1
|
Demler C, Lawlor JC, Yelin R, Llivichuzcha-Loja D, Shaulov L, Kim D, Stewart M, Lee FK, Shylo N, Trainor PA, Schultheiss TM, Kurpios NA. An atypical basement membrane forms a midline barrier during left-right asymmetric gut development in the chicken embryo. eLife 2025; 12:RP89494. [PMID: 40298919 PMCID: PMC12040318 DOI: 10.7554/elife.89494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025] Open
Abstract
Correct intestinal morphogenesis depends on the early embryonic process of gut rotation, an evolutionarily conserved program in which a straight gut tube elongates and forms into its first loops. However, the gut tube requires guidance to loop in a reproducible manner. The dorsal mesentery (DM) connects the gut tube to the body and directs the lengthening gut into stereotypical loops via left-right (LR) asymmetric cellular and extracellular behavior. The LR asymmetry of the DM also governs blood and lymphatic vessel formation for the digestive tract, which is essential for prenatal organ development and postnatal vital functions including nutrient absorption. Although the genetic LR asymmetry of the DM has been extensively studied, a divider between the left and right DM has yet to be identified. Setting up LR asymmetry for the entire body requires a Lefty1+ midline barrier to separate the two sides of the embryo, without it, embryos have lethal or congenital LR patterning defects. Individual organs including the brain, heart, and gut also have LR asymmetry, and while the consequences of left and right signals mixing are severe or even lethal, organ-specific mechanisms for separating these signals remain poorly understood. Here, we uncover a midline structure composed of a transient double basement membrane, which separates the left and right halves of the embryonic chick DM during the establishment of intestinal and vascular asymmetries. Unlike other basement membranes of the DM, the midline is resistant to disruption by intercalation of Netrin4 (Ntn4). We propose that this atypical midline forms the boundary between left and right sides and functions as a barrier necessary to establish and protect organ asymmetry.
Collapse
Affiliation(s)
- Cora Demler
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell UniversityIthacaUnited States
| | - John C Lawlor
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell UniversityIthacaUnited States
| | - Ronit Yelin
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion – Israel Institute of TechnologyHaifaIsrael
| | - Dhana Llivichuzcha-Loja
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell UniversityIthacaUnited States
| | - Lihi Shaulov
- Rappaport Faculty of Medicine, Technion – Israel Institute of TechnologyHaifaIsrael
| | - David Kim
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell UniversityIthacaUnited States
| | - Megan Stewart
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell UniversityIthacaUnited States
| | - Frank K Lee
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell UniversityIthacaUnited States
| | - Natalia Shylo
- Stowers Institute for Medical ResearchKansas CityUnited States
| | - Paul A Trainor
- Stowers Institute for Medical ResearchKansas CityUnited States
- Department of Anatomy and Cell Biology, University of Kansas Medical CenterKansas CityUnited States
| | - Thomas M Schultheiss
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion – Israel Institute of TechnologyHaifaIsrael
| | - Natasza A Kurpios
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell UniversityIthacaUnited States
| |
Collapse
|
2
|
Zhu S, Loo YT, Veerapathiran S, Loo TYJ, Tran BN, Teh C, Zhong J, Matsudaira P, Saunders TE, Wohland T. Receptor binding and tortuosity explain morphogen local-to-global diffusion coefficient transition. Biophys J 2025; 124:963-979. [PMID: 39049492 PMCID: PMC11947475 DOI: 10.1016/j.bpj.2024.07.024] [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: 04/26/2024] [Revised: 06/28/2024] [Accepted: 07/19/2024] [Indexed: 07/27/2024] Open
Abstract
Morphogens are intercellular signaling molecules providing spatial information to cells in developing tissues to coordinate cell fate decisions. The spatial information is encoded within long-ranged concentration gradients of the morphogen. Direct measurement of morphogen dynamics in a range of systems suggests that local and global diffusion coefficients can differ by orders of magnitude. Further, local diffusivity can be large, which would potentially abolish any concentration gradient rapidly. Such observations have led to alternative transport models being proposed, including transcytosis and cytonemes. Here, we show that accounting for tissue architecture combined with receptor binding is sufficient to hinder the diffusive dynamics of morphogens, leading to an order of magnitude decrease in the effective diffusion coefficient from local to global scales. In particular, we built a realistic in silico architecture of the extracellular spaces of the zebrafish brain using light and electron microscopy data. Simulations on realistic architectures demonstrate that tortuosity and receptor binding within these spaces are sufficient to reproduce experimentally measured morphogen dynamics. Importantly, this work demonstrates that hindered diffusion is a viable mechanism for gradient formation, without requiring additional regulatory control.
Collapse
Affiliation(s)
- Shiwen Zhu
- NUS Centre for BioImaging Science, Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Yi Ting Loo
- Mathematics Institute, University of Warwick, Coventry, United Kingdom; Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Sapthaswaran Veerapathiran
- NUS Centre for BioImaging Science, Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Tricia Y J Loo
- Warwick Medical School, University of Warwick, Coventry, United Kingdom; Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Bich Ngoc Tran
- NUS Centre for BioImaging Science, Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Cathleen Teh
- NUS Centre for BioImaging Science, Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Jun Zhong
- NUS Centre for BioImaging Science, Department of Biological Sciences, National University of Singapore, Singapore, Singapore; Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Paul Matsudaira
- NUS Centre for BioImaging Science, Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Timothy E Saunders
- Warwick Medical School, University of Warwick, Coventry, United Kingdom; Mechanobiology Institute, National University of Singapore, Singapore, Singapore; Institute of Molecular and Cell Biology, A(∗)STAR, Singapore, Singapore.
| | - Thorsten Wohland
- NUS Centre for BioImaging Science, Department of Biological Sciences, National University of Singapore, Singapore, Singapore; Department of Chemistry, National University of Singapore, Singapore, Singapore; Institute of Digital Molecular Analytics and Science, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
3
|
Hutchins NT, Meziane M, Lu C, Mitalipova M, Fischer D, Li P. Reconstructing signaling histories of single cells via perturbation screens and transfer learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.16.643448. [PMID: 40166200 PMCID: PMC11957020 DOI: 10.1101/2025.03.16.643448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Manipulating the signaling environment is an effective approach to alter cellular states for broad-ranging applications, from engineering tissues to treating diseases. Such manipulation requires knowing the signaling states and histories of the cells in situ , for which high-throughput discovery methods are lacking. Here, we present an integrated experimental-computational framework that learns signaling response signatures from a high-throughput in vitro perturbation atlas and infers combinatorial signaling activities in in vivo cell types with high accuracy and temporal resolution. Specifically, we generated signaling perturbation atlas across diverse cell types/states through multiplexed sequential combinatorial screens on human pluripotent stem cells. Using the atlas to train IRIS, a neural network-based model, and predicting on mouse embryo scRNAseq atlas, we discovered global features of combinatorial signaling code usage over time, identified biologically meaningful heterogeneity of signaling states within each cell type, and reconstructed signaling histories along diverse cell lineages. We further demonstrated that IRIS greatly accelerates the optimization of stem cell differentiation protocols by drastically reducing the combinatorial space that needs to be tested. This framework leads to the revelation that different cell types share robust signal response signatures, and provides a scalable solution for mapping complex signaling interactions in vivo to guide targeted interventions.
Collapse
|
4
|
Yang Y, Qu Y, Wang J, Wang Y, Zhao J, Wang M, Hu W, Zhao J, Lin B, Zhang X, Luo Y. Exploring microfluidics-based organoid interactions through analysis of albumin secretion. LAB ON A CHIP 2025; 25:487-499. [PMID: 39840425 DOI: 10.1039/d4lc01085j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
Organoids-on-a-chip exhibit significant potential for advancing disease modeling, drug screening, and precision medicine, largely due to their capacity to facilitate interactions among organoids. However, the influence of chip design on these interactions remains poorly understood, primarily due to our limited knowledge of the mediators of communication and the complexity of interaction dynamics. This study demonstrates that analyzing albumin secretion from liver organoids within an organoids-on-a-chip system can provide a measure of the interaction intensity among organoids, offering valuable insights into how chip design influences these interactions. Our findings reveal that the interaction dynamics of target organoids is primarily affected by the types of neighboring organoids positioned upstream. For instance, adipose organoids located upstream and adjacent to liver organoids considerably stimulate functional improvements in the liver organoids, whereas adipose organoids in other arrangements do not produce similar effects. Importantly, both theoretical and experimental evidence indicate that the interaction dynamics is independent of the physical distance between organoids. Instead, it can be adjusted by flow rate, well depth, introducing a vascular barrier, or the media volume within the system. However, it is crucial to note that the influence of these factors is not linear. Finally, the exosome was identified as one of key mediators of communication within the organoids-on-a-chip system.
Collapse
Affiliation(s)
- Yang Yang
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, #2 Linggong Road, Dalian, 116024, China
| | - Yueyang Qu
- Jiangsu Key Laboratory of Neuropsychiatric Disease and College of Pharmaceutical Science, Suzhou Medical College, Soochow University, #199 Renai Road, Suzhou, 215127, China.
| | - Jing Wang
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, #2 Linggong Road, Dalian, 116024, China
| | - Yuxiu Wang
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, #2 Linggong Road, Dalian, 116024, China
| | - Jiamin Zhao
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, #2 Linggong Road, Dalian, 116024, China
| | - Miaomiao Wang
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, #2 Linggong Road, Dalian, 116024, China
| | - Wanqing Hu
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, #2 Linggong Road, Dalian, 116024, China
| | - Jiaqi Zhao
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, #2 Linggong Road, Dalian, 116024, China
| | - Bingcheng Lin
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, #457 Zhongshan Road, Dalian, 116023, China
| | - Xiuli Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Disease and College of Pharmaceutical Science, Suzhou Medical College, Soochow University, #199 Renai Road, Suzhou, 215127, China.
| | - Yong Luo
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, #2 Linggong Road, Dalian, 116024, China
| |
Collapse
|
5
|
Eissa T, Voronina L, Huber M, Fleischmann F, Žigman M. The Perils of Molecular Interpretations from Vibrational Spectra of Complex Samples. Angew Chem Int Ed Engl 2024:e202411596. [PMID: 39508580 DOI: 10.1002/anie.202411596] [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: 06/20/2024] [Indexed: 11/15/2024]
Abstract
Vibrational spectroscopy is a widely used technique for chemical characterizations across various analytical sciences. Its applications are increasingly extending to the analysis of complex samples such as biofluids, providing high-throughput molecular profiling. While powerful, the technique suffers from an inherent limitation: The overlap of absorption information across different spectral domains hinders the capacity to identify individual molecular substances contributing to measured signals. Despite the awareness of this challenge, the difficulty of analyzing multi-molecular spectra is often underestimated, leading to unsubstantiated molecular interpretations. Here, we examine the prevalent overreliance on spectral band assignment and illuminate the pitfalls of correlating spectral signals to discrete molecular entities or physiological states without rigorous validation. Focusing on blood-based infrared spectroscopy, we provide examples illustrating how peak overlap among different substances, relative substance concentrations, and preprocessing steps can lead to erroneous interpretations. We advocate for a viewpoint shift towards a more careful understanding of complex spectra, which shall lead to either accepting their fingerprinting nature and leveraging machine learning analysis - or involving additional measurement modalities for robust molecular interpretations. Aiming to help translate and improve analytical practices within the field, we highlight the limitations of molecular interpretations and feature their viable applications.
