1
|
Le PM, Quinn JM, Flozak AS, Steffeck AWT, Huang CF, Gottardi CJ. α-catenin phosphorylation is elevated during mitosis to resist apical rounding and epithelial barrier leak. Biol Open 2025; 14:bio061726. [PMID: 39782767 PMCID: PMC11744050 DOI: 10.1242/bio.061726] [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: 09/06/2024] [Accepted: 12/03/2024] [Indexed: 01/12/2025] Open
Abstract
Epithelial cell cohesion and barrier function critically depend on α-catenin, an actin-binding protein and essential constituent of cadherin-catenin-based adherens junctions. α-catenin undergoes actomyosin force-dependent unfolding of both actin-binding and middle domains to strongly engage actin filaments and its various effectors; this mechanosensitivity is critical for adherens junction function. We previously showed that α-catenin is highly phosphorylated in an unstructured region that links the mechanosensitive middle and actin-binding domains (known as the P-linker region), but the cellular processes that promote α-catenin phosphorylation have remained elusive. Here, we leverage a previously published phospho-proteomic data set to show that the α-catenin P-linker region is maximally phosphorylated during mitosis. By reconstituting α-catenin CRISPR knockout MDCK cells with wild-type, phospho-mutant and phospho-mimic forms of α-catenin, we show that full phosphorylation restrains mitotic cell rounding in the apical direction, strengthening the interactions between dividing and non-dividing neighbors to limit epithelial barrier leak. As the major scaffold components of adherens junctions, tight junctions and desmosomes are also differentially phosphorylated during mitosis, we reason that epithelial cell division may be a tractable system to understand how junction complexes are coordinately regulated to sustain barrier function under tension-generating morphogenetic processes.
Collapse
Affiliation(s)
- Phuong M. Le
- Department of Pulmonary Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jeanne M. Quinn
- Department of Pulmonary Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Driskill Graduate Program in the Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Annette S. Flozak
- Department of Pulmonary Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Adam W. T. Steffeck
- Driskill Graduate Program in the Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Che-Fan Huang
- Proteomics Center of Excellence, Northwestern University, Evanston, IL 60208, USA
| | - Cara J. Gottardi
- Department of Pulmonary Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Cell & Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
2
|
Gonçalves M, Lopes C, Alégot H, Osswald M, Bosveld F, Ramos C, Richard G, Bellaiche Y, Mirouse V, Morais-de-Sá E. The Dystrophin-Dystroglycan complex ensures cytokinesis efficiency in Drosophila epithelia. EMBO Rep 2025; 26:307-328. [PMID: 39548266 PMCID: PMC11772804 DOI: 10.1038/s44319-024-00319-y] [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: 03/04/2024] [Revised: 10/21/2024] [Accepted: 10/24/2024] [Indexed: 11/17/2024] Open
Abstract
Cytokinesis physically separates daughter cells at the end of cell division. This step is particularly challenging for epithelial cells, which are connected to their neighbors and to the extracellular matrix by transmembrane protein complexes. To systematically evaluate the impact of the cell adhesion machinery on epithelial cytokinesis efficiency, we performed an RNAi-based modifier screen in the Drosophila follicular epithelium. Strikingly, this unveiled adhesion molecules and transmembrane receptors that facilitate cytokinesis completion. Among these is Dystroglycan, which connects the extracellular matrix to the cytoskeleton via Dystrophin. Live imaging revealed that Dystrophin and Dystroglycan become enriched in the ingressing membrane, below the cytokinetic ring, during and after ring constriction. Using multiple alleles, including Dystrophin isoform-specific mutants, we show that Dystrophin/Dystroglycan localization is linked with unanticipated roles in regulating cytokinetic ring contraction and in preventing membrane regression during the abscission period. Altogether, we provide evidence that, rather than opposing cytokinesis completion, the machinery involved in cell-cell and cell-matrix interactions has also evolved functions to ensure cytokinesis efficiency in epithelial tissues.
Collapse
Affiliation(s)
- Margarida Gonçalves
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135, Porto, Portugal
- Programa Doutoral em Biologia Molecular e Celular (MCBiology), Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Catarina Lopes
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135, Porto, Portugal
| | - Hervé Alégot
- Université Clermont Auvergne - iGReD (Institute of Genetics, Reproduction and Development), UMR CNRS 6293 - INSERM U1103, Faculté de Médecine, Clermont-Ferrand, France
| | - Mariana Osswald
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135, Porto, Portugal
| | - Floris Bosveld
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005, Paris, France
| | - Carolina Ramos
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135, Porto, Portugal
| | - Graziella Richard
- Université Clermont Auvergne - iGReD (Institute of Genetics, Reproduction and Development), UMR CNRS 6293 - INSERM U1103, Faculté de Médecine, Clermont-Ferrand, France
| | - Yohanns Bellaiche
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005, Paris, France
| | - Vincent Mirouse
- Université Clermont Auvergne - iGReD (Institute of Genetics, Reproduction and Development), UMR CNRS 6293 - INSERM U1103, Faculté de Médecine, Clermont-Ferrand, France
| | - Eurico Morais-de-Sá
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135, Porto, Portugal.
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135, Porto, Portugal.
| |
Collapse
|
3
|
Walther N, Anantakrishnan S, Graham TGW, Dailey GM, Tjian R, Darzacq X. Automated live-cell single-molecule tracking in enteroid monolayers reveals transcription factor dynamics probing lineage-determining function. Cell Rep 2024; 43:114914. [PMID: 39480809 DOI: 10.1016/j.celrep.2024.114914] [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: 04/22/2024] [Revised: 08/29/2024] [Accepted: 10/10/2024] [Indexed: 11/02/2024] Open
Abstract
Lineage transcription factors (TFs) provide one regulatory level of differentiation crucial for the generation and maintenance of healthy tissues. To probe TF function by measuring their dynamics during adult intestinal homeostasis, we established HILO-illumination-based live-cell single-molecule tracking (SMT) in mouse small intestinal enteroid monolayers recapitulating tissue differentiation hierarchies in vitro. To increase the throughput, capture cellular features, and correlate morphological characteristics with diffusion parameters, we developed an automated imaging and analysis pipeline, broadly applicable to two-dimensional culture systems. Studying two absorptive lineage-determining TFs, we found an expression level-independent contrasting diffusive behavior: while Hes1, key determinant of absorptive lineage commitment, displays a large cell-to-cell variability and an average fraction of DNA-bound molecules of ∼32%, Hnf4g, conferring enterocyte identity, exhibits more uniform dynamics and a bound fraction of ∼56%. Our results suggest that TF diffusive behavior could indicate the progression of differentiation and modulate early versus late differentiation within a lineage.
Collapse
Affiliation(s)
- Nike Walther
- Department of Molecular and Cell Biology, Li Ka Shing Center for Biomedical and Health Sciences, California Institute for Regenerative Medicine (CIRM) Center of Excellence, University of California, Berkeley, Berkeley, CA 94720, USA.
| | - Sathvik Anantakrishnan
- Department of Molecular and Cell Biology, Li Ka Shing Center for Biomedical and Health Sciences, California Institute for Regenerative Medicine (CIRM) Center of Excellence, University of California, Berkeley, Berkeley, CA 94720, USA; Biophysics Graduate Group, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Thomas G W Graham
- Department of Molecular and Cell Biology, Li Ka Shing Center for Biomedical and Health Sciences, California Institute for Regenerative Medicine (CIRM) Center of Excellence, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Gina M Dailey
- Department of Molecular and Cell Biology, Li Ka Shing Center for Biomedical and Health Sciences, California Institute for Regenerative Medicine (CIRM) Center of Excellence, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Robert Tjian
- Department of Molecular and Cell Biology, Li Ka Shing Center for Biomedical and Health Sciences, California Institute for Regenerative Medicine (CIRM) Center of Excellence, University of California, Berkeley, Berkeley, CA 94720, USA; Howard Hughes Medical Institute, Berkeley, CA 94720, USA.
| | - Xavier Darzacq
- Department of Molecular and Cell Biology, Li Ka Shing Center for Biomedical and Health Sciences, California Institute for Regenerative Medicine (CIRM) Center of Excellence, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
4
|
Graham AJ, Khoo MW, Srivastava V, Viragova S, Kim H, Parekh K, Hennick KM, Bird M, Goldhammer N, Yu JZ, Morley CD, Lebel P, Kumar S, Rosenbluth JM, Nowakowski TJ, Klein O, Gómez-Sjöberg R, Gartner ZJ. MAGIC matrices: freeform bioprinting materials to support complex and reproducible organoid morphogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.01.578324. [PMID: 38370663 PMCID: PMC10871257 DOI: 10.1101/2024.02.01.578324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Organoids are powerful models of tissue physiology, yet their applications remain limited due to their relatively simple morphology and high organoid-to-organoid structural variability. To address these limitations we developed a soft, composite yield-stress extracellular matrix that supports optimal organoid morphogenesis following freeform 3D bioprinting of cell slurries at tissue-like densities. The material is designed with two temperature regimes: at 4 °C it exhibits reversible yield-stress behavior to support long printing times without compromising cell viability. When transferred to cell culture at 37 °C, the material cross-links and exhibits similar viscoelasticity and plasticity to basement membrane extracts such as Matrigel. We first characterize the rheological properties of MAGIC matrices that optimize organoid morphogenesis, including low stiffness and high stress relaxation. Next, we combine this material with a custom piezoelectric printhead that allows more reproducible and robust self-organization from uniform and spatially organized tissue "seeds." We apply MAGIC matrix bioprinting for high-throughput generation of intestinal, mammary, vascular, salivary gland, and brain organoid arrays that are structurally similar to those grown in pure Matrigel, but exhibit dramatically improved homogeneity in organoid size, shape, maturation time, and efficiency of morphogenesis. The flexibility of this method and material enabled fabrication of fully 3D microphysiological systems, including perfusable organoid tubes that experience cyclic 3D strain in response to pressurization. Furthermore, the reproducibility of organoid structure increased the statistical power of a drug response assay by up to 8 orders-of-magnitude for a given number of comparisons. Combined, these advances lay the foundation for the efficient fabrication of complex tissue morphologies by canalizing their self-organization in both space and time.
Collapse
Affiliation(s)
- Austin J. Graham
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA
- Chan Zuckerberg Biohub SF, San Francisco, CA
| | | | - Vasudha Srivastava
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA
| | - Sara Viragova
- Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA
| | - Honesty Kim
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA
- Chan Zuckerberg Biohub SF, San Francisco, CA
| | - Kavita Parekh
- Department of Bioengineering, University of California Berkeley, Berkeley, CA
| | - Kelsey M. Hennick
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Malia Bird
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA
| | - Nadine Goldhammer
- Department of Medicine, University of California San Francisco, San Francisco, CA
| | - Jie Zeng Yu
- Department of Medicine, University of California San Francisco, San Francisco, CA
| | - Cameron D. Morley
- Department of Bioengineering, University of California Berkeley, Berkeley, CA
| | - Paul Lebel
- Chan Zuckerberg Biohub SF, San Francisco, CA
| | - Sanjay Kumar
- Department of Bioengineering, University of California Berkeley, Berkeley, CA
- Department of Chemical and Biomolecular Engineering, University of California Berkeley, Berkeley, CA
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA
| | - Jennifer M. Rosenbluth
- Chan Zuckerberg Biohub SF, San Francisco, CA
- Department of Medicine, University of California San Francisco, San Francisco, CA
| | - Tomasz J. Nowakowski
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Ophir Klein
- Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA
- Department of Pediatrics, Cedars-Sinai Guerin Children’s, Los Angeles, CA
| | | | - Zev J. Gartner
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA
- Chan Zuckerberg Biohub SF, San Francisco, CA
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA
- Center for Cellular Construction, University of California San Francisco, San Francisco, CA
| |
Collapse
|
5
|
Lee MJ, Lee J, Ha J, Kim G, Kim HJ, Lee S, Koo BK, Park Y. Long-term three-dimensional high-resolution imaging of live unlabeled small intestinal organoids via low-coherence holotomography. Exp Mol Med 2024; 56:2162-2170. [PMID: 39349827 PMCID: PMC11541879 DOI: 10.1038/s12276-024-01312-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 10/03/2024] Open
Abstract
Organoids, which are miniature in vitro versions of organs, possess significant potential for studying human diseases and elucidating their underlying mechanisms. Live imaging techniques play a crucial role in organoid research and contribute to elucidating the complex structure and dynamic biological phenomena of organoids. However, live, unlabeled high-resolution imaging of native organoids is challenging, primarily owing to the complexities of sample handling and optical scattering inherent in three-dimensional (3D) structures. Additionally, conventional imaging methods fail to capture the real-time dynamic processes of growing organoids. In this study, we introduce low-coherence holotomography as an advanced, label-free, quantitative imaging modality designed to overcome several technical obstacles for long-term live imaging of 3D organoids. We demonstrate the efficacy of low-coherence holotomography by capturing high-resolution morphological details and dynamic activities within mouse small intestinal organoids at subcellular resolution. Moreover, our approach facilitates the distinction between viable and nonviable organoids, significantly enhancing its utility in organoid-based research. This advancement underscores the critical role of live imaging in organoid studies, offering a more comprehensive understanding of these complex systems.
