78 results on '"David Q. Matus"'
Search Results
2. A light sheet fluorescence microscopy protocol for Caenorhabditis elegans larvae and adults
- Author
-
Jayson J. Smith, Isabel W. Kenny, Carsten Wolff, Rachel Cray, Abhishek Kumar, David R. Sherwood, and David Q. Matus
- Subjects
C. elegans ,light sheet fluorescence microscopy ,BIO-133 ,postembryonic development ,timelapse ,Biology (General) ,QH301-705.5 - Abstract
Light sheet fluorescence microscopy (LSFM) has become a method of choice for live imaging because of its fast acquisition and reduced photobleaching and phototoxicity. Despite the strengths and growing availability of LSFM systems, no generalized LSFM mounting protocol has been adapted for live imaging of post-embryonic stages of C. elegans. A major challenge has been to develop methods to limit animal movement using a mounting media that matches the refractive index of the optical system. Here, we describe a simple mounting and immobilization protocol using a refractive-index matched UV-curable hydrogel within fluorinated ethylene propylene (FEP) tubes for efficient and reliable imaging of larval and adult C. elegans stages.
- Published
- 2022
- Full Text
- View/download PDF
3. Rapid Degradation of Caenorhabditis elegans Proteins at Single-Cell Resolution with a Synthetic Auxin
- Author
-
Michael A. Q. Martinez, Brian A. Kinney, Taylor N. Medwig-Kinney, Guinevere Ashley, James M. Ragle, Londen Johnson, Joseph Aguilera, Christopher M. Hammell, Jordan D. Ward, and David Q. Matus
- Subjects
c. elegans ,aid system ,synthetic auxin ,microfluidics ,scf complex ,nhr-25 ,Genetics ,QH426-470 - Published
- 2020
- Full Text
- View/download PDF
4. Imaging multicellular specimens with real-time optimized tiling light-sheet selective plane illumination microscopy
- Author
-
Qinyi Fu, Benjamin L. Martin, David Q. Matus, and Liang Gao
- Subjects
Science - Abstract
Selective plane illumination microscopy (SPIM) is capable of high-resolution, high-speed 3D imaging of single cells, but application to multicellular samples is challenging. Here the authors develop tiling light sheet SPIM to image large multicellular specimens in 3D with subcellular resolution.
- Published
- 2016
- Full Text
- View/download PDF
5. The SWI/SNF chromatin remodeling assemblies BAF and PBAF differentially regulate cell cycle exit and cellular invasion in vivo.
- Author
-
Jayson J Smith, Yutong Xiao, Nithin Parsan, Taylor N Medwig-Kinney, Michael A Q Martinez, Frances E Q Moore, Nicholas J Palmisano, Abraham Q Kohrman, Mana Chandhok Delos Reyes, Rebecca C Adikes, Simeiyun Liu, Sydney A Bracht, Wan Zhang, Kailong Wen, Paschalis Kratsios, and David Q Matus
- Subjects
Genetics ,QH426-470 - Abstract
Chromatin remodelers such as the SWI/SNF complex coordinate metazoan development through broad regulation of chromatin accessibility and transcription, ensuring normal cell cycle control and cellular differentiation in a lineage-specific and temporally restricted manner. Mutations in genes encoding the structural subunits of chromatin, such as histone subunits, and chromatin regulating factors are associated with a variety of disease mechanisms including cancer metastasis, in which cancer co-opts cellular invasion programs functioning in healthy cells during development. Here we utilize Caenorhabditis elegans anchor cell (AC) invasion as an in vivo model to identify the suite of chromatin agents and chromatin regulating factors that promote cellular invasiveness. We demonstrate that the SWI/SNF ATP-dependent chromatin remodeling complex is a critical regulator of AC invasion, with pleiotropic effects on both G0 cell cycle arrest and activation of invasive machinery. Using targeted protein degradation and enhanced RNA interference (RNAi) vectors, we show that SWI/SNF contributes to AC invasion in a dose-dependent fashion, with lower levels of activity in the AC corresponding to aberrant cell cycle entry and increased loss of invasion. Our data specifically implicate the SWI/SNF BAF assembly in the regulation of the G0 cell cycle arrest in the AC, whereas the SWI/SNF PBAF assembly promotes AC invasion via cell cycle-independent mechanisms, including attachment to the basement membrane (BM) and activation of the pro-invasive fos-1/FOS gene. Together these findings demonstrate that the SWI/SNF complex is necessary for two essential components of AC invasion: arresting cell cycle progression and remodeling the BM. The work here provides valuable single-cell mechanistic insight into how the SWI/SNF assemblies differentially contribute to cellular invasion and how SWI/SNF subunit-specific disruptions may contribute to tumorigeneses and cancer metastasis.
- Published
- 2022
- Full Text
- View/download PDF
6. Visualizing the metazoan proliferation-quiescence decision in vivo
- Author
-
Rebecca C Adikes, Abraham Q Kohrman, Michael A Q Martinez, Nicholas J Palmisano, Jayson J Smith, Taylor N Medwig-Kinney, Mingwei Min, Maria D Sallee, Ononnah B Ahmed, Nuri Kim, Simeiyun Liu, Robert D Morabito, Nicholas Weeks, Qinyun Zhao, Wan Zhang, Jessica L Feldman, Michalis Barkoulas, Ariel M Pani, Sabrina L Spencer, Benjamin L Martin, and David Q Matus
- Subjects
cell cycle ,quiescence ,cell proliferation ,G1/G0 ,CDK sensor ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Cell proliferation and quiescence are intimately coordinated during metazoan development. Here, we adapt a cyclin-dependent kinase (CDK) sensor to uncouple these key events of the cell cycle in Caenorhabditis elegans and zebrafish through live-cell imaging. The CDK sensor consists of a fluorescently tagged CDK substrate that steadily translocates from the nucleus to the cytoplasm in response to increasing CDK activity and consequent sensor phosphorylation. We show that the CDK sensor can distinguish cycling cells in G1 from quiescent cells in G0, revealing a possible commitment point and a cryptic stochasticity in an otherwise invariant C. elegans cell lineage. Finally, we derive a predictive model of future proliferation behavior in C. elegans based on a snapshot of CDK activity in newly born cells. Thus, we introduce a live-cell imaging tool to facilitate in vivo studies of cell-cycle control in a wide-range of developmental contexts.
