1
|
Schnider ST, Vigano MA, Affolter M, Aguilar G. Functionalized Protein Binders in Developmental Biology. Annu Rev Cell Dev Biol 2024; 40:119-142. [PMID: 39038471 DOI: 10.1146/annurev-cellbio-112122-025214] [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] [Indexed: 07/24/2024]
Abstract
Developmental biology has greatly profited from genetic and reverse genetic approaches to indirectly studying protein function. More recently, nanobodies and other protein binders derived from different synthetic scaffolds have been used to directly dissect protein function. Protein binders have been fused to functional domains, such as to lead to protein degradation, relocalization, visualization, or posttranslational modification of the target protein upon binding. The use of such functionalized protein binders has allowed the study of the proteome during development in an unprecedented manner. In the coming years, the advent of the computational design of protein binders, together with further advances in scaffold engineering and synthetic biology, will fuel the development of novel protein binder-based technologies. Studying the proteome with increased precision will contribute to a better understanding of the immense molecular complexities hidden in each step along the way to generate form and function during development.
Collapse
Affiliation(s)
| | | | | | - Gustavo Aguilar
- Current affiliation: Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA;
- Biozentrum, Universität Basel, Basel, Switzerland;
| |
Collapse
|
2
|
Mii Y. Understanding and manipulating extracellular behaviors of Wnt ligands. In Vitro Cell Dev Biol Anim 2024; 60:441-448. [PMID: 38379096 DOI: 10.1007/s11626-024-00856-9] [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: 11/13/2023] [Accepted: 01/17/2024] [Indexed: 02/22/2024]
Abstract
Wnt, a family of secreted signaling proteins, serves diverse functions in embryogenesis, organogenesis, cancer, and stem cell functions. In the context of development, Wnt has been considered a representative morphogen, forming concentration gradients to give positional information to cells or tissues. However, although gradients are often illustrated in schemata, the reality of concentration gradients, or in other words, actual spatial distribution of Wnt ligands, and their behaviors in the extracellular space still remain poorly known. To understand extracellular behavior of Wnt ligands, quantitative analyses such as fluorescence correlation spectroscopy (FCS) and fluorescence recovery after photobleaching (FRAP) are highly informative because Wnt dispersal involves physical and biochemical processes, such as diffusion and binding to or dissociation from cell surface molecules, including heparan sulfate proteoglycans (HSPGs). Here, I briefly discuss representative methods to quantify morphogen dynamics. In addition, I discuss molecular manipulations of morphogens, mainly focusing on use of protein binders, and synthetic biology of morphogens as indicators of current and future directions in this field.
Collapse
Affiliation(s)
- Yusuke Mii
- National Institute for Basic Biology (NIBB) and Exploratory Research Center On Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, Aichi, 444-8787, Japan.
- The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Aichi, 444-8787, Japan.
| |
Collapse
|
3
|
Ridwan SM, Twillie A, Poursaeid S, Beard EK, Bener MB, Antel M, Cowan AE, Matsuda S, Inaba M. Diffusible fraction of niche BMP ligand safeguards stem-cell differentiation. Nat Commun 2024; 15:1166. [PMID: 38326318 PMCID: PMC10850516 DOI: 10.1038/s41467-024-45408-7] [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: 03/01/2023] [Accepted: 01/22/2024] [Indexed: 02/09/2024] Open
Abstract
Drosophila male germline stem cells (GSCs) reside at the tip of the testis and surround a cluster of niche cells. Decapentaplegic (Dpp) is one of the well-established ligands and has a major role in maintaining stem cells located in close proximity. However, the existence and the role of the diffusible fraction of Dpp outside of the niche have been unclear. Here, using genetically-encoded nanobodies called Morphotraps, we physically block Dpp diffusion without interfering with niche-stem cell signaling and find that a diffusible fraction of Dpp is required to ensure differentiation of GSC daughter cells, opposite of its role in maintenance of GSC in the niche. Our work provides an example in which a soluble niche ligand induces opposed cellular responses in stem cells versus in differentiating descendants to ensure spatial control of the niche. This may be a common mechanism to regulate tissue homeostasis.
Collapse
Affiliation(s)
- Sharif M Ridwan
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, USA
| | - Autumn Twillie
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, USA
| | - Samaneh Poursaeid
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, USA
| | - Emma Kristine Beard
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, USA
| | - Muhammed Burak Bener
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, USA
| | - Matthew Antel
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, USA
| | - Ann E Cowan
- Richard D. Berlin Center for Cell Analysis and Modeling, University of Connecticut Health Center, Farmington, CT, USA
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT, USA
| | - Shinya Matsuda
- Biozentrum, University of Basel, Basel, Switzerland.
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan.
| | - Mayu Inaba
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, USA.
| |
Collapse
|
4
|
Matamoro-Vidal A, Cumming T, Davidović A, Levillayer F, Levayer R. Patterned apoptosis has an instructive role for local growth and tissue shape regulation in a fast-growing epithelium. Curr Biol 2024; 34:376-388.e7. [PMID: 38215743 PMCID: PMC10808510 DOI: 10.1016/j.cub.2023.12.031] [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: 04/21/2022] [Revised: 07/13/2023] [Accepted: 12/11/2023] [Indexed: 01/14/2024]
Abstract
What regulates organ size and shape remains one fundamental mystery of modern biology. Research in this area has primarily focused on deciphering the regulation in time and space of growth and cell division, while the contribution of cell death has been overall neglected. This includes studies of the Drosophila wing, one of the best-characterized systems for the study of growth and patterning, undergoing massive growth during larval stage and important morphogenetic remodeling during pupal stage. So far, it has been assumed that cell death was relatively neglectable in this tissue both during larval stage and pupal stage, and as a result, the pattern of growth was usually attributed to the distribution of cell division. Here, using systematic mapping and registration combined with quantitative assessment of clone size and disappearance as well as live imaging, we outline a persistent pattern of cell death and clone elimination emerging in the larval wing disc and persisting during pupal wing morphogenesis. Local variation of cell death is associated with local variation of clone size, pointing to an impact of cell death on local growth that is not fully compensated by proliferation. Using morphometric analyses of adult wing shape and genetic perturbations, we provide evidence that patterned death locally and globally affects adult wing shape and size. This study describes a roadmap for precise assessment of the contribution of cell death to tissue shape and outlines an important instructive role of cell death in modulating quantitatively local growth and morphogenesis of a fast-growing tissue.
Collapse
Affiliation(s)
- Alexis Matamoro-Vidal
- Department of Developmental and Stem Cell Biology, Institut Pasteur, CNRS UMR 3738, Université Paris Cité, Cell Death and Epithelial Homeostasis Unit, 75015 Paris, France
| | - Tom Cumming
- Department of Developmental and Stem Cell Biology, Institut Pasteur, CNRS UMR 3738, Université Paris Cité, Cell Death and Epithelial Homeostasis Unit, 75015 Paris, France; PPU program Institut Pasteur, Sorbonne Université, Collège Doctoral, 75005 Paris, France
| | - Anđela Davidović
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, 75015 Paris, France
| | - Florence Levillayer
- Department of Developmental and Stem Cell Biology, Institut Pasteur, CNRS UMR 3738, Université Paris Cité, Cell Death and Epithelial Homeostasis Unit, 75015 Paris, France
| | - Romain Levayer
- Department of Developmental and Stem Cell Biology, Institut Pasteur, CNRS UMR 3738, Université Paris Cité, Cell Death and Epithelial Homeostasis Unit, 75015 Paris, France.
| |
Collapse
|
5
|
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: 3] [Impact Index Per Article: 3.0] [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
|
6
|
Matsuda S, Affolter M. Is Drosophila Dpp/BMP morphogen spreading required for wing patterning and growth? Bioessays 2023; 45:e2200218. [PMID: 37452394 DOI: 10.1002/bies.202200218] [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: 11/09/2022] [Revised: 06/02/2023] [Accepted: 06/15/2023] [Indexed: 07/18/2023]
Abstract
Secreted signaling molecules act as morphogens to control patterning and growth in many developing tissues. Since locally produced morphogens spread to form a concentration gradient in the surrounding tissue, spreading is generally thought to be the key step in the non-autonomous actions. Here, we review recent advances in tool development to investigate morphogen function using the role of decapentaplegic (Dpp)/bone morphogenetic protein (BMP)-type ligand in the Drosophila wing disc as an example. By applying protein binder tools to distinguish between the roles of Dpp spreading and local Dpp signaling, we found that Dpp signaling in the source cells is important for wing patterning and growth but Dpp spreading from this source cells is not as strictly required as previously thought. Given recent studies showing unexpected requirements of long-range action of different morphogens, manipulating endogenous morphogen gradients by synthetic protein binder tools could shed more light on how morphogens act in developing tissues.
Collapse
Affiliation(s)
- Shinya Matsuda
- Growth & Development, Biozentrum, University of Basel, Basel, Switzerland
| | - Markus Affolter
- Growth & Development, Biozentrum, University of Basel, Basel, Switzerland
| |
Collapse
|
7
|
Ramezani A, Britton S, Zandi R, Alber M, Nematbakhsh A, Chen W. A multiscale chemical-mechanical model predicts impact of morphogen spreading on tissue growth. NPJ Syst Biol Appl 2023; 9:16. [PMID: 37210381 DOI: 10.1038/s41540-023-00278-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 05/03/2023] [Indexed: 05/22/2023] Open
Abstract
The exact mechanism controlling cell growth remains a grand challenge in developmental biology and regenerative medicine. The Drosophila wing disc tissue serves as an ideal biological model to study mechanisms involved in growth regulation. Most existing computational models for studying tissue growth focus specifically on either chemical signals or mechanical forces. Here we developed a multiscale chemical-mechanical model to investigate the growth regulation mechanism based on the dynamics of a morphogen gradient. By comparing the spatial distribution of dividing cells and the overall tissue shape obtained in model simulations with experimental data of the wing disc, it is shown that the size of the domain of the Dpp morphogen is critical in determining tissue size and shape. A larger tissue size with a faster growth rate and more symmetric shape can be achieved if the Dpp gradient spreads in a larger domain. Together with Dpp absorbance at the peripheral zone, the feedback regulation that downregulates Dpp receptors on the cell membrane allows for further spreading of the morphogen away from its source region, resulting in prolonged tissue growth at a more spatially homogeneous growth rate.
Collapse
Affiliation(s)
- Alireza Ramezani
- Department of Physics and Astronomy, University of California, Riverside, CA, 92521, USA
- Interdisciplinary Center for Quantitative Modeling in Biology, University of California, Riverside, CA, 92521, USA
| | - Samuel Britton
- Department of Mathematics, University of California, Riverside, CA, 92521, USA
| | - Roya Zandi
- Department of Physics and Astronomy, University of California, Riverside, CA, 92521, USA
- Interdisciplinary Center for Quantitative Modeling in Biology, University of California, Riverside, CA, 92521, USA
| | - Mark Alber
- Interdisciplinary Center for Quantitative Modeling in Biology, University of California, Riverside, CA, 92521, USA
- Department of Mathematics, University of California, Riverside, CA, 92521, USA
| | - Ali Nematbakhsh
- Department of Mathematics, University of California, Riverside, CA, 92521, USA.
| | - Weitao Chen
- Interdisciplinary Center for Quantitative Modeling in Biology, University of California, Riverside, CA, 92521, USA.