Collapse
Affiliation(s)
- Mahn Jae Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
- KAIST Institute for Health Science and Technology, Daejeon, 34141, Republic of Korea
| | | | - Jeongmin Ha
- Center for Genome Engineering, Institute for Basic Science, Daejeon, 34126, Republic of Korea
| | - Geon Kim
- KAIST Institute for Health Science and Technology, Daejeon, 34141, Republic of Korea
- Department of Physics, KAIST, Daejeon, 34141, Republic of Korea
| | | | - Sumin Lee
- Tomocube Inc., Daejeon, Republic of Korea
| | - Bon-Kyoung Koo
- Center for Genome Engineering, Institute for Basic Science, Daejeon, 34126, Republic of Korea.
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea.
| | - YongKeun Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.
- KAIST Institute for Health Science and Technology, Daejeon, 34141, Republic of Korea.
- Tomocube Inc., Daejeon, Republic of Korea.
- Department of Physics, KAIST, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
6
|
Le PM, Quinn JM, Flozak AS, Steffeck AWT, Huang CF, Gottardi CJ. α -catenin phosphorylation is elevated during mitosis to resist apical rounding and epithelial barrier leak. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.06.611639. [PMID: 39282345 PMCID: PMC11398478 DOI: 10.1101/2024.09.06.611639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/29/2024]
Abstract
Epithelial cell cohesion and barrier function critically depend on α -catenin, an actin-binding protein and essential constituent of cadherin-catenin-based adherens junctions. α -catenin undergoes actomyosin force-dependent unfolding of both actin-binding and middle domains to strongly engage actin filaments and its various effectors, where this mechanosensitivity is critical for adherens junction function. We previously showed that α -catenin is highly phosphorylated in an unstructured region that links mechanosensitive middle- and actin-binding domains (known as the P-linker region), but the cellular processes that promote α -catenin phosphorylation have remained elusive. Here, we leverage a previously published phosphor-proteomic data set to show that the α -catenin P-linker region is maximally phosphorylated during mitosis. By reconstituting α -catenin Crispr KO MDCK with wild-type, phospho- mutant and mimic forms of α -catenin, we show that full phosphorylation restrains mitotic cell rounding in the apical direction, strengthening interactions between dividing and non-dividing neighbors to limit epithelial barrier leak. Since major scaffold components of adherens junctions, tight junctions and desmosomes are also differentially phosphorylated during mitosis, we reason that epithelial cell division may be a tractable system to understand how junction complexes are coordinately regulated to sustain barrier function under tension-generating morphogenetic processes.
Collapse
Affiliation(s)
- Phuong M. Le
- Department of Pulmonary Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Jeanne M. Quinn
- Department of Pulmonary Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Driskill Graduate Program in the Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Annette S. Flozak
- Department of Pulmonary Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Adam WT Steffeck
- Driskill Graduate Program in the Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Che-Fan Huang
- Proteomics Center of Excellence, Northwestern University, Evanston, IL 60208, USA
| | - Cara J. Gottardi
- Department of Pulmonary Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Cell & Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| |
Collapse
|
7
|
Cong J, Wu J, Fang Y, Wang J, Kong X, Wang L, Duan Z. Application of organoid technology in the human health risk assessment of microplastics: A review of progresses and challenges. ENVIRONMENT INTERNATIONAL 2024; 188:108744. [PMID: 38761429 DOI: 10.1016/j.envint.2024.108744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/16/2024] [Accepted: 05/10/2024] [Indexed: 05/20/2024]
Abstract
Microplastic (MP) pollution has become a global environmental issue, and increasing concern has been raised about its impact on human health. Current studies on the toxic effects and mechanisms of MPs have mostly been conducted in animal models or in vitro cell cultures, which have limitations regarding inter-species differences or stimulation of cellular functions. Organoid technology derived from human pluripotent or adult stem cells has broader prospects for predicting the potential health risks of MPs to humans. Herein, we reviewed the current application advancements and opportunities for different organoids, including brain, retinal, intestinal, liver, and lung organoids, to assess the human health risks of MPs. Organoid techniques accurately simulate the complex processes of MPs and reflect phenotypes related to diseases caused by MPs such as liver fibrosis, neurodegeneration, impaired intestinal barrier and cardiac hypertrophy. Future perspectives were also proposed for technological innovation in human risk assessment of MPs using organoids, including extending the lifespan of organoids to assess the chronic toxicity of MPs, and reconstructing multi-organ interactions to explore their potential in studying the microbiome-gut-brainaxis effect of MPs.
Collapse
Affiliation(s)
- Jiaoyue Cong
- School of Environmental Science and Safety Engineering, Tianjin University of Technology, Tianjin 300384, China
| | - Jin Wu
- Tianjin Institute of Environment and Operational Medicine, Tianjin 300050, China
| | - Yanjun Fang
- Tianjin Institute of Environment and Operational Medicine, Tianjin 300050, China
| | - Jing Wang
- School of Environmental Science and Safety Engineering, Tianjin University of Technology, Tianjin 300384, China
| | - Xiaoyan Kong
- School of Environmental Science and Safety Engineering, Tianjin University of Technology, Tianjin 300384, China
| | - Lei Wang
- College of Environmental Science and Engineering, Nankai University, Tianjin 300071, China
| | - Zhenghua Duan
- School of Environmental Science and Safety Engineering, Tianjin University of Technology, Tianjin 300384, China.
| |
Collapse
|
8
|
Finegan TM, Cammarota C, Mendoza Andrade O, Garoutte AM, Bergstralh DT. Fas2EB112: a tale of two chromosomes. G3 (BETHESDA, MD.) 2024; 14:jkae047. [PMID: 38447284 PMCID: PMC11075550 DOI: 10.1093/g3journal/jkae047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 02/29/2024] [Accepted: 02/29/2024] [Indexed: 03/08/2024]
Abstract
The cell-cell adhesion molecule Fasciclin II (Fas2) has long been studied for its evolutionarily conserved role in axon guidance. It is also expressed in the follicular epithelium, where together with a similar protein, Neuroglian (Nrg), it helps to drive the reintegration of cells born out of the tissue plane. Remarkably, one Fas2 protein null allele, Fas2G0336, demonstrates a mild reintegration phenotype, whereas work with the classic null allele Fas2EB112 showed more severe epithelial disorganization. These observations raise the question of which allele (if either) causes a bona fide loss of Fas2 protein function. The problem is not only relevant to reintegration but fundamentally important to understanding what this protein does and how it works: Fas2EB112 has been used in at least 37 research articles, and Fas2G0336 in at least three. An obvious solution is that one of the two chromosomes carries a modifier that either suppresses (Fas2G0336) or enhances (Fas2EB112) phenotypic severity. We find not only the latter to be the case, but identify the enhancing mutation as Nrg14, also a classic null allele.
Collapse
Affiliation(s)
- Tara M Finegan
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
- Division of Biological Sciences, University of Missouri, Columbia, MO 65203, USA
| | - Christian Cammarota
- Department of Physics and Astronomy, University of Rochester, Rochester, NY 14627, USA
| | | | - Audrey M Garoutte
- Division of Biological Sciences, University of Missouri, Columbia, MO 65203, USA
| | - Dan T Bergstralh
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
- Division of Biological Sciences, University of Missouri, Columbia, MO 65203, USA
- Department of Physics and Astronomy, University of Rochester, Rochester, NY 14627, USA
| |
Collapse
|
9
|
Ko J, Hyung S, Cheong S, Chung Y, Li Jeon N. Revealing the clinical potential of high-resolution organoids. Adv Drug Deliv Rev 2024; 207:115202. [PMID: 38336091 DOI: 10.1016/j.addr.2024.115202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 01/01/2024] [Accepted: 02/02/2024] [Indexed: 02/12/2024]
Abstract
The symbiotic interplay of organoid technology and advanced imaging strategies yields innovative breakthroughs in research and clinical applications. Organoids, intricate three-dimensional cell cultures derived from pluripotent or adult stem/progenitor cells, have emerged as potent tools for in vitro modeling, reflecting in vivo organs and advancing our grasp of tissue physiology and disease. Concurrently, advanced imaging technologies such as confocal, light-sheet, and two-photon microscopy ignite fresh explorations, uncovering rich organoid information. Combined with advanced imaging technologies and the power of artificial intelligence, organoids provide new insights that bridge experimental models and real-world clinical scenarios. This review explores exemplary research that embodies this technological synergy and how organoids reshape personalized medicine and therapeutics.
Collapse
Affiliation(s)
- Jihoon Ko
- Department of BioNano Technology, Gachon University, Gyeonggi 13120, Republic of Korea
| | - Sujin Hyung
- Precision Medicine Research Institute, Samsung Medical Center, Seoul 08826, Republic of Korea; Division of Hematology-Oncology, Department of Medicine, Sungkyunkwan University, Samsung Medical Center, Seoul 08826, Republic of Korea
| | - Sunghun Cheong
- Interdisciplinary Program in Bioengineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Yoojin Chung
- Division of Computer Engineering, Hankuk University of Foreign Studies, Yongin 17035, Republic of Korea
| | - Noo Li Jeon
- Interdisciplinary Program in Bioengineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea; Department of Mechanical Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea; Institute of Advanced Machines and Design, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea; Qureator, Inc., San Diego, CA, USA.
| |
Collapse
|
10
|
Kim C, Robitaille M, Christodoulides J, Ng Y, Raphael M, Kang W. Hs27 fibroblast response to contact guidance cues. Sci Rep 2023; 13:21691. [PMID: 38066191 PMCID: PMC10709656 DOI: 10.1038/s41598-023-48913-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 12/01/2023] [Indexed: 12/18/2023] Open
Abstract
Contact guidance is the phenomena of how cells respond to the topography of their external environment. The morphological and dynamic cell responses are strongly influenced by topographic features such as lateral and vertical dimensions, namely, ridge and groove widths and groove depth ([Formula: see text], respectively). However, experimental studies that independently quantify the effect of the individual dimensions as well as their coupling on cellular function are still limited. In this work, we perform extensive parametric studies in the dimensional space-well beyond the previously studied range in the literature-to explore topographical effects on morphology and migration of Hs27 fibroblasts via static and dynamic analyses of live cell images. Our static analysis reveals that the [Formula: see text] is most significant, followed by the [Formula: see text]. The fibroblasts appear to be more elongated and aligned in the groove direction as the [Formula: see text] increases, but their trend changes after 725 nm. Interestingly, the cell shape and alignment show a very strong correlation regardless of [Formula: see text]. Our dynamic analysis confirms that directional cell migration is also strongly influenced by the [Formula: see text], while the effect of the [Formula: see text] and [Formula: see text] is statistically insignificant. Directional cell migration, as observed in the static cell behavior, shows the statistically significant transition when the [Formula: see text] is 725 nm, showing the intimate links between cell morphology and migration. We propose possible scenarios to offer mechanistic explanations of the observed cell behavior.