- Published
- 2020
- Full Text
- View/download PDF
7. The Caenorhabditis elegans anchor cell transcriptome: ribosome biogenesis drives cell invasion through basement membrane
- Author
-
Daniel S. Costa, Isabel W. Kenny-Ganzert, Qiuyi Chi, Kieop Park, Laura C. Kelley, Aastha Garde, David Q. Matus, Junhyun Park, Shaul Yogev, Bob Goldstein, Theresa V. Gibney, Ariel M. Pani, and David R. Sherwood
- Subjects
Molecular Biology ,Developmental Biology - Abstract
Cell invasion through basement membrane (BM) barriers is important in development, immune function and cancer progression. As invasion through BM is often stochastic, capturing gene expression profiles of actively invading cells in vivo remains elusive. Using the stereotyped timing of Caenorhabditis elegans anchor cell (AC) invasion, we generated an AC transcriptome during BM breaching. Through a focused RNAi screen of transcriptionally enriched genes, we identified new invasion regulators, including translationally controlled tumor protein (TCTP). We also discovered gene enrichment of ribosomal proteins. AC-specific RNAi, endogenous ribosome labeling and ribosome biogenesis analysis revealed that a burst of ribosome production occurs shortly after AC specification, which drives the translation of proteins mediating BM removal. Ribosomes also enrich near the AC endoplasmic reticulum (ER) Sec61 translocon and the endomembrane system expands before invasion. We show that AC invasion is sensitive to ER stress, indicating a heightened requirement for translation of ER-trafficked proteins. These studies reveal key roles for ribosome biogenesis and endomembrane expansion in cell invasion through BM and establish the AC transcriptome as a resource to identify mechanisms underlying BM transmigration.
- Published
- 2023
- Full Text
- View/download PDF
8. A simple method to dramatically increase C. elegans germline microinjection efficiency
- Author
-
Theresa V. Gibney, Michelle Favichia, Laila Latifi, Taylor N. Medwig-Kinney, David Q. Matus, Daniel C. McIntyre, Angelo B. Arrigo, Kendall R. Branham, Louis T. Bubrig, Abbas Ghaddar, Juliana A. Jiranek, Kendra E. Liu, Charles G. Marcucci, Robert J. Porter, and Ariel M. Pani
- Subjects
Article - Abstract
Genome manipulation methods inC. elegansrequire microinjecting DNA or ribonucleoprotein complexes into the microscopic core of the gonadal syncytium. These microinjections are technically demanding and represent a key bottleneck for all genome engineering and transgenic approaches inC. elegans. While there have been steady improvements in the ease and efficiency of genetic methods forC. elegansgenome manipulation, there have not been comparable advances in the physical process of microinjection. Here, we report a simple and inexpensive method for handling worms using a paintbrush during the injection process that nearly tripled average microinjection rates compared to traditional worm handling methods. We found that the paintbrush increased injection throughput by substantially increasing both injection speeds and post-injection survival rates. In addition to dramatically and universally increasing injection efficiency for experienced personnel, the paintbrush method also significantly improved the abilities of novice investigators to perform key steps in the microinjection process. We expect that this method will benefit theC. eleganscommunity by increasing the speed at which new strains can be generated and will also make microinjection-based approaches less challenging and more accessible to personnel and labs without extensive experience.
- Published
- 2023
9. Breaking theC. elegansinvasion/proliferation dichotomy
- Author
-
Michael A. Q. Martinez, Chris Z. Zhao, Frances E. Q. Moore, Callista Yee, Wan Zhang, Kang Shen, Benjamin L. Martin, and David Q. Matus
- Abstract
The acquisition of the post-mitotic state is crucial for the execution of many terminally differentiated cell behaviors during organismal development. However, the mechanisms that maintain the post-mitotic state in this context remain poorly understood. To gain insight into these mechanisms, we used the genetically and visually accessible model ofC. elegansanchor cell (AC) invasion into the vulval epithelium. The AC is a terminally differentiated uterine cell that must exit the cell cycle and enter a post-mitotic to initiate contact between the uterus and vulva through a cell invasion event. Here, we set out to identify the negative cell cycle regulators that maintain the AC in a post-mitotic, invasive state. Although our findings revealed a critical role for CKI-1 (p21CIP1/p27KIP1) in maintaining the post-mitotic state of the AC, loss of CKI-1 alone or in combination with other negative cell cycle regulators—including CKI-2 (p21CIP1/p27KIP1), LIN-35 (pRb/p107/p130), FZR-1 (Cdh1/Hct1), and LIN-23 (β-TrCP)—resulted in proliferating ACs that retained their invasive abilities. Upon examination of the gene regulatory network controlling AC invasion, we determined that proliferating, invasive ACs maintain pro-invasive gene expression. We therefore report that maintenance of the post-mitotic state is not necessary for AC invasion, breaking the previously establishedC. elegansinvasion/proliferation dichotomy.
