 94:JVI.01370-20. [PMID: 32669337 PMCID: PMC7527038 DOI: 10.1128/jvi.01370-20] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 07/08/2020] [Indexed: 12/20/2022] Open
Abstract
Herpes simplex virus 2 (HSV-2) is a prevalent human pathogen that establishes lifelong latency in neurons of the peripheral nervous system. Colonization of neurons is required for HSV-2 persistence and pathogenesis. The viral and cellular factors required for efficient infection of neurons are not fully understood. We show here that nonneuronal cells repel neurite outgrowth of sensory neurons, while HSV-2 infection overcomes this inhibition and, rather, stimulates neurite outgrowth. HSV-2 glycoprotein G and nerve growth factor contribute to this phenotype, which may attract neurites to sites of infection and facilitate virus spread to neurons. Understanding the mechanisms that modulate neurite outgrowth and facilitate HSV-2 infection of neurons might foster the development of therapeutics to reduce HSV-2 colonization of the nervous system and provide insights on neurite outgrowth and regeneration. During primary infection, herpes simplex virus 2 (HSV-2) replicates in epithelial cells and enters neurites to infect neurons of the peripheral nervous system. Growth factors and attractive and repulsive directional cues influence neurite outgrowth and neuronal survival. We hypothesized that HSV-2 modulates the activity of such cues to increase neurite outgrowth. To test this hypothesis, we exposed sensory neurons to nerve growth factor (NGF) and mock- or HSV-2-infected HEK-293T cells, since they express repellents of neurite outgrowth. We show that HEK-293T cells secrete factors that inhibit neurite outgrowth, while infection with HSV-2 strains MS and 333 reduces this repelling phenotype, increasing neurite numbers. The HSV-2-mediated restoration of neurite outgrowth required the activity of NGF. In the absence of infection, however, NGF did not overcome the repulsion mediated by HEK-293T cells. We previously showed that recombinant, soluble glycoprotein G of HSV-2 (rSgG2) binds and enhances NGF activity, increasing neurite outgrowth. However, the effect of gG2 during infection has not been investigated. Therefore, we addressed whether gG2 contributes to overcoming neurite outgrowth repulsion. To do so, we generated viruses lacking gG2 expression and complemented them by exogenous expression of gG2. Overall, our results suggest that HSV-2 infection of nonneuronal cells reduces their repelling effect on neurite outgrowth in an NGF-dependent manner. gG2 contributed to this phenotype, but it was not the only factor. The enhanced neurite outgrowth may facilitate HSV-2 spread from epithelial cells into neurons expressing NGF receptors and increase HSV-2-mediated pathogenesis. IMPORTANCE Herpes simplex virus 2 (HSV-2) is a prevalent human pathogen that establishes lifelong latency in neurons of the peripheral nervous system. Colonization of neurons is required for HSV-2 persistence and pathogenesis. The viral and cellular factors required for efficient infection of neurons are not fully understood. We show here that nonneuronal cells repel neurite outgrowth of sensory neurons, while HSV-2 infection overcomes this inhibition and, rather, stimulates neurite outgrowth. HSV-2 glycoprotein G and nerve growth factor contribute to this phenotype, which may attract neurites to sites of infection and facilitate virus spread to neurons. Understanding the mechanisms that modulate neurite outgrowth and facilitate HSV-2 infection of neurons might foster the development of therapeutics to reduce HSV-2 colonization of the nervous system and provide insights on neurite outgrowth and regeneration.