Collapse
Affiliation(s)
- C Kim
- Mechanical Engineering, School for Engineering of Matter, Transport and Energy, Arizona State University, Tempe, AZ, 85281, USA
| | - M Robitaille
- US Naval Research Laboratory, Washington, DC, 20375, USA
| | | | - Y Ng
- Mechanical Engineering, School for Engineering of Matter, Transport and Energy, Arizona State University, Tempe, AZ, 85281, USA
| | - M Raphael
- US Naval Research Laboratory, Washington, DC, 20375, USA
| | - W Kang
- Mechanical Engineering, School for Engineering of Matter, Transport and Energy, Arizona State University, Tempe, AZ, 85281, USA.
| |
Collapse
|
11
|
Sun W, Liang X, Lei J, Jiang C, Sheng D, Zhang S, Liu X, Chen H. Regulating cell behavior via regional patterned distribution of heparin-like polymers. BIOMATERIALS ADVANCES 2023; 154:213664. [PMID: 37866231 DOI: 10.1016/j.bioadv.2023.213664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 10/11/2023] [Accepted: 10/15/2023] [Indexed: 10/24/2023]
Abstract
Molecular patterning on biomaterial surfaces is an effective strategy to regulate biomaterial properties. Among the specific molecules, due to their biological functions, such as regulating cell behavior, heparin-like polymers (HLPs) have attracted much attention. In this study, HLP-distributed regional patterned surfaces (300 μm diameter circular array) were prepared by the combination of visible light-induced graft polymerization, transfer imprinting, and self-assembly to regulate the behavior of human umbilical vein endothelial cells (HUVECs) and human umbilical vein smooth muscle cells (HUVSMCs). The introduction of the regional pattern on HLP-modified surfaces enhanced the promotion effect of sulfonate-containing polymer (pSS) and sulfonate-, and glyco-containing copolymer (pS-co-pM), and slightly weakened the inhibition effect of glyco-containing polymer (pMAG) on the growth of HUVECs and HUVSMCs. Compared with flat surfaces, it was found that the unmodified regional patterned surfaces inhibit the spreading of HUVECs and HUVSMCs, while significantly promoting the spreading of HUVECs and HUVSMCs on all the HLP-distributed regional patterned surfaces. The patterned surface modified with pS-co-pM had the highest average spread area of HUVECs (∼10,554 μm2), which was 193 % higher than that of the unmodified flat surface. This trend was somewhat related to surface VEGF adsorption. The combination of regional divisive patterns and different HLP distributions enriched the potential of further exploring the influences of HLP chemical distributions and complex surface environments on cell-material interactions.
Collapse
Affiliation(s)
- Wei Sun
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren-Ai Road, Suzhou 215123, PR China
| | - Xinyi Liang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren-Ai Road, Suzhou 215123, PR China
| | - Jiao Lei
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren-Ai Road, Suzhou 215123, PR China
| | - Chi Jiang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren-Ai Road, Suzhou 215123, PR China
| | - Denghai Sheng
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren-Ai Road, Suzhou 215123, PR China
| | - Sulei Zhang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren-Ai Road, Suzhou 215123, PR China
| | - Xiaoli Liu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren-Ai Road, Suzhou 215123, PR China.
| | - Hong Chen
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren-Ai Road, Suzhou 215123, PR China
| |
Collapse
|
12
|
Ozdemir M, Ozdil B, Abdikan CSA, Erisik D, Yesin TK, Avci CB, Kurkutçu Y, Guler G, Aktug H. HDAC9/p300/F-actin immunoexpression and migration analysis for malignant melanoma stem cell. Pathol Res Pract 2023; 250:154829. [PMID: 37748211 DOI: 10.1016/j.prp.2023.154829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/08/2023] [Accepted: 09/21/2023] [Indexed: 09/27/2023]
Abstract
Melanoma is an aggressive tumor with a poor prognosis that worsens in the metastatic phase. Distruptions of epigenetic mechanisms is known to effect cancer stem cells (CSCs) activity. Malignant melanoma (MM) progression may be promoted by changes in the genetic structure of CSC. Thus, treatments that target epigenetic modifications could be a promising weapon, especially in melanoma. Here, we compared p300, HDAC9, and F-actin proteins in melanoma CSCs (CD133+), non-CSCs (CD133-) and CHL-1 cell line, as well as cell migration and division rates. At 4 and 6 h, P300 protein levels in CHL-1 and CD133 + were remarkably similar, and the CD133- showed increases in expression levels as the incubation period lengthened. HDAC9 protein intensity decreased in CHL-1, increased in the CD133-, and remained relatively unchanged in the CD133+ as the incubation period lengthened. The mean value of F-actin expression level increased in all cell group with time, when the highest increase observed in CHL-1. In conclusion, our studies contribute to the management of metastatic diseases in the future and offer new insight into the molecular basis of the initiation and progression of MM.
Collapse
Affiliation(s)
- Merve Ozdemir
- Department of Histology and Embryology, Faculty of Medicine, Ege University, Izmir 35100, Turkey
| | - Berrin Ozdil
- Department of Histology and Embryology, Faculty of Medicine, Ege University, Izmir 35100, Turkey; Department of Histology and Embryology, Faculty of Medicine, Suleyman Demirel University, Isparta 32260, Turkey
| | | | - Derya Erisik
- Department of Histology and Embryology, Faculty of Medicine, Ege University, Izmir 35100, Turkey
| | - Taha Kadir Yesin
- Department of Histology and Embryology, Faculty of Medicine, Ege University, Izmir 35100, Turkey
| | - Cıgır Biray Avci
- Department of Medical Biology, Faculty of Medicine, Ege University, Izmir 35100, Turkey
| | - Yesim Kurkutçu
- Department of Histology and Embryology, Faculty of Medicine, Ege University, Izmir 35100, Turkey
| | - Gunnur Guler
- Department of Physics, Biophysics Laboratory, Izmir Institute of Technology, Izmir 35430, Turkey
| | - Huseyin Aktug
- Department of Histology and Embryology, Faculty of Medicine, Ege University, Izmir 35100, Turkey.
| |
Collapse
|
13
|
Saleh J, Fardin MA, Barai A, Soleilhac M, Frenoy O, Gaston C, Cui H, Dang T, Gaudin N, Vincent A, Minc N, Delacour D. Length limitation of astral microtubules orients cell divisions in murine intestinal crypts. Dev Cell 2023; 58:1519-1533.e6. [PMID: 37419117 DOI: 10.1016/j.devcel.2023.06.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 05/25/2023] [Accepted: 06/14/2023] [Indexed: 07/09/2023]
Abstract
Planar spindle orientation is critical for epithelial tissue organization and is generally instructed by the long cell-shape axis or cortical polarity domains. We introduced mouse intestinal organoids in order to study spindle orientation in a monolayered mammalian epithelium. Although spindles were planar, mitotic cells remained elongated along the apico-basal (A-B) axis, and polarity complexes were segregated to basal poles, so that spindles oriented in an unconventional manner, orthogonal to both polarity and geometric cues. Using high-resolution 3D imaging, simulations, and cell-shape and cytoskeleton manipulations, we show that planar divisions resulted from a length limitation in astral microtubules (MTs) which precludes them from interacting with basal polarity, and orient spindles from the local geometry of apical domains. Accordingly, lengthening MTs affected spindle planarity, cell positioning, and crypt arrangement. We conclude that MT length regulation may serve as a key mechanism for spindles to sense local cell shapes and tissue forces to preserve mammalian epithelial architecture.
Collapse
Affiliation(s)
- Jad Saleh
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013 Paris, France
| | | | - Amlan Barai
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013 Paris, France
| | - Matis Soleilhac
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013 Paris, France
| | - Olivia Frenoy
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013 Paris, France
| | - Cécile Gaston
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013 Paris, France
| | - Hongyue Cui
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013 Paris, France
| | - Tien Dang
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013 Paris, France
| | - Noémie Gaudin
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013 Paris, France
| | - Audrey Vincent
- Université de Lille, CNRS, INSERM, CHU Lille, UMR9020-U1277, 59000 Lille, France; ORGALille Core Facility, CANTHER, Université de Lille, CNRS, INSERM, CHU Lille, UMR9020-U1277, 59000 Lille, France
| | - Nicolas Minc
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013 Paris, France; Equipe Labellisée La Ligue Contre le Cancer, France.
| | - Delphine Delacour
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013 Paris, France.
| |
Collapse
|
14
|
Zheng X, Betjes MA, Ender P, Goos YJ, Huelsz-Prince G, Clevers H, van Zon JS, Tans SJ. Organoid cell fate dynamics in space and time. SCIENCE ADVANCES 2023; 9:eadd6480. [PMID: 37595032 PMCID: PMC10438469 DOI: 10.1126/sciadv.add6480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 07/20/2023] [Indexed: 08/20/2023]
Abstract
Organoids are a major new tool to study tissue renewal. However, characterizing the underlying differentiation dynamics remains challenging. Here, we developed TypeTracker, which identifies cell fates by AI-enabled cell tracking and propagating end point fates back along the branched lineage trees. Cells that ultimately migrate to the villus commit to their new type early, when still deep inside the crypt, with important consequences: (i) Secretory cells commit before terminal division, with secretory fates emerging symmetrically in sister cells. (ii) Different secretory types descend from distinct stem cell lineages rather than an omnipotent secretory progenitor. (iii) The ratio between secretory and absorptive cells is strongly affected by proliferation after commitment. (iv) Spatial patterning occurs after commitment through type-dependent cell rearrangements. This "commit-then-sort" model contrasts with the conventional conveyor belt picture, where cells differentiate by moving up the crypt-villus axis and hence raises new questions about the underlying commitment and sorting mechanisms.
Collapse
Affiliation(s)
| | | | | | | | | | - Hans Clevers
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, Uppsalalaan 8, Utrecht 3584 CT, Netherlands
| | | | - Sander J Tans
- Bionanoscience Department, Kavli Institute of Nanoscience Delft, Delft University of Technology, Delft, Netherlands
- AMOLF, Amsterdam, Netherlands.
| |
Collapse
|
15
|
Srivastava V, Hu JL, Garbe JC, Veytsman B, Shalabi SF, Yllanes D, Thomson M, LaBarge MA, Huber G, Gartner ZJ. Configurational entropy is an intrinsic driver of tissue structural heterogeneity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.01.546933. [PMID: 37425903 PMCID: PMC10327153 DOI: 10.1101/2023.07.01.546933] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Tissues comprise ordered arrangements of cells that can be surprisingly disordered in their details. How the properties of single cells and their microenvironment contribute to the balance between order and disorder at the tissue-scale remains poorly understood. Here, we address this question using the self-organization of human mammary organoids as a model. We find that organoids behave like a dynamic structural ensemble at the steady state. We apply a maximum entropy formalism to derive the ensemble distribution from three measurable parameters - the degeneracy of structural states, interfacial energy, and tissue activity (the energy associated with positional fluctuations). We link these parameters with the molecular and microenvironmental factors that control them to precisely engineer the ensemble across multiple conditions. Our analysis reveals that the entropy associated with structural degeneracy sets a theoretical limit to tissue order and provides new insight for tissue engineering, development, and our understanding of disease progression.
Collapse
Affiliation(s)
- Vasudha Srivastava
- Dept. of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, USA
| | - Jennifer L. Hu
- UC Berkeley-UC San Francisco Graduate Program in Bioengineering, Berkeley, CA 94720, USA
| | - James C. Garbe
- Dept. of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, USA
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Boris Veytsman
- Chan Zuckerberg Initiative, Redwood City, CA 94963, USA
- School of Systems Biology, George Mason University, Fairfax, VA 22030, USA
| | | | - David Yllanes
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
- Instituto de Biocomputaciòn y Fìsica de Sistemas Complejos (BIFI), 50018 Zaragoza, Spain
| | - Matt Thomson
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Mark A. LaBarge
- Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Greg Huber
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Zev J. Gartner
- Dept. of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
- Center for Cellular Construction, University of California, San Francisco, CA 94158, USA
| |
Collapse
|
16
|
Lee HW, Adachi T, Pak B, Park S, Hu X, Choi W, Kowalski PS, Chang CH, Clapham KR, Lee A, Papangeli I, Kim J, Han O, Park J, Anderson DG, Simons M, Jin SW, Chun HJ. BMPR1A promotes ID2-ZEB1 interaction to suppress excessive endothelial to mesenchymal transition. Cardiovasc Res 2023; 119:813-825. [PMID: 36166408 PMCID: PMC10409893 DOI: 10.1093/cvr/cvac159] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 07/25/2022] [Accepted: 09/14/2022] [Indexed: 11/14/2022] Open
Abstract
AIMS Components of bone morphogenetic protein (BMP) signalling have been implicated in both pathogenesis of pulmonary arterial hypertension (PAH) and endothelial-mesenchymal transition (EndoMT). In particular, the importance of BMP type 2 receptor in these processes has been extensively analysed. However, the contribution of BMP type 1 receptors (BMPR1s) to the onset of PAH and EndoMT remains poorly understood. BMPR1A, one of BMPR1s, was recently implicated in the pathogenesis of PAH, and was found to be down-regulated in the lungs of PAH patients, neither the downstream mechanism nor its contribution to EndoMT has been described. Therefore, we aim to delineate the role of endothelial BMPR1A in modulating EndoMT and pathogenesis of PAH. METHODS AND RESULTS We find that BMPR1A knockdown in endothelial cells (ECs) induces hallmarks of EndoMT, and deletion of endothelial Bmpr1a in adult mice (Bmpr1aiECKO) leads to development of PAH-like symptoms due to excessive EndoMT. By lineage tracing, we show that endothelial-derived smooth muscle cells are increased in endothelial Bmpr1a-deleted mice. Mechanistically, we identify ZEB1 as a primary target for BMPR1A in this setting; upon BMPR1A activation, ID2 physically interacts and sequesters ZEB1 to attenuate transcription of Tgfbr2, which in turn lowers the responses of ECs towards transforming growth factor beta (TGFβ) stimulation and prevents excessive EndoMT. In Bmpr1aiECKO mice, administering endothelial targeting lipid nanoparticles containing siRNA against Tgfbr2 effectively ameliorate PAH, reiterating the importance of BMPR1A-ID2/ZEB1-TGFBR2 axis in modulating progression of EndoMT and pathogenesis of PAH. CONCLUSIONS We demonstrate that BMPR1A is key to maintain endothelial identity and to prevent excessive EndoMT. We identify BMPR1A-induced interaction between ID2 and ZEB1 is the key regulatory step for onset of EndoMT and pathogenesis of PAH. Our findings indicate that BMPR1A-ID2/ZEB1-TGFBR2 signalling axis could serve as a potential novel therapeutic target for PAH and other EndoMT-related vascular disorders.