- Published
- 2023
- Full Text
- View/download PDF
10. An expandable FLP-ON::TIR1 system for precise spatiotemporal protein degradation in Caenorhabditis elegans
- Author
-
Yutong Xiao, Callista Yee, Chris Z Zhao, Michael A Q Martinez, Wan Zhang, Kang Shen, David Q Matus, and Christopher Hammell
- Subjects
Genetics - Abstract
The auxin-inducible degradation system has been widely adopted in the Caenorhabditis elegans research community for its ability to empirically control the spatiotemporal expression of target proteins. This system can efficiently degrade auxin-inducible degron (AID)-tagged proteins via the expression of a ligand-activatable AtTIR1 protein derived from A. thaliana that adapts target proteins to the endogenous C. elegans proteasome. While broad expression of AtTIR1 using strong, ubiquitous promoters can lead to rapid degradation of AID-tagged proteins, cell type-specific expression of AtTIR1 using spatially restricted promoters often results in less efficient target protein degradation. To circumvent this limitation, we have developed an FLP/FRT3-based system that functions to reanimate a dormant, high-powered promoter that can drive sufficient AtTIR1 expression in a cell type-specific manner. We benchmark the utility of this system by generating a number of tissue-specific FLP-ON::TIR1 drivers to reveal genetically separable cell type-specific phenotypes for several target proteins. We also demonstrate that the FLP-ON::TIR1 system is compatible with enhanced degron epitopes. Finally, we provide an expandable toolkit utilizing the basic FLP-ON::TIR1 system that can be adapted to drive optimized AtTIR1 expression in any tissue or cell type of interest.
- Published
- 2023
- Full Text
- View/download PDF
11. Loss of the E3 ubiquitin ligases UBR-5 or HECD-1 restores Caenorhabditis elegans development in the absence of SWI/SNF function
- Author
-
Lisa Lampersberger, Francesca Conte, Subhanita Ghosh, Yutong Xiao, Jonathan Price, David Jordan, David Q Matus, Peter Sarkies, Petra Beli, Eric A Miska, Nicholas O Burton, Xiao, Yutong [0000-0001-7279-3423], Matus, David Q [0000-0002-1570-5025], Miska, Eric A [0000-0002-4450-576X], and Apollo - University of Cambridge Repository
- Subjects
SWI/SNF ,Multidisciplinary ,Ubiquitin-Protein Ligases ,C. elegans ,Animals ,Humans ,HECD-1 ,Caenorhabditis elegans ,Caenorhabditis elegans Proteins ,development ,Ubiquitins ,UBR-5 - Abstract
SWItch/sucrose non-fermenting (SWI/SNF) complexes are a family of chromatin remodelers that are conserved across eukaryotes. Mutations in subunits of SWI/SNF cause a multitude of different developmental disorders in humans, most of which have no current treatment options. Here, we identify an alanine-to-valine–causing mutation in the SWI/SNF subunit snfc-5 ( SMARCB1 in humans) that prevents embryonic lethality in Caenorhabditis elegans nematodes harboring a loss-of-function mutation in the SWI/SNF subunit swsn-1 ( SMARCC1/2 in humans). Furthermore, we found that the combination of this specific mutation in snfc-5 and a loss-of-function mutation in either of the E3 ubiquitin ligases ubr-5 ( UBR5 in humans) or hecd-1 ( HECTD1 in humans) can restore development to adulthood in swsn-1 loss-of-function mutants that otherwise die as embryos. Using these mutant models, we established a set of 335 genes that are dysregulated in SWI/SNF mutants that arrest their development embryonically but exhibit near wild-type levels of expression in the presence of suppressor mutations that prevent embryonic lethality, suggesting that SWI/SNF promotes development by regulating some subset of these 335 genes. In addition, we show that SWI/SNF protein levels are reduced in swsn-1; snfc-5 double mutants and partly restored to wild-type levels in swsn-1; snfc-5; ubr-5 triple mutants, consistent with a model in which UBR-5 regulates SWI/SNF levels by tagging the complex for proteasomal degradation. Our findings establish a link between two E3 ubiquitin ligases and SWI/SNF function and suggest that UBR5 and HECTD1 could be potential therapeutic targets for the many developmental disorders caused by missense mutations in SWI/SNF subunits.
- Published
- 2023
12. TheC. elegansAnchor Cell Transcriptome: Ribosome Biogenesis Drives Cell Invasion through Basement Membrane
- Author
-
Daniel S. Costa, Isabel W. Kenny-Ganzert, Qiuyi Chi, Kieop Park, Laura C. Kelley, Aastha Garde, David Q. Matus, Junhyun Park, Shaul Yogev, Bob Goldstein, Theresa V. Gibney, Ariel M. Pani, and David R. Sherwood
- Abstract
Cell invasion through basement membrane (BM) barriers is important in development, immune function, and cancer progression. As invasion through BM is often stochastic, capturing gene expression profiles of cells actively transmigrating BMin vivoremains elusive. Using the stereotyped timing ofC. elegansanchor cell (AC) invasion, we generated an AC transcriptome during BM breaching. Through a focused RNAi screen of transcriptionally enriched genes, we identified new invasion regulators, including TCTP (Translationally Controlled Tumor Protein). We also discovered gene enrichment of ribosomal proteins. AC-specific RNAi, endogenous ribosome labeling, and ribosome biogenesis analysis revealed a burst of ribosome production occurs shortly after AC specification, which drives the translation of proteins mediating BM removal. Ribosomes also strongly localize to the AC’s endoplasmic reticulum (ER) and the endomembrane system expands prior to invasion. We show that AC invasion is sensitive to ER stress, indicating a heightened requirement for translation of ER trafficked proteins. These studies reveal key roles for ribosome biogenesis and endomembrane expansion in cell invasion through BM and establish the AC transcriptome as a resource to identify mechanisms underlying BM transmigration.