- Department of Mathematics, University of California, Riverside, CA, 92521, USA.
| |
Collapse
|
8
|
Bauer M, Aguilar G, Wharton KA, Matsuda S, Affolter M. Heterodimerization-dependent secretion of bone morphogenetic proteins in Drosophila. Dev Cell 2023; 58:645-659.e4. [PMID: 37054707 PMCID: PMC10303954 DOI: 10.1016/j.devcel.2023.03.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 01/13/2023] [Accepted: 03/09/2023] [Indexed: 04/15/2023]
Abstract
Combinatorial signaling is key to instruct context-dependent cell behaviors. During embryonic development, adult homeostasis, and disease, bone morphogenetic proteins (BMPs) act as dimers to instruct specific cellular responses. BMP ligands can form both homodimers or heterodimers; however, obtaining direct evidence of the endogenous localization and function of each form has proven challenging. Here, we make use of precise genome editing and direct protein manipulation via protein binders to dissect the existence and functional relevance of BMP homodimers and heterodimers in the Drosophila wing imaginal disc. This approach identified in situ the existence of Dpp (BMP2/4)/Gbb (BMP5/6/7/8) heterodimers. We found that Gbb is secreted in a Dpp-dependent manner in the wing imaginal disc. Dpp and Gbb form a gradient of heterodimers, whereas neither Dpp nor Gbb homodimers are evident under endogenous physiological conditions. We find that the formation of heterodimers is critical for obtaining optimal signaling and long-range BMP distribution.
Collapse
Affiliation(s)
- Milena Bauer
- Biozentrum, University of Basel, Spitalstrasse 41, 4056 Basel, Switzerland
| | - Gustavo Aguilar
- Biozentrum, University of Basel, Spitalstrasse 41, 4056 Basel, Switzerland
| | | | - Shinya Matsuda
- Biozentrum, University of Basel, Spitalstrasse 41, 4056 Basel, Switzerland.
| | - Markus Affolter
- Biozentrum, University of Basel, Spitalstrasse 41, 4056 Basel, Switzerland.
| |
Collapse
|
9
|
Qian W, Guo M, Peng J, Zhao T, Li Z, Yang Y, Li H, Zhang X, King-Jones K, Cheng D. Decapentaplegic retards lipolysis during metamorphosis in Bombyx mori and Drosophila melanogaster. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2023; 155:103928. [PMID: 36870515 DOI: 10.1016/j.ibmb.2023.103928] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 01/29/2023] [Accepted: 03/01/2023] [Indexed: 05/10/2023]
Abstract
Insect morphogen decapentaplegic (Dpp) functions as one of the key extracellular ligands of the Bone Morphogenetic Protein (BMP) signaling pathway. Previous studies in insects mainly focused on the roles of Dpp during embryonic development and the formation of adult wings. In this study, we demonstrate a new role for Dpp in retarding lipolysis during metamorphosis in both Bombyx mori and Drosophila melanogaster. CRISPR/Cas9-mediated mutation of Bombyx dpp causes pupal lethality, induces an excessive and premature breakdown of lipids in the fat body, and upregulates the expressions of several lipolytic enzyme genes, including brummer (bmm), lipase 3 (lip3), and hormone-sensitive lipase (hsl), and lipid storage droplet 1 (lsd1), a lipid droplets (LD)-associated protein gene. Further investigation in Drosophila reveals that salivary gland-specific knockdown of the dpp gene and fat body-specific knockdown of Mad involved in Dpp signaling phenocopy the effects of Bombyx dpp mutation on pupal development and lipolysis. Taken together, our data indicate that the Dpp-mediated BMP signaling in the fat body maintains lipid homeostasis by retarding lipolysis, which is necessary for pupa-adult transition during insect metamorphosis.
Collapse
Affiliation(s)
- Wenliang Qian
- State Key Laboratory of Silkworm Genome Biology, Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City & Southwest University, Biological Science Research Center, Southwest University, Chongqing, 400715, China; Chongqing Key Laboratory of Sericultural Science, Biological Science Research Center, Southwest University, Chongqing, 400715, China
| | - Mengge Guo
- State Key Laboratory of Silkworm Genome Biology, Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City & Southwest University, Biological Science Research Center, Southwest University, Chongqing, 400715, China; Chongqing Key Laboratory of Sericultural Science, Biological Science Research Center, Southwest University, Chongqing, 400715, China
| | - Jian Peng
- State Key Laboratory of Silkworm Genome Biology, Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City & Southwest University, Biological Science Research Center, Southwest University, Chongqing, 400715, China; Chongqing Key Laboratory of Sericultural Science, Biological Science Research Center, Southwest University, Chongqing, 400715, China
| | - Tujing Zhao
- State Key Laboratory of Silkworm Genome Biology, Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City & Southwest University, Biological Science Research Center, Southwest University, Chongqing, 400715, China; Chongqing Key Laboratory of Sericultural Science, Biological Science Research Center, Southwest University, Chongqing, 400715, China
| | - Zheng Li
- State Key Laboratory of Silkworm Genome Biology, Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City & Southwest University, Biological Science Research Center, Southwest University, Chongqing, 400715, China; Chongqing Key Laboratory of Sericultural Science, Biological Science Research Center, Southwest University, Chongqing, 400715, China
| | - Yan Yang
- State Key Laboratory of Silkworm Genome Biology, Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City & Southwest University, Biological Science Research Center, Southwest University, Chongqing, 400715, China; Chongqing Key Laboratory of Sericultural Science, Biological Science Research Center, Southwest University, Chongqing, 400715, China
| | - Hao Li
- State Key Laboratory of Silkworm Genome Biology, Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City & Southwest University, Biological Science Research Center, Southwest University, Chongqing, 400715, China; Chongqing Key Laboratory of Sericultural Science, Biological Science Research Center, Southwest University, Chongqing, 400715, China
| | - Xing Zhang
- State Key Laboratory of Silkworm Genome Biology, Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City & Southwest University, Biological Science Research Center, Southwest University, Chongqing, 400715, China; Chongqing Key Laboratory of Sericultural Science, Biological Science Research Center, Southwest University, Chongqing, 400715, China
| | - Kirst King-Jones
- Department of Biological Sciences, University of Alberta, G-504 Biological Sciences Bldg., Edmonton, Alberta, T6G 2E9, Canada.
| | - Daojun Cheng
- State Key Laboratory of Silkworm Genome Biology, Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City & Southwest University, Biological Science Research Center, Southwest University, Chongqing, 400715, China; Chongqing Key Laboratory of Sericultural Science, Biological Science Research Center, Southwest University, Chongqing, 400715, China.
| |
Collapse
|
10
|
Merino MM, Garcia-Sanz JA. Stemming Tumoral Growth: A Matter of Grotesque Organogenesis. Cells 2023; 12:872. [PMID: 36980213 PMCID: PMC10047265 DOI: 10.3390/cells12060872] [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: 02/17/2023] [Revised: 03/07/2023] [Accepted: 03/09/2023] [Indexed: 03/16/2023] Open
Abstract
The earliest metazoans probably evolved from single-celled organisms which found the colonial system to be a beneficial organization. Over the course of their evolution, these primary colonial organisms increased in size, and division of labour among the cells became a remarkable feature, leading to a higher level of organization: the biological organs. Primitive metazoans were the first organisms in evolution to show organ-type structures, which set the grounds for complex organs to evolve. Throughout evolution, and concomitant with organogenesis, is the appearance of tissue-specific stem cells. Tissue-specific stem cells gave rise to multicellular living systems with distinct organs which perform specific physiological functions. This setting is a constructive role of evolution; however, rebel cells can take over the molecular mechanisms for other purposes: nowadays we know that cancer stem cells, which generate aberrant organ-like structures, are at the top of a hierarchy. Furthermore, cancer stem cells are the root of metastasis, therapy resistance, and relapse. At present, most therapeutic drugs are unable to target cancer stem cells and therefore, treatment becomes a challenging issue. We expect that future research will uncover the mechanistic "forces" driving organ growth, paving the way to the implementation of new strategies to impair human tumorigenesis.
Collapse
Affiliation(s)
- Marisa M. Merino
- Department of Biochemistry, Faculty of Sciences, University of Geneva, 1205 Geneva, Switzerland
| | - Jose A. Garcia-Sanz
- Department of Molecular Biomedicine, Centro de Investigaciones Biologicas Margarita Salas, Spanish National Research Council (CIB-CSIC), 28040 Madrid, Spain
| |
Collapse
|
11
|
Growth anisotropy of the extracellular matrix shapes a developing organ. Nat Commun 2023; 14:1220. [PMID: 36869053 PMCID: PMC9984492 DOI: 10.1038/s41467-023-36739-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 02/15/2023] [Indexed: 03/05/2023] Open
Abstract
Final organ size and shape result from volume expansion by growth and shape changes by contractility. Complex morphologies can also arise from differences in growth rate between tissues. We address here how differential growth guides the morphogenesis of the growing Drosophila wing imaginal disc. We report that 3D morphology results from elastic deformation due to differential growth anisotropy between the epithelial cell layer and its enveloping extracellular matrix (ECM). While the tissue layer grows in plane, growth of the bottom ECM occurs in 3D and is reduced in magnitude, thereby causing geometric frustration and tissue bending. The elasticity, growth anisotropy and morphogenesis of the organ are fully captured by a mechanical bilayer model. Moreover, differential expression of the Matrix metalloproteinase MMP2 controls growth anisotropy of the ECM envelope. This study shows that the ECM is a controllable mechanical constraint whose intrinsic growth anisotropy directs tissue morphogenesis in a developing organ.
Collapse
|
12
|
Chen YL, Xie XX, Zhong N, Sun LC, Lin D, Zhang LJ, Weng L, Jin T, Cao MJ. Research Progresses and Applications of Fluorescent Protein Antibodies: A Review Focusing on Nanobodies. Int J Mol Sci 2023; 24:4307. [PMID: 36901737 PMCID: PMC10002328 DOI: 10.3390/ijms24054307] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 02/16/2023] [Accepted: 02/18/2023] [Indexed: 02/24/2023] Open
Abstract
Since the discovery of fluorescent proteins (FPs), their rich fluorescence spectra and photochemical properties have promoted widespread biological research applications. FPs can be classified into green fluorescent protein (GFP) and its derivates, red fluorescent protein (RFP) and its derivates, and near-infrared FPs. With the continuous development of FPs, antibodies targeting FPs have emerged. The antibody, a class of immunoglobulin, is the main component of humoral immunity that explicitly recognizes and binds antigens. Monoclonal antibody, originating from a single B cell, has been widely applied in immunoassay, in vitro diagnostics, and drug development. The nanobody is a new type of antibody entirely composed of the variable domain of a heavy-chain antibody. Compared with conventional antibodies, these small and stable nanobodies can be expressed and functional in living cells. In addition, they can easily access grooves, seams, or hidden antigenic epitopes on the surface of the target. This review provides an overview of various FPs, the research progress of their antibodies, particularly nanobodies, and advanced applications of nanobodies targeting FPs. This review will be helpful for further research on nanobodies targeting FPs, making FPs more valuable in biological research.