Collapse
Affiliation(s)
- Heon-Woo Lee
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Takaomi Adachi
- Division of Nephrology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Boryeong Pak
- School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, Korea
| | - Saejeong Park
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Xiaoyue Hu
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Woosoung Choi
- School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, Korea
| | - Piotr S Kowalski
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - C Hong Chang
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Katharine R Clapham
- Division of Pulmonary and Critical Care, Brigham and Women’s Hospital, Boston, MA 02127, USA
| | - Aram Lee
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA
- Division of Biological Sciences, Sookmyung Women's University, Seoul 04310, Korea
| | - Irinna Papangeli
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Jongmin Kim
- Division of Biological Sciences, Sookmyung Women's University, Seoul 04310, Korea
| | - Orjin Han
- School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, Korea
| | - Jihwan Park
- School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, Korea
| | - Daniel G Anderson
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Michael Simons
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Suk-Won Jin
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA
- School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, Korea
| | - Hyung J Chun
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA
- VA Connecticut Healthcare System, 950 Campbell Ave, 111B, West Haven, CT 06516, USA
| |
Collapse
|
17
|
Galenza A, Moreno-Roman P, Su YH, Acosta-Alvarez L, Debec A, Guichet A, Knapp JM, Kizilyaprak C, Humbel BM, Kolotuev I, O'Brien LE. Basal stem cell progeny establish their apical surface in a junctional niche during turnover of an adult barrier epithelium. Nat Cell Biol 2023; 25:658-671. [PMID: 36997641 PMCID: PMC10317055 DOI: 10.1038/s41556-023-01116-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 02/23/2023] [Indexed: 04/01/2023]
Abstract
Barrier epithelial organs face the constant challenge of sealing the interior body from the external environment while simultaneously replacing the cells that contact this environment. New replacement cells-the progeny of basal stem cells-are born without barrier-forming structures such as a specialized apical membrane and occluding junctions. Here, we investigate how new progeny acquire barrier structures as they integrate into the intestinal epithelium of adult Drosophila. We find they gestate their future apical membrane in a sublumenal niche created by a transitional occluding junction that envelops the differentiating cell and enables it to form a deep, microvilli-lined apical pit. The transitional junction seals the pit from the intestinal lumen until differentiation-driven, basal-to-apical remodelling of the niche opens the pit and integrates the now-mature cell into the barrier. By coordinating junctional remodelling with terminal differentiation, stem cell progeny integrate into a functional, adult epithelium without jeopardizing barrier integrity.
Collapse
Affiliation(s)
- Anthony Galenza
- Department of Molecular & Cellular Physiology and Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Paola Moreno-Roman
- Department of Molecular & Cellular Physiology and Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Foldscope Instruments, Inc., Palo Alto, CA, USA
| | - Yu-Han Su
- Department of Molecular & Cellular Physiology and Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Lehi Acosta-Alvarez
- Department of Molecular & Cellular Physiology and Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Alain Debec
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris, France
- Institute of Ecology and Environmental Sciences, iEES, Sorbonne University, UPEC, CNRS, IRD, INRA, Paris, France
| | - Antoine Guichet
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris, France
| | | | - Caroline Kizilyaprak
- Université de Lausanne, Bâtiment Biophore, Quartier Sorge, Lausanne, Switzerland
| | - Bruno M Humbel
- Université de Lausanne, Bâtiment Biophore, Quartier Sorge, Lausanne, Switzerland
- Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Provost's Office, Okinawa Institute of Science and Technology, Tancha, Japan
| | - Irina Kolotuev
- Université de Lausanne, Bâtiment Biophore, Quartier Sorge, Lausanne, Switzerland
| | - Lucy Erin O'Brien
- Department of Molecular & Cellular Physiology and Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Chan-Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
18
|
Lboukili I, Stamatas GN, Descombes X. Age-dependent changes in epidermal architecture explored using an automated image analysis algorithm on in vivo reflectance confocal microscopy images. Skin Res Technol 2023; 29:e13343. [PMID: 37231922 PMCID: PMC10177282 DOI: 10.1111/srt.13343] [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: 11/30/2022] [Accepted: 04/25/2023] [Indexed: 05/27/2023]
Abstract
BACKGROUND Reflectance confocal microscopy (RCM) allows for real-time in vivo visualization of the epidermis at the cellular level noninvasively. Parameters relating to tissue architecture can be extracted from RCM images, however, analysis of such images requires manual identification of cells to derive these parameters, which can be time-consuming and subject to human error, highlighting the need for an automated cell identification method. METHODS First, the region-of-interest (ROI) containing cells needs to be identified, followed by the identification of individual cells within the ROI. To perform this task, we use successive applications of Sato and Gabor filters. The final step is post-processing improvement of cell detection and removal of size outliers. The proposed algorithm is evaluated on manually annotated real data. It is then applied to 5345 images to study the evolution of epidermal architecture in children and adults. The images were acquired on the volar forearm of healthy children (3 months to 10 years) and women (25-80 years), and on the volar forearm and cheek of women (40-80 years). Following the identification of cell locations, parameters such as cell area, cell perimeter, and cell density are calculated, as well as the probability distribution of the number of nearest neighbors per cell. The thicknesses of the Stratum Corneum and supra-papillary epidermis are also calculated using a hybrid deep-learning method. RESULTS Epidermal keratinocytes are significantly larger (area and perimeter) in the granular layer than in the spinous layer and they get progressively larger with a child's age. Skin continues to mature dynamically during adulthood, as keratinocyte size continues to increase with age on both the cheeks and volar forearm, but the topology and cell aspect ratio remain unchanged across different epidermal layers, body sites, and age. Stratum Corneum and supra-papillary epidermis thicknesses increase with age, at a faster rate in children than in adults. CONCLUSIONS The proposed methodology can be applied to large datasets to automate image analysis and the calculation of parameters relevant to skin physiology. These data validate the dynamic nature of skin maturation during childhood and skin aging in adulthood.
Collapse
Affiliation(s)
- Imane Lboukili
- R&D Essential HealthJohnson & Johnson Santé Beauté FranceIssy‐les‐moulineauxFrance
- MorphemeUCA–INRIA–I3S/CNRSSophia AntipolisFrance
| | - Georgios N. Stamatas
- R&D Essential HealthJohnson & Johnson Santé Beauté FranceIssy‐les‐moulineauxFrance
| | | |
Collapse
|
19
|
Bury S, Kratochvíl L, Starostová Z. Scaling of erythrocyte shape and nucleus size among squamate reptiles: reanalysis points to constrained, proportional rather than adaptive changes. ROYAL SOCIETY OPEN SCIENCE 2023; 10:221513. [PMID: 37122952 PMCID: PMC10130710 DOI: 10.1098/rsos.221513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 04/04/2023] [Indexed: 05/03/2023]
Abstract
Small erythrocytes might be beneficial for blood rheology, as they contribute less to blood viscosity than large erythrocytes. We predicted that rheological disadvantages of larger erythrocytes could be alleviated by relatively smaller nucleus size in larger cells allowing higher flexibility and by more elongated shape. Across squamate reptiles, we found that species with larger erythrocytes tend to have smaller ratio of nucleus size to cell size (N : C ratio), but that larger erythrocytes tend to be rounder, not more elongated. Nevertheless, we document that in fact nucleus area changes with erythrocyte area more or less linearly, which is also true for the relationship between cell length and cell width. These linear relationships suggest that nucleus size and cell size, and cell width and cell length, might be constrained to largely proportional mutual changes. The shifts in widely used N : C ratio and elongation ratio (cell length/cell width) with cell size might be misleading, as they do not reflect adaptive or maladaptive changes of erythrocytes, but rather mathematically trivial scaling of the ratios of two variables with a linear relationship with non-zero intercepts. We warn that ratio scaling without analyses of underlying patterns of evolutionary changes can lead to misinterpretation of evolutionary processes.
Collapse
Affiliation(s)
- Stanisław Bury
- Department of Comparative Anatomy, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387 Kraków, Poland
| | - Lukáš Kratochvíl
- Department of Ecology, Faculty of Science, Charles University, Viničná 7, 12844 Prague, Czech Republic
| | - Zuzana Starostová
- Department of Zoology, Faculty of Science, Charles University, Viničná 7, 12844 Prague, Czech Republic
| |
Collapse
|
20
|
Cohen PJR, Luquet E, Pletenka J, Leonard A, Warter E, Gurchenkov B, Carrere J, Rieu C, Hardouin J, Moncaubeig F, Lanero M, Quelennec E, Wurtz H, Jamet E, Demarco M, Banal C, Van Liedekerke P, Nassoy P, Feyeux M, Lefort N, Alessandri K. Engineering 3D micro-compartments for highly efficient and scale-independent expansion of human pluripotent stem cells in bioreactors. Biomaterials 2023; 295:122033. [PMID: 36764194 DOI: 10.1016/j.biomaterials.2023.122033] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 12/12/2022] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
Human pluripotent stem cells (hPSCs) have emerged as the most promising cellular source for cell therapies. To overcome the scale-up limitations of classical 2D culture systems, suspension cultures have been developed to meet the need for large-scale culture in regenerative medicine. Despite constant improvements, current protocols that use microcarriers or generate cell aggregates only achieve moderate amplification performance. Here, guided by reports showing that hPSCs can self-organize in vitro into cysts reminiscent of the epiblast stage in embryo development, we developed a physio-mimetic approach for hPSC culture. We engineered stem cell niche microenvironments inside microfluidics-assisted core-shell microcapsules. We demonstrate that lumenized three-dimensional colonies significantly improve viability and expansion rates while maintaining pluripotency compared to standard hPSC culture platforms such as 2D cultures, microcarriers, and aggregates. By further tuning capsule size and culture conditions, we scale up this method to industrial-scale stirred tank bioreactors and achieve an unprecedented hPSC amplification rate of 277-fold in 6.5 days. In brief, our findings indicate that our 3D culture system offers a suitable strategy both for basic stem cell biology experiments and for clinical applications.
Collapse
Affiliation(s)
- Philippe J R Cohen
- Université Paris Cité, Imagine Institute, IPSC Core Facility, INSERM UMR U1163, F-75015, Paris, France; Treefrog Therapeutics, F-33600, Pessac, France.
| | | | | | | | | | | | | | | | | | | | | | - Eddy Quelennec
- Université Paris Cité, Imagine Institute, IPSC Core Facility, INSERM UMR U1163, F-75015, Paris, France; Treefrog Therapeutics, F-33600, Pessac, France
| | | | | | | | - Celine Banal
- Université Paris Cité, Imagine Institute, IPSC Core Facility, INSERM UMR U1163, F-75015, Paris, France
| | - Paul Van Liedekerke
- Inria Paris & Sorbonne Université LJLL, 2 Rue Simone IFF, F-75012, Paris, France
| | - Pierre Nassoy
- LP2N, Laboratoire Photonique Numérique et Nanosciences, Univ. Bordeaux, F-33400, Talence, France; Institut D'Optique Graduate School & CNRS UMR 5298, F-33400, Talence, France
| | | | - Nathalie Lefort
- Université Paris Cité, Imagine Institute, IPSC Core Facility, INSERM UMR U1163, F-75015, Paris, France
| | | |
Collapse
|
21
|
di Pietro F, Osswald M, De Las Heras JM, Cristo I, López-Gay J, Wang Z, Pelletier S, Gaugué I, Leroy A, Martin C, Morais-de-Sá E, Bellaïche Y. Systematic analysis of RhoGEF/GAP localizations uncovers regulators of mechanosensing and junction formation during epithelial cell division. Curr Biol 2023; 33:858-874.e7. [PMID: 36917931 PMCID: PMC10017266 DOI: 10.1016/j.cub.2023.01.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/30/2022] [Accepted: 01/16/2023] [Indexed: 02/17/2023]
Abstract
Cell proliferation is central to epithelial tissue development, repair, and homeostasis. During cell division, small RhoGTPases control both actomyosin dynamics and cell-cell junction remodeling to faithfully segregate the genome while maintaining tissue polarity and integrity. To decipher the mechanisms of RhoGTPase spatiotemporal regulation during epithelial cell division, we generated a transgenic fluorescently tagged library for the 48 Drosophila Rho guanine exchange factors (RhoGEFs) and GTPase-activating proteins (GAPs), and we systematically characterized their endogenous distributions by time-lapse microscopy. Therefore, we unveiled candidate regulators of the interplay between actomyosin and junctional dynamics during epithelial cell division. Building on these findings, we established that the conserved RhoGEF Cysts and RhoGEF4 play sequential and distinct roles to couple cytokinesis with de novo junction formation. During ring contraction, Cysts via Rho1 participates in the neighbor mechanosensing response, promoting daughter-daughter cell membrane juxtaposition in preparation to de novo junction formation. Subsequently and upon midbody formation, RhoGEF4 via Rac acts in the dividing cell to ensure the withdrawal of the neighboring cell membranes, thus controlling de novo junction length and cell-cell arrangements upon cytokinesis. Altogether, our findings delineate how the RhoGTPases Rho and Rac are locally and temporally activated during epithelial cytokinesis, highlighting the RhoGEF/GAP library as a key resource to understand the broad range of biological processes regulated by RhoGTPases.