Collapse
Affiliation(s)
- Tarek Eissa
- Ludwig-Maximilians-Universität München (LMU), Chair of Exper-imental Physics - Laser Physics, Garching, Germany
- Max Planck Institute of Quantum Optics (MPQ), Laboratory for Attosecond Physics, Garching, Germany
- Technical University of Munich (TUM), School of Computation, Information and Technology, Garching, Germany
| | - Liudmila Voronina
- Ludwig-Maximilians-Universität München (LMU), Chair of Exper-imental Physics - Laser Physics, Garching, Germany
| | - Marinus Huber
- Ludwig-Maximilians-Universität München (LMU), Chair of Exper-imental Physics - Laser Physics, Garching, Germany
- Max Planck Institute of Quantum Optics (MPQ), Laboratory for Attosecond Physics, Garching, Germany
| | - Frank Fleischmann
- Ludwig-Maximilians-Universität München (LMU), Chair of Exper-imental Physics - Laser Physics, Garching, Germany
- Max Planck Institute of Quantum Optics (MPQ), Laboratory for Attosecond Physics, Garching, Germany
| | - Mihaela Žigman
- Ludwig-Maximilians-Universität München (LMU), Chair of Exper-imental Physics - Laser Physics, Garching, Germany
- Max Planck Institute of Quantum Optics (MPQ), Laboratory for Attosecond Physics, Garching, Germany
| |
Collapse
|
6
|
Takahara Y, Sumitomo T, Kono M, Takemura M, Akamatsu Y, Hirose Y, Yamaguchi M, Nakata M, Hotomi M, Kawabata S. Pneumolysin contributes to dysfunction of nasal epithelial barrier for promotion of pneumococcal dissemination into brain tissue. mSphere 2024; 9:e0065524. [PMID: 39345124 PMCID: PMC11520308 DOI: 10.1128/msphere.00655-24] [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: 08/19/2024] [Accepted: 09/10/2024] [Indexed: 10/01/2024] Open
Abstract
Streptococcus pneumoniae is one of the major pathogens responsible for bacterial meningitis and neurological sequelae. The present study was conducted to identify a non-hematogenous route used by S. pneumoniae to gain access to brain tissue without causing bacteremia or pneumonia, as well as bacterial and host factors involved in this process. To investigate the molecular mechanisms and dissemination pathways of pneumococcal infection in brain tissue, mice were intranasally inoculated with S. pneumoniae strain EF3030, a clinical isolate from a patient with otitis media. Pneumococci were isolated from the frontal olfactory bulb, caudal cerebrum, and cerebellum, with neither bacteremia nor pneumonia observed in the present model. Immunostaining imaging revealed the presence of S. pneumoniae organisms in olfactory nerve fibers. Knockout of the ply gene encoding pneumolysin (PLY) markedly compromised the ability of the bacterial organisms to disseminate into brain tissue, whereas the dissemination efficiency of the complemented strain was restored to nearly the same level as the wild type. Notably, distinct upregulation of Gli1 and Snail1, which are involved in the transcriptional repression of junctional proteins, along with downregulation of E-cadherin, was detected in nasal lavage samples from mice infected with the wild-type or complemented strain, but not in those from mice infected with the ply mutant. Taken together, the present findings indicate that PLY induces Gli1-Snail1-dependent dysfunction of the nasal epithelial barrier, thus allowing pneumococcal dissemination to brain tissue that occurs in a non-hematogenous manner.IMPORTANCEBacterial meningitis, considered to be caused by bacteremia, can lead to blood-brain barrier disruption and bacterial dissemination into the central nervous system. Despite the availability of intravenously administered antibiotics with cerebrospinal fluid transferability, bacterial meningitis remains associated with high rates of morbidity and mortality. Here, we utilized Streptococcus pneumoniae strain EF3030, clinically isolated from otitis media, for the construction of a murine infection model to investigate the molecular mechanisms by which nasally colonized pneumococci disseminate into brain tissue. The obtained findings indicate that pneumolysin (PLY) induces Gli1-Snail1-dependent dysfunction of the nasal epithelial barrier, which facilitates pneumococcal dissemination to brain tissue in a non-hematogenous manner. Our results support the existence of an alternative route by which S. pneumoniae can reach the central nervous system and indicate the need for the development of novel therapeutic strategies, which would be an important contribution to the clinical management of bacterial meningitis.
Collapse
Affiliation(s)
- Yuki Takahara
- Department of Microbiology, Osaka University Graduate School of Dentistry, Osaka, Japan
- Department of Fixed Prosthodontics and Orofacial Function, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Tomoko Sumitomo
- Department of Microbiology, Osaka University Graduate School of Dentistry, Osaka, Japan
- Department of Oral Microbiology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Masamitsu Kono
- Department of Otorhinolaryngology—Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| | - Moe Takemura
- Department of Microbiology, Osaka University Graduate School of Dentistry, Osaka, Japan
- Department of Oral Surgery, Rinku General Medical Center, Izumisano, Osaka, Japan
| | - Yukako Akamatsu
- Department of Microbiology, Osaka University Graduate School of Dentistry, Osaka, Japan
- Division of Special Care Dentistry, Osaka University Dental Hospital, Osaka, Japan
| | - Yujiro Hirose
- Department of Microbiology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Masaya Yamaguchi
- Department of Microbiology, Osaka University Graduate School of Dentistry, Osaka, Japan
- Bioinformatics Research Unit, Osaka University Graduate School of Dentistry, Osaka, Japan
- Bioinformatics Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Center for Infectious Diseases Education and Research, Osaka University, Osaka, Japan
| | - Masanobu Nakata
- Department of Oral Microbiology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Muneki Hotomi
- Department of Otorhinolaryngology—Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| | - Shigetada Kawabata
- Department of Microbiology, Osaka University Graduate School of Dentistry, Osaka, Japan
- Center for Infectious Diseases Education and Research, Osaka University, Osaka, Japan
| |
Collapse
|
7
|
Schlissel G, Meziane M, Narducci D, Hansen AS, Li P. Diffusion barriers imposed by tissue topology shape Hedgehog morphogen gradients. Proc Natl Acad Sci U S A 2024; 121:e2400677121. [PMID: 39190357 PMCID: PMC11388384 DOI: 10.1073/pnas.2400677121] [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: 01/11/2024] [Accepted: 07/15/2024] [Indexed: 08/28/2024] Open
Abstract
Animals use a small number of morphogens to pattern tissues, but it is unclear how evolution modulates morphogen signaling range to match tissues of varying sizes. Here, we used single-molecule imaging in reconstituted morphogen gradients and in tissue explants to determine that Hedgehog diffused extracellularly as a monomer, and rapidly transitioned between membrane-confined and -unconfined states. Unexpectedly, the vertebrate-specific protein SCUBE1 expanded Hedgehog gradients by accelerating the transition rates between states without affecting the relative abundance of molecules in each state. This observation could not be explained under existing models of morphogen diffusion. Instead, we developed a topology-limited diffusion model in which cell-cell gaps create diffusion barriers, which morphogens can only overcome by passing through a membrane-unconfined state. Under this model, SCUBE1 promoted Hedgehog secretion and diffusion by allowing it to transiently overcome diffusion barriers. This multiscale understanding of morphogen gradient formation unified prior models and identified knobs that nature can use to tune morphogen gradient sizes across tissues and organisms.
Collapse
Affiliation(s)
- Gavin Schlissel
- Whitehead Institute for Biomedical Research, Cambridge, MA02142
| | - Miram Meziane
- Whitehead Institute for Biomedical Research, Cambridge, MA02142
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Domenic Narducci
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
- Gene Regulation Observatory, The Broad Institute of MIT and Harvard, Cambridge, MA02142
- Koch Institute for Integrative Cancer Research, Cambridge, MA02139
| | - Anders S. Hansen
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
- Gene Regulation Observatory, The Broad Institute of MIT and Harvard, Cambridge, MA02142
- Koch Institute for Integrative Cancer Research, Cambridge, MA02139
| | - Pulin Li
- Whitehead Institute for Biomedical Research, Cambridge, MA02142
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA02139
| |
Collapse
|
8
|
Caldarelli P, Chamolly A, Villedieu A, Alegria-Prévot O, Phan C, Gros J, Corson F. Self-organized tissue mechanics underlie embryonic regulation. Nature 2024; 633:887-894. [PMID: 39261736 PMCID: PMC11424473 DOI: 10.1038/s41586-024-07934-8] [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: 03/31/2022] [Accepted: 08/09/2024] [Indexed: 09/13/2024]
Abstract
Early amniote development is highly self-organized, capable of adapting to interference through local and long-range cell-cell interactions. This process, called embryonic regulation1, has been well illustrated in experiments on avian embryos, in which subdividing the epiblast disk into different parts not only redirects cell fates to eventually form a complete and well-proportioned embryo at its original location, but also leads to the self-organization of additional, fully formed embryos2,3 in the other separated parts. The cellular interactions underlying embryonic self-organization are widely believed to be mediated by molecular signals, yet the identity of such signals is unclear. Here, by analysing intact and mechanically perturbed quail embryos, we show that the mechanical forces that drive embryogenesis self-organize, with contractility locally self-activating and the ensuing tension acting as a long-range inhibitor. This mechanical feedback governs the persistent pattern of tissue flows that shape the embryo4-6 and also steers the concomitant emergence of embryonic territories by modulating gene expression, ensuring the formation of a single embryo under normal conditions, yet allowing the emergence of multiple, well-proportioned embryos after perturbations. Thus, mechanical forces act at the core of embryonic self-organization, shaping both tissues and gene expression to robustly yet plastically canalize early development.
Collapse
Affiliation(s)
- Paolo Caldarelli
- Developmental and Stem Cell Biology Department, Institut Pasteur, Université de Paris, CNRS UMR3738, Paris, France
- Collège Doctoral, Sorbonne Université, Paris, France
| | - Alexander Chamolly
- Developmental and Stem Cell Biology Department, Institut Pasteur, Université de Paris, CNRS UMR3738, Paris, France
- Laboratoire de Physique de l'Ecole Normale Supérieure, CNRS, ENS, Université PSL, Sorbonne Université, Université de Paris, Paris, France
| | - Aurélien Villedieu
- Developmental and Stem Cell Biology Department, Institut Pasteur, Université de Paris, CNRS UMR3738, Paris, France
| | - Olinda Alegria-Prévot
- Developmental and Stem Cell Biology Department, Institut Pasteur, Université de Paris, CNRS UMR3738, Paris, France
| | - Carole Phan
- Developmental and Stem Cell Biology Department, Institut Pasteur, Université de Paris, CNRS UMR3738, Paris, France
| | - Jerome Gros
- Developmental and Stem Cell Biology Department, Institut Pasteur, Université de Paris, CNRS UMR3738, Paris, France.
| | - Francis Corson
- Laboratoire de Physique de l'Ecole Normale Supérieure, CNRS, ENS, Université PSL, Sorbonne Université, Université de Paris, Paris, France.
- Center for Studies in Physics and Biology, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
9
|
Demler C, Lawlor JC, Yelin R, Llivichuzcha-Loja D, Shaulov L, Kim D, Stewart M, Lee F, Shylo NA, Trainor PA, Schultheiss T, Kurpios NA. An atypical basement membrane forms a midline barrier during left-right asymmetric gut development in the chicken embryo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.15.553395. [PMID: 37645918 PMCID: PMC10461973 DOI: 10.1101/2023.08.15.553395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Correct intestinal morphogenesis depends on the early embryonic process of gut rotation, an evolutionarily conserved program in which a straight gut tube elongates and forms into its first loops. However, the gut tube requires guidance to loop in a reproducible manner. The dorsal mesentery (DM) connects the gut tube to the body and directs the lengthening gut into stereotypical loops via left-right (LR) asymmetric cellular and extracellular behavior. The LR asymmetry of the DM also governs blood and lymphatic vessel formation for the digestive tract, which is essential for prenatal organ development and postnatal vital functions including nutrient absorption. Although the genetic LR asymmetry of the DM has been extensively studied, a divider between the left and right DM has yet to be identified. Setting up LR asymmetry for the entire body requires a Lefty1+ midline barrier to separate the two sides of the embryo, without it, embryos have lethal or congenital LR patterning defects. Individual organs including the brain, heart, and gut also have LR asymmetry, and while the consequences of left and right signals mixing are severe or even lethal, organ-specific mechanisms for separating these signals are poorly understood. Here, we uncover a midline structure composed of a transient double basement membrane, which separates the left and right halves of the embryonic chick DM during the establishment of intestinal and vascular asymmetries. Unlike other basement membranes of the DM, the midline is resistant to disruption by intercalation of Netrin4 (Ntn4). We propose that this atypical midline forms the boundary between left and right sides and functions as a barrier necessary to establish and protect organ asymmetry.
Collapse
Affiliation(s)
- Cora Demler
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - John Coates Lawlor
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Ronit Yelin
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Dhana Llivichuzcha-Loja
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Lihi Shaulov
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - David Kim
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Megan Stewart
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | | | | | - Paul A. Trainor
- Stowers Institute for Medical Research, Kansas City, MO, USA
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Missouri, USA
| | - Thomas Schultheiss
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Natasza A. Kurpios
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
10
|
Quach H, Farrell S, Wu MJM, Kanagarajah K, Leung JWH, Xu X, Kallurkar P, Turinsky AL, Bear CE, Ratjen F, Kalish B, Goyal S, Moraes TJ, Wong AP. Early human fetal lung atlas reveals the temporal dynamics of epithelial cell plasticity. Nat Commun 2024; 15:5898. [PMID: 39003323 PMCID: PMC11246468 DOI: 10.1038/s41467-024-50281-5] [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: 11/16/2023] [Accepted: 07/05/2024] [Indexed: 07/15/2024] Open
Abstract
Studying human fetal lungs can inform how developmental defects and disease states alter the function of the lungs. Here, we sequenced >150,000 single cells from 19 healthy human pseudoglandular fetal lung tissues ranging between gestational weeks 10-19. We capture dynamic developmental trajectories from progenitor cells that express abundant levels of the cystic fibrosis conductance transmembrane regulator (CFTR). These cells give rise to multiple specialized epithelial cell types. Combined with spatial transcriptomics, we show temporal regulation of key signalling pathways that may drive the temporal and spatial emergence of specialized epithelial cells including ciliated and pulmonary neuroendocrine cells. Finally, we show that human pluripotent stem cell-derived fetal lung models contain CFTR-expressing progenitor cells that capture similar lineage developmental trajectories as identified in the native tissue. Overall, this study provides a comprehensive single-cell atlas of the developing human lung, outlining the temporal and spatial complexities of cell lineage development and benchmarks fetal lung cultures from human pluripotent stem cell differentiations to similar developmental window.
Collapse
Affiliation(s)
- Henry Quach
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Spencer Farrell
- Department of Physics, University of Toronto, Toronto, Ontario, Canada
| | - Ming Jia Michael Wu
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Kayshani Kanagarajah
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Joseph Wai-Hin Leung
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Xiaoqiao Xu
- Centre for Computational Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Prajkta Kallurkar
- Centre for Computational Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Andrei L Turinsky
- Centre for Computational Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Christine E Bear
- Program in Molecular Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Felix Ratjen
- Program in Translational Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Brian Kalish
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Division of Neonatology, Department of Paediatrics, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Sidhartha Goyal
- Department of Physics, University of Toronto, Toronto, Ontario, Canada
| | - Theo J Moraes
- Program in Translational Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Amy P Wong
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada.