Collapse
|
39
|
A20 promotes melanoma progression via the activation of Akt pathway. Cell Death Dis 2020; 11:794. [PMID: 32968045 PMCID: PMC7511359 DOI: 10.1038/s41419-020-03001-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 09/05/2020] [Accepted: 09/09/2020] [Indexed: 12/12/2022]
Abstract
Melanoma is the most life-threatening skin cancer with increasing incidence around the world. Although recent advances in targeted therapy and immunotherapy have brought revolutionary progress of the treatment outcome, the survival of patients with advanced melanoma remains unoptimistic, and metastatic melanoma is still an incurable disease. Therefore, to further understand the mechanism underlying melanoma pathogenesis could be helpful for developing novel therapeutic strategy. A20 is a crucial ubiquitin-editing enzyme implicated immunity regulation, inflammatory responses and cancer pathogenesis. Herein, we report that A20 played an oncogenic role in melanoma. We first found that the expression of A20 was significantly up-regulated in melanoma cell lines. Then, we showed that knockdown of A20 suppressed melanoma cell proliferation in vitro and melanoma growth in vivo through the regulation of cell-cycle progression. Moreover, A20 could potentiate the invasive and migratory capacities of melanoma cell in vitro and melanoma metastasis in vivo by promoting epithelial–mesenchymal transition (EMT). Mechanistically, we found that Akt activation mediated the oncogenic effect of A20 on melanoma development, with the involvement of glycolysis. What’s more, the up-regulation of A20 conferred the acquired resistance to Vemurafenib in BRAF-mutant melanoma. Taken together, we demonstrated that up-regulated A20 promoted melanoma progression via the activation of Akt pathway, and that A20 could be exploited as a potential therapeutic target for melanoma treatment.
Collapse
|
40
|
Omidinia-Anarkoli A, Ephraim JW, Rimal R, De Laporte L. Hierarchical fibrous guiding cues at different scales influence linear neurite extension. Acta Biomater 2020; 113:350-359. [PMID: 32663661 DOI: 10.1016/j.actbio.2020.07.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 07/04/2020] [Accepted: 07/07/2020] [Indexed: 12/22/2022]
Abstract
Surface topographies at micro- and nanoscales can influence different cellular behavior, such as their growth rate and directionality. While different techniques have been established to fabricate 2-dimensional flat substrates with nano- and microscale topographies, most of them are prone to high costs and long preparation times. The 2.5-dimensional fiber platform presented here provides knowledge on the effect of the combination of fiber alignment, inter-fiber distance (IFD), and fiber surface topography on contact guidance to direct neurite behavior from dorsal root ganglia (DRGs) or dissociated primary neurons. For the first time, the interplay of the micro-/nanoscale topography and IFD is studied to induce linear nerve growth, while controlling branching. The results demonstrate that grooved fibers promote a higher percentage of aligned neurite extension, compensating the adverse effect of increased IFD. Accordingly, maximum neurite extension from primary neurons is achieved on grooved fibers separated by an IFD of 30 μm, with a higher percentage of aligned neurons on grooved fibers at a large IFD compared to porous fibers with the smallest IFD of 10 µm. We further demonstrate that the neurite "decision-making" behavior on whether to cross a fiber or grow along it is not only dependent on the IFD but also on the fiber surface topography. In addition, axons growing in between the fibers seem to have a memory after leaving grooved fibers, resulting in higher linear growth and higher IFDs lead to more branching. Such information is of great importance for new material development for several tissue engineering applications. STATEMENT OF SIGNIFICANCE: One of the key aspects of tissue engineering is controlling cell behavior using hierarchical structures. Compared to 2D surfaces, fibers are an important class of materials, which can emulate the native ECM architecture of tissues. Despite the importance of both fiber surface topography and alignment to direct growing neurons, the current state of the art did not yet study the synergy between both scales of guidance. To achieve this, we established a solvent assisted spinning process to combine these two crucial features and control neuron growth, alignment, and branching. Rational design of new platforms for various tissue engineering and drug discovery applications can benefit from such information as it allows for fabrication of functional materials, which selectively influence neurite behavior.
Collapse
|
41
|
Rangel Olguin AG, Rochon PL, Krishnaswamy A. New Optical Tools to Study Neural Circuit Assembly in the Retina. Front Neural Circuits 2020; 14:44. [PMID: 32848633 PMCID: PMC7424070 DOI: 10.3389/fncir.2020.00044] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 06/23/2020] [Indexed: 12/17/2022] Open
Abstract
During development, neurons navigate a tangled thicket of thousands of axons and dendrites to synapse with just a few specific targets. This phenomenon termed wiring specificity, is critical to the assembly of neural circuits and the way neurons manage this feat is only now becoming clear. Recent studies in the mouse retina are shedding new insight into this process. They show that specific wiring arises through a series of stages that include: directed axonal and dendritic growth, the formation of neuropil layers, positioning of such layers, and matching of co-laminar synaptic partners. Each stage appears to be directed by a distinct family of recognition molecules, suggesting that the combinatorial expression of such family members might act as a blueprint for retinal connectivity. By reviewing the evidence in support of each stage, and by considering their underlying molecular mechanisms, we attempt to synthesize these results into a wiring model which generates testable predictions for future studies. Finally, we conclude by highlighting new optical methods that could be used to address such predictions and gain further insight into this fundamental process.