Collapse
Affiliation(s)
- Florencia di Pietro
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Mariana Osswald
- IBMC - Instituto de Biologia Molecular e Celular; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - José M De Las Heras
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Inês Cristo
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Jesús López-Gay
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Zhimin Wang
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Stéphane Pelletier
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Isabelle Gaugué
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Adrien Leroy
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Charlotte Martin
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Eurico Morais-de-Sá
- IBMC - Instituto de Biologia Molecular e Celular; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
| | - Yohanns Bellaïche
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France.
| |
Collapse
|
22
|
Castillo-Azofeifa D, Wald T, Reyes EA, Gallagher A, Schanin J, Vlachos S, Lamarche-Vane N, Bomidi C, Blutt S, Estes MK, Nystul T, Klein OD. A DLG1-ARHGAP31-CDC42 axis is essential for the intestinal stem cell response to fluctuating niche Wnt signaling. Cell Stem Cell 2023; 30:188-206.e6. [PMID: 36640764 PMCID: PMC9922544 DOI: 10.1016/j.stem.2022.12.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 10/13/2022] [Accepted: 12/12/2022] [Indexed: 01/15/2023]
Abstract
A central factor in the maintenance of tissue integrity is the response of stem cells to variations in the levels of niche signals. In the gut, intestinal stem cells (ISCs) depend on Wnt ligands for self-renewal and proliferation. Transient increases in Wnt signaling promote regeneration after injury or in inflammatory bowel diseases, whereas constitutive activation of this pathway leads to colorectal cancer. Here, we report that Discs large 1 (Dlg1), although dispensable for polarity and cellular turnover during intestinal homeostasis, is required for ISC survival in the context of increased Wnt signaling. RNA sequencing (RNA-seq) and genetic mouse models demonstrated that DLG1 regulates the cellular response to increased canonical Wnt ligands. This occurs via the transcriptional regulation of Arhgap31, a GTPase-activating protein that deactivates CDC42, an effector of the non-canonical Wnt pathway. These findings reveal a DLG1-ARHGAP31-CDC42 axis that is essential for the ISC response to increased niche Wnt signaling.
Collapse
Affiliation(s)
- David Castillo-Azofeifa
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA; Department of Regenerative Medicine, Genentech, Inc., South San Francisco, CA, USA
| | - Tomas Wald
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Efren A Reyes
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA; Department of Pharmaceutical Chemistry and TETRAD Program, University of California, San Francisco, San Francisco, CA, USA
| | - Aaron Gallagher
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Julia Schanin
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Stephanie Vlachos
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
| | - Nathalie Lamarche-Vane
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada; Department of Anatomy and Cell Biology, McGill University, Montréal, QC, Canada
| | - Carolyn Bomidi
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Sarah Blutt
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Mary K Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Todd Nystul
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
| | - Ophir D Klein
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA; Department of Pediatrics and Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA; Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
23
|
Guevara-Garcia A, Soleilhac M, Minc N, Delacour D. Regulation and functions of cell division in the intestinal tissue. Semin Cell Dev Biol 2023:S1084-9521(23)00004-6. [PMID: 36702722 DOI: 10.1016/j.semcdb.2023.01.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 12/16/2022] [Accepted: 01/06/2023] [Indexed: 01/26/2023]
Abstract
In multicellular organisms, epithelial cells are key elements of tissue organization. In developing epithelial tissues, cellular proliferation and differentiation are under the tight regulation of morphogenetic programs to ensure correct organ formation and functioning. In these processes, proliferation rates and division orientation regulate the speed, timing and direction of tissue expansion but also its proper patterning. Moreover, tissue homeostasis relies on spatio-temporal modulations of daughter cell behavior and arrangement. These aspects are particularly crucial in the intestine, which is one of the most proliferative tissues in adults, making it a very attractive adult organ system to study the role of cell division on epithelial morphogenesis and organ function. Although epithelial cell division has been the subject of intense research for many years in multiple models, it still remains in its infancy in the context of the intestinal tissue. In this review, we focus on the current knowledge on cell division and regulatory mechanisms at play in the intestinal epithelial tissue, as well as their importance in developmental biology and physiopathology.
Collapse
Affiliation(s)
| | - Matis Soleilhac
- Université de Paris, CNRS, Institut Jacques Monod, F-75006 Paris, France
| | - Nicolas Minc
- Université de Paris, CNRS, Institut Jacques Monod, F-75006 Paris, France
| | - Delphine Delacour
- Université de Paris, CNRS, Institut Jacques Monod, F-75006 Paris, France.
| |
Collapse
|
24
|
Stemness potency and structural characteristics of thyroid cancer cell lines. Pathol Res Pract 2023; 241:154262. [PMID: 36527836 DOI: 10.1016/j.prp.2022.154262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/02/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Thyroid cancer is the most frequent type of endocrine malignancy. Thyroid carcinomas are derived from the follicular epithelium and classified as papillary (PTC) (85%), follicular (FTC) (12%), and anaplastic (ATC) (<3%). Thyroid cancer could arise from thyroid cancer stem-like cells (CSCs). CSCs are cancer cells that feature stem-like properties. Kruppel-like factor (KLF4) and Stage-spesific embryonic antigen 1 (SSEA-1) are types of stem cell markers. Filamentous actin (F-actin) is an essential part of the cellular cytoskeleton. The purpose of this study was to evaluate the stem cell potency and the spatial distribution of the cytoskeletal element F-actin in PTC, FTC, and ATC cell lines. MATERIALS AND METHODS Normal thyroid cell line (NTC) Nthy-ori-3-1, PTC cell line BCPAP, FTC cell line FTC-133 and ATC cell line 8505c were stained with SSEA-1 and KLF4 for stem cell potency and F-actin for cytoskeleton. The morphological properties of cells were assessed by a scanning electron microscope (SEM) and elemental ratios were compared with EDS. RESULTS PTCs had greater percentages of SSEA-1 and KLF4 protein intensity (0.32% and 0.49%, respectively) than NTCs. ATCs had a greater proportion of KLF4 expression (0.8%) than NTCs. NTCs and FTCs had increased F-actin intensity across the cell, but PTCs had the lowest among these four cell lines. NTCs and PTCs, as well as NTCs and FTCs, have statistically identical aspect ratios and round values. These values, however, were statistically different in ATCs. CONCLUSION The study of stem cell markers and the cytoskeletal element F-actin in cancer and normal thyroid cell lines may assist in the identification of new therapeutic targets and contribute in the understanding of treatment resistance mechanisms.
Collapse
|
25
|
Sadhukhan S, Nandi SK. On the origin of universal cell shape variability in confluent epithelial monolayers. eLife 2022; 11:e76406. [PMID: 36563034 PMCID: PMC9833828 DOI: 10.7554/elife.76406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 12/22/2022] [Indexed: 12/24/2022] Open
Abstract
Cell shape is fundamental in biology. The average cell shape can influence crucial biological functions, such as cell fate and division orientation. But cell-to-cell shape variability is often regarded as noise. In contrast, recent works reveal that shape variability in diverse epithelial monolayers follows a nearly universal distribution. However, the origin and implications of this universality remain unclear. Here, assuming contractility and adhesion are crucial for cell shape, characterized via aspect ratio (r), we develop a mean-field analytical theory for shape variability. We find that all the system-specific details combine into a single parameter α that governs the probability distribution function (PDF) of r; this leads to a universal relation between the standard deviation and the average of r. The PDF for the scaled r is not strictly but nearly universal. In addition, we obtain the scaled area distribution, described by the parameter μ. Information of α and μ together can distinguish the effects of changing physical conditions, such as maturation, on different system properties. We have verified the theory via simulations of two distinct models of epithelial monolayers and with existing experiments on diverse systems. We demonstrate that in a confluent monolayer, average shape determines both the shape variability and dynamics. Our results imply that cell shape distribution is inevitable, where a single parameter describes both statics and dynamics and provides a framework to analyze and compare diverse epithelial systems. In contrast to existing theories, our work shows that the universal properties are consequences of a mathematical property and should be valid in general, even in the fluid regime.
Collapse
|
26
|
Pentinmikko N, Lozano R, Scharaw S, Andersson S, Englund JI, Castillo-Azofeifa D, Gallagher A, Broberg M, Song KY, Sola Carvajal A, Speidel AT, Sundstrom M, Allbritton N, Stevens MM, Klein OD, Teixeira A, Katajisto P. Cellular shape reinforces niche to stem cell signaling in the small intestine. SCIENCE ADVANCES 2022; 8:eabm1847. [PMID: 36240269 PMCID: PMC9565803 DOI: 10.1126/sciadv.abm1847] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 08/30/2022] [Indexed: 06/06/2023]
Abstract
Niche-derived factors regulate tissue stem cells, but apart from the mechanosensory pathways, the effect of niche geometry is not well understood. We used organoids and bioengineered tissue culture platforms to demonstrate that the conical shape of Lgr5+ small intestinal stem cells (ISCs) facilitate their self-renewal and function. Inhibition of non-muscle myosin II (NM II)-driven apical constriction altered ISC shape and reduced niche curvature and stem cell capacity. Niche curvature is decreased in aged mice, suggesting that suboptimal interactions between old ISCs and their niche develop with age. We show that activation of NM IIC or physical restriction to young topology improves in vitro regeneration by old epithelium. We propose that the increase in lateral surface area of ISCs induced by apical constriction promotes interactions between neighboring cells, and the curved topology of the intestinal niche has evolved to maximize signaling between ISCs and neighboring cells.
Collapse
Affiliation(s)
- Nalle Pentinmikko
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Rodrigo Lozano
- Department of Cell and Molecular Biology (CMB), Karolinska Institutet, Stockholm, Sweden
- Division of Rheumatology, Department of Medicine, Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden
| | - Sandra Scharaw
- Department of Cell and Molecular Biology (CMB), Karolinska Institutet, Stockholm, Sweden
| | - Simon Andersson
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Johanna I. Englund
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - David Castillo-Azofeifa
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA
- Immunology Discovery, Genentech Inc., South San Francisco, CA, USA
| | - Aaron Gallagher
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Martin Broberg
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Ki-Young Song
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Agustín Sola Carvajal
- Department of Cell and Molecular Biology (CMB), Karolinska Institutet, Stockholm, Sweden
| | - Alessondra T. Speidel
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Michael Sundstrom
- Division of Rheumatology, Department of Medicine, Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden
| | - Nancy Allbritton
- University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Molly M. Stevens
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Department of Materials and Department of Bioengineering, Imperial College London, UK
| | - Ophir D. Klein
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA
- Department of Pediatrics and Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ana Teixeira
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Pekka Katajisto
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
- Department of Cell and Molecular Biology (CMB), Karolinska Institutet, Stockholm, Sweden
- Molecular and Integrative Bioscience Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| |
Collapse
|
27
|
Gupta VK, Chaudhuri O. Mechanical regulation of cell-cycle progression and division. Trends Cell Biol 2022; 32:773-785. [PMID: 35491306 PMCID: PMC9378598 DOI: 10.1016/j.tcb.2022.03.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/28/2022] [Accepted: 03/30/2022] [Indexed: 10/18/2022]
Abstract
Cell-cycle progression and division are fundamental biological processes in animal cells, and their biochemical regulation has been extensively studied. An emerging body of work has revealed how mechanical interactions of cells with their microenvironment in tissues, including with the extracellular matrix (ECM) and neighboring cells, also plays a crucial role in regulating cell-cycle progression and division. We review recent work on how cells interpret physical cues and alter their mechanics to promote cell-cycle progression and initiate cell division, and then on how dividing cells generate forces on their surrounding microenvironment to successfully divide. Finally, the article ends by discussing how force generation during division potentially contributes to larger tissue-scale processes involved in development and homeostasis.