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
11
|
Yang Y, Li S, Luo L. Responses of organ precursors to correct and incorrect inductive signals. Trends Cell Biol 2024; 34:484-495. [PMID: 37739814 DOI: 10.1016/j.tcb.2023.08.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/14/2023] [Accepted: 08/31/2023] [Indexed: 09/24/2023]
Abstract
During embryonic development, the inductive molecules produced by local origins normally arrive at their target tissues in a nondirectional, diffusion manner. The target organ precursor cells must correctly interpret these inductive signals to ensure proper specification/differentiation, which is dependent on two prerequisites: (i) obtaining cell-intrinsic competence; and (ii) receiving correct inductive signals while resisting incorrect ones. Gain of intrinsic competence could avoid a large number of misinductions because the incompetent cells are nonresponsive to inductive signals. However, in cases of different precursor cells with similar competence and located in close proximity, resistance to incorrect inductive signals is essential for accurate determination of cell fate. Here we outline the mechanisms of how organ precursors respond to correct and incorrect inductive signals.
Collapse
Affiliation(s)
- Yun Yang
- Institute of Development Biology and Regenerative Medicine, Southwest University, Chongqing, China
| | - Shuang Li
- Institute of Development Biology and Regenerative Medicine, Southwest University, Chongqing, China
| | - Lingfei Luo
- Institute of Development Biology and Regenerative Medicine, Southwest University, Chongqing, China; School of Life Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
12
|
Wattenberg MM, Colby S, Garrido-Laguna I, Xue Y, Chang R, Delman D, Lee J, Affolter K, Mulvihill SJ, Beg MS, Wang-Gillam A, Wade JL, Guthrie KA, Chiorean EG, Ahmad SA, Lowy AM, Philip PA, Sohal DPS, Beatty GL. Intratumoral Cell Neighborhoods Coordinate Outcomes in Pancreatic Ductal Adenocarcinoma. Gastroenterology 2024; 166:1114-1129. [PMID: 38244727 PMCID: PMC11102852 DOI: 10.1053/j.gastro.2024.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 01/22/2024]
Abstract
BACKGROUND & AIMS Pancreatic ductal adenocarcinoma (PDA) is a highly lethal disease characterized by a spatially heterogeneous tumor microenvironment. Within the PDA microenvironment, cells organize into communities where cell fate is influenced by neighboring cells of diverse ontogeny and function. However, it remains unclear how cell neighborhoods in the tumor microenvironment evolve with treatment and impact clinical outcomes. METHODS Here, using automated chromogenic multiplex immunohistochemistry and unsupervised computational image analysis of human PDA tumors, we investigated cell neighborhoods in surgically resected tumors from patients with chemotherapy-naïve PDA (n = 59) and neoadjuvant chemotherapy-treated PDA (n = 57). Single cells were defined by lineage markers (CD3, CD8, Foxp3, CD68, CK19), proliferation (Ki67), and neighboring cells. RESULTS Distinct intratumoral immune and tumor cell subsets were defined by neighboring cells. Higher content of stromal-associated macrophages was seen in chemotherapy-naïve tumors from long-term survivors (overall survival >3 years) compared with short-term survivors (overall survival <1 year), whereas immune-excluded tumor cells were higher in short-term survivors. Chemotherapy-treated vs -naïve tumors showed lower content of tumor-associated T cells and macrophages but similar densities of stromal-associated immune cells. However, proliferating tumor cell subsets with immune-rich neighborhoods were higher in chemotherapy-treated tumors. In a blinded analysis of tumors from patients treated with neoadjuvant chemotherapy, a composite index comprising lower quantities of immune-excluded tumor cells and higher spatially distinct immune cell subsets was associated with prolonged survival. CONCLUSIONS Together, these data provide new insights into discrete cell communities in PDA and show their clinical relevance.
Collapse
Affiliation(s)
- Max M Wattenberg
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sarah Colby
- SWOG Statistics and Data Management Center, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Ignacio Garrido-Laguna
- Division of Oncology, Department of Internal Medicine, University of Utah School of Medicine, University of Utah, Salt Lake City, Utah
| | - Yuqing Xue
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Renee Chang
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Devora Delman
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jesse Lee
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kajsa Affolter
- Department of Pathology, University of Utah School of Medicine, University of Utah, Salt Lake City, Utah
| | - Sean J Mulvihill
- Department of Surgery, University of Utah School of Medicine, University of Utah, Salt Lake City, Utah
| | | | | | | | - Katherine A Guthrie
- SWOG Statistics and Data Management Center, Fred Hutchinson Cancer Center, Seattle, Washington
| | - E Gabriela Chiorean
- University of Washington, Fred Hutchinson Cancer Center, Seattle, Washington
| | | | - Andrew M Lowy
- Division of Surgical Oncology, Department of Surgery, UC San Diego, La Jolla, California
| | - Philip Agop Philip
- Henry Ford Health, Wayne State University, Oncology and Pharmacology, Detroit, Michigan
| | | | - Gregory L Beatty
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
13
|
Zhou Z, Liu J, Xiong T, Liu Y, Tuan RS, Li ZA. Engineering Innervated Musculoskeletal Tissues for Regenerative Orthopedics and Disease Modeling. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310614. [PMID: 38200684 DOI: 10.1002/smll.202310614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 12/28/2023] [Indexed: 01/12/2024]
Abstract
Musculoskeletal (MSK) disorders significantly burden patients and society, resulting in high healthcare costs and productivity loss. These disorders are the leading cause of physical disability, and their prevalence is expected to increase as sedentary lifestyles become common and the global population of the elderly increases. Proper innervation is critical to maintaining MSK function, and nerve damage or dysfunction underlies various MSK disorders, underscoring the potential of restoring nerve function in MSK disorder treatment. However, most MSK tissue engineering strategies have overlooked the significance of innervation. This review first expounds upon innervation in the MSK system and its importance in maintaining MSK homeostasis and functions. This will be followed by strategies for engineering MSK tissues that induce post-implantation in situ innervation or are pre-innervated. Subsequently, research progress in modeling MSK disorders using innervated MSK organoids and organs-on-chips (OoCs) is analyzed. Finally, the future development of engineering innervated MSK tissues to treat MSK disorders and recapitulate disease mechanisms is discussed. This review provides valuable insights into the underlying principles, engineering methods, and applications of innervated MSK tissues, paving the way for the development of targeted, efficacious therapies for various MSK conditions.
Collapse
Affiliation(s)
- Zhilong Zhou
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
| | - Jun Liu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
| | - Tiandi Xiong
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
| | - Yuwei Liu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, 518000, P. R. China
| | - Rocky S Tuan
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
| | - Zhong Alan Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Key Laboratory of Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518057, P. R. China
| |
Collapse
|
14
|
Recouvreux P, Pai P, Dunsing V, Torro R, Ludanyi M, Mélénec P, Boughzala M, Bertrand V, Lenne PF. Transfer of polarity information via diffusion of Wnt ligands in C. elegans embryos. Curr Biol 2024; 34:1853-1865.e6. [PMID: 38604167 DOI: 10.1016/j.cub.2024.03.030] [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: 01/18/2023] [Revised: 10/26/2023] [Accepted: 03/18/2024] [Indexed: 04/13/2024]
Abstract
Different signaling mechanisms concur to ensure robust tissue patterning and cell fate instruction during animal development. Most of these mechanisms rely on signaling proteins that are produced, transported, and detected. The spatiotemporal dynamics of signaling molecules are largely unknown, yet they determine signal activity's spatial range and time frame. Here, we use the Caenorhabditis elegans embryo to study how Wnt ligands, an evolutionarily conserved family of signaling proteins, dynamically organize to establish cell polarity in a developing tissue. We identify how Wnt ligands, produced in the posterior half of the embryos, spread extracellularly to transmit information to distant target cells in the anterior half. With quantitative live imaging and fluorescence correlation spectroscopy, we show that Wnt ligands diffuse through the embryo over a timescale shorter than the cell cycle, in the intercellular space, and outside the tissue below the eggshell. We extracted diffusion coefficients of Wnt ligands and their receptor Frizzled and characterized their co-localization. Integrating our different measurements and observations in a simple computational framework, we show how fast diffusion in the embryo can polarize individual cells through a time integration of the arrival of the ligands at the target cells. The polarity established at the tissue level by a posterior Wnt source can be transferred to the cellular level. Our results support a diffusion-based long-range Wnt signaling, which is consistent with the dynamics of developing processes.
Collapse
Affiliation(s)
- Pierre Recouvreux
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living Systems, Marseille, France.
| | - Pritha Pai
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living Systems, Marseille, France
| | - Valentin Dunsing
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living Systems, Marseille, France
| | - Rémy Torro
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living Systems, Marseille, France
| | - Monika Ludanyi
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living Systems, Marseille, France
| | - Pauline Mélénec
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living Systems, Marseille, France
| | - Mariem Boughzala
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living Systems, Marseille, France
| | - Vincent Bertrand
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living Systems, Marseille, France
| | - Pierre-François Lenne
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living Systems, Marseille, France
| |
Collapse
|
15
|
Schlissel G, Meziane M, Narducci D, Hansen AS, Li P. Diffusion barriers imposed by tissue topology shape morphogen gradients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.01.592050. [PMID: 38746265 PMCID: PMC11092646 DOI: 10.1101/2024.05.01.592050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Animals use a small number of morphogens to pattern tissues, but it is unclear how evolution modulates morphogen signaling range to match tissues of varying sizes. Here, we used single molecule imaging in reconstituted morphogen gradients and in tissue explants to determine that Hedgehog diffused extra-cellularly as a monomer, and rapidly transitioned between membrane-confined and -unconfined states. Unexpectedly, the vertebrate-specific protein SCUBE1 expanded Hedgehog gradients by accelerating the transition rates between states without affecting the relative abundance of molecules in each state. This observation could not be explained under existing models of morphogen diffusion. Instead, we developed a topology-limited diffusion model in which cell-cell gaps create diffusion barriers, and morphogens can only overcome the barrier by passing through a membrane-unconfined state. Under this model, SCUBE1 promotes Hedgehog secretion and diffusion by allowing it to transiently overcome diffusion barriers. This multiscale understanding of morphogen gradient formation unified prior models and discovered novel knobs that nature can use to tune morphogen gradient sizes across tissues and organisms.
Collapse
|
16
|
Lingam M. Information Transmission via Molecular Communication in Astrobiological Environments. ASTROBIOLOGY 2024; 24:84-99. [PMID: 38109216 DOI: 10.1089/ast.2023.0069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2023]
Abstract
The ubiquity of information transmission via molecular communication between cells is comprehensively documented on Earth; this phenomenon might even have played a vital role in the origin(s) and early evolution of life. Motivated by these considerations, a simple model for molecular communication entailing the diffusion of signaling molecules from transmitter to receiver is elucidated. The channel capacity C (maximal rate of information transmission) and an optimistic heuristic estimate of the actual information transmission rate ℐ are derived for this communication system; the two quantities, especially the latter, are demonstrated to be broadly consistent with laboratory experiments and more sophisticated theoretical models. The channel capacity exhibits a potentially weak dependence on environmental parameters, whereas the actual information transmission rate may scale with the intercellular distance d as ℐ ∝ d-4 and could vary substantially across settings. These two variables are roughly calculated for diverse astrobiological environments, ranging from Earth's upper oceans (C ∼ 3.1 × 103 bits/s; ℐ ∼ 4.7 × 10-2 bits/s) and deep sea hydrothermal vents (C ∼ 4.2 × 103 bits/s; ℐ ∼ 1.2 × 10-1 bits/s) to the hydrocarbon lakes and seas of Titan (C ∼ 3.8 × 103 bits/s; ℐ ∼ 2.6 × 10-1 bits/s).
Collapse
Affiliation(s)
- Manasvi Lingam
- Department of Aerospace, Physics and Space Sciences, Florida Institute of Technology, Melbourne, Florida, USA
- Department of Physics, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
17
|
Arif S, Moulin VJ. Extracellular vesicles on the move: Traversing the complex matrix of tissues. Eur J Cell Biol 2023; 102:151372. [PMID: 37972445 DOI: 10.1016/j.ejcb.2023.151372] [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: 06/22/2023] [Revised: 11/10/2023] [Accepted: 11/10/2023] [Indexed: 11/19/2023] Open
Abstract
Extracellular vesicles are small particles involved in intercellular signaling. They are produced by virtually all cell types, transport biological molecules, and are released into the extracellular space. Studies on extracellular vesicles have become more numerous in recent years, leading to promising research on their potential impact on health and disease. Despite significant progress in understanding the bioactivity of extracellular vesicles, most in vitro and in vivo studies overlook their transport through the extracellular matrix in tissues. The interaction or free diffusion of extracellular vesicles in their environment can provide valuable insights into their efficacy and function. Therefore, understanding the factors that influence the transport of extracellular vesicles in the extracellular matrix is essential for the development of new therapeutic approaches that involve the use of these extracellular vesicles. This review discusses the importance of the interaction between extracellular vesicles and the extracellular matrix and the different factors that influence their diffusion. In addition, we evaluate their role in tissue homeostasis, pathophysiology, and potential clinical applications. Understanding the complex interaction between extracellular vesicles and the extracellular matrix is critical in order to develop effective strategies to target specific cells and tissues in a wide range of clinical applications.