- Published
- 2022
- Full Text
- View/download PDF
13. An expandable FLP-ON::TIR1 system for precise spatiotemporal protein degradation inC. elegans
- Author
-
Yutong Xiao, Callista Yee, Michael A. Q. Martinez, Chris Z. Zhao, Wan Zhang, Kang Shen, David Q. Matus, and Christopher Hammell
- Abstract
The auxin-inducible degradation system has been widely adopted in theC. elegansresearch community for its ability to empirically control the spatiotemporal expression of target proteins. This system can efficiently degradeauxin-inducibledegron (AID)-tagged proteins via the expression of a ligand-activatableAtTIR1 protein derived fromA. thalianathat adapts target proteins to the endogenousC. elegansproteosome. While broad expression ofAtTIR1 using strong, ubiquitous promoters can lead to rapid degradation of AID-tagged proteins, cell type-specific expression ofAtTIR1 using spatially restricted promoters often results in less efficient target protein degradation. To circumvent this limitation, we have developed a FLP/FRT3-based system that functions to reanimate a dormant, high-powered promoter that can drive sufficientAtTIR1expression in a cell type-specific manner. We benchmark the utility of this system by generating a number of tissue specific FLP-ON::TIR1 drivers to reveal genetically separable cell type-specific phenotypes for several target proteins. We also demonstrate that the FLP-ON::TIR1 system is compatible with enhanced degron epitopes. Finally, we provide an expandable toolkit utilizing the basic FLP-ON::TIR1 system that can be adapted to drive optimizedAtTIR1expression in any tissue or cell type of interest.
- Published
- 2022
- Full Text
- View/download PDF
14. A reevaluation of the relationship between EGL-43 (EVI1/MECOM) and LIN-12 (Notch) duringC. elegansanchor cell invasion
- Author
-
Michael A. Q. Martinez, Angelina A. Mullarkey, Callista Yee, Chris Z. Zhao, Wan Zhang, Kang Shen, and David Q. Matus
- Abstract
Development of theC. elegansreproductive tract is orchestrated by the anchor cell (AC). Among other things, this occurs through a cell invasion event that connects the uterine and vulval tissue. Several key transcription factors regulate AC invasion, such as EGL-43, HLH-2, and NHR-67. Specifically, these transcription factors function together to maintain the post-mitotic state of the AC, a requirement for AC invasion. EGL-43 is theC. eleganshomolog of the human EVI1/MECOM proto-oncogene, and recently, a mechanistic connection has been made between its loss and AC cell-cycle entry. The current model states that EGL-43 represses LIN-12 (Notch) expression to prevent AC proliferation, suggesting that Notch signaling is mitogenic in the absence of EGL-43. To reevaluate the relationship between EGL-43 and LIN-12, we designed and implemented a heterologous co-expression system called AIDHB that combines the auxin-inducible degron (AID) system of plants with a live cell-cycle sensor based on human DNA helicase B (DHB). After validating the AIDHB approach using AID-tagged GFP, we sought to test this approach using AID-tagged alleles ofegl-43andlin-12. Auxin-inducible degradation of either EGL-43 or LIN-12 resulted in the expected AC phenotypes. Lastly, we seized the opportunity to pair AIDHB with RNAi to co-deplete LIN-12 and EGL-43, respectively. This combined approach revealed that LIN-12 is not required for AC proliferation following loss of EGL-43, which contrasts with a double RNAi experiment directed against these same targets. The addition of AIDHB to theC. eleganstransgenic toolkit should facilitate functionalin vivoimaging of cell-cycle associated phenomena.
- Published
- 2022
- Full Text
- View/download PDF
15. CDK activity sensors: genetically encoded ratiometric biosensors for live analysis of the cell cycle
- Author
-
Michael A. Q. Martinez and David Q. Matus
- Subjects
Cyclins ,Cell Cycle ,Animals ,Biosensing Techniques ,Biochemistry ,Article ,Cell Division ,Cyclin-Dependent Kinases - Abstract
Cyclin-dependent kinase (CDK) sensors have facilitated investigations of the cell cycle in living cells. These genetically encoded fluorescent biosensors change their subcellular location upon activation of CDKs. Activation is primarily regulated by their association with cyclins, which in turn trigger cell-cycle progression. In the absence of CDK activity, cells exit the cell cycle and become quiescent, a key step in stem cell maintenance and cancer cell dormancy. The evolutionary conservation of CDKs has allowed for the rapid development of CDK activity sensors for cell lines and several research organisms, including nematodes, fish, and flies. CDK activity sensors are utilized for their ability to visualize the exact moment of cell-cycle commitment. This has provided a breakthrough in understanding the proliferation-quiescence decision. Further adoption of these biosensors will usher in new discoveries focused on the cell-cycle regulation of development, ageing, and cancer.