Collapse
Affiliation(s)
- Vivek K Gupta
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA
| | - Ovijit Chaudhuri
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA..
| |
Collapse
|
28
|
Donà F, Eli S, Mapelli M. Insights Into Mechanisms of Oriented Division From Studies in 3D Cellular Models. Front Cell Dev Biol 2022; 10:847801. [PMID: 35356279 PMCID: PMC8959941 DOI: 10.3389/fcell.2022.847801] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 02/17/2022] [Indexed: 12/13/2022] Open
Abstract
In multicellular organisms, epithelial cells are key elements of tissue organization. In developing tissues, cellular proliferation and differentiation are under the tight regulation of morphogenetic programs, that ensure the correct organ formation and functioning. In these processes, mitotic rates and division orientation are crucial in regulating the velocity and the timing of the forming tissue. Division orientation, specified by mitotic spindle placement with respect to epithelial apico-basal polarity, controls not only the partitioning of cellular components but also the positioning of the daughter cells within the tissue, and hence the contacts that daughter cells retain with the surrounding microenvironment. Daughter cells positioning is important to determine signal sensing and fate, and therefore the final function of the developing organ. In this review, we will discuss recent discoveries regarding the mechanistics of planar divisions in mammalian epithelial cells, summarizing technologies and model systems used to study oriented cell divisions in vitro such as three-dimensional cysts of immortalized cells and intestinal organoids. We also highlight how misorientation is corrected in vivo and in vitro, and how it might contribute to the onset of pathological conditions.
Collapse
Affiliation(s)
- Federico Donà
- IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Susanna Eli
- IEO, European Institute of Oncology IRCCS, Milan, Italy
| | | |
Collapse
|
29
|
Fei K, Zhang J, Yuan J, Xiao P. Present Application and Perspectives of Organoid Imaging Technology. Bioengineering (Basel) 2022; 9:121. [PMID: 35324810 PMCID: PMC8945799 DOI: 10.3390/bioengineering9030121] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/23/2022] [Accepted: 03/13/2022] [Indexed: 11/18/2022] Open
Abstract
An organoid is a miniaturized and simplified in vitro model with a similar structure and function to a real organ. In recent years, the use of organoids has increased explosively in the field of growth and development, disease simulation, drug screening, cell therapy, etc. In order to obtain necessary information, such as morphological structure, cell function and dynamic signals, it is necessary and important to directly monitor the culture process of organoids. Among different detection technologies, imaging technology is a simple and convenient choice and can realize direct observation and quantitative research. In this review, the principle, advantages and disadvantages of imaging technologies that have been applied in organoids research are introduced. We also offer an overview of prospective technologies for organoid imaging. This review aims to help biologists find appropriate imaging techniques for different areas of organoid research, and also contribute to the development of organoid imaging systems.
Collapse
Affiliation(s)
| | | | - Jin Yuan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou 510060, China; (K.F.); (J.Z.)
| | - Peng Xiao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou 510060, China; (K.F.); (J.Z.)
| |
Collapse
|
30
|
Cambra HM, Tallapragada NP, Mannam P, Breault DT, Klein AM. Triple-Decker Sandwich Cultures of Intestinal Organoids for Long-Term Live Imaging, Uniform Perturbation, and Statistical Sampling. Curr Protoc 2022; 2:e330. [PMID: 35030297 PMCID: PMC9006308 DOI: 10.1002/cpz1.330] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Three-dimensional organoid cultures enable the study of stem cell and tissue biology ex vivo, providing improved access to cells for perturbation and live imaging. Typically, organoids are grown in hydrogel domes that are simple to prepare but that lead to non-uniform tissue growth and viability. We recently developed a simple alternative culture method to embed intestinal organoids in multilayered hydrogels, called "triple-decker sandwiches," that align organoids in a common z-plane with uniform access to medium. This culture configuration improves the growth and survival of organoids over a wide working area and facilitates long-term confocal imaging and molecular perturbation. Here, we present protocols for preparing organoids in triple-decker sandwich cultures and using them for live imaging, immunostaining, and single-cell RNA sequencing. We have tested our methods on mouse and human intestinal organoids and expect them to be useful for other highly proliferative three-dimensional cell cultures. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Pre-coating plates with PolyHEMA to prepare them for triple-decker sandwich culture Support Protocol 1: Preparing PolyHEMA solution to coat glass-bottom dishes Basic Protocol 2: Embedding intestinal organoids in triple-decker sandwiches Alternate Protocol 1: Seeding single cells or organoids at low density in triple-decker sandwiches Support Protocol 2: Embedding intestinal organoids in hydrogel domes Support Protocol 3: Production of Wnt3a-conditioned medium Support Protocol 4: Production of Rspo1-conditioned medium Basic Protocol 3: Live imaging of mouse intestinal organoids in triple-decker sandwich cultures Alternate Protocol 2: Live imaging of vital dye-treated mouse intestinal organoids in triple-decker sandwich cultures Basic Protocol 4: Immunofluorescence imaging of mouse organoids liberated from triple-decker sandwich cultures Alternate Protocol 3: Liberating and fixing mouse intestinal organoids from dome cultures Support Protocol 5: Measuring cell proliferation by EdU staining of mouse intestinal organoids Basic Protocol 5: Single-cell RNA sequencing and analysis of mouse intestinal organoids.
Collapse
Affiliation(s)
- Hailey M. Cambra
- Department of Systems Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Naren P. Tallapragada
- Department of Systems Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Prabhath Mannam
- Division of Endocrinology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - David T. Breault
- Division of Endocrinology, Boston Children’s Hospital, Boston, MA 02115, USA,Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA,Harvard Stem Cell Institute, Harvard University, Boston, MA 02139, USA
| | - Allon M. Klein
- Department of Systems Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA,Lead contact,Correspondence:
| |
Collapse
|
31
|
He X, Smith SE, Chen S, Li H, Wu D, Meneses-Giles PI, Wang Y, Hembree M, Yi K, Zhao X, Guo F, Unruh JR, Maddera LE, Yu Z, Scott A, Perera A, Wang Y, Zhao C, Bae K, Box A, Haug JS, Tao F, Hu D, Hansen DM, Qian P, Saha S, Dixon D, Anant S, Zhang D, Lin EH, Sun W, Wiedemann LM, Li L. Tumor-initiating stem cell shapes its microenvironment into an immunosuppressive barrier and pro-tumorigenic niche. Cell Rep 2021; 36:109674. [PMID: 34496236 PMCID: PMC8451448 DOI: 10.1016/j.celrep.2021.109674] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 04/30/2021] [Accepted: 08/13/2021] [Indexed: 01/10/2023] Open
Abstract
Tumor-initiating stem cells (TSCs) are critical for drug resistance and immune escape. However, the mutual regulations between TSC and tumor microenvironment (TME) remain unclear. Using DNA-label retaining, single-cell RNA sequencing (scRNA-seq), and other approaches, we investigated intestinal adenoma in response to chemoradiotherapy (CRT), thus identifying therapy-resistant TSCs (TrTSCs). We find bidirectional crosstalk between TSCs and TME using CellPhoneDB analysis. An intriguing finding is that TSCs shape TME into a landscape that favors TSCs for immunosuppression and propagation. Using adenoma-organoid co-cultures, niche-cell depletion, and lineaging tracing, we characterize a functional role of cyclooxygenase-2 (Cox-2)-dependent signaling, predominantly occurring between tumor-associated monocytes and macrophages (TAMMs) and TrTSCs. We show that TAMMs promote TrTSC proliferation through prostaglandin E2 (PGE2)-PTGER4(EP4) signaling, which enhances β-catenin activity via AKT phosphorylation. Thus, our study shows that the bidirectional crosstalk between TrTSC and TME results in a pro-tumorigenic and immunosuppressive contexture.
Collapse
Affiliation(s)
- Xi He
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Sarah E Smith
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Shiyuan Chen
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Hua Li
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Di Wu
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | | | - Yongfu Wang
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Mark Hembree
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Kexi Yi
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Xia Zhao
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Fengli Guo
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Jay R Unruh
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | | | - Zulin Yu
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Allison Scott
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Anoja Perera
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Yan Wang
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Chongbei Zhao
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - KyeongMin Bae
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Andrew Box
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Jeffrey S Haug
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Fang Tao
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Deqing Hu
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Darrick M Hansen
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Pengxu Qian
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Subhrajit Saha
- Department of Cancer Biology/Radiation Oncology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Dan Dixon
- Department of Molecular Biosciences, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Shrikant Anant
- Department of Cancer Biology/Radiation Oncology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Da Zhang
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 661607, USA
| | - Edward H Lin
- Seattle Cancer Care Alliance, University of Washington, Seattle, WA 98109, USA
| | - Weijing Sun
- Division of Medical Oncology, University of Kansas Medical Center, Kansas City, KS 66205, USA
| | - Leanne M Wiedemann
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA; Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 661607, USA
| | - Linheng Li
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA; Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 661607, USA.
| |
Collapse
|
32
|
Abstract
During salivary gland development, branches arise through formation of new end buds (budding) and division of existing buds (clefting). In a recent Cell study, Wang et al., (2021) report that mouse salivary gland budding is reliant on a delicate interplay between epithelial cells and the extracellular matrix surrounding them.
Collapse
Affiliation(s)
- Oscar Cazares
- Program in Craniofacial Biology and Division of Craniofacial Anomalies, Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Ameera S Haque
- Program in Craniofacial Biology and Division of Craniofacial Anomalies, Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Ophir D Klein
- Program in Craniofacial Biology and Division of Craniofacial Anomalies, Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA, USA; Division of Medical Genetics, Department of Pediatrics and Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
33
|
Mechanical compartmentalization of the intestinal organoid enables crypt folding and collective cell migration. Nat Cell Biol 2021; 23:745-757. [PMID: 34155382 PMCID: PMC7611697 DOI: 10.1038/s41556-021-00699-6] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 05/12/2021] [Indexed: 02/06/2023]
Abstract
Intestinal organoids capture essential features of the intestinal epithelium such as crypt folding, cellular compartmentalization and collective movements. Each of these processes and their coordination require patterned forces that are at present unknown. Here we map three-dimensional cellular forces in mouse intestinal organoids grown on soft hydrogels. We show that these organoids exhibit a non-monotonic stress distribution that defines mechanical and functional compartments. The stem cell compartment pushes the extracellular matrix and folds through apical constriction, whereas the transit amplifying zone pulls the extracellular matrix and elongates through basal constriction. The size of the stem cell compartment depends on the extracellular-matrix stiffness and endogenous cellular forces. Computational modelling reveals that crypt shape and force distribution rely on cell surface tensions following cortical actomyosin density. Finally, cells are pulled out of the crypt along a gradient of increasing tension. Our study unveils how patterned forces enable compartmentalization, folding and collective migration in the intestinal epithelium.
Collapse
|
34
|
Louey A, Hernández D, Pébay A, Daniszewski M. Automation of Organoid Cultures: Current Protocols and Applications. SLAS DISCOVERY 2021; 26:1138-1147. [PMID: 34167363 DOI: 10.1177/24725552211024547] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
GRAPHICAL ABSTRACT [Formula: see text].
Collapse
Affiliation(s)
- Alexandra Louey
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, Victoria, Australia
| | - Damián Hernández
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, Victoria, Australia
| | - Alice Pébay
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, Victoria, Australia.,Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, Victoria, Australia
| | - Maciej Daniszewski
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
35
|
Gupta VK, Nam S, Yim D, Camuglia J, Martin JL, Sanders EN, O'Brien LE, Martin AC, Kim T, Chaudhuri O. The nature of cell division forces in epithelial monolayers. J Cell Biol 2021; 220:212389. [PMID: 34132746 PMCID: PMC8240854 DOI: 10.1083/jcb.202011106] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 04/05/2021] [Accepted: 05/19/2021] [Indexed: 02/06/2023] Open
Abstract
Epithelial cells undergo striking morphological changes during division to ensure proper segregation of genetic and cytoplasmic materials. These morphological changes occur despite dividing cells being mechanically restricted by neighboring cells, indicating the need for extracellular force generation. Beyond driving cell division itself, forces associated with division have been implicated in tissue-scale processes, including development, tissue growth, migration, and epidermal stratification. While forces generated by mitotic rounding are well understood, forces generated after rounding remain unknown. Here, we identify two distinct stages of division force generation that follow rounding: (1) Protrusive forces along the division axis that drive division elongation, and (2) outward forces that facilitate postdivision spreading. Cytokinetic ring contraction of the dividing cell, but not activity of neighboring cells, generates extracellular forces that propel division elongation and contribute to chromosome segregation. Forces from division elongation are observed in epithelia across many model organisms. Thus, division elongation forces represent a universal mechanism that powers cell division in confining epithelia.