Collapse
|
42
|
Bernier LP, Bohlen CJ, York EM, Choi HB, Kamyabi A, Dissing-Olesen L, Hefendehl JK, Collins HY, Stevens B, Barres BA, MacVicar BA. Nanoscale Surveillance of the Brain by Microglia via cAMP-Regulated Filopodia. Cell Rep 2020; 27:2895-2908.e4. [PMID: 31167136 DOI: 10.1016/j.celrep.2019.05.010] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 03/14/2019] [Accepted: 04/30/2019] [Indexed: 02/07/2023] Open
Abstract
Microglia, the brain's immune cells, maintain homeostasis and sense pathological changes by continuously surveying the parenchyma with highly motile large processes. Here, we demonstrate that microglia also use thin actin-dependent filopodia that allow fast nanoscale sensing within discrete regions. Filopodia are distinct from large processes by their size, speed, and regulation mechanism. Increasing cyclic AMP (cAMP) by activating norepinephrine Gs-coupled receptors, applying nitric oxide, or inhibiting phosphodiesterases rapidly increases filopodia but collapses large processes. Alternatively, Gi-coupled P2Y12 receptor activation collapses filopodia but triggers large processes extension with bulbous tips. Similar control of cytoskeletal dynamics and microglial morphology by cAMP is observed in ramified primary microglia, suggesting that filopodia are intrinsically generated sensing structures. Therefore, nanoscale surveillance of brain parenchyma by microglia requires localized cAMP increases that drive filopodia formation. Shifting intracellular cAMP levels controls the polarity of microglial responses to changes in brain homeostasis and alters the scale of immunosurveillance.
Collapse
Affiliation(s)
- Louis-Philippe Bernier
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC V6T 1Z3, Canada.
| | - Christopher J Bohlen
- Stanford University School of Medicine, Department of Neurobiology, Stanford, CA 94305, USA
| | - Elisa M York
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC V6T 1Z3, Canada
| | - Hyun B Choi
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC V6T 1Z3, Canada
| | - Alireza Kamyabi
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC V6T 1Z3, Canada
| | - Lasse Dissing-Olesen
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC V6T 1Z3, Canada; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School Boston, MA 02115, USA
| | - Jasmin K Hefendehl
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC V6T 1Z3, Canada
| | - Hannah Y Collins
- Stanford University School of Medicine, Department of Neurobiology, Stanford, CA 94305, USA
| | - Beth Stevens
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School Boston, MA 02115, USA
| | - Ben A Barres
- Stanford University School of Medicine, Department of Neurobiology, Stanford, CA 94305, USA
| | - Brian A MacVicar
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
43
|
Li ZL, Müller-Greven J, Kim S, Tamagnone L, Buck M. Plexin-Bs enhance their GAP activity with a novel activation switch loop generating a cooperative enzyme. Cell Mol Life Sci 2020; 78:1101-1112. [PMID: 32601713 DOI: 10.1007/s00018-020-03571-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 05/24/2020] [Accepted: 06/12/2020] [Indexed: 01/01/2023]
Abstract
Plexins receive guidance cues from semaphorin ligands and transmit their signal through the plasma membrane. This family of proteins is unique amongst single-pass transmembrane receptors as their intracellular regions interact directly with several small GTPases, which regulate cytoskeletal dynamics and cell adhesion. Here, we characterize the GTPase Activating Protein (GAP) function of Plexin-B1 and find that a cooperative GAP activity towards the substrate GTPase, Rap1b, is associated with the N-terminal Juxtamembrane region of Plexin-B1. Importantly, we unveil an activation mechanism of Plexin-B1 by identifying a novel functional loop which partially blocks Rap1b entry into the plexin GAP domain. Consistent with the concept of allokairy developed for other systems, Plexin-B activity is increased by an apparent substrate-mediated cooperative effect. Simulations and mutagenesis suggest the repositioned JM conformation is stabilized by the new activation switch loop when the active site is occupied, giving rise to faster enzymatic turnover and cooperative behavior. The biological implications, essentially those of a threshold behavior for cell migration, are discussed.