Collapse
Affiliation(s)
- Syrine Arif
- Faculté de Médecine, Université Laval, Quebec City, QC G1V 0A6, Canada; Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC G1S 4L8, Canada; Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Quebec City, QC G1J 1Z4, Canada
| | - Véronique J Moulin
- Faculté de Médecine, Université Laval, Quebec City, QC G1V 0A6, Canada; Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC G1S 4L8, Canada; Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Quebec City, QC G1J 1Z4, Canada; Department of Surgery, Faculty of Medicine, Université Laval, Quebec City, QC, Canada.
| |
Collapse
|
18
|
Thrane K, Winge MCG, Wang H, Chen L, Guo MG, Andersson A, Abalo XM, Yang X, Kim DS, Longo SK, Soong BY, Meyers JM, Reynolds DL, McGeever A, Demircioglu D, Hasson D, Mirzazadeh R, Rubin AJ, Bae GH, Karkanias J, Rieger K, Lundeberg J, Ji AL. Single-Cell and Spatial Transcriptomic Analysis of Human Skin Delineates Intercellular Communication and Pathogenic Cells. J Invest Dermatol 2023; 143:2177-2192.e13. [PMID: 37142187 PMCID: PMC10592679 DOI: 10.1016/j.jid.2023.02.040] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 01/26/2023] [Accepted: 02/16/2023] [Indexed: 05/06/2023]
Abstract
Epidermal homeostasis is governed by a balance between keratinocyte proliferation and differentiation with contributions from cell-cell interactions, but conserved or divergent mechanisms governing this equilibrium across species and how an imbalance contributes to skin disease are largely undefined. To address these questions, human skin single-cell RNA sequencing and spatial transcriptomics data were integrated and compared with mouse skin data. Human skin cell-type annotation was improved using matched spatial transcriptomics data, highlighting the importance of spatial context in cell-type identity, and spatial transcriptomics refined cellular communication inference. In cross-species analyses, we identified a human spinous keratinocyte subpopulation that exhibited proliferative capacity and a heavy metal processing signature, which was absent in mouse and may account for species differences in epidermal thickness. This human subpopulation was expanded in psoriasis and zinc-deficiency dermatitis, attesting to disease relevance and suggesting a paradigm of subpopulation dysfunction as a hallmark of the disease. To assess additional potential subpopulation drivers of skin diseases, we performed cell-of-origin enrichment analysis within genodermatoses, nominating pathogenic cell subpopulations and their communication pathways, which highlighted multiple potential therapeutic targets. This integrated dataset is encompassed in a publicly available web resource to aid mechanistic and translational studies of normal and diseased skin.
Collapse
Affiliation(s)
- Kim Thrane
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Solna, Sweden
| | - Mårten C G Winge
- Program in Epithelial Biology, Department of Dermatology, Stanford University School of Medicine, Stanford, California, USA
| | - Hongyu Wang
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Black Family Stem Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA; School of Computer Science and Engineering, Northwestern Polytechnical University, Xi'an, China
| | - Larry Chen
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Black Family Stem Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Margaret G Guo
- Program in Epithelial Biology, Department of Dermatology, Stanford University School of Medicine, Stanford, California, USA; Biomedical Informatics Program, Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, California, USA
| | - Alma Andersson
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Solna, Sweden
| | - Xesús M Abalo
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Solna, Sweden
| | - Xue Yang
- Program in Epithelial Biology, Department of Dermatology, Stanford University School of Medicine, Stanford, California, USA
| | - Daniel S Kim
- Program in Epithelial Biology, Department of Dermatology, Stanford University School of Medicine, Stanford, California, USA; Biomedical Informatics Program, Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, California, USA
| | - Sophia K Longo
- Program in Epithelial Biology, Department of Dermatology, Stanford University School of Medicine, Stanford, California, USA
| | - Brian Y Soong
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Black Family Stem Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jordan M Meyers
- Program in Epithelial Biology, Department of Dermatology, Stanford University School of Medicine, Stanford, California, USA
| | - David L Reynolds
- Program in Epithelial Biology, Department of Dermatology, Stanford University School of Medicine, Stanford, California, USA
| | - Aaron McGeever
- Chan Zuckerberg Biohub San Francisco, San Francisco, California, USA
| | - Deniz Demircioglu
- Bioinformatics for Next Generation Sequencing Core, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Dan Hasson
- Bioinformatics for Next Generation Sequencing Core, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Reza Mirzazadeh
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Solna, Sweden
| | - Adam J Rubin
- Program in Epithelial Biology, Department of Dermatology, Stanford University School of Medicine, Stanford, California, USA
| | - Gordon H Bae
- Program in Epithelial Biology, Department of Dermatology, Stanford University School of Medicine, Stanford, California, USA
| | - Jim Karkanias
- Chan Zuckerberg Biohub San Francisco, San Francisco, California, USA
| | - Kerri Rieger
- Program in Epithelial Biology, Department of Dermatology, Stanford University School of Medicine, Stanford, California, USA
| | - Joakim Lundeberg
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Solna, Sweden
| | - Andrew L Ji
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Black Family Stem Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| |
Collapse
|
19
|
Harish RK, Gupta M, Zöller D, Hartmann H, Gheisari A, Machate A, Hans S, Brand M. Real-time monitoring of an endogenous Fgf8a gradient attests to its role as a morphogen during zebrafish gastrulation. Development 2023; 150:dev201559. [PMID: 37665167 PMCID: PMC10565248 DOI: 10.1242/dev.201559] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 08/29/2023] [Indexed: 09/05/2023]
Abstract
Morphogen gradients impart positional information to cells in a homogenous tissue field. Fgf8a, a highly conserved growth factor, has been proposed to act as a morphogen during zebrafish gastrulation. However, technical limitations have so far prevented direct visualization of the endogenous Fgf8a gradient and confirmation of its morphogenic activity. Here, we monitor Fgf8a propagation in the developing neural plate using a CRISPR/Cas9-mediated EGFP knock-in at the endogenous fgf8a locus. By combining sensitive imaging with single-molecule fluorescence correlation spectroscopy, we demonstrate that Fgf8a, which is produced at the embryonic margin, propagates by diffusion through the extracellular space and forms a graded distribution towards the animal pole. Overlaying the Fgf8a gradient curve with expression profiles of its downstream targets determines the precise input-output relationship of Fgf8a-mediated patterning. Manipulation of the extracellular Fgf8a levels alters the signaling outcome, thus establishing Fgf8a as a bona fide morphogen during zebrafish gastrulation. Furthermore, by hindering Fgf8a diffusion, we demonstrate that extracellular diffusion of the protein from the source is crucial for it to achieve its morphogenic potential.
Collapse
Affiliation(s)
- Rohit Krishnan Harish
- CRTD – Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
- PoL – Cluster of Excellence Physics of Life, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| | - Mansi Gupta
- CRTD – Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
- PoL – Cluster of Excellence Physics of Life, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| | - Daniela Zöller
- CRTD – Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
- PoL – Cluster of Excellence Physics of Life, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| | - Hella Hartmann
- CRTD – Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
- CMCB Technology Platform, Technische Universität Dresden, Tatzberg 47-51, 01307 Dresden, Germany
| | - Ali Gheisari
- CRTD – Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
- CMCB Technology Platform, Technische Universität Dresden, Tatzberg 47-51, 01307 Dresden, Germany
| | - Anja Machate
- CRTD – Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
- PoL – Cluster of Excellence Physics of Life, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| | - Stefan Hans
- CRTD – Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
- PoL – Cluster of Excellence Physics of Life, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| | - Michael Brand
- CRTD – Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
- PoL – Cluster of Excellence Physics of Life, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| |
Collapse
|
20
|
Martin L, Schumacher L, Chandra T. Modelling the dynamics of senescence spread. Aging Cell 2023; 22:e13892. [PMID: 37288475 PMCID: PMC10410058 DOI: 10.1111/acel.13892] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/04/2023] [Accepted: 05/09/2023] [Indexed: 06/09/2023] Open
Abstract
Cellular senescence is a cell surveillance mechanism that arrests the cell cycle in damaged cells. The senescent phenotype can spread from cell to cell through paracrine and juxtacrine signalling, but the dynamics of this process are not well understood. Although senescent cells are important in ageing, wound healing and cancer, it is unclear how the spread of senescence is contained in senescent lesions. In the absence of the immune system, senescence could theoretically spread infinitely from one cell to another, but this contradicts experimental evidence. To investigate this issue, we developed both a minimal mathematical model and a stochastic simulation of senescence spread. Our results suggest that differences in the number of signalling molecules secreted between subtypes of senescent cells can limit the spread of senescence. We found that dynamic, time-dependent paracrine signalling prevents the uncontrolled spread of senescence, and we demonstrate how model parameters can be determined using Bayesian inference in a proposed experiment.
Collapse
Affiliation(s)
- Lucy Martin
- Institute of Genetics and CancerUniversity of EdinburghEdinburghUK
| | - Linus Schumacher
- Centre for Regenerative MedicineUniversity of EdinburghEdinburghUK
| | - Tamir Chandra
- Institute of Genetics and CancerUniversity of EdinburghEdinburghUK
| |
Collapse
|
21
|
Bowman RL, Wang D, Eom DS. A macrophage subpopulation promotes airineme-mediated intercellular communication in a matrix metalloproteinase-9 dependent manner. Cell Rep 2023; 42:112818. [PMID: 37454294 PMCID: PMC10530396 DOI: 10.1016/j.celrep.2023.112818] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 06/05/2023] [Accepted: 06/30/2023] [Indexed: 07/18/2023] Open
Abstract
Tissue-resident macrophages are heterogeneous and perform location-dependent functions. Skin resident macrophages play intriguing roles in long-distance intercellular signaling by mediating cellular protrusions called airinemes in zebrafish. These macrophages relay signaling molecules containing airineme vesicles between pigment cells, and their absence disrupts airineme-mediated signaling and pigment pattern formation. It is unknown if the same macrophages control both these signaling and typical immune functions or if a separate subpopulation functions in intercellular communication. With high-resolution imaging and genetic ablation approaches, we identify a macrophage subpopulation responsible for airineme-mediated signaling. These seem to be distinct from conventional skin-resident macrophages by their ameboid morphology and faster or expansive migratory behaviors. They resemble ectoderm-derived macrophages termed metaphocytes. Metaphocyte ablation markedly decreases airineme extension and signaling. In addition, these ameboid/metaphocytes require matrix metalloproteinase-9 for their migration and airineme-mediated signaling. These results reveal a macrophage subpopulation with specialized functions in airineme-mediated signaling, which may play roles in other aspects of intercellular communication.
Collapse
Affiliation(s)
- Raquel Lynn Bowman
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Daoqin Wang
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Dae Seok Eom
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
22
|
Ullo MF, Case LB. How cells sense and integrate information from different sources. WIREs Mech Dis 2023:e1604. [PMID: 36781396 DOI: 10.1002/wsbm.1604] [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: 06/24/2022] [Revised: 01/06/2023] [Accepted: 01/24/2023] [Indexed: 02/15/2023]
Abstract
Cell signaling is a fundamental cellular process that enables cells to sense and respond to information in their surroundings. At the molecular level, signaling is primarily carried out by transmembrane protein receptors that can initiate complex downstream signal transduction cascades to alter cellular behavior. In the human body, different cells can be exposed to a wide variety of environmental conditions, and cells express diverse classes of receptors capable of sensing and integrating different signals. Furthermore, different receptors and signaling pathways can crosstalk with each other to calibrate the cellular response. Crosstalk occurs through multiple mechanisms at different levels of signaling pathways. In this review, we discuss how cells sense and integrate different chemical, mechanical, and spatial signals as well as the mechanisms of crosstalk between pathways. To illustrate these concepts, we use a few well-studied signaling pathways, including receptor tyrosine kinases and integrin receptors. Finally, we discuss the implications of dysregulated cellular sensing on driving diseases such as cancer. This article is categorized under: Cancer > Molecular and Cellular Physiology Metabolic Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Maria F Ullo
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Lindsay B Case
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
23
|
Mahboubi-Rabbani M, Abbasi M, Zarghi A. Natural-Derived COX-2 Inhibitors as Anticancer Drugs: A Review of their Structural Diversity and Mechanism of Action. Anticancer Agents Med Chem 2023; 23:15-36. [PMID: 35638275 DOI: 10.2174/1389450123666220516153915] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/07/2022] [Accepted: 03/01/2022] [Indexed: 02/08/2023]
Abstract
Cyclooxygenase-2 (COX-2) is a key-type enzyme playing a crucial role in cancer development, making it a target of high interest for drug designers. In the last two decades, numerous selective COX-2 inhibitors have been approved for various clinical conditions. However, data from clinical trials propose that the prolonged use of COX-2 inhibitors is associated with life-threatening cardiovascular side effects. The data indicate that a slight structural modification can help develop COX-2 selective inhibitors with comparative efficacy and limited side effects. In this regard, secondary metabolites from natural sources offer great hope for developing novel COX-2 inhibitors with potential anticancer activity. In recent years, various nature-derived organic scaffolds are being explored as leads for developing new COX-2 inhibitors. The current review attempts to highlight the COX-2 inhibition activity of some naturally occurring secondary metabolites, concerning their capacity to inhibit COX-1 and COX-2 enzymes and inhibit cancer development, aiming to establish a structure-activity relationship.