Collapse
Affiliation(s)
- Vivek K Gupta
- Department of Mechanical Engineering, Stanford University, Stanford, CA
| | - Sungmin Nam
- Department of Mechanical Engineering, Stanford University, Stanford, CA.,Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA.,Wyss Institute for Biologically Inspired Engineering, Cambridge, MA
| | - Donghyun Yim
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN
| | - Jaclyn Camuglia
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA
| | - Judy Lisette Martin
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA
| | - Erin Nicole Sanders
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA
| | - Lucy Erin O'Brien
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA
| | - Adam C Martin
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA
| | - Taeyoon Kim
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN
| | - Ovijit Chaudhuri
- Department of Mechanical Engineering, Stanford University, Stanford, CA
| |
Collapse
|
36
|
Betjes MA, Zheng X, Kok RNU, van Zon JS, Tans SJ. Cell Tracking for Organoids: Lessons From Developmental Biology. Front Cell Dev Biol 2021; 9:675013. [PMID: 34150770 PMCID: PMC8209328 DOI: 10.3389/fcell.2021.675013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 05/03/2021] [Indexed: 12/20/2022] Open
Abstract
Organoids have emerged as powerful model systems to study organ development and regeneration at the cellular level. Recently developed microscopy techniques that track individual cells through space and time hold great promise to elucidate the organizational principles of organs and organoids. Applied extensively in the past decade to embryo development and 2D cell cultures, cell tracking can reveal the cellular lineage trees, proliferation rates, and their spatial distributions, while fluorescent markers indicate differentiation events and other cellular processes. Here, we review a number of recent studies that exemplify the power of this approach, and illustrate its potential to organoid research. We will discuss promising future routes, and the key technical challenges that need to be overcome to apply cell tracking techniques to organoid biology.
Collapse
Affiliation(s)
| | | | | | | | - Sander J Tans
- AMOLF, Amsterdam, Netherlands.,Bionanoscience Department, Kavli Institute of Nanoscience Delft, Delft University of Technology, Delft, Netherlands
| |
Collapse
|
37
|
Abstract
Whilst tissues form during development, some cells are extruded from epithelial monolayers. Rather than dying or differentiating, a new study shows that displaced cells can reintegrate after dividing. Surprisingly, this 'intrusion' pathway shares common features with axon guidance.
Collapse
Affiliation(s)
- Scott E Williams
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA.
| | - Kendall J Lough
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| |
Collapse
|
38
|
Huang Q, Garrett A, Bose S, Blocker S, Rios AC, Clevers H, Shen X. The frontier of live tissue imaging across space and time. Cell Stem Cell 2021; 28:603-622. [PMID: 33798422 PMCID: PMC8034393 DOI: 10.1016/j.stem.2021.02.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
What you see is what you get-imaging techniques have long been essential for visualization and understanding of tissue development, homeostasis, and regeneration, which are driven by stem cell self-renewal and differentiation. Advances in molecular and tissue modeling techniques in the last decade are providing new imaging modalities to explore tissue heterogeneity and plasticity. Here we describe current state-of-the-art imaging modalities for tissue research at multiple scales, with a focus on explaining key tradeoffs such as spatial resolution, penetration depth, capture time/frequency, and moieties. We explore emerging tissue modeling and molecular tools that improve resolution, specificity, and throughput.
Collapse
Affiliation(s)
- Qiang Huang
- Department of Pediatric Surgery, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004 Shaanxi, China; Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA
| | - Aliesha Garrett
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA
| | - Shree Bose
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA
| | - Stephanie Blocker
- Center for In Vitro Microscopy, Duke University, Durham, NC 27708, USA
| | - Anne C Rios
- Princess Máxima Center for Pediatric Oncology, Utrecht 3584, the Netherlands; Department of Cancer Research, Oncode Institute, Hubrecht Institute-KNAW Utrecht, Utrecht 3584, the Netherlands
| | - Hans Clevers
- Princess Máxima Center for Pediatric Oncology, Utrecht 3584, the Netherlands; Department of Cancer Research, Oncode Institute, Hubrecht Institute-KNAW Utrecht, Utrecht 3584, the Netherlands; Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center (UMC) Utrecht, Utrecht 3584, the Netherlands
| | - Xiling Shen
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA.
| |
Collapse
|
39
|
Das A, Adhikary S, Chowdhury AR, Barui A. Substrate-dependent control of the chiral orientation of mesenchymal stem cells: image-based quantitative profiling. Biomed Mater 2021; 16:034102. [PMID: 33657017 DOI: 10.1088/1748-605x/abce4e] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Stem-cell (SC) chirality or left-right (LR) asymmetry is an essential attribute, observed during tissue regeneration. The ability to control the LR orientation of cells by biophysical manipulation is a promising approach for recapitulating their inherent function. Despite remarkable progress in tissue engineering, the development of LR chirality in SCs has been largely unexplored. Here, we demonstrate the role of substrate stiffness on the LR asymmetry of cultured mesenchymal stem cells (MSCs). We found that MSCs acquired higher asymmetricity when cultured on stiffer PCL/collagen matrices. To confirm cellular asymmetry, different parameters such as the aspect ratio, orientation angle and intensity of polarized proteins (Par) were investigated. The results showed a significant (p < 0.01) difference in the average orientation angle, the cellular aspect ratio, and the expression of actin and Par proteins in MSCs cultured on matrices with different stiffnesses. Furthermore, a Gaussian support-vector machine was applied to classify cells cultured on both (2% and 10% PCL/Collagen) matrices, with a resulting accuracy of 96.2%. To the best of our knowledge, this study is the first that interrelates and quantifies MSC asymmetricity with matrix properties using a simple 2D model.
Collapse
Affiliation(s)
- Ankita Das
- Centre for Healthcare Science and Technology, IIEST, Shibpur, Howrah, West Bengal 711103, India
| | | | | | | |
Collapse
|
40
|
Live Imaging Reveals Listeria Hijacking of E-Cadherin Recycling as It Crosses the Intestinal Barrier. Curr Biol 2020; 31:1037-1047.e4. [PMID: 33333010 DOI: 10.1016/j.cub.2020.11.041] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/13/2020] [Accepted: 11/17/2020] [Indexed: 12/19/2022]
Abstract
Listeria monocytogenes is a foodborne bacterial pathogen that causes human listeriosis, a severe systemic infection.1 Its translocation across the intestinal epithelium is mediated by the interaction of internalin (InlA), a Listeria surface protein, with its host-species-specific receptor E-cadherin (Ecad).2-5 It occurs through goblet cells, on which Ecad is luminally accessible,6 via an unknown mechanism. In the absence of cell lines recapitulating this phenotype in vitro, we developed an ex vivo experimental system, based on the intraluminal microinjection of Listeria in untreated, pharmacologically treated, and genetically modified intestinal organoids. Using both live light-sheet microscopy and confocal imaging, we show that Listeria translocates through goblet cells within a membrane vacuole in an InlA- and microtubule-dependent manner. As Ecad undergoes constant apical-basal recycling,7,8 we hypothesized that Lm may transit through goblet cells by hijacking Ecad recycling pathway. Indeed, Listeria is stuck at goblet cell apex when Ecad endocytosis is blocked and remains trapped intracellularly at the basolateral pole of goblet cells when Rab11-dependent Ecad recycling is compromised. Together, these results show that Listeria, upon docking onto its luminally accessible receptor Ecad, hijacks its recycling pathway to be transferred by transcytosis across goblet cells. Live imaging of host-pathogen interactions in organoids is a promising approach to dissect their underlying cell and molecular biology.
Collapse
|
41
|
Chen H, Qian W, Good MC. Integrating cellular dimensions with cell differentiation during early development. Curr Opin Cell Biol 2020; 67:109-117. [PMID: 33152556 DOI: 10.1016/j.ceb.2020.08.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/14/2020] [Accepted: 08/03/2020] [Indexed: 11/25/2022]
Abstract
Early embryo development is characterized by alteration of cellular dimensions and fating of blastomeres. An emerging concept is that cell size and shape drive cellular differentiation during early embryogenesis in a variety of model organisms. In this review, we summarize recent advances that elucidate the contribution of the physical dimensions of a cell to major embryonic transitions and cell fate specification in vivo. We also highlight techniques and newly evolving methods for manipulating the sizes and shapes of cells and whole embryos in situ and ex vivo. Finally, we provide an outlook for addressing fundamental questions in the field and more broadly uncovering how changes to cell size control decision making in a variety of biological contexts.
Collapse
Affiliation(s)
- Hui Chen
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wenchao Qian
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Matthew C Good
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
42
|
Kok RNU, Hebert L, Huelsz-Prince G, Goos YJ, Zheng X, Bozek K, Stephens GJ, Tans SJ, van Zon JS. OrganoidTracker: Efficient cell tracking using machine learning and manual error correction. PLoS One 2020; 15:e0240802. [PMID: 33091031 PMCID: PMC7580893 DOI: 10.1371/journal.pone.0240802] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 10/05/2020] [Indexed: 12/30/2022] Open
Abstract
Time-lapse microscopy is routinely used to follow cells within organoids, allowing direct study of division and differentiation patterns. There is an increasing interest in cell tracking in organoids, which makes it possible to study their growth and homeostasis at the single-cell level. As tracking these cells by hand is prohibitively time consuming, automation using a computer program is required. Unfortunately, organoids have a high cell density and fast cell movement, which makes automated cell tracking difficult. In this work, a semi-automated cell tracker has been developed. To detect the nuclei, we use a machine learning approach based on a convolutional neural network. To form cell trajectories, we link detections at different time points together using a min-cost flow solver. The tracker raises warnings for situations with likely errors. Rapid changes in nucleus volume and position are reported for manual review, as well as cases where nuclei divide, appear and disappear. When the warning system is adjusted such that virtually error-free lineage trees can be obtained, still less than 2% of all detected nuclei positions are marked for manual analysis. This provides an enormous speed boost over manual cell tracking, while still providing tracking data of the same quality as manual tracking.
Collapse
Affiliation(s)
| | - Laetitia Hebert
- Okinawa Institute of Science and Technology Graduate University (OIST), Onna-son, Okinawa, Japan
| | | | | | | | - Katarzyna Bozek
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Greg J. Stephens
- Okinawa Institute of Science and Technology Graduate University (OIST), Onna-son, Okinawa, Japan
- Department of Physics and Astronomy, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Sander J. Tans
- AMOLF, Amsterdam, The Netherlands
- Bionanoscience Department, Kavli Institute of Nanoscience Delft, Delft University of Technology, Delft, The Netherlands
| | | |
Collapse
|
43
|
Cammarota C, Finegan TM, Wilson TJ, Yang S, Bergstralh DT. An Axon-Pathfinding Mechanism Preserves Epithelial Tissue Integrity. Curr Biol 2020; 30:5049-5057.e3. [PMID: 33065006 DOI: 10.1016/j.cub.2020.09.061] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 08/21/2020] [Accepted: 09/18/2020] [Indexed: 01/09/2023]
Abstract
Epithelial tissues form the boundaries of organs, where they perform a range of functions, including secretion, absorption, and protection. These tissues are commonly composed of discrete cell layers-sheets of cells that are one-cell thick. In multiple systems examined, epithelial cells round up and move in the apical direction before dividing, likely in response to neighbor-cell crowding [1-6]. Because of this movement, daughter cells may be born displaced from the tissue layer. Reintegration of these displaced cells supports tissue growth and maintains tissue architecture [4]. Two conserved IgCAMs (immunoglobulin superfamily cell adhesion molecules), neuroglian (Nrg) and fasciclin 2 (Fas2), participate in cell reintegration in the Drosophila follicular epithelium [4]. Like their vertebrate orthologs L1CAM and NCAM1/2, respectively, Nrg and Fas2 are cell adhesion molecules primarily studied in the context of nervous system development [7-10]. Consistent with this, we identify another neural IgCAM, Fasciclin 3 (Fas3), as a reintegration factor. Nrg, Fas2, and Fas3 are components of the insect septate junction, the functional equivalent of the vertebrate tight junction, but proliferating follicle cells do not have mature septate junctions, and we find that the septate junction protein neurexin IV does not participate in reintegration [11, 12]. Here, we show that epithelial reintegration works in the same way as IgCAM-mediated axon growth and pathfinding; it relies not only on extracellular adhesion but also mechanical coupling between IgCAMs and the lateral spectrin-based membrane skeleton. Our work indicates that reintegration is mediated by a distinct epithelial adhesion assembly that is compositionally and functionally equivalent to junctions made between axons.