Collapse
Affiliation(s)
- Zhen-Lu Li
- Department of Physiology and Biophysics, Case Western Reserve University, School of Medicine, 10900 Euclid Avenue, Cleveland, OH, 44106, USA
| | - Jeannine Müller-Greven
- Department of Physiology and Biophysics, Case Western Reserve University, School of Medicine, 10900 Euclid Avenue, Cleveland, OH, 44106, USA
| | - SoonJeung Kim
- Department of Physiology and Biophysics, Case Western Reserve University, School of Medicine, 10900 Euclid Avenue, Cleveland, OH, 44106, USA
| | - Luca Tamagnone
- School of Medicine, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Matthias Buck
- Department of Physiology and Biophysics, Case Western Reserve University, School of Medicine, 10900 Euclid Avenue, Cleveland, OH, 44106, USA.
- Department of Pharmacology, Case Western Reserve University, School of Medicine, 10900 Euclid Avenue, Cleveland, OH, 44106, USA.
- Department of Neurosciences, Case Western Reserve University, School of Medicine, 10900 Euclid Avenue, Cleveland, OH, 44106, USA.
- Case Comprehensive Cancer Center, Case Western Reserve University, School of Medicine, 10900 Euclid Avenue, Cleveland, OH, 44106, USA.
| |
Collapse
|
44
|
Modular and Distinct Plexin-A4/FARP2/Rac1 Signaling Controls Dendrite Morphogenesis. J Neurosci 2020; 40:5413-5430. [PMID: 32499377 DOI: 10.1523/jneurosci.2730-19.2020] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 04/29/2020] [Accepted: 05/26/2020] [Indexed: 12/26/2022] Open
Abstract
Diverse neuronal populations with distinct cellular morphologies coordinate the complex function of the nervous system. Establishment of distinct neuronal morphologies critically depends on signaling pathways that control axonal and dendritic development. The Sema3A-Nrp1/PlxnA4 signaling pathway promotes cortical neuron basal dendrite arborization but also repels axons. However, the downstream signaling components underlying these disparate functions of Sema3A signaling are unclear. Using the novel PlxnA4KRK-AAA knock-in male and female mice, generated by CRISPR/cas9, we show here that the KRK motif in the PlxnA4 cytoplasmic domain is required for Sema3A-mediated cortical neuron dendritic elaboration but is dispensable for inhibitory axon guidance. The RhoGEF FARP2, which binds to the KRK motif, shows identical functional specificity as the KRK motif in the PlxnA4 receptor. We find that Sema3A activates the small GTPase Rac1, and that Rac1 activity is required for dendrite elaboration but not axon growth cone collapse. This work identifies a novel Sema3A-Nrp1/PlxnA4/FARP2/Rac1 signaling pathway that specifically controls dendritic morphogenesis but is dispensable for repulsive guidance events. Overall, our results demonstrate that the divergent signaling output from multifunctional receptor complexes critically depends on distinct signaling motifs, highlighting the modular nature of guidance cue receptors and its potential to regulate diverse cellular responses.SIGNIFICANCE STATEMENT The proper formation of axonal and dendritic morphologies is crucial for the precise wiring of the nervous system that ultimately leads to the generation of complex functions in an organism. The Semaphorin3A-Neuropilin1/Plexin-A4 signaling pathway has been shown to have multiple key roles in neurodevelopment, from axon repulsion to dendrite elaboration. This study demonstrates that three specific amino acids, the KRK motif within the Plexin-A4 receptor cytoplasmic domain, are required to coordinate the downstream signaling molecules to promote Sema3A-mediated cortical neuron dendritic elaboration, but not inhibitory axon guidance. Our results unravel a novel Semaphorin3A-Plexin-A4 downstream signaling pathway and shed light on how the disparate functions of axon guidance and dendritic morphogenesis are accomplished by the same extracellular ligand in vivo.