- Published
- 2022
16. An
- Author
-
Taylor N, Medwig-Kinney, Sydney S, Sirota, Theresa V, Gibney, Ariel M, Pani, and David Q, Matus
- Abstract
Notch/Delta signaling regulates numerous cell-cell interactions that occur during development, homeostasis, and in disease states. In many cases, the Notch/Delta pathway mediates lateral inhibition between cells to specify alternative fates. Here, we provide new tools for use in
- Published
- 2022
17. A new toolkit to visualize and perturb endogenous LIN-12/Notch signaling in
- Author
-
Ariel M, Pani, Theresa V, Gibney, Taylor N, Medwig-Kinney, David Q, Matus, and Bob, Goldstein
- Abstract
Notch signaling mediates cell-cell interactions during development and homeostasis. Methods for visualizing and manipulating Notch activity
- Published
- 2022
18. The Caenorhabditis elegans centrosome is surrounded by a membrane reticulum, the centriculum, that affects centrosome size and function
- Author
-
Richa Maheshwari, Mohammad M. Rahman, Seth Drey, Megan Onyundo, Gunar Fabig, Michael A. Q. Martinez, David Q. Matus, Thomas Müller-Reichert, and Orna Cohen-Fix
- Abstract
SummaryCentrosomes are membraneless organelles that nucleate microtubules. At their core is a pair of centrioles that recruit pericentriolar material (PCM), a phase-separated condensate. In many cell types, including human cells, centrosomes are surrounded by endoplasmic reticulum-derived membranes of unknown structure and function. Using volume electron microscopy, we show that the C. elegans centrosome is surrounded by a membrane reticulum that we call the centriculum, for centrosome-associated membrane reticulum. Increasing centriculum size by genetic means led to expansion of the PCM and increased microtubule nucleation capacity, an unexpected finding given that the PCM is a membraneless condensate. We provide evidence that the centriculum serves as a microtubule “filter” by limiting the number of microtubules that can elongate fully. We also show the centriculum fuses with the nuclear envelope during mitosis. We propose that this fusion contributes to nuclear envelope breakdown by transducing forces from the elongating spindle to the nuclear membranes.
- Published
- 2022
- Full Text
- View/download PDF
19. An engineered, orthogonal auxin analog/AtTIR1(F79G) pairing improves both specificity and efficacy of the auxin degradation system in Caenorhabditis elegans
- Author
-
David Q. Matus, Shilpa Hebbar, Christopher M. Hammell, Maria Ivanova, Jordan D. Ward, Eric G. Moss, Sevinc Ercan, Taylor N. Medwig-Kinney, Ana Karina Morao, Frances E. Q. Moore, Kelly Hills-Muckey, Natalia Stec, Anna Y. Zinovyeva, Michael A. Q. Martinez, Joanne Saldanha, and Buelow, H
- Subjects
Mutant ,Arabidopsis ,Bioengineering ,Biology ,Protein degradation ,Auxin ,Genetics ,Animals ,heterocyclic compounds ,Caenorhabditis elegans ,Caenorhabditis elegans Proteins ,Cas9 ,CRISPR/Cas9 ,chemistry.chemical_classification ,Indoleacetic Acids ,Arabidopsis Proteins ,AID system ,F-Box Proteins ,elegans ,targeted degradation ,food and beverages ,biology.organism_classification ,Transport inhibitor ,Cell biology ,chemistry ,Proteasome ,heterochronic ,CRISPR ,C. elegans ,Heterologous expression ,Degron ,RNA pol II inhibition ,auxin ,Biotechnology ,Developmental Biology - Abstract
The auxin-inducible degradation system in C. elegans allows for spatial and temporal control of protein degradation via heterologous expression of a single Arabidopsis thaliana F-box protein, transport inhibitor response 1 (AtTIR1). In this system, exogenous auxin (Indole-3-acetic acid; IAA) enhances the ability of AtTIR1 to function as a substrate recognition component that adapts engineered degron-tagged proteins to the endogenous C. elegans E3 ubiquitin ligases complex [SKR-1/2-CUL-1-F-box (SCF)], targeting them for degradation by the proteosome. While this system has been employed to dissect the developmental functions of many C. elegans proteins, we have found that several auxin-inducible degron (AID)-tagged proteins are constitutively degraded by AtTIR1 in the absence of auxin, leading to undesired loss-of-function phenotypes. In this manuscript, we adapt an orthogonal auxin derivative/mutant AtTIR1 pair [C. elegans AID version 2 (C.e.AIDv2)] that transforms the specificity of allosteric regulation of TIR1 from IAA to one that is dependent on an auxin derivative harboring a bulky aryl group (5-Ph-IAA). We find that a mutant AtTIR1(F79G) allele that alters the ligand-binding interface of TIR1 dramatically reduces ligand-independent degradation of multiple AID*-tagged proteins. In addition to solving the ectopic degradation problem for some AID-targets, the addition of 5-Ph-IAA to culture media of animals expressing AtTIR1(F79G) leads to more penetrant loss-of-function phenotypes for AID*-tagged proteins than those elicited by the AtTIR1-IAA pairing at similar auxin analog concentrations. The improved specificity and efficacy afforded by the mutant AtTIR1(F79G) allele expand the utility of the AID system and broaden the number of proteins that can be effectively targeted with it.
- Published
- 2022
20. A laboratory module that explores RNA interference and codon optimization through fluorescence microscopy using Caenorhabditis elegans
- Author
-
Maryam A. Azmi, David Q. Matus, Wan Zhang, Taylor N. Medwig-Kinney, Rebecca C. Adikes, Rumana Rahman, Frances E. Q. Moore, and Nicholas J. Palmisano
- Subjects
Critical thinking ,biology ,Human–computer interaction ,RNA interference ,Computer science ,Codon optimization ,General Medicine ,biology.organism_classification ,Gene ,Organism ,Caenorhabditis elegans - Abstract
Scientific research experiences are beneficial to students allowing them to gain laboratory and problem-solving skills, as well as foundational research skills in a team-based setting. We designed a laboratory module to provide a guided research experience to stimulate curiosity, introduce students to experimental techniques, and provide students with foundational skills needed for higher levels of guided inquiry. In this laboratory module, students learn about RNA interference (RNAi) and codon optimization using the research organism Caenorhabditis elegans (C. elegans). Students are given the opportunity to perform a commonly used method of gene downregulation in C. elegans where they visualize gene depletion using fluorescence microscopy and quantify the efficacy of depletion using quantitative image analysis. The module presented here educates students on how to report their results and findings by generating publication quality figures and figure legends. The activities outlined exemplify ways by which students can improve their critical thinking, data interpretation, and technical skills, all of which are beneficial for future laboratory classes, independent inquiry-based research projects, and careers in the life sciences and beyond.