Collapse
Affiliation(s)
- Mohammad Mahboubi-Rabbani
- Department of Medicinal Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Abbasi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Afshin Zarghi
- Department of Medicinal Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
Nieves Torres D, Lee SH. Inter-neuronal signaling mediated by small extracellular vesicles: wireless communication? Front Mol Neurosci 2023; 16:1187300. [PMID: 37181650 PMCID: PMC10172472 DOI: 10.3389/fnmol.2023.1187300] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 04/03/2023] [Indexed: 05/16/2023] Open
Abstract
Conventional inter-neuronal communication conceptualizes the wired method of chemical synapses that physically connect pre-and post-synaptic neurons. In contrast, recent studies indicate that neurons also utilize synapse-independent, hence "wireless" broadcasting-type communications via small extracellular vesicles (EVs). Small EVs including exosomes are secreted vesicles released by cells and contain a variety of signaling molecules including mRNAs, miRNAs, lipids, and proteins. Small EVs are subsequently absorbed by local recipient cells via either membrane fusion or endocytic processes. Therefore, small EVs enable cells to exchange a "packet" of active biomolecules for communication purposes. It is now well established that central neurons also secrete and uptake small EVs, especially exosomes, a type of small EVs that are derived from the intraluminal vesicles of multivesicular bodies. Specific molecules carried by neuronal small EVs are shown to affect a variety of neuronal functions including axon guidance, synapse formation, synapse elimination, neuronal firing, and potentiation. Therefore, this type of volume transmission mediated by small EVs is thought to play important roles not only in activity-dependent changes in neuronal function but also in the maintenance and homeostatic control of local circuitry. In this review, we summarize recent discoveries, catalog neuronal small EV-specific biomolecules, and discuss the potential scope of small EV-mediated inter-neuronal signaling.
Collapse
Affiliation(s)
- Damaris Nieves Torres
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Sang H Lee
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States
- Neuroscience Research Institute, Medical College of Wisconsin, Milwaukee, WI, United States
- *Correspondence: Sang H. Lee,
| |
Collapse
|
25
|
Shiah JV, Johnson DE, Grandis JR. Transcription Factors and Cancer: Approaches to Targeting. Cancer J 2023; 29:38-46. [PMID: 36693157 PMCID: PMC9881838 DOI: 10.1097/ppo.0000000000000639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
ABSTRACT Cancer is defined by the presence of uncontrollable cell growth, whereby improper proliferative signaling has overcome regulation by cellular mechanisms. Transcription factors are uniquely situated at the helm of signaling, merging extracellular stimuli with intracellular responses. Therefore, this class of proteins plays a pivotal role in coordinating the correct gene expression levels for maintaining normal cellular functions. Dysregulation of transcription factor activity unsurprisingly drives tumorigenesis and oncogenic transformation. Although this imparts considerable therapeutic potential to targeting transcription factors, their lack of enzymatic activity renders intervention challenging and has contributed to a sense that transcription factors are "undruggable." Yet, enduring efforts to elucidate strategies for targeting transcription factors as well as a deeper understanding of their interactions with binding partners have led to advancements that are emerging to counter this narrative. Here, we highlight some of these approaches, focusing primarily on therapeutics that have advanced to the clinic.
Collapse
Affiliation(s)
- Jamie V Shiah
- From the Department Otolaryngology-Head and Neck Surgery, University of California at San Francisco, San Francisco, CA
| | | | | |
Collapse
|
26
|
Malingen SA, Rangamani P. Modelling membrane curvature generation using mechanics and machine learning. J R Soc Interface 2022; 19:20220448. [PMID: 36128706 PMCID: PMC9490339 DOI: 10.1098/rsif.2022.0448] [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: 06/15/2022] [Accepted: 08/24/2022] [Indexed: 11/12/2022] Open
Abstract
The deformation of cellular membranes regulates trafficking processes, such as exocytosis and endocytosis. Classically, the Helfrich continuum model is used to characterize the forces and mechanical parameters that cells tune to accomplish membrane shape changes. While this classical model effectively captures curvature generation, one of the core challenges in using it to approximate a biological process is selecting a set of mechanical parameters (including bending modulus and membrane tension) from a large set of reasonable values. We used the Helfrich model to generate a large synthetic dataset from a random sampling of realistic mechanical parameters and used this dataset to train machine-learning models. These models produced promising results, accurately classifying model behaviour and predicting membrane shape from mechanical parameters. We also note emerging methods in machine learning that can leverage the physical insight of the Helfrich model to improve performance and draw greater insight into how cells control membrane shape change.
Collapse
Affiliation(s)
- S. A. Malingen
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA 92093, USA
| | - P. Rangamani
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
27
|
Narasimhan M, Simon R. Spatial range, temporal span, and promiscuity of CLE-RLK signaling. FRONTIERS IN PLANT SCIENCE 2022; 13:906087. [PMID: 36092449 PMCID: PMC9459042 DOI: 10.3389/fpls.2022.906087] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 08/08/2022] [Indexed: 06/15/2023]
Abstract
CLAVATA3/EMBRYO SURROUNDING REGION-RELATED (CLE) signaling through receptor-like kinases (RLKs) regulates developmental transitions and responses to biotic and abiotic inputs by communicating the physiological state of cells and tissues. CLE peptides have varying signaling ranges, which can be defined as the distance between the source, i.e., the cells or tissue that secrete the peptide, and their destination, i.e., cells or tissue where the RLKs that bind the peptide and/or respond are expressed. Case-by-case analysis substantiates that CLE signaling is predominantly autocrine or paracrine, and rarely endocrine. Furthermore, upon CLE reception, the ensuing signaling responses extend from cellular to tissue, organ and whole organism level as the downstream signal gets amplified. CLE-RLK-mediated effects on tissue proliferation and differentiation, or on subsequent primordia and organ development have been widely studied. However, studying how CLE-RLK regulates different stages of proliferation and differentiation at cellular level can offer additional insights into these processes. Notably, CLE-RLK signaling also mediates diverse non-developmental effects, which are less often observed; however, this could be due to biased experimental approaches. In general, CLEs and RLKs, owing to the sequence or structural similarity, are prone to promiscuous interactions at least under experimental conditions in which they are studied. Importantly, there are regulatory mechanisms that suppress CLE-RLK cross-talk in vivo, thereby eliminating the pressure for co-evolving binding specificity. Alternatively, promiscuity in signaling may also offer evolutionary advantages and enable different CLEs to work in combination to activate or switch off different RLK signaling pathways.
Collapse
Affiliation(s)
- Madhumitha Narasimhan
- Institute for Developmental Genetics, Heinrich-Heine University, Düsseldorf, Germany
| | - Rüdiger Simon
- Institute for Developmental Genetics and Cluster of Excellence in Plant Sciences, Heinrich-Heine University, Düsseldorf, Germany
| |
Collapse
|
28
|
Mendieta-Serrano MA, Dhar S, Ng BH, Narayanan R, Lee JJY, Ong HT, Toh PJY, Röllin A, Roy S, Saunders TE. Slow muscles guide fast myocyte fusion to ensure robust myotome formation despite the high spatiotemporal stochasticity of fusion events. Dev Cell 2022; 57:2095-2110.e5. [PMID: 36027918 DOI: 10.1016/j.devcel.2022.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 06/07/2022] [Accepted: 08/05/2022] [Indexed: 11/03/2022]
Abstract
Skeletal myogenesis is dynamic, and it involves cell-shape changes together with cell fusion and rearrangements. However, the final muscle arrangement is highly organized with striated fibers. By combining live imaging with quantitative analyses, we dissected fast-twitch myocyte fusion within the zebrafish myotome in toto. We found a strong mediolateral bias in fusion timing; however, at a cellular scale, there was heterogeneity in cell shape and the relationship between initial position of fast myocytes and resulting fusion partners. We show that the expression of the fusogen myomaker is permissive, but not instructive, in determining the spatiotemporal fusion pattern. Rather, we observed a close coordination between slow muscle rearrangements and fast myocyte fusion. In mutants that lack slow fibers, the spatiotemporal fusion pattern is substantially noisier. We propose a model in which slow muscles guide fast myocytes by funneling them close together, enhancing fusion probability. Thus, despite fusion being highly stochastic, a robust myotome structure emerges at the tissue scale.
Collapse
Affiliation(s)
| | - Sunandan Dhar
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore; Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Boon Heng Ng
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Rachna Narayanan
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore; Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Jorge J Y Lee
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Hui Ting Ong
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Pearlyn Jia Ying Toh
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Adrian Röllin
- Department of Statistics and Data Science, National University of Singapore, Singapore 117546, Singapore
| | - Sudipto Roy
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Proteos, Singapore 138673, Singapore; Department of Biological Sciences, National University of Singapore, Singapore 117558, Singapore; Department of Paediatrics, National University of Singapore, Singapore 119228, Singapore.
| | - Timothy E Saunders
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore; Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK; Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Proteos, Singapore 138673, Singapore; Department of Biological Sciences, National University of Singapore, Singapore 117558, Singapore.
| |
Collapse
|
29
|
Jin T, Wu H, Deng Z, Cai T, Li J, Liu Z, Waterhouse PM, White RG, Liang D. Control of root-to-shoot long-distance flow by a key ROS-regulating factor in Arabidopsis. PLANT, CELL & ENVIRONMENT 2022; 45:2476-2491. [PMID: 35689480 DOI: 10.1111/pce.14375] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 05/09/2022] [Accepted: 05/29/2022] [Indexed: 06/15/2023]
Abstract
Inter-tissue communication is instrumental to coordinating the whole-body level behaviour for complex multicellular organisms. However, little is known about the regulation of inter-tissue information exchange. Here we carried out genetic screens for root-to-shoot mobile silencing in Arabidopsis plants with a compromised small RNA-mediated gene silencing movement rate and identified radical-induced cell death 1 (RCD1) as a critical regulator of root-shoot communication. RCD1 belongs to a family of poly (ADP-ribose) polymerase proteins, which are highly conserved across land plants. We found that RCD1 coordinates symplastic and apoplastic movement by modulating the sterol level of lipid rafts. The higher superoxide production in rcd1-knockout plants resulted in lower plasmodesmata (PD) frequency and altered PD structure in the symplasm of the hypocotyl cortex. Furthermore, the mutants showed increased lateral area of tracheary pits, which reduced axial movement. Our study highlights a novel mechanism through which root-to-shoot long-distance signalling can be modulated both symplastically and apoplastically.
Collapse
Affiliation(s)
- Tianling Jin
- Hubei Collaborative Innovation Center for Grain Industry, School of Agriculture, Yangtze University, Jingzhou, Hubei Province, China
| | - Huiyan Wu
- Hubei Collaborative Innovation Center for Grain Industry, School of Agriculture, Yangtze University, Jingzhou, Hubei Province, China
| | - Zhuying Deng
- Hubei Collaborative Innovation Center for Grain Industry, School of Agriculture, Yangtze University, Jingzhou, Hubei Province, China
| | - Tingting Cai
- Hubei Collaborative Innovation Center for Grain Industry, School of Agriculture, Yangtze University, Jingzhou, Hubei Province, China
| | - Junkai Li
- Hubei Collaborative Innovation Center for Grain Industry, School of Agriculture, Yangtze University, Jingzhou, Hubei Province, China
| | - Zhangyong Liu
- Engineering Research Center of Ecology and Agricultural Use of Wetlandy, Ministry of Education/Hubei Key Laboratory of Waterlogging Disaster and Wetland Agriculture, Yangtze University, Jingzhou, Hubei Province, China
| | - Peter M Waterhouse
- Centre for Tropical Crops and Biocommodities, Queensland University of Technology, Brisbane, QLD, Australia
| | - Rosemary G White
- Department of Plant Sciences, Australian National University, Canberra, ACT, Australia
| | - Dacheng Liang
- Hubei Collaborative Innovation Center for Grain Industry, School of Agriculture, Yangtze University, Jingzhou, Hubei Province, China
- Engineering Research Center of Ecology and Agricultural Use of Wetlandy, Ministry of Education/Hubei Key Laboratory of Waterlogging Disaster and Wetland Agriculture, Yangtze University, Jingzhou, Hubei Province, China
| |
Collapse
|
30
|
Thompson CJ, Kienle DF, Schwartz DK. Enhanced Facilitated Diffusion of Membrane-Associating Proteins under Symmetric Confinement. J Phys Chem Lett 2022; 13:2901-2907. [PMID: 35333540 DOI: 10.1021/acs.jpclett.2c00227] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The facilitated surface diffusion of transiently adsorbing molecules in a planar confined microenvironment (i.e., slit-like confinement) is highly relevant to biological phenomena, such as extracellular signaling, as well as numerous biotechnology systems. Here, we studied the surface diffusion of individual proteins confined between two symmetric lipid bilayer membranes, under a continuum of confinement heights, using single-molecule tracking and convex lens-induced confinement as well as hybrid, kinetic Monte Carlo simulations of a generalized continuous time random walk process. Surface diffusion was observed to vary non-monotonically with confinement height, exhibiting a maximum at a height of ∼750 nm, where diffusion was nearly 40% greater than that for a semi-infinite system. This demonstrated that planar confinement can, in fact, increase surface diffusion, qualitatively validating previous theoretical predictions. Simulations reproduced the experimental results and suggested that confinement enhancement of surface diffusion for symmetric systems is limited to cases where the adsorbate exhibits weak surface sticking.