Collapse
|
45
|
Cohen S, Sazan H, Kenigsberg A, Schori H, Piperno S, Shpaisman H, Shefi O. Large-scale acoustic-driven neuronal patterning and directed outgrowth. Sci Rep 2020; 10:4932. [PMID: 32188875 PMCID: PMC7080736 DOI: 10.1038/s41598-020-60748-2] [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: 10/07/2019] [Accepted: 01/31/2020] [Indexed: 11/09/2022] Open
Abstract
Acoustic manipulation is an emerging non-invasive method enabling precise spatial control of cells in their native environment. Applying this method for organizing neurons is invaluable for neural tissue engineering applications. Here, we used surface and bulk standing acoustic waves for large-scale patterning of Dorsal Root Ganglia neurons and PC12 cells forming neuronal cluster networks, organized biomimetically. We showed that by changing parameters such as voltage intensity or cell concentration we were able to affect cluster properties. We examined the effects of acoustic arrangement on cells atop 3D hydrogels for up to 6 days and showed that assembled cells spontaneously grew branches in a directed manner towards adjacent clusters, infiltrating the matrix. These findings have great relevance for tissue engineering applications as well as for mimicking architectures and properties of native tissues.
Collapse
Affiliation(s)
- Sharon Cohen
- Faculty of Engineering, Bar-Ilan University, Ramat Gan, Israel
- Bar-Ilan Institute of Nanotechnologies and Advanced Materials, Bar-Ilan University, Ramat Gan, Israel
- Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan, Israel
| | - Haim Sazan
- Bar-Ilan Institute of Nanotechnologies and Advanced Materials, Bar-Ilan University, Ramat Gan, Israel
- Department of Chemistry, Bar-Ilan University, Ramat Gan, 5290002, Israel
| | - Avraham Kenigsberg
- Bar-Ilan Institute of Nanotechnologies and Advanced Materials, Bar-Ilan University, Ramat Gan, Israel
- Department of Chemistry, Bar-Ilan University, Ramat Gan, 5290002, Israel
| | - Hadas Schori
- Faculty of Engineering, Bar-Ilan University, Ramat Gan, Israel
- Bar-Ilan Institute of Nanotechnologies and Advanced Materials, Bar-Ilan University, Ramat Gan, Israel
| | - Silvia Piperno
- Bar-Ilan Institute of Nanotechnologies and Advanced Materials, Bar-Ilan University, Ramat Gan, Israel
- Department of Chemistry, Bar-Ilan University, Ramat Gan, 5290002, Israel
| | - Hagay Shpaisman
- Bar-Ilan Institute of Nanotechnologies and Advanced Materials, Bar-Ilan University, Ramat Gan, Israel.
- Department of Chemistry, Bar-Ilan University, Ramat Gan, 5290002, Israel.
| | - Orit Shefi
- Faculty of Engineering, Bar-Ilan University, Ramat Gan, Israel.
- Bar-Ilan Institute of Nanotechnologies and Advanced Materials, Bar-Ilan University, Ramat Gan, Israel.
- Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan, Israel.
| |
Collapse
|
46
|
Biram A, Davidzohn N, Shulman Z. T cell interactions with B cells during germinal center formation, a three-step model. Immunol Rev 2019; 288:37-48. [PMID: 30874355 DOI: 10.1111/imr.12737] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Accepted: 01/04/2019] [Indexed: 12/25/2022]
Abstract
Establishment of effective immunity against invading microbes depends on continuous generation of antibodies that facilitate pathogen clearance. Long-lived plasma cells with the capacity to produce high affinity antibodies evolve in germinal centers (GCs), where B cells undergo somatic hypermutation and are subjected to affinity-based selection. Here, we focus on the cellular interactions that take place early in the antibody immune response during GC colonization. Clones bearing B-cell receptors with different affinities and specificities compete for entry to the GC, at the boundary between the B-cell and T-cell zones in lymphoid organs. During this process, B cells compete for interactions with T follicular helper cells, which provide selection signals required for differentiation into GC cells and antibody secreting cells. These cellular engagements are long-lasting and depend on activation of adhesion molecules that support persistent interactions and promote transmission of signals between the cells. Here, we discuss how interactions between cognate T and B cells are primarily maintained by three types of molecular interactions: homophilic signaling lymphocytic activation molecule (SLAM) interactions, T-cell receptor: peptide-loaded major histocompatibility class II (pMHCII), and LFA-1:ICAMs. These essential components support a three-step process that controls clonal selection for entry into the antibody affinity maturation response in the GC, and establishment of long-lasting antibody-mediated immunity.