- Published
- 2022
- Full Text
- View/download PDF
21. A membrane reticulum, the centriculum, affects centrosome size and function in Caenorhabditis elegans
- Author
-
Richa Maheshwari, Mohammad M. Rahman, Seth Drey, Megan Onyundo, Gunar Fabig, Michael A.Q. Martinez, David Q. Matus, Thomas Müller-Reichert, and Orna Cohen-Fix
- Subjects
General Agricultural and Biological Sciences ,General Biochemistry, Genetics and Molecular Biology - Published
- 2023
- Full Text
- View/download PDF
22. Cyclin-Dependent Kinase Sensor Transgenic Zebrafish Lines for Improved Cell Cycle State Visualization in Live Animals
- Author
-
Robert D Morabito, David Q. Matus, Benjamin L. Martin, and Rebecca C. Adikes
- Subjects
biology ,Cell Cycle ,Cyclin-Dependent Kinase 4 ,Cell cycle ,Zebrafish Proteins ,biology.organism_classification ,Cyclin-Dependent Kinases ,Cell biology ,Animals, Genetically Modified ,Cyclin-dependent kinase ,Transgenic zebrafish ,biology.protein ,Animals ,Animal Science and Zoology ,TechnoFish ,Zebrafish ,Developmental Biology - Published
- 2021
23. Deletion of a putative HDA-1 binding site in the
- Author
-
Taylor N, Medwig-Kinney, Nicholas J, Palmisano, and David Q, Matus
- Subjects
endocrine system ,New Finding ,hemic and lymphatic diseases ,fungi ,Interaction Data ,musculoskeletal system ,Phenotype Data ,Expression Data ,C. Elegans - Abstract
The helix-loop-helix transcription factor hlh-2 (E/Daughterless) has been shown to play an important role in regulating cell fate patterning, cell cycle, and basement membrane invasion in the context of the development of the C. elegans somatic gonad. Here, using CRISPR/Cas9 genome engineering, we generated a new hlh-2 allele (hlh-2(Δ-1303-702)) in the endogenous, GFP-tagged hlh-2 locus. This allele represents a deletion of a 601 bp region in the hlh-2 promoter that contains a putative binding site of the histone deacetylase hda-1 (HDAC) according to publicly available ChIP-sequencing data. Strikingly, we find that HLH-2 expression is virtually absent in the dorsal uterine cells of hlh-2(Δ-1303-702) animals compared to wild type controls. Levels of HLH-2 in the anchor cell and ventral uterine cells are only modestly reduced in the mutant; however, this does not seem to be functionally significant based on the lack of relevant phenotypes and expression levels of a downstream gene, NHR-67 (TLX/Tailless/NR2E1), in these cells. Taken together, these results support growing evidence that HDACs can potentially positively regulate transcription and provide a new reagent for studying hlh-2 regulation.
- Published
- 2021
24. An engineered, orthogonal auxin analog/AtTIR1(F79G) pairing improves both specificity and efficacy of the auxin degradation system inCaenorhabditis elegans
- Author
-
Shilpa Hebbar, Natalia Stec, Jordan D. Ward, David Q. Matus, Kelly Hills-Muckey, Taylor N. Medwig-Kinney, Michael A. Q. Martinez, Mariia Ivanova, Eric G. Moss, Frances E. Q. Moore, Joanne Saldanha, Anna Y. Zinovyeva, Christopher M. Hammell, Ana Moraro, and Sevinc Ercan
- Subjects
chemistry.chemical_classification ,biology ,Mutant ,food and beverages ,Protein degradation ,biology.organism_classification ,Transport inhibitor ,Cell biology ,chemistry ,Ubiquitin ,Proteasome ,Auxin ,biology.protein ,Degron ,Caenorhabditis elegans - Abstract
The auxin-inducible degradation system inC. elegansallows for spatial and temporal control of protein degradation via heterologous expression of a singleArabidopsis thalianaF-box protein, transport inhibitor response 1 (AtTIR1). In this system, exogenous auxin (Indole-3-acetic acid; IAA) enhances the ability ofAtTIR1 to function as a substrate recognition component that adapts engineered degron-tagged proteins to the endogenousC. elegansE3 ubiquitin ligases complex (SKR-1/2-CUL-1-F-box (SCF)), targeting them for degradation by the proteosome. While this system has been employed to dissect the developmental functions of manyC. elegansproteins, we have found that several auxin-inducible degron (AID)-tagged proteins are constitutively degraded byAtTIR1 in the absence of auxin, leading to undesired loss-of-function phenotypes. In this manuscript, we adapt an orthogonal auxin-derivative/mutantAtTIR1 pair (C. elegansAID version 2 (C.e.AIDv2)) that transforms the specificity of allosteric regulation of TIR1 from IAA to one that is dependent on an auxin derivative harboring a bulky aryl group (5-Ph-IAA). We find that a mutantAtTIR1(F79G) allele that alters the ligand binding interface of TIR1 dramatically reduces ligand-independent degradation of multiple AID*-tagged proteins. In addition to solving the ectopic degradation problem for some AID targets, addition of 5-Ph-IAA to culture media of animals expressingAtTIR1(F79G) leads to more penetrant loss-of-function phenotypes for AID*-tagged proteins than those elicited by theAtTIR1-IAA pairing at similar auxin analog concentrations. The improved specificity and efficacy afforded by the mutantAtTIR1(F79G) allele expands the utility of the AID system and broadens the number of proteins that can be effectively targeted with it.ARITCLE SUMMARYImplementation of the auxin induced degradation (AID) system has increased the power if theC. elegansmodel through its ability to rapidly degrade target proteins in the presence of the plant hormone auxin (IAA). The currentC.e.AID system is limited in that a substantial level of target degradation occurs in the absence of ligand and full levels of target protein degradation require high levels of auxin inducer. In this manuscript, we modify the AID system to solve these problems.