Collapse
Affiliation(s)
- Yu-Lei Chen
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Xin-Xin Xie
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Ning Zhong
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Le-Chang Sun
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Duanquan Lin
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Ling-Jing Zhang
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Ling Weng
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Tengchuan Jin
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science & Technology of China, Hefei 230007, China
| | - Min-Jie Cao
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
| |
Collapse
|
13
|
Loreau V, Rees R, Chan EH, Taxer W, Gregor K, Mußil B, Pitaval C, Luis NM, Mangeol P, Schnorrer F, Görlich D. A nanobody toolbox to investigate localisation and dynamics of Drosophila titins and other key sarcomeric proteins. eLife 2023; 12:79343. [PMID: 36645120 PMCID: PMC9886281 DOI: 10.7554/elife.79343] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 12/16/2022] [Indexed: 01/17/2023] Open
Abstract
Measuring the positions and dynamics of proteins in intact tissues or whole animals is key to understanding protein function. However, to date, this is challenging, as the accessibility of large antibodies to dense tissues is often limited, and fluorescent proteins inserted close to a domain of interest may affect protein function. These complications apply in particular to muscle sarcomeres, arguably one of the most protein-dense assemblies in nature, which complicates studying sarcomere morphogenesis at molecular resolution. Here, we introduce a toolbox of nanobodies recognising various domains of the two Drosophila titin homologs, Sallimus and Projectin, as well as the key sarcomeric proteins Obscurin, α-Actinin, and Zasp52. We verified the superior labelling qualities of our nanobodies in muscle tissue as compared to antibodies. By applying our toolbox to larval muscles, we found a gigantic Sallimus isoform stretching more than 2 µm to bridge the sarcomeric I-band, while Projectin covers almost the entire myosin filaments in a polar orientation. Transgenic expression of tagged nanobodies confirmed their high affinity-binding without affecting target protein function. Finally, adding a degradation signal to anti-Sallimus nanobodies suggested that it is difficult to fully degrade Sallimus in mature sarcomeres; however, expression of these nanobodies caused developmental lethality. These results may inspire the generation of similar toolboxes for other large protein complexes in Drosophila or mammals.
Collapse
Affiliation(s)
- Vincent Loreau
- Turing Centre for Living Systems, Aix-Marseille University, CNRS, IDBMMarseilleFrance
| | - Renate Rees
- Department of Cellular Logistics, Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Eunice HoYee Chan
- Turing Centre for Living Systems, Aix-Marseille University, CNRS, IDBMMarseilleFrance
| | - Waltraud Taxer
- Department of Cellular Logistics, Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Kathrin Gregor
- Department of Cellular Logistics, Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Bianka Mußil
- Department of Cellular Logistics, Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Christophe Pitaval
- Turing Centre for Living Systems, Aix-Marseille University, CNRS, IDBMMarseilleFrance
| | - Nuno Miguel Luis
- Turing Centre for Living Systems, Aix-Marseille University, CNRS, IDBMMarseilleFrance
| | - Pierre Mangeol
- Turing Centre for Living Systems, Aix-Marseille University, CNRS, IDBMMarseilleFrance
| | - Frank Schnorrer
- Turing Centre for Living Systems, Aix-Marseille University, CNRS, IDBMMarseilleFrance
| | - Dirk Görlich
- Department of Cellular Logistics, Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
| |
Collapse
|
14
|
Schueder F, Mangeol P, Chan EH, Rees R, Schünemann J, Jungmann R, Görlich D, Schnorrer F. Nanobodies combined with DNA-PAINT super-resolution reveal a staggered titin nanoarchitecture in flight muscles. eLife 2023; 12:e79344. [PMID: 36645127 PMCID: PMC9886278 DOI: 10.7554/elife.79344] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 11/22/2022] [Indexed: 01/17/2023] Open
Abstract
Sarcomeres are the force-producing units of all striated muscles. Their nanoarchitecture critically depends on the large titin protein, which in vertebrates spans from the sarcomeric Z-disc to the M-band and hence links actin and myosin filaments stably together. This ensures sarcomeric integrity and determines the length of vertebrate sarcomeres. However, the instructive role of titins for sarcomeric architecture outside of vertebrates is not as well understood. Here, we used a series of nanobodies, the Drosophila titin nanobody toolbox, recognising specific domains of the two Drosophila titin homologs Sallimus and Projectin to determine their precise location in intact flight muscles. By combining nanobodies with DNA-PAINT super-resolution microscopy, we found that, similar to vertebrate titin, Sallimus bridges across the flight muscle I-band, whereas Projectin is located at the beginning of the A-band. Interestingly, the ends of both proteins overlap at the I-band/A-band border, revealing a staggered organisation of the two Drosophila titin homologs. This architecture may help to stably anchor Sallimus at the myosin filament and hence ensure efficient force transduction during flight.
Collapse
Affiliation(s)
- Florian Schueder
- Faculty of Physics and Center for Nanoscience, Ludwig Maximilian UniversityMunichGermany
- Max Planck Institute of BiochemistryMartinsriedGermany
| | - Pierre Mangeol
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living SystemsMarseilleFrance
| | - Eunice HoYee Chan
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living SystemsMarseilleFrance
| | - Renate Rees
- Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | | | - Ralf Jungmann
- Faculty of Physics and Center for Nanoscience, Ludwig Maximilian UniversityMunichGermany
- Max Planck Institute of BiochemistryMartinsriedGermany
| | - Dirk Görlich
- Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Frank Schnorrer
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living SystemsMarseilleFrance
| |
Collapse
|
15
|
Alvarez-Rodrigo I, Willnow D, Vincent JP. The logistics of Wnt production and delivery. Curr Top Dev Biol 2023; 153:1-60. [PMID: 36967191 DOI: 10.1016/bs.ctdb.2023.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Wnts are secreted proteins that control stem cell maintenance, cell fate decisions, and growth during development and adult homeostasis. Wnts carry a post-translational modification not seen in any other secreted protein: during biosynthesis, they are appended with a palmitoleoyl moiety that is required for signaling but also impairs solubility and hence diffusion in the extracellular space. In some contexts, Wnts act only in a juxtacrine manner but there are also instances of long range action. Several proteins and processes ensure that active Wnts reach the appropriate target cells. Some, like Porcupine, Wntless, and Notum are dedicated to Wnt function; we describe their activities in molecular detail. We also outline how the cell infrastructure (secretory, endocytic, and retromer pathways) contribute to the progression of Wnts from production to delivery. We then address how Wnts spread in the extracellular space and form a signaling gradient despite carrying a hydrophobic moiety. We highlight particularly the role of lipid-binding Wnt interactors and heparan sulfate proteoglycans. Finally, we briefly discuss how evolution might have led to the emergence of this unusual signaling pathway.
Collapse
|
16
|
Abstract
Metazoan embryos develop from a single cell into three-dimensional structured organisms while groups of genetically identical cells attain specialized identities. Cells of the developing embryo both create and accurately interpret morphogen gradients to determine their positions and make specific decisions in response. Here, we first cover intellectual roots of morphogen and positional information concepts. Focusing on animal embryos, we then provide a review of current understanding on how morphogen gradients are established and how their spans are controlled. Lastly, we cover how gradients evolve in time and space during development, and how they encode information to control patterning. In sum, we provide a list of patterning principles for morphogen gradients and review recent advances in quantitative methodologies elucidating information provided by morphogens.
Collapse
Affiliation(s)
- M. Fethullah Simsek
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Ertuğrul M. Özbudak
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| |
Collapse
|
17
|
Peterson AJ, Murphy SJ, Mundt MG, Shimell M, Leof EB, O’Connor MB. A juxtamembrane basolateral targeting motif regulates signaling through a TGF-β pathway receptor in Drosophila. PLoS Biol 2022; 20:e3001660. [PMID: 35594316 PMCID: PMC9162340 DOI: 10.1371/journal.pbio.3001660] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 06/02/2022] [Accepted: 05/04/2022] [Indexed: 11/23/2022] Open
Abstract
In polarized epithelial cells, receptor-ligand interactions can be restricted by different spatial distributions of the 2 interacting components, giving rise to an underappreciated layer of regulatory complexity. We explored whether such regulation occurs in the Drosophila wing disc, an epithelial tissue featuring the TGF-β family member Decapentaplegic (Dpp) as a morphogen controlling growth and patterning. Dpp protein has been observed in an extracellular gradient within the columnar cell layer of the disc, but also uniformly in the disc lumen, leading to the question of how graded signaling is achieved in the face of 2 distinctly localized ligand pools. We find the Dpp Type II receptor Punt, but not the Type I receptor Tkv, is enriched at the basolateral membrane and depleted at the junctions and apical surface. Wit, a second Type II receptor, shows a markedly different behavior, with the protein detected on all membrane regions but enriched at the apical side. Mutational studies identified a short juxtamembrane sequence required for basolateral restriction of Punt in both wing discs and mammalian Madin-Darby canine kidney (MDCK) cells. This basolateral targeting (BLT) determinant can dominantly confer basolateral localization on an otherwise apical receptor. Rescue of punt mutants with transgenes altered in the targeting motif showed that flies expressing apicalized Punt due to the lack of a functional BLT displayed developmental defects, female sterility, and significant lethality. We also show that apicalized Punt does not produce an ectopic signal, indicating that the apical pool of Dpp is not a significant signaling source even when presented with Punt. Instead, we find that basolateral presentation of Punt is required for optimal signaling. Finally, we present evidence that the BLT acts through polarized sorting machinery that differs between types of epithelia. This suggests a code whereby each epithelial cell type may differentially traffic common receptors to enable distinctive responses to spatially localized pools of extracellular ligands.
Collapse
Affiliation(s)
- Aidan J. Peterson
- Department of Genetics, Cell Biology & Development and the Developmental Biology Center, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Stephen J. Murphy
- Center for Individualized Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota, United States of America
| | - Melinda G. Mundt
- Department of Genetics, Cell Biology & Development and the Developmental Biology Center, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - MaryJane Shimell
- Department of Genetics, Cell Biology & Development and the Developmental Biology Center, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Edward B. Leof
- Thoracic Diseases Research Unit, Department of Pulmonary and Critical Care Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota, United States of America
| | - Michael B. O’Connor
- Department of Genetics, Cell Biology & Development and the Developmental Biology Center, University of Minnesota, Minneapolis, Minnesota, United States of America
| |
Collapse
|
18
|
Xu J, Kim AR, Cheloha RW, Fischer FA, Li JSS, Feng Y, Stoneburner E, Binari R, Mohr SE, Zirin J, Ploegh HL, Perrimon N. Protein visualization and manipulation in Drosophila through the use of epitope tags recognized by nanobodies. eLife 2022; 11:74326. [PMID: 35076390 PMCID: PMC8853664 DOI: 10.7554/elife.74326] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Abstract
Expansion of the available repertoire of reagents for visualization and manipulation of proteins will help understand their function. Short epitope tags linked to proteins of interest and recognized by existing binders such as nanobodies facilitate protein studies by obviating the need to isolate new antibodies directed against them. Nanobodies have several advantages over conventional antibodies, as they can be expressed and used as tools for visualization and manipulation of proteins in vivo. Here, we characterize two short (<15aa) NanoTag epitopes, 127D01 and VHH05, and their corresponding high-affinity nanobodies. We demonstrate their use in Drosophila for in vivo protein detection and re-localization, direct and indirect immunofluorescence, immunoblotting, and immunoprecipitation. We further show that CRISPR-mediated gene targeting provides a straightforward approach to tagging endogenous proteins with the NanoTags. Single copies of the NanoTags, regardless of their location, suffice for detection. This versatile and validated toolbox of tags and nanobodies will serve as a resource for a wide array of applications, including functional studies in Drosophila and beyond.