Collapse
Affiliation(s)
- Adi Biram
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Natalia Davidzohn
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Ziv Shulman
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
47
|
Basso JMV, Yurchenko I, Wiens MR, Staii C. Neuron dynamics on directional surfaces. SOFT MATTER 2019; 15:9931-9941. [PMID: 31764921 DOI: 10.1039/c9sm01769k] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Geometrical features play a very important role in neuronal growth and the formation of functional connections between neuronal cells. Here, we analyze the dynamics of axonal growth for neuronal cells cultured on micro-patterned polydimethylsiloxane surfaces. We utilize fluorescence microscopy to image axons, quantify their dynamics, and demonstrate that periodic geometrical patterns impart strong directional bias to neuronal growth. We quantify axonal alignment and present a general stochastic approach that quantitatively describes the dynamics of the growth cones. Neuronal growth is described by a general phenomenological model, based on a simple automatic controller with a closed-loop feedback system. We demonstrate that axonal alignment on these substrates is determined by the surface geometry, and it is quantified by the deterministic part of the stochastic (Langevin and Fokker-Planck) equations. We also show that the axonal alignment with the surface patterns is greatly suppressed by the neuron treatment with Blebbistatin, a chemical compound that inhibits the activity of myosin II. These results give new insight into the role played by the molecular motors and external geometrical cues in guiding axonal growth, and could lead to novel approaches for bioengineering neuronal regeneration platforms.
Collapse
Affiliation(s)
- Joao Marcos Vensi Basso
- Department of Physics and Astronomy, Center for Nanoscopic Physics, Tufts University, Medford, Massachusetts 02155, USA.
| | | | | | | |
Collapse
|
48
|
Maternal valproic acid exposure leads to neurogenesis defects and autism-like behaviors in non-human primates. Transl Psychiatry 2019; 9:267. [PMID: 31636273 PMCID: PMC6803711 DOI: 10.1038/s41398-019-0608-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 07/17/2019] [Indexed: 02/05/2023] Open
Abstract
Despite the substantial progress made in identifying genetic defects in autism spectrum disorder (ASD), the etiology for majority of ASD individuals remains elusive. Maternal exposure to valproic acid (VPA), a commonly prescribed antiepileptic drug during pregnancy in human, has long been considered a risk factor to contribute to ASD susceptibility in offspring from epidemiological studies in humans. The similar exposures in murine models have provided tentative evidence to support the finding from human epidemiology. However, the apparent difference between rodent and human poses a significant challenge to extrapolate the findings from rodent models to humans. Here we report for the first time the neurodevelopmental and behavioral outcomes of maternal VPA exposure in non-human primates. Monkey offspring from the early maternal VPA exposure have significantly reduced NeuN-positive mature neurons in prefrontal cortex (PFC) and cerebellum and the Ki67-positive proliferating neuronal precursors in the cerebellar external granular layer, but increased GFAP-positive astrocytes in PFC. Transcriptome analyses revealed that maternal VPA exposure disrupted the expression of genes associated with neurodevelopment in embryonic brain in offspring. VPA-exposed juvenile offspring have variable presentations of impaired social interaction, pronounced stereotypies, and more attention on nonsocial stimuli by eye tracking analysis. Our findings in non-human primates provide the best evidence so far to support causal link between maternal VPA exposure and neurodevelopmental defects and ASD susceptibility in humans.
Collapse
|
49
|
Xu C, Theisen E, Maloney R, Peng J, Santiago I, Yapp C, Werkhoven Z, Rumbaut E, Shum B, Tarnogorska D, Borycz J, Tan L, Courgeon M, Griffin T, Levin R, Meinertzhagen IA, de Bivort B, Drugowitsch J, Pecot MY. Control of Synaptic Specificity by Establishing a Relative Preference for Synaptic Partners. Neuron 2019; 103:865-877.e7. [PMID: 31300277 PMCID: PMC6728174 DOI: 10.1016/j.neuron.2019.06.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 04/19/2019] [Accepted: 06/11/2019] [Indexed: 02/07/2023]
Abstract
The ability of neurons to identify correct synaptic partners is fundamental to the proper assembly and function of neural circuits. Relative to other steps in circuit formation such as axon guidance, our knowledge of how synaptic partner selection is regulated is severely limited. Drosophila Dpr and DIP immunoglobulin superfamily (IgSF) cell-surface proteins bind heterophilically and are expressed in a complementary manner between synaptic partners in the visual system. Here, we show that in the lamina, DIP mis-expression is sufficient to promote synapse formation with Dpr-expressing neurons and that disrupting DIP function results in ectopic synapse formation. These findings indicate that DIP proteins promote synapses to form between specific cell types and that in their absence, neurons synapse with alternative partners. We propose that neurons have the capacity to synapse with a broad range of cell types and that synaptic specificity is achieved by establishing a preference for specific partners.