Collapse
Affiliation(s)
- Connor J Thompson
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, Colorado 80309, United States
| | - Daniel F Kienle
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, Colorado 80309, United States
| | - Daniel K Schwartz
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, Colorado 80309, United States
| |
Collapse
|
31
|
Bandzerewicz A, Gadomska-Gajadhur A. Into the Tissues: Extracellular Matrix and Its Artificial Substitutes: Cell Signalling Mechanisms. Cells 2022; 11:914. [PMID: 35269536 PMCID: PMC8909573 DOI: 10.3390/cells11050914] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/02/2022] [Accepted: 03/04/2022] [Indexed: 02/06/2023] Open
Abstract
The existence of orderly structures, such as tissues and organs is made possible by cell adhesion, i.e., the process by which cells attach to neighbouring cells and a supporting substance in the form of the extracellular matrix. The extracellular matrix is a three-dimensional structure composed of collagens, elastin, and various proteoglycans and glycoproteins. It is a storehouse for multiple signalling factors. Cells are informed of their correct connection to the matrix via receptors. Tissue disruption often prevents the natural reconstitution of the matrix. The use of appropriate implants is then required. This review is a compilation of crucial information on the structural and functional features of the extracellular matrix and the complex mechanisms of cell-cell connectivity. The possibilities of regenerating damaged tissues using an artificial matrix substitute are described, detailing the host response to the implant. An important issue is the surface properties of such an implant and the possibilities of their modification.
Collapse
|
32
|
Derrick CJ, Sánchez-Posada J, Hussein F, Tessadori F, Pollitt EJG, Savage AM, Wilkinson RN, Chico TJ, van Eeden FJ, Bakkers J, Noël ES. Asymmetric Hapln1a drives regionalized cardiac ECM expansion and promotes heart morphogenesis in zebrafish development. Cardiovasc Res 2022; 118:226-240. [PMID: 33616638 PMCID: PMC8752364 DOI: 10.1093/cvr/cvab004] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 01/08/2021] [Indexed: 01/24/2023] Open
Abstract
AIMS Vertebrate heart development requires the complex morphogenesis of a linear tube to form the mature organ, a process essential for correct cardiac form and function, requiring coordination of embryonic laterality, cardiac growth, and regionalized cellular changes. While previous studies have demonstrated broad requirements for extracellular matrix (ECM) components in cardiac morphogenesis, we hypothesized that ECM regionalization may fine tune cardiac shape during heart development. METHODS AND RESULTS Using live in vivo light sheet imaging of zebrafish embryos, we describe a left-sided expansion of the ECM between the myocardium and endocardium prior to the onset of heart looping and chamber ballooning. Analysis using an ECM sensor revealed the cardiac ECM is further regionalized along the atrioventricular axis. Spatial transcriptomic analysis of gene expression in the heart tube identified candidate genes that may drive ECM expansion. This approach identified regionalized expression of hapln1a, encoding an ECM cross-linking protein. Validation of transcriptomic data by in situ hybridization confirmed regionalized hapln1a expression in the heart, with highest levels of expression in the future atrium and on the left side of the tube, overlapping with the observed ECM expansion. Analysis of CRISPR-Cas9-generated hapln1a mutants revealed a reduction in atrial size and reduced chamber ballooning. Loss-of-function analysis demonstrated that ECM expansion is dependent upon Hapln1a, together supporting a role for Hapln1a in regionalized ECM modulation and cardiac morphogenesis. Analysis of hapln1a expression in zebrafish mutants with randomized or absent embryonic left-right asymmetry revealed that laterality cues position hapln1a-expressing cells asymmetrically in the left side of the heart tube. CONCLUSION We identify a regionalized ECM expansion in the heart tube which promotes correct heart development, and propose a novel model whereby embryonic laterality cues orient the axis of ECM asymmetry in the heart, suggesting these two pathways interact to promote robust cardiac morphogenesis.
Collapse
Affiliation(s)
- Christopher J Derrick
- Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Juliana Sánchez-Posada
- Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Farah Hussein
- Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Federico Tessadori
- Hubrecht Institute for Developmental and Stem Cell Biology, Uppsalalaan 8, 3584 CT, Utrecht, Netherlands
| | - Eric J G Pollitt
- Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Aaron M Savage
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Medical School, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Robert N Wilkinson
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Medical School, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Timothy J Chico
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Medical School, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Fredericus J van Eeden
- Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Jeroen Bakkers
- Hubrecht Institute for Developmental and Stem Cell Biology, Uppsalalaan 8, 3584 CT, Utrecht, Netherlands
| | - Emily S Noël
- Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| |
Collapse
|
33
|
Wrenn E, Huang Y, Cheung K. Collective metastasis: coordinating the multicellular voyage. Clin Exp Metastasis 2021; 38:373-399. [PMID: 34254215 PMCID: PMC8346286 DOI: 10.1007/s10585-021-10111-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 06/14/2021] [Indexed: 12/16/2022]
Abstract
The metastatic process is arduous. Cancer cells must escape the confines of the primary tumor, make their way into and travel through the circulation, then survive and proliferate in unfavorable microenvironments. A key question is how cancer cells overcome these multiple barriers to orchestrate distant organ colonization. Accumulating evidence in human patients and animal models supports the hypothesis that clusters of tumor cells can complete the entire metastatic journey in a process referred to as collective metastasis. Here we highlight recent studies unraveling how multicellular coordination, via both physical and biochemical coupling of cells, induces cooperative properties advantageous for the completion of metastasis. We discuss conceptual challenges and unique mechanisms arising from collective dissemination that are distinct from single cell-based metastasis. Finally, we consider how the dissection of molecular transitions regulating collective metastasis could offer potential insight into cancer therapy.
Collapse
Affiliation(s)
- Emma Wrenn
- Translational Research Program, Public Health Sciences and Human Biology Divisions, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA, 98195, USA
| | - Yin Huang
- Translational Research Program, Public Health Sciences and Human Biology Divisions, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Kevin Cheung
- Translational Research Program, Public Health Sciences and Human Biology Divisions, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA.
| |
Collapse
|
34
|
Sherman CD, Lodha S, Sahoo S. EV Cargo Sorting in Therapeutic Development for Cardiovascular Disease. Cells 2021; 10:1500. [PMID: 34203713 PMCID: PMC8232200 DOI: 10.3390/cells10061500] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/24/2020] [Accepted: 10/28/2020] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease remains the leading cause of morbidity and mortality in the world. Thus, therapeutic interventions to circumvent this growing burden are of utmost importance. Extracellular vesicles (EVs) actively secreted by most living cells, play a key role in paracrine and endocrine intercellular communication via exchange of biological molecules. As the content of secreted EVs reflect the physiology and pathology of the cell of their origin, EVs play a significant role in cellular homeostasis, disease pathogenesis and diagnostics. Moreover, EVs are gaining popularity in clinics as therapeutic and drug delivery vehicles, transferring bioactive molecules such as proteins, genes, miRNAs and other therapeutic agents to target cells to treat diseases and deter disease progression. Despite our limited but growing knowledge of EV biology, it is imperative to understand the complex mechanisms of EV cargo sorting in pursuit of designing next generation EV-based therapeutic delivery systems. In this review, we highlight the mechanisms of EV cargo sorting and methods of EV bioengineering and discuss engineered EVs as a potential therapeutic delivery system to treat cardiovascular disease.
Collapse
Affiliation(s)
| | | | - Susmita Sahoo
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, One Gustav L. Levy, P.O. Box 1030, New York, NY 10029, USA; (C.D.S.); (S.L.)
| |
Collapse
|
35
|
Symmetry Breaking as an Interdisciplinary Concept Unifying Cell and Developmental Biology. Cells 2021; 10:cells10010086. [PMID: 33430209 PMCID: PMC7825676 DOI: 10.3390/cells10010086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 12/25/2020] [Indexed: 12/16/2022] Open
|
36
|
Yang S, Pieters PA, Joesaar A, Bögels BWA, Brouwers R, Myrgorodska I, Mann S, de Greef TFA. Light-Activated Signaling in DNA-Encoded Sender-Receiver Architectures. ACS NANO 2020; 14:15992-16002. [PMID: 33078948 PMCID: PMC7690052 DOI: 10.1021/acsnano.0c07537] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 10/14/2020] [Indexed: 05/22/2023]
Abstract
Collective decision making by living cells is facilitated by exchange of diffusible signals where sender cells release a chemical signal that is interpreted by receiver cells. A variety of nonliving artificial cell models have been developed in recent years that mimic various aspects of diffusion-based intercellular communication. However, localized secretion of diffusive signals from individual protocells, which is critical for mimicking biological sender-receiver systems, has remained challenging to control precisely. Here, we engineer light-responsive, DNA-encoded sender-receiver architectures, where protein-polymer microcapsules act as cell mimics and molecular communication occurs through diffusive DNA signals. We prepare spatial distributions of sender and receiver protocells using a microfluidic trapping array and set up a signaling gradient from a single sender cell using light, which activates surrounding receivers through DNA strand displacement. Our systematic analysis reveals how the effective signal range of a single sender is determined by various factors including the density and permeability of receivers, extracellular signal degradation, signal consumption, and catalytic regeneration. In addition, we construct a three-population configuration where two sender cells are embedded in a dense array of receivers that implement Boolean logic and investigate spatial integration of nonidentical input cues. The results offer a means for studying diffusion-based sender-receiver topologies and present a strategy to achieve the congruence of reaction-diffusion and positional information in chemical communication systems that have the potential to reconstitute collective cellular patterns.
Collapse
Affiliation(s)
- Shuo Yang
- Laboratory
of Chemical Biology, Department of Biomedical Engineering, Computational
Biology Group, Department of Biomedical Engineering and Institute
for Complex Molecular Systems, Eindhoven
University of Technology, P.O. Box 513, Eindhoven 5600 MB, The
Netherlands
| | - Pascal A. Pieters
- Laboratory
of Chemical Biology, Department of Biomedical Engineering, Computational
Biology Group, Department of Biomedical Engineering and Institute
for Complex Molecular Systems, Eindhoven
University of Technology, P.O. Box 513, Eindhoven 5600 MB, The
Netherlands
| | - Alex Joesaar
- Laboratory
of Chemical Biology, Department of Biomedical Engineering, Computational
Biology Group, Department of Biomedical Engineering and Institute
for Complex Molecular Systems, Eindhoven
University of Technology, P.O. Box 513, Eindhoven 5600 MB, The
Netherlands
| | - Bas W. A. Bögels
- Laboratory
of Chemical Biology, Department of Biomedical Engineering, Computational
Biology Group, Department of Biomedical Engineering and Institute
for Complex Molecular Systems, Eindhoven
University of Technology, P.O. Box 513, Eindhoven 5600 MB, The
Netherlands
| | - Rens Brouwers
- Laboratory
of Chemical Biology, Department of Biomedical Engineering, Computational
Biology Group, Department of Biomedical Engineering and Institute
for Complex Molecular Systems, Eindhoven
University of Technology, P.O. Box 513, Eindhoven 5600 MB, The
Netherlands
| | - Iuliia Myrgorodska
- Centre
for Protolife Research and Max Planck Bristol Centre for Minimal Biology,
School of Chemistry, University of Bristol, Bristol BS8 1TS, United Kingdom
| | - Stephen Mann
- Centre
for Protolife Research and Max Planck Bristol Centre for Minimal Biology,
School of Chemistry, University of Bristol, Bristol BS8 1TS, United Kingdom
| | - Tom F. A. de Greef
- Laboratory
of Chemical Biology, Department of Biomedical Engineering, Computational
Biology Group, Department of Biomedical Engineering and Institute
for Complex Molecular Systems, Eindhoven
University of Technology, P.O. Box 513, Eindhoven 5600 MB, The
Netherlands
- Institute
for Molecules and Materials, Radboud University, Heyendaalseweg 135, Nijmegen 6525 MB, The Netherlands
| |
Collapse
|
37
|
Roy S, Cha JN, Goodwin AP. Nongenetic Bioconjugation Strategies for Modifying Cell Membranes and Membrane Proteins: A Review. Bioconjug Chem 2020; 31:2465-2475. [PMID: 33146010 DOI: 10.1021/acs.bioconjchem.0c00529] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The cell membrane possesses an extensive library of proteins, carbohydrates, and lipids that control a significant portion of inter- and intracellular functions, including signaling, proliferation, migration, and adhesion, among others. Augmenting the cell surface composition would open possibilities for advances in therapy, tissue engineering, and probing fundamental cell processes. While genetic engineering has proven effective for many in vitro applications, these techniques result in irreversible changes to cells and are difficult to apply in vivo. Another approach is to instead attach exogenous functional groups to the cell membrane without changing the genetic nature of the cell. This review focuses on more recent approaches of nongenetic methods of cell surface modification through metabolic pathways, anchorage by hydrophobic interactions, and chemical conjugation. Benefits and drawbacks of each approach are considered, followed by a discussion of potential applications for nongenetic cell surface modification and an outlook on the future of the field.