Collapse
Affiliation(s)
- Jun Xu
- Department of Genetics, Harvard Medical School
| | - Ah-Ram Kim
- Department of Genetics, Harvard Medical School
| | | | | | | | - Yuan Feng
- Department of Genetics, Harvard Medical School
| | | | | | | | | | | | | |
Collapse
|
19
|
Avellaneda J, Schnorrer F. Tagging Drosophila Proteins with Genetically Encoded Fluorophores. Methods Mol Biol 2022; 2540:251-268. [PMID: 35980582 DOI: 10.1007/978-1-0716-2541-5_12] [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] [Indexed: 06/15/2023]
Abstract
Proteins are typically not expressed homogeneously in all cells of a complex organism. Within cells, proteins can dynamically change locations, be transported to their destinations, or be degraded upon external signals. Thus, revealing the cellular and subcellular localizations as well as the temporal dynamics of a protein provides important insights into the possible function of the studied protein. Tagging a protein of interest with a genetically encoded fluorophore enables us to follow its expression dynamics in the living organism. Here, we summarize the genetic resources available for tagged Drosophila proteins that assist in studying protein expression and dynamics. We also review the various techniques used in the past and at present to tag a protein of interest with a genetically encoded fluorophore. Comparing the pros and cons of the various techniques guides the reader to judge the suitable applications possible with these tagged proteins in Drosophila.
Collapse
Affiliation(s)
- Jerome Avellaneda
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living Systems, Marseille, France
| | - Frank Schnorrer
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living Systems, Marseille, France.
| |
Collapse
|
20
|
Matsuda S, Aguilar G, Vigano MA, Affolter M. Nanobody-Based GFP Traps to Study Protein Localization and Function in Developmental Biology. Methods Mol Biol 2022; 2446:581-593. [PMID: 35157295 DOI: 10.1007/978-1-0716-2075-5_30] [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] [Indexed: 06/14/2023]
Abstract
Synthetic protein-binding tools based on anti-green fluorescent protein (GFP) nanobodies have recently emerged as useful resources to study developmental biology. By fusing GFP-targeting nanobodies to well-characterized protein domains residing in discrete sub-cellular locations, it is possible to directly and acutely manipulate the localization of GFP-tagged proteins-of-interest in a predictable manner. Here, we describe a detailed protocol for the application of nanobody-based GFP-binding tools, namely Morphotrap and GrabFP, to study the localization and function of extracellular and intracellular proteins in the Drosophila wing imaginal disc. Given the generality of these methods, they are easily applicable for use in other tissues and model organisms.
Collapse
|
21
|
Lepeta K, Bauer M, Aguilar G, Vigano MA, Matsuda S, Affolter M. Studying Protein Function Using Nanobodies and Other Protein Binders in Drosophila. Methods Mol Biol 2022; 2540:219-237. [PMID: 35980580 DOI: 10.1007/978-1-0716-2541-5_10] [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] [Indexed: 06/15/2023]
Abstract
The direct manipulation of proteins by nanobodies and other protein binders has become an additional and valuable approach to investigate development and homeostasis in Drosophila. In contrast to other techniques, that indirectly interfere with proteins via their nucleic acids (CRISPR, RNAi, etc.), protein binders permit direct and acute protein manipulation. Since the first use of a nanobody in Drosophila a decade ago, many different applications exploiting protein binders have been introduced. Most of these applications use nanobodies against GFP to regulate GFP fusion proteins. In order to exert specific protein manipulations, protein binders are linked to domains that confer them precise biochemical functions. Here, we reflect on the use of tools based on protein binders in Drosophila. We describe their key features and provide an overview of the available reagents. Finally, we briefly explore the future avenues that protein binders might open up and thus further contribute to better understand development and homeostasis of multicellular organisms.
Collapse
Affiliation(s)
| | - Milena Bauer
- Biozentrum der Universität Basel, Basel, Switzerland
| | | | | | | | | |
Collapse
|
22
|
Michailidi MR, Hadjivasiliou Z, Aguilar-Hidalgo D, Basagiannis D, Seum C, Dubois M, Jülicher F, Gonzalez-Gaitan M. Morphogen gradient scaling by recycling of intracellular Dpp. Nature 2021; 602:287-293. [PMID: 34937053 DOI: 10.1038/s41586-021-04346-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 12/10/2021] [Indexed: 11/09/2022]
Abstract
Morphogen gradients are fundamental to establish morphological patterns in developing tissues1. During development, gradients scale to remain proportional to the size of growing organs2,3. Scaling is a universal gear adjusting patterns to size in living organisms3-8, yet its mechanisms remain unclear. Here, focusing on the Dpp gradient in the Drosophila wing disc, we unravel a cell biological basis behind scaling. From small to large discs, scaling of the Dpp gradient is achieved by increasing the contribution of the internalized Dpp molecules to Dpp transport: to expand the gradient, endocytosed molecules are re-exocytosed to spread extracellularly. To regulate the contribution of endocytosed Dpp to the spreading extracellular pool during tissue growth, it is the Dpp binding rates that are progressively modulated by the extracellular factor Pentagone, driving scaling. Thus, for some morphogens, evolution may act on endocytic trafficking to regulate the range of the gradient and its scaling, which could allow adaptation of shape and pattern to different sizes of organs in different species.
Collapse
Affiliation(s)
| | - Zena Hadjivasiliou
- Department of Biochemistry, Faculty of Sciences, University of Geneva, Geneva, Switzerland.,Max Planck Institute for the Physics of Complex Systems, Dresden, Germany
| | - Daniel Aguilar-Hidalgo
- Department of Biochemistry, Faculty of Sciences, University of Geneva, Geneva, Switzerland.,Max Planck Institute for the Physics of Complex Systems, Dresden, Germany
| | - Dimitris Basagiannis
- Department of Biochemistry, Faculty of Sciences, University of Geneva, Geneva, Switzerland
| | - Carole Seum
- Department of Biochemistry, Faculty of Sciences, University of Geneva, Geneva, Switzerland
| | - Marine Dubois
- Department of Biochemistry, Faculty of Sciences, University of Geneva, Geneva, Switzerland
| | - Frank Jülicher
- Max Planck Institute for the Physics of Complex Systems, Dresden, Germany.
| | - Marcos Gonzalez-Gaitan
- Department of Biochemistry, Faculty of Sciences, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
23
|
Asaadi Y, Jouneghani FF, Janani S, Rahbarizadeh F. A comprehensive comparison between camelid nanobodies and single chain variable fragments. Biomark Res 2021; 9:87. [PMID: 34863296 PMCID: PMC8642758 DOI: 10.1186/s40364-021-00332-6] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/22/2021] [Indexed: 12/24/2022] Open
Abstract
By the emergence of recombinant DNA technology, many antibody fragments have been developed devoid of undesired properties of natural immunoglobulins. Among them, camelid heavy-chain variable domains (VHHs) and single-chain variable fragments (scFvs) are the most favored ones. While scFv is used widely in various applications, camelid antibodies (VHHs) can serve as an alternative because of their superior chemical and physical properties such as higher solubility, stability, smaller size, and lower production cost. Here, these two counterparts are compared in structure and properties to identify which one is more suitable for each of their various therapeutic, diagnosis, and research applications.
Collapse
Affiliation(s)
- Yasaman Asaadi
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Fatemeh Fazlollahi Jouneghani
- Department of Cell & Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Sara Janani
- Department of Cell & Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Fatemeh Rahbarizadeh
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
- Research and Development Center of Biotechnology, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
24
|
Matsuda S, Schaefer JV, Mii Y, Hori Y, Bieli D, Taira M, Plückthun A, Affolter M. Asymmetric requirement of Dpp/BMP morphogen dispersal in the Drosophila wing disc. Nat Commun 2021; 12:6435. [PMID: 34750371 PMCID: PMC8576045 DOI: 10.1038/s41467-021-26726-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 10/20/2021] [Indexed: 11/26/2022] Open
Abstract
How morphogen gradients control patterning and growth in developing tissues remains largely unknown due to lack of tools manipulating morphogen gradients. Here, we generate two membrane-tethered protein binders that manipulate different aspects of Decapentaplegic (Dpp), a morphogen required for overall patterning and growth of the Drosophila wing. One is "HA trap" based on a single-chain variable fragment (scFv) against the HA tag that traps HA-Dpp to mainly block its dispersal, the other is "Dpp trap" based on a Designed Ankyrin Repeat Protein (DARPin) against Dpp that traps Dpp to block both its dispersal and signaling. Using these tools, we found that, while posterior patterning and growth require Dpp dispersal, anterior patterning and growth largely proceed without Dpp dispersal. We show that dpp transcriptional refinement from an initially uniform to a localized expression and persistent signaling in transient dpp source cells render the anterior compartment robust against the absence of Dpp dispersal. Furthermore, despite a critical requirement of dpp for the overall wing growth, neither Dpp dispersal nor direct signaling is critical for lateral wing growth after wing pouch specification. These results challenge the long-standing dogma that Dpp dispersal is strictly required to control and coordinate overall wing patterning and growth.
Collapse
Affiliation(s)
| | - Jonas V Schaefer
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Yusuke Mii
- National Institute for Basic Biology and Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi, Japan
- JST PRESTO, Kawaguchi, Saitama, Japan
| | - Yutaro Hori
- Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
| | | | - Masanori Taira
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
- Department of Biological Sciences, Faculty of Science and Engineering, Chuo University, Tokyo, Japan
| | - Andreas Plückthun
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | | |
Collapse
|
25
|
Juarez-Carreño S, Vallejo DM, Carranza-Valencia J, Palomino-Schätzlein M, Ramon-Cañellas P, Santoro R, de Hartog E, Ferres-Marco D, Romero A, Peterson HP, Ballesta-Illan E, Pineda-Lucena A, Dominguez M, Morante J. Body-fat sensor triggers ribosome maturation in the steroidogenic gland to initiate sexual maturation in Drosophila. Cell Rep 2021; 37:109830. [PMID: 34644570 DOI: 10.1016/j.celrep.2021.109830] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 06/25/2021] [Accepted: 09/23/2021] [Indexed: 12/18/2022] Open
Abstract
Fat stores are critical for reproductive success and may govern maturation initiation. Here, we report that signaling and sensing fat sufficiency for sexual maturation commitment requires the lipid carrier apolipophorin in fat cells and Sema1a in the neuroendocrine prothoracic gland (PG). Larvae lacking apolpp or Sema1a fail to initiate maturation despite accruing sufficient fat stores, and they continue gaining weight until death. Mechanistically, sensing peripheral body-fat levels via the apolipophorin/Sema1a axis regulates endocytosis, endoplasmic reticulum remodeling, and ribosomal maturation for the acquisition of the PG cells' high biosynthetic and secretory capacity. Downstream of apolipophorin/Sema1a, leptin-like upd2 triggers the cessation of feeding and initiates sexual maturation. Human Leptin in the insect PG substitutes for upd2, preventing obesity and triggering maturation downstream of Sema1a. These data show how peripheral fat levels regulate the control of the maturation decision-making process via remodeling of endomembranes and ribosomal biogenesis in gland cells.