Collapse
Affiliation(s)
- Christian Cammarota
- Department of Physics & Astronomy, University of Rochester, Rochester, NY 14627, USA
| | - Tara M Finegan
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| | - Tyler J Wilson
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| | - Sifan Yang
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| | - Dan T Bergstralh
- Department of Physics & Astronomy, University of Rochester, Rochester, NY 14627, USA; Department of Biology, University of Rochester, Rochester, NY 14627, USA; Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14627, USA.
| |
Collapse
|
44
|
The role of stem cell niche in intestinal aging. Mech Ageing Dev 2020; 191:111330. [DOI: 10.1016/j.mad.2020.111330] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 08/05/2020] [Accepted: 08/07/2020] [Indexed: 12/16/2022]
|
45
|
Fumagalli A, Bruens L, Scheele CLGJ, van Rheenen J. Capturing Stem Cell Behavior Using Intravital and Live Cell Microscopy. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a035949. [PMID: 31767651 DOI: 10.1101/cshperspect.a035949] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Stem cells maintain tissue homeostasis by driving cellular turnover and regeneration upon damage. They reside within specialized niches that provide the signals required for stem cell maintenance. Stem cells have been identified in many tissues and cancer types, but their behavior within the niche and their reaction to microenvironmental signals were inferred from limited static observations. Recent advances in live imaging techniques, such as live cell imaging and intravital microscopy, have allowed the visualization of stem cell behavior and dynamics over time in their (near) native environment. Through these recent technological advances, it is now evident that stem cells are much more dynamic than previously anticipated, resulting in a model in which stemness is a state that can be gained or lost over time. In this review, we will highlight how live imaging and intravital microscopy have unraveled previously unanticipated stem cell dynamics and plasticity during development, homeostasis, regeneration, and tumor formation.
Collapse
Affiliation(s)
- Arianna Fumagalli
- Molecular Pathology, Oncode Institute, the Netherlands Cancer Institute, Amsterdam 1066CX, Netherlands
| | - Lotte Bruens
- Molecular Pathology, Oncode Institute, the Netherlands Cancer Institute, Amsterdam 1066CX, Netherlands
| | - Colinda L G J Scheele
- Molecular Pathology, Oncode Institute, the Netherlands Cancer Institute, Amsterdam 1066CX, Netherlands
| | - Jacco van Rheenen
- Molecular Pathology, Oncode Institute, the Netherlands Cancer Institute, Amsterdam 1066CX, Netherlands
| |
Collapse
|
46
|
Ge Z, Zhao J, Yu H, Yang W, Zhou P, Wang Z, Liu L. Biomimetic construction of peritoneum to imitate peritoneal metastasis using digital micromirror device-based optical projection lithography. LAB ON A CHIP 2020; 20:3109-3119. [PMID: 32661538 DOI: 10.1039/d0lc00361a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Currently, the mechanisms underlying the peritoneal metastasis of gastric cancer cells and the function of mesothelial cells during this process are unclear, primarily due to the absence of an effective in vitro peritoneal model. In this study, we constructed a biomimetic peritoneal model using a digital micromirror device-based optical projection lithography system. This model enabled the simulation of a damaged peritoneum, which allowed for a comparison of the characteristics of an undamaged peritoneum, such as porosity, mechanical properties, and surface morphology, with those of a damaged peritoneum. Biological inertness and removability of the polyethylene glycol dimethacrylate hydrogel were exploited to fabricate an arrayed heterogeneous interface that imitated a damaged human peritoneum. The porous structure of the peritoneum was achieved by adjusting the ratio of collagen I to gelatin methacryloyl; this structure of the peritoneum might contribute to its shock absorption property. Atomic force microscopy characterization showed that the outermost layers of the model peritoneum and real peritoneum were similar in surface morphology and mechanical properties. Furthermore, we reproduced the process of peritoneal metastasis in vitro. The numbers of gastric cancer cells that adhered to the heterogeneous interface were different, and mesothelial cells played an essential role in peritoneal metastasis. Our findings indicate that this model can be utilized in preclinical drug screening and personalized therapy.
Collapse
Affiliation(s)
- Zhixing Ge
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, China. and Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110016, China and University of Chinese Academy of Sciences, Beijing 100049, China
| | - Junhua Zhao
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang 110016, China. and Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang 110016, China
| | - Haibo Yu
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, China. and Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110016, China
| | - Wenguang Yang
- School of Electromechanical and Automotive Engineering, Yantai University, Yantai 264005, China
| | - Peilin Zhou
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, China. and Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110016, China and University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhenning Wang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang 110016, China. and Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang 110016, China
| | - Lianqing Liu
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, China. and Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang 110016, China
| |
Collapse
|
47
|
Liu X, Xu L, Li J, Yao PY, Wang W, Ismail H, Wang H, Liao B, Yang Z, Ward T, Ruan K, Zhang J, Wu Q, He P, Ding X, Wang D, Fu C, Dou Z, Yan F, Wang W, Liu X, Yao X. Mitotic motor CENP-E cooperates with PRC1 in temporal control of central spindle assembly. J Mol Cell Biol 2020; 12:654-665. [PMID: 31174204 PMCID: PMC7683015 DOI: 10.1093/jmcb/mjz051] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 04/21/2019] [Accepted: 04/30/2019] [Indexed: 01/10/2023] Open
Abstract
Error-free cell division depends on the accurate assembly of the spindle midzone from dynamic spindle microtubules to ensure chromatid segregation during metaphase-anaphase transition. However, the mechanism underlying the key transition from the mitotic spindle to central spindle before anaphase onset remains elusive. Given the prevalence of chromosome instability phenotype in gastric tumorigenesis, we developed a strategy to model context-dependent cell division using a combination of light sheet microscope and 3D gastric organoids. Light sheet microscopic image analyses of 3D organoids showed that CENP-E inhibited cells undergoing aberrant metaphase-anaphase transition and exhibiting chromosome segregation errors during mitosis. High-resolution real-time imaging analyses of 2D cell culture revealed that CENP-E inhibited cells undergoing central spindle splitting and chromosome instability phenotype. Using biotinylated syntelin as an affinity matrix, we found that CENP-E forms a complex with PRC1 in mitotic cells. Chemical inhibition of CENP-E in metaphase by syntelin prevented accurate central spindle assembly by perturbing temporal assembly of PRC1 to the midzone. Thus, CENP-E-mediated PRC1 assembly to the central spindle constitutes a temporal switch to organize dynamic kinetochore microtubules into stable midzone arrays. These findings reveal a previously uncharacterized role of CENP-E in temporal control of central spindle assembly. Since CENP-E is absent from yeast, we reasoned that metazoans evolved an elaborate central spindle organization machinery to ensure accurate sister chromatid segregation during anaphase and cytokinesis.
Collapse
Affiliation(s)
- Xu Liu
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Leilei Xu
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
| | - Junying Li
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
| | - Phil Y Yao
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Wanjuan Wang
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
- School of Basic Medical Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Hazrat Ismail
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
| | - Haowei Wang
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
| | - Bryce Liao
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
- Department of Biology, Duke University Durham, NC 27708, USA
| | - Zhihong Yang
- Institute of ProteoGenomics, Beijing 100029, China
| | - Tarsha Ward
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Ke Ruan
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
| | - Jianchun Zhang
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
| | - Quan Wu
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
| | - Ping He
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
| | - Xia Ding
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
- School of Basic Medical Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Dongmei Wang
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Chuanhai Fu
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
| | - Zhen Dou
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Feng Yan
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
| | - Wenwen Wang
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Xing Liu
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Xuebiao Yao
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science, Hefei National Science Center for Physical Sciences at Microscale & University of Science and Technology of China, Hefei 230027, China
| |
Collapse
|
48
|
Abstract
New work reveals that interkinetic nuclear migration - the movement of nuclei towards the apical surface of dividing epithelial cells - is mechanically regulated, relying on a balance of forces between the mitotic cell and the surrounding tissue.
Collapse
Affiliation(s)
- Christian M Cammarota
- Department of Physics and Astronomy, University of Rochester, Rochester, NY 14627, USA
| | - Dan Bergstralh
- Department of Physics and Astronomy, University of Rochester, Rochester, NY 14627, USA; Department of Biology, University of Rochester, Rochester, NY 14627, USA; Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14627, USA.
| |
Collapse
|
49
|
McKinley KL, Castillo-Azofeifa D, Klein OD. Tools and Concepts for Interrogating and Defining Cellular Identity. Cell Stem Cell 2020; 26:632-656. [PMID: 32386555 PMCID: PMC7250495 DOI: 10.1016/j.stem.2020.03.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Defining the mechanisms that generate specialized cell types and coordinate their functions is critical for understanding organ development and renewal. New tools and discoveries are challenging and refining our definitions of a cell type. A rapidly growing toolkit for single-cell analyses has expanded the number of markers that can be assigned to a cell simultaneously, revealing heterogeneity within cell types that were previously regarded as homogeneous populations. Additionally, cell types defined by specific molecular markers can exhibit distinct, context-dependent functions; for example, between tissues in homeostasis and those responding to damage. Here we review the current technologies used to identify and characterize cells, and we discuss how experimental and pathological perturbations are adding increasing complexity to our definitions of cell identity.
Collapse
Affiliation(s)
- Kara L McKinley
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
| | - David Castillo-Azofeifa
- Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, USA; Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Ophir D Klein
- Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, USA; Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA, USA; Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA; Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
50
|
Lough KJ, Byrd KM, Descovich CP, Spitzer DC, Bergman AJ, Beaudoin GM, Reichardt LF, Williams SE. Telophase correction refines division orientation in stratified epithelia. eLife 2019; 8:49249. [PMID: 31833472 PMCID: PMC6959978 DOI: 10.7554/elife.49249] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 12/12/2019] [Indexed: 02/06/2023] Open
Abstract
During organogenesis, precise control of spindle orientation balances proliferation and differentiation. In the developing murine epidermis, planar and perpendicular divisions yield symmetric and asymmetric fate outcomes, respectively. Classically, division axis specification involves centrosome migration and spindle rotation, events occurring early in mitosis. Here, we identify a novel orientation mechanism which corrects erroneous anaphase orientations during telophase. The directionality of reorientation correlates with the maintenance or loss of basal contact by the apical daughter. While the scaffolding protein LGN is known to determine initial spindle positioning, we show that LGN also functions during telophase to reorient oblique divisions toward perpendicular. The fidelity of telophase correction also relies on the tension-sensitive adherens junction proteins vinculin, α-E-catenin, and afadin. Failure of this corrective mechanism impacts tissue architecture, as persistent oblique divisions induce precocious, sustained differentiation. The division orientation plasticity provided by telophase correction may enable progenitors to adapt to local tissue needs.
Collapse
Affiliation(s)
- Kendall J Lough
- Department of Pathology and Laboratory Medicine, Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, United States.,Department of Biology, Lineberger Comprehensive Cancer Centre, The University of North Carolina, Chapel Hill, United States
| | - Kevin M Byrd
- Department of Pathology and Laboratory Medicine, Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, United States.,Department of Biology, Lineberger Comprehensive Cancer Centre, The University of North Carolina, Chapel Hill, United States.,Department of Oral & Craniofacial Health Sciences, The University of North Carolina School of Dentistry, Chapel Hill, United States
| | - Carlos P Descovich
- Department of Pathology and Laboratory Medicine, Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, United States.,Department of Biology, Lineberger Comprehensive Cancer Centre, The University of North Carolina, Chapel Hill, United States
| | - Danielle C Spitzer
- Department of Pathology and Laboratory Medicine, Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, United States.,Department of Biology, Lineberger Comprehensive Cancer Centre, The University of North Carolina, Chapel Hill, United States
| | - Abby J Bergman
- Department of Pathology and Laboratory Medicine, Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, United States.,Department of Biology, Lineberger Comprehensive Cancer Centre, The University of North Carolina, Chapel Hill, United States
| | - Gerard Mj Beaudoin
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, United States.,Department of Physiology, University of California, San Francisco, San Francisco, United States
| | - Louis F Reichardt
- Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, United States.,Department of Physiology, University of California, San Francisco, San Francisco, United States
| | - Scott E Williams
- Department of Pathology and Laboratory Medicine, Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, United States.,Department of Biology, Lineberger Comprehensive Cancer Centre, The University of North Carolina, Chapel Hill, United States
| |
Collapse
|