Collapse
|
38
|
van Neel TL, Berry SB, Berthier E, Theberge AB. Localized Cell-Surface Sampling of a Secreted Factor Using Cell-Targeting Beads. Anal Chem 2020; 92:13634-13640. [PMID: 32941013 DOI: 10.1021/acs.analchem.0c02578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Intercellular communication through the secretion of soluble factors plays a vital role in a wide range of biological processes (e.g., homeostasis, immune response), yet identification and quantification of many of these factors can be challenging due to their degradation or sequestration in cell culture media prior to analysis. Here, we present a customizable bead-based system capable of simultaneously binding to live cells (through antibody-mediated cell tethering) and capturing cell-secreted molecules. Our functionalized beads capture secreted molecules (e.g., hepatocyte growth factor secreted by fibroblasts) that are diminished when sampled via traditional supernatant analysis techniques (p < 0.05), effectively rescuing a reduced signal in the presence of neutralizing components in the cell culture media. Our system enables capture and analysis of molecules integral to chemical communication that would otherwise be markedly decreased prior to analysis.
Collapse
Affiliation(s)
- Tammi L van Neel
- Department of Chemistry, University of Washington, Box 351700, Seattle, Washington 98195, United States
| | - Samuel B Berry
- Department of Chemistry, University of Washington, Box 351700, Seattle, Washington 98195, United States
| | - Erwin Berthier
- Department of Chemistry, University of Washington, Box 351700, Seattle, Washington 98195, United States
| | - Ashleigh B Theberge
- Department of Chemistry, University of Washington, Box 351700, Seattle, Washington 98195, United States.,Department of Urology, University of Washington, Seattle, Washington 98105, United States
| |
Collapse
|
39
|
Abstract
Understanding the mechanisms of cell-to-cell communication is one of the fundamental questions in biology and medicine. In particular, long-range signalling where cells communicate over several cell diameters is vital during development and homeostasis. The major morphogens, their receptors and intracellular signalling cascades have largely been identified; however, there is a gap in our knowledge of how such signalling factors are propagated over a long distance. In addition to the diffusion-based propagation model, new modalities of disseminating signalling molecules have been identified. It has been shown that cells can communicate with direct contact through long, thin cellular protrusions between signal sending and receiving cells at a distance. Recent studies have revealed a type of cellular protrusion termed 'airinemes' in zebrafish pigment cell types. They share similarities with previously reported cellular protrusions; however, they also exhibit distinct morphology and features. Airinemes are indispensable for pigment pattern development by mediating long-distance Delta-Notch signalling between different pigment cell types. Notably, airineme-mediated signalling is dependent on skin-resident macrophages. Key findings of airineme-mediated intercellular signalling in pattern development, their interplay with macrophages and their implications for the understanding of cellular protrusion-mediated intercellular communication will be discussed.
Collapse
Affiliation(s)
- Dae Seok Eom
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697, USA
| |
Collapse
|
40
|
Marcińska I, Dziurka K, Waligórski P, Janowiak F, Skrzypek E, Warchoł M, Juzoń K, Kapłoniak K, Czyczyło-Mysza IM. Exogenous Polyamines Only Indirectly Induce Stress Tolerance in Wheat Growing in Hydroponic Culture under Polyethylene Glycol-Induced Osmotic Stress. Life (Basel) 2020; 10:life10080151. [PMID: 32823849 PMCID: PMC7459500 DOI: 10.3390/life10080151] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/10/2020] [Accepted: 08/12/2020] [Indexed: 12/21/2022] Open
Abstract
The aim of the present study was to evaluate the effect of osmotic stress caused by polyethylene glycol (PEG) 6000 in hydroponic culture on wheat seedlings of drought-resistant Chinese Spring (CS) and drought-susceptible SQ1 cultivar, and to examine the alleviative role of exogenous polyamines (PAs) applied to the medium. The assessment was based on physiological (chlorophyll a fluorescence kinetics, chlorophyll and water content) as well as biochemical (content of carbohydrates, phenols, proline, salicylic and abscisic acid, activity of low molecular weight antioxidants) parameters, measured after supplementation with PAs (putrescine, spermidine and spermine) on the 3rd, 5th and 7th day of the treatment. The results indicate that PAs ameliorate the effects of stress, indirectly and conditionally inducing stress tolerance of wheat seedlings. In contrast to the susceptible SQ1, the resistant CS cultivar activated its protective mechanisms, adjusting the degree of their activation to the level of the stress, depending on the genetic resources of the plant. Increased accumulation of antioxidants in the resistant CS in response to stress after the application of PAs confirms the hypothesis that PAs are involved in the signaling pathway determining the antioxidative response and the tolerance of wheat plants to drought stress.
Collapse
|
41
|
Matos AM, Gonçalves AI, El Haj AJ, Gomes ME. Magnetic biomaterials and nano-instructive tools as mediators of tendon mechanotransduction. NANOSCALE ADVANCES 2020; 2:140-148. [PMID: 36133967 PMCID: PMC9417540 DOI: 10.1039/c9na00615j] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 11/29/2019] [Indexed: 05/29/2023]
Abstract
Tendon tissues connect muscle to bone allowing the transmission of forces resulting in joint movement. Tendon injuries are prevalent in society and the impact on public health is of utmost concern. Thus, clinical options for tendon treatments are in demand, and tissue engineering aims to provide reliable and successful long-term regenerative solutions. Moreover, the possibility of regulating cell fate by triggering intracellular pathways is a current challenge in regenerative medicine. In the last decade, the use of magnetic nanoparticles as nano-instructive tools has led to great advances in diagnostics and therapeutics. Recent advances using magnetic nanomaterials for regenerative medicine applications include the incorporation of magnetic biomaterials within 3D scaffolds resulting in mechanoresponsive systems with unprecedented properties and the use of nanomagnetic actuators to control cell signaling. Mechano-responsive scaffolds and nanomagnetic systems can act as mechanostimulation platforms to apply forces directly to single cells and multicellular biological tissues. As transmitters of forces in a localized manner, the approaches enable the downstream activation of key tenogenic signaling pathways. In this minireview, we provide a brief outlook on the tenogenic signaling pathways which are most associated with the conversion of mechanical input into biochemical signals, the novel bio-magnetic approaches which can activate these pathways, and the efforts to translate magnetic biomaterials into regenerative platforms for tendon repair.
Collapse
Affiliation(s)
- Ana M Matos
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine Avepark - Zona Industrial da Gandra, 4805-017 Barco Guimarães Portugal
- ICVS/3B's - PT Government Associate Laboratory Braga/Guimarães Portugal
| | - Ana I Gonçalves
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine Avepark - Zona Industrial da Gandra, 4805-017 Barco Guimarães Portugal
- ICVS/3B's - PT Government Associate Laboratory Braga/Guimarães Portugal
| | - Alicia J El Haj
- Healthcare Technologies Institute, Birmingham University B15 2TT Birmingham UK
| | - Manuela E Gomes
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine Avepark - Zona Industrial da Gandra, 4805-017 Barco Guimarães Portugal
- ICVS/3B's - PT Government Associate Laboratory Braga/Guimarães Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at the University of Minho Avepark, 4805-017 Barco Guimarães Portugal
| |
Collapse
|
42
|
Jansa J, Šmilauer P, Borovička J, Hršelová H, Forczek ST, Slámová K, Řezanka T, Rozmoš M, Bukovská P, Gryndler M. Dead Rhizophagus irregularis biomass mysteriously stimulates plant growth. MYCORRHIZA 2020; 30:63-77. [PMID: 32062707 DOI: 10.1007/s00572-020-00937-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 02/05/2020] [Indexed: 05/26/2023]
Abstract
Arbuscular mycorrhizal (AM) fungi establish symbiotic associations with many plant species, transferring significant amounts of soil nutrients such as phosphorus to plants and receiving photosynthetically fixed carbon in return. Functioning of AM symbiosis is thus based on interaction between two living partners. The importance of dead AM fungal biomass (necromass) in ecosystem processes remains unclear. Here, we applied either living biomass or necromass (0.0004 potting substrate weight percent) of monoxenically produced AM fungus (Rhizophagus irregularis) into previously sterilized potting substrate planted with Andropogon gerardii. Plant biomass production significantly improved in both treatments as compared to non-amended controls. Living AM fungus, in contrast to the necromass, specifically improved plant acquisition of nutrients normally supplied to the plants by AM fungal networks, such as phosphorus and zinc. There was, however, no difference between the two amendment treatments with respect to plant uptake of other nutrients such as nitrogen and/or magnesium, indicating that the effect on plants of the AM fungal necromass was not primarily nutritional. Plant growth stimulation by the necromass could thus be either due to AM fungal metabolites directly affecting the plants, indirectly due to changes in soil/root microbiomes or due to physicochemical modifications of the potting substrate. In the necromass, we identified several potentially bioactive molecules. We also provide experimental evidence for significant differences in underground microbiomes depending on the amendment with living or dead AM fungal biomass. This research thus provides the first glimpse into possible mechanisms responsible for observed plant growth stimulation by the AM fungal necromass.
Collapse
Affiliation(s)
- Jan Jansa
- Laboratory of Fungal Biology, Institute of Microbiology, Czech Academy of Sciences, Vídeňská 1083, 142 20, Prague 4, Czech Republic.
| | - Petr Šmilauer
- Department of Ecosystem Biology, Faculty of Science, University of South Bohemia, Branišovská 1760, 370 05, České Budějovice, Czech Republic
| | - Jan Borovička
- Institute of Geology, Czech Academy of Sciences, Rozvojová 269, 165 00, Prague 6, Czech Republic
| | - Hana Hršelová
- Laboratory of Fungal Biology, Institute of Microbiology, Czech Academy of Sciences, Vídeňská 1083, 142 20, Prague 4, Czech Republic
| | - Sándor T Forczek
- Laboratory of Fungal Biology, Institute of Microbiology, Czech Academy of Sciences, Vídeňská 1083, 142 20, Prague 4, Czech Republic
- Isotope Laboratory, Institute of Experimental Botany, Czech Academy of Sciences, Vídeňská 1083, 142 20, Prague 4, Czech Republic
| | - Kristýna Slámová
- Laboratory of Biotransformation, Institute of Microbiology, Czech Academy of Sciences, Vídeňská 1083, 142 20, Prague 4, Czech Republic
| | - Tomáš Řezanka
- Laboratory of Fungal Genetics and Metabolism, Institute of Microbiology, Czech Academy of Sciences, Vídeňská 1083, 142 20, Prague 4, Czech Republic
| | - Martin Rozmoš
- Laboratory of Fungal Biology, Institute of Microbiology, Czech Academy of Sciences, Vídeňská 1083, 142 20, Prague 4, Czech Republic
| | - Petra Bukovská
- Laboratory of Fungal Biology, Institute of Microbiology, Czech Academy of Sciences, Vídeňská 1083, 142 20, Prague 4, Czech Republic
| | - Milan Gryndler
- Laboratory of Fungal Biology, Institute of Microbiology, Czech Academy of Sciences, Vídeňská 1083, 142 20, Prague 4, Czech Republic
- Faculty of Science, Jan Evangelista Purkyně University in Ústí nad Labem, České mládeže 8, 400 96, Ústí nad Labem, Czech Republic
| |
Collapse
|
43
|
Čapek D, Müller P. Positional information and tissue scaling during development and regeneration. Development 2019; 146:146/24/dev177709. [PMID: 31862792 DOI: 10.1242/dev.177709] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In order to contribute to the appropriate tissues during development, cells need to know their position within the embryo. This positional information is conveyed by gradients of signaling molecules, termed morphogens, that are produced in specific regions of the embryo and induce concentration-dependent responses in target tissues. Positional information is remarkably robust, and embryos often develop with the correct proportions even if large parts of the embryo are removed. In this Review, we discuss classical embryological experiments and modern quantitative analyses that have led to mechanistic insights into how morphogen gradients adapt, scale and properly pattern differently sized domains. We analyze these experimental findings in the context of mathematical models and synthesize general principles that apply to multiple systems across species and developmental stages.
Collapse
Affiliation(s)
- Daniel Čapek
- Systems Biology of Development Group, Friedrich Miescher Laboratory of the Max Planck Society, Max-Planck-Ring 9, 72076 Tübingen Germany
| | - Patrick Müller
- Systems Biology of Development Group, Friedrich Miescher Laboratory of the Max Planck Society, Max-Planck-Ring 9, 72076 Tübingen Germany .,Modeling Tumorigenesis Group, Translational Oncology Division, Eberhard Karls University Tübingen, Otfried-Müller-Strasse 10, 72076 Tübingen Germany
| |
Collapse
|
44
|
Rogers KW, Müller P. Optogenetic approaches to investigate spatiotemporal signaling during development. Curr Top Dev Biol 2019; 137:37-77. [PMID: 32143750 DOI: 10.1016/bs.ctdb.2019.11.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Embryogenesis is coordinated by signaling pathways that pattern the developing organism. Many aspects of this process are not fully understood, including how signaling molecules spread through embryonic tissues, how signaling amplitude and dynamics are decoded, and how multiple signaling pathways cooperate to pattern the body plan. Optogenetic approaches can be used to address these questions by providing precise experimental control over a variety of biological processes. Here, we review how these strategies have provided new insights into developmental signaling and discuss how they could contribute to future investigations.