Collapse
Affiliation(s)
- Sergio Juarez-Carreño
- Instituto de Neurociencias, Consejo Superior de Investigaciones Cientificas (CSIC), and Universidad Miguel Hernández (UMH), Campus de Sant Joan, Apartado 18, 03550 Sant Joan, Alicante, Spain
| | - Diana Marcela Vallejo
- Instituto de Neurociencias, Consejo Superior de Investigaciones Cientificas (CSIC), and Universidad Miguel Hernández (UMH), Campus de Sant Joan, Apartado 18, 03550 Sant Joan, Alicante, Spain
| | - Juan Carranza-Valencia
- Instituto de Neurociencias, Consejo Superior de Investigaciones Cientificas (CSIC), and Universidad Miguel Hernández (UMH), Campus de Sant Joan, Apartado 18, 03550 Sant Joan, Alicante, Spain
| | | | - Pol Ramon-Cañellas
- Instituto de Neurociencias, Consejo Superior de Investigaciones Cientificas (CSIC), and Universidad Miguel Hernández (UMH), Campus de Sant Joan, Apartado 18, 03550 Sant Joan, Alicante, Spain
| | - Roberto Santoro
- Instituto de Neurociencias, Consejo Superior de Investigaciones Cientificas (CSIC), and Universidad Miguel Hernández (UMH), Campus de Sant Joan, Apartado 18, 03550 Sant Joan, Alicante, Spain
| | - Emily de Hartog
- Instituto de Neurociencias, Consejo Superior de Investigaciones Cientificas (CSIC), and Universidad Miguel Hernández (UMH), Campus de Sant Joan, Apartado 18, 03550 Sant Joan, Alicante, Spain
| | - Dolors Ferres-Marco
- Instituto de Neurociencias, Consejo Superior de Investigaciones Cientificas (CSIC), and Universidad Miguel Hernández (UMH), Campus de Sant Joan, Apartado 18, 03550 Sant Joan, Alicante, Spain
| | - Aitana Romero
- Instituto de Neurociencias, Consejo Superior de Investigaciones Cientificas (CSIC), and Universidad Miguel Hernández (UMH), Campus de Sant Joan, Apartado 18, 03550 Sant Joan, Alicante, Spain
| | - Hannah Payette Peterson
- Instituto de Neurociencias, Consejo Superior de Investigaciones Cientificas (CSIC), and Universidad Miguel Hernández (UMH), Campus de Sant Joan, Apartado 18, 03550 Sant Joan, Alicante, Spain
| | - Esther Ballesta-Illan
- Instituto de Neurociencias, Consejo Superior de Investigaciones Cientificas (CSIC), and Universidad Miguel Hernández (UMH), Campus de Sant Joan, Apartado 18, 03550 Sant Joan, Alicante, Spain
| | - Antonio Pineda-Lucena
- Instituto de Investigación Sanitaria La Fe, Hospital Universitario y Politécnico La Fe, Avenida Fernando Abril Martorell, 106, 46026 Valencia, Spain; Programa de Terapias Moleculares, Centro de Investigación Médica Aplicada, Universidad de Navarra, Avenida Pío XII, 55, 31008 Pamplona, Spain
| | - Maria Dominguez
- Instituto de Neurociencias, Consejo Superior de Investigaciones Cientificas (CSIC), and Universidad Miguel Hernández (UMH), Campus de Sant Joan, Apartado 18, 03550 Sant Joan, Alicante, Spain.
| | - Javier Morante
- Instituto de Neurociencias, Consejo Superior de Investigaciones Cientificas (CSIC), and Universidad Miguel Hernández (UMH), Campus de Sant Joan, Apartado 18, 03550 Sant Joan, Alicante, Spain.
| |
Collapse
|
26
|
Ákos Z, Dunipace L, Stathopoulos A. NaNuTrap: a technique for in vivo cell nucleus labelling using nanobodies. Development 2021; 148:dev199822. [PMID: 34328170 PMCID: PMC10656463 DOI: 10.1242/dev.199822] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/12/2021] [Indexed: 11/20/2022]
Abstract
In vivo cell labelling is challenging in fast developmental processes because many cell types differentiate more quickly than the maturation time of fluorescent proteins, making visualization of these tissues impossible with standard techniques. Here, we present a nanobody-based method, Nanobody Nuclear Trap (NaNuTrap), which works with the existing Gal4/UAS system in Drosophila and allows for early in vivo cell nuclei labelling independently of the maturation time of the fluorescent protein. This restores the utility of fluorescent proteins that have longer maturation times, such as those used in two-photon imaging, for live imaging of fast or very early developmental processes. We also present a more general application of this system, whereby NaNuTrap can convert cytoplasmic GFP expressed in any existing transgenic fly line into a nuclear label. This nuclear re-localization of the fluorescent signal can improve the utility of the GFP label, e.g. in cell counting, as well as resulting in a general increase in intensity of the live fluorescent signal. We demonstrate these capabilities of NaNuTrap by effectively tracking subsets of cells during the fast movements associated with gastrulation.
Collapse
Affiliation(s)
- Zsuzsa Ákos
- California Institute of Technology, 1200 East California Blvd, Pasadena, CA 91125, USA
| | - Leslie Dunipace
- California Institute of Technology, 1200 East California Blvd, Pasadena, CA 91125, USA
| | - Angelike Stathopoulos
- California Institute of Technology, 1200 East California Blvd, Pasadena, CA 91125, USA
| |
Collapse
|
27
|
Simon E, Jiménez-Jiménez C, Seijo-Barandiarán I, Aguilar G, Sánchez-Hernández D, Aguirre-Tamaral A, González-Méndez L, Ripoll P, Guerrero I. Glypicans define unique roles for the Hedgehog co-receptors boi and ihog in cytoneme-mediated gradient formation. eLife 2021; 10:64581. [PMID: 34355694 PMCID: PMC8410076 DOI: 10.7554/elife.64581] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 08/04/2021] [Indexed: 02/06/2023] Open
Abstract
The conserved family of Hedgehog (Hh) signaling proteins plays a key role in cell–cell communication in development, tissue repair, and cancer progression, inducing distinct concentration-dependent responses in target cells located at short and long distances. One simple mechanism for long distance dispersal of the lipid modified Hh is the direct contact between cell membranes through filopodia-like structures known as cytonemes. Here we have analyzed in Drosophila the interaction between the glypicans Dally and Dally-like protein, necessary for Hh signaling, and the adhesion molecules and Hh coreceptors Ihog and Boi. We describe that glypicans are required to maintain the levels of Ihog, but not of Boi. We also show that the overexpression of Ihog, but not of Boi, regulates cytoneme dynamics through their interaction with glypicans, the Ihog fibronectin III domains being essential for this interaction. Our data suggest that the regulation of glypicans over Hh signaling is specifically given by their interaction with Ihog in cytonemes. Contrary to previous data, we also show that there is no redundancy of Ihog and Boi functions in Hh gradient formation, being Ihog, but not of Boi, essential for the long-range gradient.
Collapse
Affiliation(s)
- Eléanor Simon
- Tissue and Organ Homeostasis, Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Nicolás Cabrera 1, Universidad Autónoma de Madrid, Cantoblanco, Spain
| | - Carlos Jiménez-Jiménez
- Tissue and Organ Homeostasis, Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Nicolás Cabrera 1, Universidad Autónoma de Madrid, Cantoblanco, Spain
| | - Irene Seijo-Barandiarán
- Tissue and Organ Homeostasis, Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Nicolás Cabrera 1, Universidad Autónoma de Madrid, Cantoblanco, Spain
| | - Gustavo Aguilar
- Tissue and Organ Homeostasis, Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Nicolás Cabrera 1, Universidad Autónoma de Madrid, Cantoblanco, Spain.,Growth and Development, University of Basel, Biozentrum, Switzerland
| | - David Sánchez-Hernández
- Tissue and Organ Homeostasis, Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Nicolás Cabrera 1, Universidad Autónoma de Madrid, Cantoblanco, Spain
| | - Adrián Aguirre-Tamaral
- Tissue and Organ Homeostasis, Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Nicolás Cabrera 1, Universidad Autónoma de Madrid, Cantoblanco, Spain
| | - Laura González-Méndez
- Tissue and Organ Homeostasis, Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Nicolás Cabrera 1, Universidad Autónoma de Madrid, Cantoblanco, Spain
| | - Pedro Ripoll
- Tissue and Organ Homeostasis, Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Nicolás Cabrera 1, Universidad Autónoma de Madrid, Cantoblanco, Spain
| | - Isabel Guerrero
- Tissue and Organ Homeostasis, Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Nicolás Cabrera 1, Universidad Autónoma de Madrid, Cantoblanco, Spain
| |
Collapse
|
28
|
Montanari MP, Tran NV, Shimmi O. Regulation of spatial distribution of BMP ligands for pattern formation. Dev Dyn 2021; 251:198-212. [PMID: 34241935 DOI: 10.1002/dvdy.397] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/15/2021] [Accepted: 07/05/2021] [Indexed: 12/25/2022] Open
Abstract
Bone morphogenetic proteins (BMPs), members of the transforming growth factor-ß (TGF-ß) family, have been shown to contribute to embryogenesis and organogenesis during animal development. Relevant studies provide support for the following concepts: (a) BMP signals are evolutionarily highly conserved as a genetic toolkit; (b) spatiotemporal distributions of BMP signals are precisely controlled at the post-translational level; and (c) the BMP signaling network has been co-opted to adapt to diversified animal development. These concepts originated from the historical findings of the Spemann-Mangold organizer and the subsequent studies about how this organizer functions at the molecular level. In this Commentary, we focus on two topics. First, we review how the BMP morphogen gradient is formed to sustain larval wing imaginal disc and early embryo growth and patterning in Drosophila. Second, we discuss how BMP signal is tightly controlled in a context-dependent manner, and how the signal and tissue dynamics are coupled to facilitate complex tissue structure formation. Finally, we argue how these concepts might be developed in the future for further understanding the significance of BMP signaling in animal development.