Collapse
Affiliation(s)
- Chundi Xu
- Department of Neurobiology, Harvard Medical School, 220 Longwood Ave, Boston, MA 02115, USA.
| | - Emma Theisen
- Department of Neurobiology, Harvard Medical School, 220 Longwood Ave, Boston, MA 02115, USA
| | - Ryan Maloney
- Department of Neurobiology, Harvard Medical School, 220 Longwood Ave, Boston, MA 02115, USA
| | - Jing Peng
- Department of Neurobiology, Harvard Medical School, 220 Longwood Ave, Boston, MA 02115, USA
| | - Ivan Santiago
- Department of Neurobiology, Harvard Medical School, 220 Longwood Ave, Boston, MA 02115, USA
| | - Clarence Yapp
- Image and Data Analysis Core, Harvard Medical School, Boston, MA 02115, USA
| | - Zachary Werkhoven
- Center for Brain Science and Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA
| | - Elijah Rumbaut
- Department of Neurobiology, Harvard Medical School, 220 Longwood Ave, Boston, MA 02115, USA
| | - Bryan Shum
- Department of Neurobiology, Harvard Medical School, 220 Longwood Ave, Boston, MA 02115, USA
| | - Dorota Tarnogorska
- Department of Psychology and Neuroscience, Life Sciences Centre, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Jolanta Borycz
- Department of Psychology and Neuroscience, Life Sciences Centre, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Liming Tan
- Department of Biological Chemistry, HHMI, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Maximilien Courgeon
- Department of Biology, New York University, 100 Washington Square East, New York, NY 10003, USA
| | - Tessa Griffin
- Department of Neurobiology, Harvard Medical School, 220 Longwood Ave, Boston, MA 02115, USA
| | - Raina Levin
- Department of Neurobiology, Harvard Medical School, 220 Longwood Ave, Boston, MA 02115, USA
| | - Ian A Meinertzhagen
- Department of Psychology and Neuroscience, Life Sciences Centre, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Benjamin de Bivort
- Center for Brain Science and Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA
| | - Jan Drugowitsch
- Department of Neurobiology, Harvard Medical School, 220 Longwood Ave, Boston, MA 02115, USA
| | - Matthew Y Pecot
- Department of Neurobiology, Harvard Medical School, 220 Longwood Ave, Boston, MA 02115, USA.
| |
Collapse
|
50
|
Sunnerberg JP, Moore P, Spedden E, Kaplan DL, Staii C. Variations of Elastic Modulus and Cell Volume with Temperature for Cortical Neurons. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:10965-10976. [PMID: 31380651 PMCID: PMC7306228 DOI: 10.1021/acs.langmuir.9b01651] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Neurons change their growth dynamics and mechanical properties in response to external stimuli such as stiffness of the local microenvironment, ambient temperature, and biochemical or geometrical guidance cues. Here we use combined atomic force microscopy (AFM) and fluorescence microscopy experiments to investigate the relationship between external temperature, soma volume, and elastic modulus for cortical neurons. We measure how changes in ambient temperature affect the volume and the mechanical properties of neuronal cells at both the bulk (elastic modulus) and local (elasticity maps) levels. The experimental data demonstrate that both the volume and the elastic modulus of the neuron soma vary with changes in temperature. Our results show a decrease by a factor of 2 in the soma elastic modulus as the ambient temperature increases from room (25 °C) to physiological (37 °C) temperature, while the volume of the soma increases by a factor of 1.3 during the same temperature sweep. Using high-resolution AFM force mapping, we measure the temperature-induced variations within different regions of the elasticity maps (low and high values of elastic modulus) and correlate these variations with the dynamics of cytoskeleton components and molecular motors. We quantify the change in soma volume with temperature and propose a simple theoretical model that relates this change with variations in soma elastic modulus. These results have significant implications for understanding neuronal development and functions, as ambient temperature, cytoskeletal dynamics, and cellular volume may change with variations in physiological conditions, for example, during tissue compression and infections in vivo as well as during cell manipulation and tissue regeneration ex vivo.
Collapse
|