Collapse
Affiliation(s)
- Katherine W Rogers
- Systems Biology of Development Group, Friedrich Miescher Laboratory of the Max Planck Society, Tübingen, Germany
| | - Patrick Müller
- Systems Biology of Development Group, Friedrich Miescher Laboratory of the Max Planck Society, Tübingen, Germany; Modeling Tumorigenesis Group, Translational Oncology Division, Eberhard Karls University Tübingen, Tübingen, Germany.
| |
Collapse
|
45
|
Ye D, Wang X, Wei C, He M, Wang H, Wang Y, Zhu Z, Sun Y. Marcksb plays a key role in the secretory pathway of zebrafish Bmp2b. PLoS Genet 2019; 15:e1008306. [PMID: 31545789 PMCID: PMC6776368 DOI: 10.1371/journal.pgen.1008306] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 10/03/2019] [Accepted: 07/11/2019] [Indexed: 12/24/2022] Open
Abstract
During vertebrate early embryogenesis, the ventral development is directed by the ventral-to-dorsal activity gradient of the bone morphogenetic protein (BMP) signaling. As secreted ligands, the extracellular traffic of BMP has been extensively studied. However, it remains poorly understood that how BMP ligands are secreted from BMP-producing cells. In this work, we show the dominant role of Marcksb controlling the secretory process of Bmp2b via interaction with Hsp70 in vivo. We firstly carefully characterized the role of Marcksb in promoting BMP signaling during dorsoventral axis formation through knockdown approach. We then showed that Marcksb cell autonomously regulates the trafficking of Bmp2b from producing cell to the extracellular space and both the total and the extracellular Bmp2b was decreased in Marcksb-deficient embryos. However, neither the zygotic mutant of marcksb (Zmarcksb) nor the maternal zygotic mutant of marcksb (MZmarcksb) showed any defects of dorsalization. In contrast, the MZmarcksb embryos even showed increased BMP signaling activity as measured by expression of BMP targets, phosphorylated Smad1/5/9 levels and imaging of Bmp2b, suggesting that a phenomenon of “genetic over-compensation” arose. Finally, we revealed that the over-compensation effects of BMP signaling in MZmarcksb was achieved through a sequential up-regulation of MARCKS-family members Marcksa, Marcksl1a and Marcksl1b, and MARCKS-interacting protein Hsp70.3. We concluded that the Marcksb modulates BMP signaling through regulating the secretory pathway of Bmp2b. Bone morphogenetic proteins (BMPs) are extracellular proteins which belong to the transforming growth factor-β (TGF-β) superfamily. BMP signaling is essential for embryonic development, organogenesis, and tissue regeneration and homeostasis, and tightly linked to various diseases and tumorigenesis. However, as secreted proteins, how BMPs are transported and secreted from BMP-producing cells remains poorly understood. In this study, we showed that Marcksb interacts with a molecular chaperon–Hsp70.3 to mediate the secretory pathway of BMP ligands during early development of zebrafish. Moreover, we discovered a novel phenomenon of “genetic over-compensation” in the genetic knock-out mutants of marcksb. To our knowledge, this is the first report that reveals the molecules and their related trafficking system mediating the secretion of BMPs. Considering the wide distribution of BMP and MARCKS within the human body, our work may shed light on the studies of BMPs secretion in organogenesis and adult tissue homeostasis. The finding of MARCKS in controlling BMP secretion may provide potential therapeutic targets for modulating the activity of BMP signaling and thus will be of interest to clinical research.
Collapse
Affiliation(s)
- Ding Ye
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Wuhan, China
| | - Xiaosi Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Wuhan, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Changyong Wei
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Wuhan, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Mudan He
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Wuhan, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Houpeng Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Wuhan, China
| | - Yanwu Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Wuhan, China
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Zuoyan Zhu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Wuhan, China
| | - Yonghua Sun
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Wuhan, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
- * E-mail:
| |
Collapse
|
46
|
Denatonium Benzoate-Induces Oxidative Stress in the Heart and Kidney of Chinese Fast Yellow Chickens by Regulating Apoptosis, Autophagy, Antioxidative Activities and Bitter Taste Receptor Gene Expressions. Animals (Basel) 2019; 9:ani9090701. [PMID: 31546822 PMCID: PMC6770773 DOI: 10.3390/ani9090701] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 09/09/2019] [Accepted: 09/09/2019] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Denatonium benzoate is a strong bitter taste receptor agonist, extensively used for its activation of different cell pathways. Taste signals have been associated to food recognition and avoidance, and bitter taste provokes an aversive reaction and is assumed to protect chickens from consuming poisons and harmful toxic substances. The results of the study revealed that dietary supplementation with medium and high doses of denatonium benzoate damaged the epithelial cells of the heart and kidneys by inducing apoptosis and autophagy and reduced the growth of chickens, respectively. However, mRNA expressions of bitter taste receptors, downstream signaling effector genes, apoptosis-, autophagy- and antioxidant-related genes were higher on day 7, while these expressions were subsequently decreased on day-28 in the heart and kidney of Chinese Fast Yellow chickens in a dose-response manner. Abstract The sense of taste which tells us which prospective foods are nutritious, poisonous and harmful is essential for the life of the organisms. Denatonium benzoate (DB) is a bitter taste agonist known for its activation of bitter taste receptors in different cells. The aim of the current study was to investigate the mRNA expressions of bitter taste, downstream signaling effectors, apoptosis-, autophagy- and antioxidant-related genes and effector signaling pathways in the heart/kidney of chickens after DB dietary exposure. We randomly assigned 240, 1-day-old Chinese Fast Yellow chicks into four groups with five replicates of 12 chicks and studied them for 28 consecutive days. The dietary treatments consisted of basal diet and feed containing DB (5, 20 and 100 mg/kg). The results revealed that dietary DB impaired (p < 0.05) the growth performance of the chickens. Haemotoxylin and eosin staining and TUNEL assays confirmed that medium and high doses of DB damaged the epithelial cells of heart/kidney and induced apoptosis and autophagy. Remarkably, the results of RT-PCR and qRT-PCR indicated that different doses of DB gradually increased (p < 0.05) mRNA expressions of bitter taste, signaling effectors, apoptosis-, autophagy- and antioxidant- related genes on day 7 in a dose-response manner, while, these expressions were decreased (p < 0.05) subsequently by day-28 but exceptional higher (P < 0.05) expressions were observed in the high-dose DB groups of chickens. In conclusion, DB exerts adverse effects on the heart/kidney of chickens in a dose-response manner via damaging the epithelium of the heart/kidney by inducing apoptosis, autophagy associated with bitter taste and effector gene expressions. Correlation analyses for apoptosis/autophagy showed agonistic relationships. Our data provide a novel perspective for understanding the interaction of bitter taste, apoptosis, autophagy and antioxidative genes with bitter taste strong activators in the heart/kidney of chicken. These insights might help the feed industries and pave the way toward innovative directions in chicken husbandry.
Collapse
|
47
|
Miller WB, Torday JS, Baluška F. The N-space Episenome unifies cellular information space-time within cognition-based evolution. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2019; 150:112-139. [PMID: 31415772 DOI: 10.1016/j.pbiomolbio.2019.08.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 07/26/2019] [Accepted: 08/09/2019] [Indexed: 02/08/2023]
Abstract
Self-referential cellular homeostasis is maintained by the measured assessment of both internal status and external conditions based within an integrated cellular information field. This cellular field attachment to biologic information space-time coordinates environmental inputs by connecting the cellular senome, as the sum of the sensory experiences of the cell, with its genome and epigenome. In multicellular organisms, individual cellular information fields aggregate into a collective information architectural matrix, termed a N-space Episenome, that enables mutualized organism-wide information management. It is hypothesized that biological organization represents a dual heritable system constituted by both its biological materiality and a conjoining N-space Episenome. It is further proposed that morphogenesis derives from reciprocations between these inter-related facets to yield coordinated multicellular growth and development. The N-space Episenome is conceived as a whole cell informational projection that is heritable, transferable via cell division and essential for the synchronous integration of the diverse self-referential cells that constitute holobionts.
Collapse
Affiliation(s)
| | - John S Torday
- Department of Pediatrics, Harbor-UCLA Medical Center, USA.
| | | |
Collapse
|
48
|
Pearsall RS, Davies MV, Cannell M, Li J, Widrick J, Mulivor AW, Wallner S, Troy ME, Spaits M, Liharska K, Sako D, Castonguay R, Keates S, Grinberg AV, Suragani RNVS, Kumar R. Follistatin-based ligand trap ACE-083 induces localized hypertrophy of skeletal muscle with functional improvement in models of neuromuscular disease. Sci Rep 2019; 9:11392. [PMID: 31388039 PMCID: PMC6684588 DOI: 10.1038/s41598-019-47818-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 07/17/2019] [Indexed: 12/13/2022] Open
Abstract
Skeletal muscle is under inhibitory homeostatic regulation by multiple ligands of the transforming growth factor-β (TGFβ) superfamily. Follistatin is a secreted protein that promotes muscle growth and function by sequestering these ligands extracellularly. In the present study, we evaluated the potential of ACE-083 – a locally acting, follistatin-based fusion protein – as a novel therapeutic agent for focal or asymmetric myopathies. Characterization of ACE-083 in vitro revealed its high affinity for heparin and extracellular matrix while surface plasmon resonance and cell-based assays confirmed that ACE-083 binds and potently neutralizes myostatin, activin A, activin B and growth differentiation factor 11 (GDF11). Intramuscular administration of ACE-083 caused localized, dose-dependent hypertrophy of the injected muscle in wild-type mice and mouse models of Charcot-Marie-Tooth disease (CMT) and Duchenne muscular dystrophy, with no evidence of systemic muscle effects or endocrine perturbation. Importantly, ACE-083 also increased the force of isometric contraction in situ by the injected tibialis anterior muscle in wild-type mice and disease models and increased ankle dorsiflexion torque in CMT mice. Our results demonstrate the potential of ACE-083 as a therapeutic agent for patients with CMT, muscular dystrophy and other disorders with focal or asymmetric muscle atrophy or weakness.
Collapse
Affiliation(s)
| | | | - M Cannell
- Acceleron Pharma, Cambridge, MA, USA
| | - J Li
- Acceleron Pharma, Cambridge, MA, USA
| | - J Widrick
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - A W Mulivor
- Acceleron Pharma, Cambridge, MA, USA.,The Hospital for Sick Children, Toronto, Ontario, Canada
| | - S Wallner
- Acceleron Pharma, Cambridge, MA, USA.,NovaRock Biotherapeutics, Princeton, NJ, USA
| | - M E Troy
- Acceleron Pharma, Cambridge, MA, USA
| | - M Spaits
- Acceleron Pharma, Cambridge, MA, USA
| | - K Liharska
- Acceleron Pharma, Cambridge, MA, USA.,Dragonfly Therapeutics, Waltham, MA, USA
| | - D Sako
- Acceleron Pharma, Cambridge, MA, USA
| | | | - S Keates
- Acceleron Pharma, Cambridge, MA, USA
| | - A V Grinberg
- Acceleron Pharma, Cambridge, MA, USA.,Dragonfly Therapeutics, Waltham, MA, USA
| | | | - R Kumar
- Acceleron Pharma, Cambridge, MA, USA
| |
Collapse
|
49
|
Nam S, Khawar IA, Park JK, Chang S, Kuh HJ. Cellular context-dependent interaction between cancer and stellate cells in hetero-type multicellular spheroids of pancreatic tumor. Biochem Biophys Res Commun 2019; 515:183-189. [DOI: 10.1016/j.bbrc.2019.05.101] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 05/13/2019] [Indexed: 11/24/2022]
|
50
|
Castonguay R, Lachey J, Wallner S, Strand J, Liharska K, Watanabe AE, Cannell M, Davies MV, Sako D, Troy ME, Krishnan L, Mulivor AW, Li H, Keates S, Alexander MJ, Pearsall RS, Kumar R. Follistatin-288-Fc Fusion Protein Promotes Localized Growth of Skeletal Muscle. J Pharmacol Exp Ther 2019; 368:435-445. [PMID: 30563942 DOI: 10.1124/jpet.118.252304] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 12/06/2018] [Indexed: 12/19/2022] Open
Abstract
Follistatin is an endogenous glycoprotein that promotes growth and repair of skeletal muscle by sequestering inhibitory ligands of the transforming growth factor-β superfamily and may therefore have therapeutic potential for neuromuscular diseases. Here, we sought to determine the suitability of a newly engineered follistatin fusion protein (FST288-Fc) to promote localized, rather than systemic, growth of skeletal muscle by capitalizing on the intrinsic heparin-binding ability of the follistatin-288 isoform. As determined by surface plasmon resonance and cell-based assays, FST288-Fc binds to activin A, activin B, myostatin (growth differentiation factor GDF8), and GDF11 with high affinity and neutralizes their activity in vitro. Intramuscular administration of FST288-Fc in mice induced robust, dose-dependent growth of the targeted muscle but not of surrounding or contralateral muscles, in contrast to the systemic effects of a locally administered fusion protein incorporating activin receptor type IIB (ActRIIB-Fc). Furthermore, systemic administration of FST288-Fc in mice did not alter muscle mass or body composition as determined by NMR, which again contrasts with the pronounced systemic activity of ActRIIB-Fc when administered by the same route. Subsequent analysis revealed that FST288-Fc in the circulation undergoes rapid proteolysis, thereby restricting its activity to individual muscles targeted by intramuscular administration. These results indicate that FST288-Fc can produce localized growth of skeletal muscle in a targeted manner with reduced potential for undesirable systemic effects. Thus, FST288-Fc and similar agents may be beneficial in the treatment of disorders with muscle atrophy that is focal, asymmetric, or otherwise heterogeneous.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Huiming Li
- Acceleron Pharma, Cambridge, Massachusetts
| | | | | | | | - Ravi Kumar
- Acceleron Pharma, Cambridge, Massachusetts
| |
Collapse
|