Collapse
Affiliation(s)
| | - Ngan Vi Tran
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Osamu Shimmi
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland.,Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| |
Collapse
|
29
|
Lord ND, Carte AN, Abitua PB, Schier AF. The pattern of nodal morphogen signaling is shaped by co-receptor expression. eLife 2021; 10:e54894. [PMID: 34036935 PMCID: PMC8266389 DOI: 10.7554/elife.54894] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 05/26/2021] [Indexed: 12/13/2022] Open
Abstract
Embryos must communicate instructions to their constituent cells over long distances. These instructions are often encoded in the concentration of signals called morphogens. In the textbook view, morphogen molecules diffuse from a localized source to form a concentration gradient, and target cells adopt fates by measuring the local morphogen concentration. However, natural patterning systems often incorporate numerous co-factors and extensive signaling feedback, suggesting that embryos require additional mechanisms to generate signaling patterns. Here, we examine the mechanisms of signaling pattern formation for the mesendoderm inducer Nodal during zebrafish embryogenesis. We find that Nodal signaling activity spans a normal range in the absence of signaling feedback and relay, suggesting that diffusion is sufficient for Nodal gradient formation. We further show that the range of endogenous Nodal ligands is set by the EGF-CFC co-receptor Oep: in the absence of Oep, Nodal activity spreads to form a nearly uniform distribution throughout the embryo. In turn, increasing Oep levels sensitizes cells to Nodal ligands. We recapitulate these experimental results with a computational model in which Oep regulates the diffusive spread of Nodal ligands by setting the rate of capture by target cells. This model predicts, and we confirm in vivo, the surprising observation that a failure to replenish Oep transforms the Nodal signaling gradient into a travelling wave. These results reveal that patterns of Nodal morphogen signaling are shaped by co-receptor-mediated restriction of ligand spread and sensitization of responding cells.
Collapse
Affiliation(s)
- Nathan D Lord
- Department of Molecular and Cellular Biology, Harvard UniversityCambridgeUnited States
| | - Adam N Carte
- Department of Molecular and Cellular Biology, Harvard UniversityCambridgeUnited States
- Systems, Synthetic, and Quantitative Biology PhD Program, Harvard UniversityCambridgeUnited States
- Biozentrum, University of BaselBaselSwitzerland
| | - Philip B Abitua
- Department of Molecular and Cellular Biology, Harvard UniversityCambridgeUnited States
| | - Alexander F Schier
- Department of Molecular and Cellular Biology, Harvard UniversityCambridgeUnited States
- Biozentrum, University of BaselBaselSwitzerland
- Allen Discovery Center for Cell Lineage Tracing, University of WashingtonSeattleUnited States
| |
Collapse
|
30
|
Mii Y, Nakazato K, Pack CG, Ikeda T, Sako Y, Mochizuki A, Taira M, Takada S. Quantitative analyses reveal extracellular dynamics of Wnt ligands in Xenopus embryos. eLife 2021; 10:55108. [PMID: 33904408 PMCID: PMC8139832 DOI: 10.7554/elife.55108] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 04/23/2021] [Indexed: 12/11/2022] Open
Abstract
The mechanism of intercellular transport of Wnt ligands is still a matter of debate. To better understand this issue, we examined the distribution and dynamics of Wnt8 in Xenopus embryos. While Venus-tagged Wnt8 was found on the surfaces of cells close to Wnt-producing cells, we also detected its dispersal over distances of 15 cell diameters. A combination of fluorescence correlation spectroscopy and quantitative imaging suggested that only a small proportion of Wnt8 ligands diffuses freely, whereas most Wnt8 molecules are bound to cell surfaces. Fluorescence decay after photoconversion showed that Wnt8 ligands bound on cell surfaces decrease exponentially, suggesting a dynamic exchange of bound forms of Wnt ligands. Mathematical modeling based on this exchange recapitulates a graded distribution of bound, but not free, Wnt ligands. Based on these results, we propose that Wnt distribution in tissues is controlled by a dynamic exchange of its abundant bound and rare free populations.
Collapse
Affiliation(s)
- Yusuke Mii
- National Institute for Basic Biology and Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Japan.,The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Japan.,Japan Science and Technology Agency (JST), PRESTO, Kawaguchi, Japan.,Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | | | - Chan-Gi Pack
- Cellular Informatics Laboratory, RIKEN, Wako, Japan.,ASAN Institute for Life Sciences, ASAN Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Takafumi Ikeda
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Yasushi Sako
- Cellular Informatics Laboratory, RIKEN, Wako, Japan
| | - Atsushi Mochizuki
- Theoretical Biology Laboratory, RIKEN, Wako, Japan.,Laboratory of Mathematical Biology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Masanori Taira
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan.,Department of Biological Sciences, Faculty of Science and Engineering, Chuo University, Tokyo, Japan
| | - Shinji Takada
- National Institute for Basic Biology and Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Japan.,The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Japan
| |
Collapse
|
31
|
Madamanchi A, Mullins MC, Umulis DM. Diversity and robustness of bone morphogenetic protein pattern formation. Development 2021; 148:dev192344. [PMID: 33795238 PMCID: PMC8034876 DOI: 10.1242/dev.192344] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Pattern formation by bone morphogenetic proteins (BMPs) demonstrates remarkable plasticity and utility in several contexts, such as early embryonic development, tissue patterning and the maintenance of stem cell niches. BMPs pattern tissues over many temporal and spatial scales: BMP gradients as short as 1-2 cell diameters maintain the stem cell niche of the Drosophila germarium over a 24-h cycle, and BMP gradients of several hundred microns establish dorsal-ventral tissue specification in Drosophila, zebrafish and Xenopus embryos in timescales between 30 min and several hours. The mechanisms that shape BMP signaling gradients are also incredibly diverse. Although ligand diffusion plays a dominant role in forming the gradient, a cast of diffusible and non-diffusible regulators modulate gradient formation and confer robustness, including scale invariance and adaptability to perturbations in gene expression and growth. In this Review, we document the diverse ways that BMP gradients are formed and refined, and we identify the core principles that they share to achieve reliable performance.
Collapse
Affiliation(s)
- Aasakiran Madamanchi
- Agricultural and Biological Engineering. Purdue University, West Lafayette, IN 47907, USA
- Polytechnic Institute, Purdue University, West Lafayette, IN 47907, USA
| | - Mary C Mullins
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - David M Umulis
- Agricultural and Biological Engineering. Purdue University, West Lafayette, IN 47907, USA
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
32
|
Zecca M, Struhl G. A unified mechanism for the control of Drosophila wing growth by the morphogens Decapentaplegic and Wingless. PLoS Biol 2021; 19:e3001111. [PMID: 33657096 PMCID: PMC8148325 DOI: 10.1371/journal.pbio.3001111] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 05/25/2021] [Accepted: 01/22/2021] [Indexed: 12/31/2022] Open
Abstract
Development of the Drosophila wing-a paradigm of organ development-is governed by 2 morphogens, Decapentaplegic (Dpp, a BMP) and Wingless (Wg, a Wnt). Both proteins are produced by defined subpopulations of cells and spread outwards, forming gradients that control gene expression and cell pattern as a function of concentration. They also control growth, but how is unknown. Most studies have focused on Dpp and yielded disparate models in which cells throughout the wing grow at similar rates in response to the grade or temporal change in Dpp concentration or to the different amounts of Dpp "equalized" by molecular or mechanical feedbacks. In contrast, a model for Wg posits that growth is governed by a progressive expansion in morphogen range, via a mechanism in which a minimum threshold of Wg sustains the growth of cells within the wing and recruits surrounding "pre-wing" cells to grow and enter the wing. This mechanism depends on the capacity of Wg to fuel the autoregulation of vestigial (vg)-the selector gene that specifies the wing state-both to sustain vg expression in wing cells and by a feed-forward (FF) circuit of Fat (Ft)/Dachsous (Ds) protocadherin signaling to induce vg expression in neighboring pre-wing cells. Here, we have subjected Dpp to the same experimental tests used to elucidate the Wg model and find that it behaves indistinguishably. Hence, we posit that both morphogens act together, via a common mechanism, to control wing growth as a function of morphogen range.
Collapse
Affiliation(s)
- Myriam Zecca
- Department of Genetics and Development, Columbia University, New York, New York, United States of America
- The Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, New York, United States of America
| | - Gary Struhl
- Department of Genetics and Development, Columbia University, New York, New York, United States of America
- The Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, New York, United States of America
| |
Collapse
|
33
|
Martin E, Theis S, Gay G, Monier B, Rouvière C, Suzanne M. Arp2/3-dependent mechanical control of morphogenetic robustness in an inherently challenging environment. Dev Cell 2021; 56:687-701.e7. [PMID: 33535069 PMCID: PMC7955168 DOI: 10.1016/j.devcel.2021.01.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 10/23/2020] [Accepted: 01/08/2021] [Indexed: 12/21/2022]
Abstract
Epithelial sheets undergo highly reproducible remodeling to shape organs. This stereotyped morphogenesis depends on a well-defined sequence of events leading to the regionalized expression of developmental patterning genes that finally triggers downstream mechanical forces to drive tissue remodeling at a pre-defined position. However, how tissue mechanics controls morphogenetic robustness when challenged by intrinsic perturbations in close proximity has never been addressed. Using Drosophila developing leg, we show that a bias in force propagation ensures stereotyped morphogenesis despite the presence of mechanical noise in the environment. We found that knockdown of the Arp2/3 complex member Arpc5 specifically affects fold directionality while altering neither the developmental nor the force generation patterns. By combining in silico modeling, biophysical tools, and ad hoc genetic tools, our data reveal that junctional myosin II planar polarity favors long-range force channeling and ensures folding robustness, avoiding force scattering and thus isolating the fold domain from surrounding mechanical perturbations. Drosophila developing leg folding is extremely robust Fold orientation becomes variable in Arp2/3 knockdown condition Arp2/3 controls junctional myosin II planar polarity Myosin II planar polarity ensures fold robustness through force channeling
Collapse
Affiliation(s)
- Emmanuel Martin
- Molecular, Cellular & Developmental Biology (MCD), Center of Integrative Biology (CBI), Toulouse University, CNRS, UPS, Toulouse, France
| | - Sophie Theis
- Molecular, Cellular & Developmental Biology (MCD), Center of Integrative Biology (CBI), Toulouse University, CNRS, UPS, Toulouse, France; Morphogénie Logiciels, 32110 St Martin d'Armagnac, France
| | - Guillaume Gay
- Morphogénie Logiciels, 32110 St Martin d'Armagnac, France; Turing Center For Living Systems, Aix-MarseilleUniversity, 13009, Marseille, France.
| | - Bruno Monier
- Molecular, Cellular & Developmental Biology (MCD), Center of Integrative Biology (CBI), Toulouse University, CNRS, UPS, Toulouse, France
| | - Christian Rouvière
- Image Processing Facility, Center of Integrative Biology (CBI), Université de Toulouse, CNRS, UPS, France
| | - Magali Suzanne
- Molecular, Cellular & Developmental Biology (MCD), Center of Integrative Biology (CBI), Toulouse University, CNRS, UPS, Toulouse, France.
| |
Collapse
|
34
|
Driesschaert B, Mergan L, Temmerman L. Conditional gene expression in invertebrate animal models. J Genet Genomics 2021; 48:14-31. [PMID: 33814307 DOI: 10.1016/j.jgg.2021.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/11/2020] [Accepted: 01/08/2021] [Indexed: 10/22/2022]
Abstract
A mechanistic understanding of biology requires appreciating spatiotemporal aspects of gene expression and its functional implications. Conditional expression allows for (ir)reversible switching of genes on or off, with the potential of spatial and/or temporal control. This provides a valuable complement to the more often used constitutive gene (in)activation through mutagenesis, providing tools to answer a wider array of research questions across biological disciplines. Spatial and/or temporal control are granted primarily by (combinations of) specific promoters, temperature regimens, compound addition, or illumination. The use of such genetic tool kits is particularly widespread in invertebrate animal models because they can be applied to study biological processes in short time frames and on large scales, using organisms amenable to easy genetic manipulation. Recent years witnessed an exciting expansion and optimization of such tools, of which we provide a comprehensive overview and discussion regarding their use in invertebrates. The mechanism, applicability, benefits, and drawbacks of each of the systems, as well as further developments to be expected in the foreseeable future, are highlighted.