- Published
- 2021
- Full Text
- View/download PDF
25. A proliferative to invasive switch is mediated by srGAP1 downregulation through the activation of TGF-β2 signaling
- Author
-
Chandrani Mondal, Majo J. Gacha-Garay, Kathryn A. Larkin, Rebecca C. Adikes, Julie S. Di Martino, Chen-Chi Chien, Madison Fraser, Ireti Eni-aganga, Esperanza Agullo-Pascual, Katarzyna Cialowicz, Umut Ozbek, Alexandra Naba, Angelo Gaitas, Tian-Ming Fu, Srigokul Upadhyayula, Eric Betzig, David Q. Matus, Benjamin L. Martin, and Jose Javier Bravo-Cordero
- Subjects
Mice ,Transforming Growth Factor beta2 ,Cell Line, Tumor ,Animals ,Down-Regulation ,General Biochemistry, Genetics and Molecular Biology ,Actins ,Zebrafish - Abstract
Many breast cancer (BC) patients suffer from complications of metastatic disease. To form metastases, cancer cells must become migratory and coordinate both invasive and proliferative programs at distant organs. Here, we identify srGAP1 as a regulator of a proliferative-to-invasive switch in BC cells. High-resolution light-sheet microscopy demonstrates that BC cells can form actin-rich protrusions during extravasation. srGAP1
- Published
- 2021
26. The SWI/SNF chromatin remodeling assemblies BAF and PBAF differentially regulate cell cycle exit and cellular invasion in vivo
- Author
-
Reyes Mcd, Paschalis Kratsios, Rebecca C. Adikes, Wen K, Parsan N, Kohrman Aq, David Q. Matus, Jayson J. Smith, Bracht Sa, Moore Feq, Taylor N. Medwig-Kinney, Xiao Y, Liu S, Wan Zhang, Nicholas J. Palmisano, and Martinez Maq
- Subjects
enzymes and coenzymes (carbohydrates) ,Histone ,Cell cycle checkpoint ,biology ,Cellular differentiation ,genetic processes ,biology.protein ,Cell cycle ,Protein degradation ,SWI/SNF ,Chromatin remodeling ,Cell biology ,Chromatin - Abstract
SUMMARYChromatin remodelers such as the SWI/SNF complex coordinate metazoan development through broad regulation of chromatin accessibility and transcription, ensuring normal cell cycle control and cellular differentiation in a lineage-specific and temporally restricted manner. Mutations in genes encoding the structural subunits of chromatin, such as histone subunits, and chromatin regulating factors (CRFs) are associated with a variety of disease mechanisms including cancer metastasis, in which cancer co-opts cellular invasion programs functioning in healthy cells during development. Here we utilize Caenorhabditis elegans anchor cell (AC) invasion as an in vivo model to identify the suite of chromatin agents and CRFs that promote cellular invasiveness. We demonstrate that the SWI/SNF ATP-dependent chromatin remodeling complex is a critical regulator of AC invasion, with pleiotropic effects on both G0 cell cycle arrest and activation of invasive machinery. Using targeted protein degradation and enhanced RNA interference (RNAi) vectors, we show that SWI/SNF contributes to AC invasion in a dose-dependent fashion, with lower levels of activity in the AC corresponding to aberrant cell cycle entry and increased loss of invasion. Our data specifically implicate the SWI/SNF BAF assembly in the regulation of the G0 cell cycle arrest in the AC, whereas the SWI/SNF PBAF assembly promotes AC invasion via cell cycle-independent mechanisms, including attachment to the basement membrane (BM) and activation of the pro-invasive fos-1/FOS gene. Together these findings demonstrate that the SWI/SNF complex is necessary for two essential components of AC invasion: arresting cell cycle progression and remodeling the BM. The work here provides valuable single-cell mechanistic insight into how the SWI/SNF assemblies differentially contribute to cellular invasion and how SWI/SNF subunit-specific disruptions may contribute to tumorigeneses and cancer metastasis.SUMMARY STATEMENTCellular invasion through the basement membrane by the C. elegans anchor cell requires both BAF and PBAF SWI/SNF assemblies to arrest the cell cycle and promote the expression of pro-invasive genes.