Collapse
Affiliation(s)
- Brecht Driesschaert
- Animal Physiology and Neurobiology, Department of Biology, University of Leuven (KU Leuven), Naamsestraat 59 - Box 2465, B-3000 Leuven, Belgium
| | - Lucas Mergan
- Animal Physiology and Neurobiology, Department of Biology, University of Leuven (KU Leuven), Naamsestraat 59 - Box 2465, B-3000 Leuven, Belgium
| | - Liesbet Temmerman
- Animal Physiology and Neurobiology, Department of Biology, University of Leuven (KU Leuven), Naamsestraat 59 - Box 2465, B-3000 Leuven, Belgium.
| |
Collapse
|
35
|
Control of Drosophila wing size by morphogen range and hormonal gating. Proc Natl Acad Sci U S A 2020; 117:31935-31944. [PMID: 33257577 DOI: 10.1073/pnas.2018196117] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The stereotyped dimensions of animal bodies and their component parts result from tight constraints on growth. Yet, the mechanisms that stop growth when organs reach the right size are unknown. Growth of the Drosophila wing-a classic paradigm-is governed by two morphogens, Decapentaplegic (Dpp, a BMP) and Wingless (Wg, a Wnt). Wing growth during larval life ceases when the primordium attains full size, concomitant with the larval-to-pupal molt orchestrated by the steroid hormone ecdysone. Here, we block the molt by genetically dampening ecdysone production, creating an experimental paradigm in which the wing stops growing at the correct size while the larva continues to feed and gain body mass. Under these conditions, we show that wing growth is limited by the ranges of Dpp and Wg, and by ecdysone, which regulates the cellular response to their signaling activities. Further, we present evidence that growth terminates because of the loss of two distinct modes of morphogen action: 1) maintenance of growth within the wing proper and 2) induced growth of surrounding "pre-wing" cells and their recruitment into the wing. Our results provide a precedent for the control of organ size by morphogen range and the hormonal gating of morphogen action.
Collapse
|
36
|
Mii Y. Heparan Sulfate Clusters Regulate Distribution and Signaling of Wnt Morphogens. TRENDS GLYCOSCI GLYC 2020. [DOI: 10.4052/tigg.2006.7j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Yusuke Mii
- National Institute for Basic Biology, National Institutes of Natural Sciences
- Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences
- Department of Basic Biology, Graduate University for Advanced Studies (SOKENDAI)
- Japan Science and Technology Agency, PRESTO
| |
Collapse
|
37
|
Mii Y. Heparan Sulfate Clusters Regulate Distribution and Signaling of Wnt Morphogens. TRENDS GLYCOSCI GLYC 2020. [DOI: 10.4052/tigg.2006.7e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Yusuke Mii
- National Institute for Basic Biology, National Institutes of Natural Sciences
- Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences
- Department of Basic Biology, Graduate University for Advanced Studies (SOKENDAI)
- Japan Science and Technology Agency, PRESTO
| |
Collapse
|
38
|
Abstract
Unique, functional, homodimeric heavy chain-only antibodies, devoid of light chains, are circulating in the blood of Camelidae. These antibodies recognize their cognate antigen via one single domain, known as VHH or Nanobody. This serendipitous discovery made three decades ago has stimulated a growing number of researchers to generate highly specific Nanobodies against a myriad of targets. The small size, strict monomeric state, robustness, and easy tailoring of these Nanobodies have inspired many groups to design innovative Nanobody-based multi-domain constructs to explore novel applications. As such, Nanobodies have been employed as an exquisite research tool in structural, cell, and developmental biology. Furthermore, Nanobodies have demonstrated their benefit for more sensitive diagnostic tests. Finally, several Nanobody-based constructs have been designed to develop new therapeutic products.
Collapse
Affiliation(s)
- Serge Muyldermans
- Cellular and Molecular Immunology, Vrije Universiteit Brussel, 1050 Brussels, Belgium; .,Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, Liaoning, People's Republic of China
| |
Collapse
|
39
|
Cheloha RW, Harmand TJ, Wijne C, Schwartz TU, Ploegh HL. Exploring cellular biochemistry with nanobodies. J Biol Chem 2020; 295:15307-15327. [PMID: 32868455 PMCID: PMC7650250 DOI: 10.1074/jbc.rev120.012960] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 08/27/2020] [Indexed: 12/21/2022] Open
Abstract
Reagents that bind tightly and specifically to biomolecules of interest remain essential in the exploration of biology and in their ultimate application to medicine. Besides ligands for receptors of known specificity, agents commonly used for this purpose are monoclonal antibodies derived from mice, rabbits, and other animals. However, such antibodies can be expensive to produce, challenging to engineer, and are not necessarily stable in the context of the cellular cytoplasm, a reducing environment. Heavy chain-only antibodies, discovered in camelids, have been truncated to yield single-domain antibody fragments (VHHs or nanobodies) that overcome many of these shortcomings. Whereas they are known as crystallization chaperones for membrane proteins or as simple alternatives to conventional antibodies, nanobodies have been applied in settings where the use of standard antibodies or their derivatives would be impractical or impossible. We review recent examples in which the unique properties of nanobodies have been combined with complementary methods, such as chemical functionalization, to provide tools with unique and useful properties.
Collapse
Affiliation(s)
- Ross W Cheloha
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA
| | - Thibault J Harmand
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA
| | - Charlotte Wijne
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA
| | - Thomas U Schwartz
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Hidde L Ploegh
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
40
|
de Beer MA, Giepmans BNG. Nanobody-Based Probes for Subcellular Protein Identification and Visualization. Front Cell Neurosci 2020; 14:573278. [PMID: 33240044 PMCID: PMC7667270 DOI: 10.3389/fncel.2020.573278] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 10/05/2020] [Indexed: 12/14/2022] Open
Abstract
Understanding how building blocks of life contribute to physiology is greatly aided by protein identification and cellular localization. The two main labeling approaches developed over the past decades are labeling with antibodies such as immunoglobulin G (IgGs) or use of genetically encoded tags such as fluorescent proteins. However, IgGs are large proteins (150 kDa), which limits penetration depth and uncertainty of target position caused by up to ∼25 nm distance of the label created by the chosen targeting approach. Additionally, IgGs cannot be easily recombinantly modulated and engineered as part of fusion proteins because they consist of multiple independent translated chains. In the last decade single domain antigen binding proteins are being explored in bioscience as a tool in revealing molecular identity and localization to overcome limitations by IgGs. These nanobodies have several potential benefits over routine applications. Because of their small size (15 kDa), nanobodies better penetrate during labeling procedures and improve resolution. Moreover, nanobodies cDNA can easily be fused with other cDNA. Multidomain proteins can thus be easily engineered consisting of domains for targeting (nanobodies) and visualization by fluorescence microscopy (fluorescent proteins) or electron microscopy (based on certain enzymes). Additional modules for e.g., purification are also easily added. These nanobody-based probes can be applied in cells for live-cell endogenous protein detection or may be purified prior to use on molecules, cells or tissues. Here, we present the current state of nanobody-based probes and their implementation in microscopy, including pitfalls and potential future opportunities.
Collapse
Affiliation(s)
- Marit A de Beer
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Ben N G Giepmans
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
41
|
McGough IJ, Vecchia L, Bishop B, Malinauskas T, Beckett K, Joshi D, O'Reilly N, Siebold C, Jones EY, Vincent JP. Glypicans shield the Wnt lipid moiety to enable signalling at a distance. Nature 2020; 585:85-90. [PMID: 32699409 PMCID: PMC7610841 DOI: 10.1038/s41586-020-2498-z] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 04/23/2020] [Indexed: 12/14/2022]
Abstract
A relatively small number of proteins have been suggested to act as morphogens-signalling molecules that spread within tissues to organize tissue repair and the specification of cell fate during development. Among them are Wnt proteins, which carry a palmitoleate moiety that is essential for signalling activity1-3. How a hydrophobic lipoprotein can spread in the aqueous extracellular space is unknown. Several mechanisms, such as those involving lipoprotein particles, exosomes or a specific chaperone, have been proposed to overcome this so-called Wnt solubility problem4-6. Here we provide evidence against these models and show that the Wnt lipid is shielded by the core domain of a subclass of glypicans defined by the Dally-like protein (Dlp). Structural analysis shows that, in the presence of palmitoleoylated peptides, these glypicans change conformation to create a hydrophobic space. Thus, glypicans of the Dlp family protect the lipid of Wnt proteins from the aqueous environment and serve as a reservoir from which Wnt proteins can be handed over to signalling receptors.
Collapse
Affiliation(s)
| | - Luca Vecchia
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Benjamin Bishop
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Tomas Malinauskas
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | | | | | | | - Christian Siebold
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - E Yvonne Jones
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK.
| | | |
Collapse
|
42
|
Wang J, Dahmann C. Establishing compartment boundaries in Drosophila wing imaginal discs: An interplay between selector genes, signaling pathways and cell mechanics. Semin Cell Dev Biol 2020; 107:161-169. [PMID: 32732129 DOI: 10.1016/j.semcdb.2020.07.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 07/13/2020] [Accepted: 07/14/2020] [Indexed: 01/02/2023]
Abstract
The partitioning of cells into groups or 'compartments' separated by straight and sharp boundaries is important for tissue formation in animal development. Cells from neighboring compartments are characterized by distinct fates and functions and their continuous separation at compartment boundaries maintains proper tissue organization. Signaling across compartment boundaries can induce the local expression of morphogens that in turn direct growth and patterning of the surrounding cells. Compartment boundaries play therefore an important role in tissue development. Compartment boundaries were first identified in the early 1970s in the Drosophila wing. Here, we review the role of compartment boundaries in growth and patterning of the developing wing and then discuss the genetic and physical mechanisms underlying cell separation at compartment boundaries in this tissue.
Collapse
Affiliation(s)
- Jing Wang
- Institute of Genetics, Technische Universität Dresden, 01062 Dresden, Germany
| | - Christian Dahmann
- Institute of Genetics, Technische Universität Dresden, 01062 Dresden, Germany; Cluster of Excellence Physics of Life, Technische Universität Dresden, 01062 Dresden, Germany.
| |
Collapse
|
43
|
Zhang C, Ötjengerdes RM, Roewe J, Mejias R, Marschall ALJ. Applying Antibodies Inside Cells: Principles and Recent Advances in Neurobiology, Virology and Oncology. BioDrugs 2020; 34:435-462. [PMID: 32301049 PMCID: PMC7391400 DOI: 10.1007/s40259-020-00419-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
To interfere with cell function, many scientists rely on methods that target DNA or RNA due to the ease with which they can be applied. Proteins are usually the final executors of function but are targeted only indirectly by these methods. Recent advances in targeted degradation of proteins based on proteolysis-targeting chimaeras (PROTACs), ubiquibodies, deGradFP (degrade Green Fluorescent Protein) and other approaches have demonstrated the potential of interfering directly at the protein level for research and therapy. Proteins can be targeted directly and very specifically by antibodies, but using antibodies inside cells has so far been considered to be challenging. However, it is possible to deliver antibodies or other proteins into the cytosol using standard laboratory equipment. Physical methods such as electroporation have been demonstrated to be efficient and validated thoroughly over time. The expression of intracellular antibodies (intrabodies) inside cells is another way to interfere with intracellular targets at the protein level. Methodological strategies to target the inside of cells with antibodies, including delivered antibodies and expressed antibodies, as well as applications in the research areas of neurobiology, viral infections and oncology, are reviewed here. Antibodies have already been used to interfere with a wide range of intracellular targets. Disease-related targets included proteins associated with neurodegenerative diseases such as Parkinson's disease (α-synuclein), Alzheimer's disease (amyloid-β) or Huntington's disease (mutant huntingtin [mHtt]). The applications of intrabodies in the context of viral infections include targeting proteins associated with HIV (e.g. HIV1-TAT, Rev, Vif, gp41, gp120, gp160) and different oncoviruses such as human papillomavirus (HPV), hepatitis B virus (HBV), hepatitis C virus (HCV) and Epstein-Barr virus, and they have been used to interfere with various targets related to different processes in cancer, including oncogenic pathways, proliferation, cell cycle, apoptosis, metastasis, angiogenesis or neo-antigens (e.g. p53, human epidermal growth factor receptor-2 [HER2], signal transducer and activator of transcription 3 [STAT3], RAS-related RHO-GTPase B (RHOB), cortactin, vascular endothelial growth factor receptor 2 [VEGFR2], Ras, Bcr-Abl). Interfering at the protein level allows questions to be addressed that may remain unanswered using alternative methods. This review addresses why direct targeting of proteins allows unique insights, what is currently feasible in vitro, and how this relates to potential therapeutic applications.
Collapse
Affiliation(s)
- Congcong Zhang
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rina M Ötjengerdes
- Hannover Medical School (MHH), Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Julian Roewe
- German Cancer Consortium (DKTK) Clinical Cooperation Unit (CCU) Neuroimmunology and Brain TumorImmunology (D170), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rebeca Mejias
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Andrea L J Marschall
- Technische Universität Braunschweig, Institute of Biochemistry, Biotechnology and Bioinformatics, Brunswick, Germany.
| |
Collapse
|
44
|
Barrio L, Milán M. Regulation of Anisotropic Tissue Growth by Two Orthogonal Signaling Centers. Dev Cell 2020; 52:659-672.e3. [PMID: 32084357 DOI: 10.1016/j.devcel.2020.01.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 11/15/2019] [Accepted: 01/21/2020] [Indexed: 11/15/2022]
Abstract
The Drosophila wing has served as a paradigm to mechanistically characterize the role of morphogens in patterning and growth. Wingless (Wg) and Decapentaplegic (Dpp) are expressed in two orthogonal signaling centers, and their gradients organize patterning by regulating the expression of well-defined target genes. By contrast, graded activity of these morphogens is not an absolute requirement for wing growth. Despite their permissive role in regulating growth, here we show that Wg and Dpp are utilized in a non-interchangeable manner by the two existing orthogonal signaling centers to promote preferential growth along the two different axes of the developing wing. Our data indicate that these morphogens promote anisotropic growth by making use of distinct and non-interchangeable molecular mechanisms. Whereas Dpp drives growth along the anterior-posterior axis by maintaining Brinker levels below a growth-repressing threshold, Wg exerts its action along the proximal-distal axis through a double repression mechanism involving T cell factor (TCF).
Collapse
Affiliation(s)
- Lara Barrio
- Institute for Research in Biomedicine, The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028 Barcelona, Spain
| | - Marco Milán
- Institute for Research in Biomedicine, The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028 Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats, Pg. Lluís Companys 23, 08010 Barcelona, Spain.
| |
Collapse
|
45
|
Gou J, Stotsky JA, Othmer HG. Growth control in the Drosophila wing disk. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2020; 12:e1478. [PMID: 31917525 DOI: 10.1002/wsbm.1478] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 12/02/2019] [Accepted: 12/17/2019] [Indexed: 12/16/2022]
Abstract
The regulation of size and shape is a fundamental requirement of biological development and has been a subject of scientific study for centuries, but we still lack an understanding of how organisms know when to stop growing. Imaginal wing disks of the fruit fly Drosophila melanogaster, which are precursors of the adult wings, are an archetypal tissue for studying growth control. The growth of the disks is dependent on many inter- and intra-organ factors such as morphogens, mechanical forces, nutrient levels, and hormones that influence gene expression and cell growth. Extracellular signals are transduced into gene-control signals via complex signal transduction networks, and since cells typically receive many different signals, a mechanism for integrating the signals is needed. Our understanding of the effect of morphogens on tissue-level growth regulation via individual pathways has increased significantly in the last half century, but our understanding of how multiple biochemical and mechanical signals are integrated to determine whether or not a cell decides to divide is still rudimentary. Numerous fundamental questions are involved in understanding the decision-making process, and here we review the major biochemical and mechanical pathways involved in disk development with a view toward providing a basis for beginning to understand how multiple signals can be integrated at the cell level, and how this translates into growth control at the level of the imaginal disk. This article is categorized under: Analytical and Computational Methods > Computational Methods Biological Mechanisms > Cell Signaling Models of Systems Properties and Processes > Cellular Models.
Collapse
Affiliation(s)
- Jia Gou
- School of Mathematics, University of Minnesota, Minneapolis, Minnesota
| | - Jay A Stotsky
- School of Mathematics, University of Minnesota, Minneapolis, Minnesota
| | - Hans G Othmer
- School of Mathematics, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
46
|
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: 5.4] [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
|
47
|
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: 2.4] [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
|
48
|
Deng M, Wang Y, Zhang L, Yang Y, Huang S, Wang J, Ge H, Ishibashi T, Yan Y. Single cell transcriptomic landscapes of pattern formation, proliferation and growth in Drosophila wing imaginal discs. Development 2019; 146:dev.179754. [PMID: 31455604 DOI: 10.1242/dev.179754] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 08/20/2019] [Indexed: 12/13/2022]
Abstract
Organ formation relies on the orchestration of pattern formation, proliferation and growth during development. How these processes are integrated at the individual cell level remains unclear. In the past decades, studies using Drosophila wing imaginal discs as a model system have provided valuable insights into pattern formation, growth control and regeneration. Here, we provide single cell transcriptomic landscapes of pattern formation, proliferation and growth of wing imaginal discs. We found that patterning information is robustly maintained in the single cell transcriptomic data and can provide reference matrices for computationally mapping single cells into discrete spatial domains. Assignment of wing disc single cells to spatial subregions facilitates examination of patterning refinement processes. We also clustered single cells into different proliferation and growth states and evaluated the correlation between cell proliferation/growth states and spatial patterning. Furthermore, single cell transcriptomic analyses allowed us to quantitatively examine disturbances of differentiation, proliferation and growth in a well-established tumor model. We provide a database to explore these datasets at http://drosophilayanlab-virtual-wingdisc.ust.hk:3838/v2/This article has an associated 'The people behind the papers' interview.
Collapse
Affiliation(s)
- Mingxi Deng
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China.,Center of Systems Biology and Human Health, School of Science and Institute for Advanced Study, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Ying Wang
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China.,Center of Systems Biology and Human Health, School of Science and Institute for Advanced Study, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Lina Zhang
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China.,Center of Systems Biology and Human Health, School of Science and Institute for Advanced Study, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Yang Yang
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China.,Center of Systems Biology and Human Health, School of Science and Institute for Advanced Study, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Shengshuo Huang
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China.,Center of Systems Biology and Human Health, School of Science and Institute for Advanced Study, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China.,Department of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Jiguang Wang
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China.,Center of Systems Biology and Human Health, School of Science and Institute for Advanced Study, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China.,Department of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Hao Ge
- Beijing International Center for Mathematical Research and Biomedical Pioneering Innovation Center, Peking University, Peking, China 100871
| | - Toyotaka Ishibashi
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Yan Yan
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China .,Center of Systems Biology and Human Health, School of Science and Institute for Advanced Study, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| |
Collapse
|
49
|
Aguilar G, Vigano MA, Affolter M, Matsuda S. Reflections on the use of protein binders to study protein function in developmental biology. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2019; 8:e356. [PMID: 31265212 PMCID: PMC6851689 DOI: 10.1002/wdev.356] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 06/03/2019] [Accepted: 06/04/2019] [Indexed: 02/01/2023]
Abstract
Studies in the field of developmental biology aim to unravel how a fertilized egg develops into an adult organism and how proteins and other macromolecules work together during this process. With regard to protein function, most of the developmental studies have used genetic and RNA interference approaches, combined with biochemical analyses, to reach this goal. However, there always remains much room for interpretation on how a given protein functions, because proteins work together with many other molecules in complex regulatory networks and it is not easy to reveal the function of one given protein without affecting the networks. Likewise, it has remained difficult to experimentally challenge and/or validate the proposed concepts derived from mutant analyses without tools that directly manipulate protein function in a predictable manner. Recently, synthetic tools based on protein binders such as scFvs, nanobodies, DARPins, and others have been applied in developmental biology to directly manipulate target proteins in a predicted manner. Although such tools would have a great impact in filling the gap of knowledge between mutant phenotypes and protein functions, careful investigations are required when applying functionalized protein binders to fundamental questions in developmental biology. In this review, we first summarize how protein binders have been used in the field, and then reflect on possible guidelines for applying such tools to study protein functions in developmental biology. This article is categorized under: Technologies > Analysis of Proteins Establishment of Spatial and Temporal Patterns > Gradients Invertebrate Organogenesis > Flies.
Collapse
|
50
|
Zi Z. Molecular Engineering of the TGF-β Signaling Pathway. J Mol Biol 2019; 431:2644-2654. [PMID: 31121181 DOI: 10.1016/j.jmb.2019.05.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 05/05/2019] [Accepted: 05/13/2019] [Indexed: 12/19/2022]
Abstract
Transforming growth factor beta (TGF-β) is an important growth factor that plays essential roles in regulating tissue development and homeostasis. Dysfunction of TGF-β signaling is a hallmark of many human diseases. Therefore, targeting TGF-β signaling presents broad therapeutic potential. Since the discovery of the TGF-β ligand, a collection of engineered signaling proteins have been developed to probe and manipulate TGF-β signaling responses. In this review, we highlight recent progress in the engineering of TGF-β signaling for different applications and discuss how molecular engineering approaches can advance our understanding of this important pathway. In addition, we provide a future outlook on the opportunities and challenges in the engineering of the TGF-β signaling pathway from a quantitative perspective.
Collapse
Affiliation(s)
- Zhike Zi
- Otto-Warburg Laboratory, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany.
| |
Collapse
|