- Published
- 2021
- Full Text
- View/download PDF
27. Visualizing the metazoan proliferation-quiescence decision in vivo
- Author
-
Robert D Morabito, Nicholas J. Palmisano, Qinyun Zhao, Mingwei Min, Nicholas Weeks, Wan Zhang, Rebecca C. Adikes, Benjamin L. Martin, Jessica L. Feldman, Taylor N. Medwig-Kinney, Nuri Kim, Jayson J. Smith, Sabrina L. Spencer, Abraham Q. Kohrman, Ariel M. Pani, Michael A. Q. Martinez, Michalis Barkoulas, Simeiyun Liu, Ononnah B. Ahmed, Maria D. Sallee, and David Q. Matus
- Subjects
0301 basic medicine ,Cell type ,Cell division ,QH301-705.5 ,Science ,Cell ,Biosensing Techniques ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Cyclin-dependent kinase ,medicine ,Animals ,quiescence ,Biology (General) ,Caenorhabditis elegans ,Caenorhabditis elegans Proteins ,Zebrafish ,General Immunology and Microbiology ,biology ,Cell growth ,General Neuroscience ,fungi ,Cell Cycle ,food and beverages ,Cell Biology ,General Medicine ,Cell cycle ,biology.organism_classification ,Cyclin-Dependent Kinases ,Cell biology ,cell proliferation ,030104 developmental biology ,medicine.anatomical_structure ,g1/g0 ,C. elegans ,biology.protein ,Medicine ,cdk sensor ,Insight ,Developmental biology ,Cell Division ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
All living things are made up of cells that form the different tissues, organs and structures of an organism. The human body, for example, is thought to consist of some 37 trillion cells and harbor over 200 cell types. To maintain a working organism, cells divide to create new cells and replace the ones that have died. Cell division is a tightly controlled process consisting of several steps, and cells continuously face a Shakespearean dilemma of deciding whether to continue dividing (also known as cell proliferation) or to halt the process (known as quiescence). This difficult balancing act is critical during all stages of life, from embryonic development to tissue growth in an adult. Problems in the underlying pathways can result in diseases such as cancer. Cell division is driven by proteins called CDKs, which help cells to complete their cell cycle in the correct sequence. To gain more insight into this complex process, scientists have developed tools for monitoring CDKs. One such tool is a fluorescent biosensor, a molecule that can be inserted into cells that glows and moves in response to CDK activity. The biosensor can be studied and measured in each cell using a microscope. Adikes, Kohrman, Martinez et al. adapted and optimized an existing CDK biosensor to help study cell division and the switch between proliferation and quiescence in two common research organisms, the nematode Caenorhabditis elegans and the zebrafish. Analysis of this biosensor showed that CDK activity at the end of cell division is higher if the cells will divide again but is low if the cells are going to become quiescent. This could suggest that the decision of a cell between proliferation and quiescence may happen earlier than expected. The optimized biosensor is sensitive enough to detect these differences and can even measure variations that influence proliferation in a region on C. elegans that was once thought to be unchanging. The development of this biosensor provides a useful research tool that could be used in other living organisms. Many research questions relate to cell division and so the applications of this tool are wide ranging.
- Published
- 2020
- Full Text
- View/download PDF
28. Author response: Visualizing the metazoan proliferation-quiescence decision in vivo
- Author
-
Qinyun Zhao, Abraham Q. Kohrman, Ariel M. Pani, Taylor N. Medwig-Kinney, Jayson J. Smith, Sabrina L. Spencer, Ononnah B. Ahmed, Nicholas Weeks, Maria D. Sallee, Nuri Kim, Robert D Morabito, David Q. Matus, Nicholas J. Palmisano, Jessica L. Feldman, Michael A. Q. Martinez, Michalis Barkoulas, Benjamin L. Martin, Rebecca C. Adikes, Wan Zhang, Mingwei Min, and Simeiyun Liu
- Subjects
In vivo ,Biology ,Cell biology - Published
- 2020
- Full Text
- View/download PDF
29. An expanded auxin-inducible degron toolkit for Caenorhabditis elegans
- Author
-
Daniel J. Dickinson, Bob Goldstein, Raquel Martinez-Mendez, Max T. Levenson, Michael A. Q. Martinez, Nicholas J. Palmisano, David J. Reiner, Ryan Doonan, Jordan D. Ward, Hannah N. Saeger, Taylor N. Medwig-Kinney, Tam Duong, Wan Zhang, Jonathan D. Hibshman, Guinevere E Ashley, Londen C. Johnson, Brittany R Davidson, Sydney S. Sirota, James Matthew Ragle, David Q. Matus, and Greenstein, D
- Subjects
AcademicSubjects/SCI01140 ,AcademicSubjects/SCI00010 ,AcademicSubjects/SCI01180 ,0302 clinical medicine ,Plasmid ,Genome editing ,Genes, Reporter ,Receptors ,CRISPR ,Transgenes ,Caenorhabditis elegans ,Genetics ,0303 health sciences ,biology ,AID system ,Experimental Technologies and Resources ,Organ Specificity ,Cell Surface ,C. elegans ,Genetic Engineering ,Biotechnology ,Cloning vector ,Receptors, Cell Surface ,Computational biology ,03 medical and health sciences ,Animals ,Caenorhabditis elegans Proteins ,Reporter ,Gene ,CRISPR/Cas9 ,030304 developmental biology ,Investigation ,Indoleacetic Acids ,Cas9 ,Arabidopsis Proteins ,F-Box Proteins ,SapTrap ,biology.organism_classification ,self-excising cassette ,Luminescent Proteins ,Genes ,Transport Inhibitor Response 1 ,Proteolysis ,AcademicSubjects/SCI00960 ,Generic health relevance ,Degron ,CRISPR-Cas Systems ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
The auxin-inducible degron (AID) system has emerged as a powerful tool to conditionally deplete proteins in a range of organisms and cell types. Here, we describe a toolkit to augment the use of the AID system in Caenorhabditis elegans. We have generated a set of single-copy, tissue-specific (germline, intestine, neuron, muscle, pharynx, hypodermis, seam cell, anchor cell) and pan-somatic TIR1-expressing strains carrying a co-expressed blue fluorescent reporter to enable use of both red and green channels in experiments. These transgenes are inserted into commonly used, well-characterized genetic loci. We confirmed that our TIR1-expressing strains produce the expected depletion phenotype for several nuclear and cytoplasmic AID-tagged endogenous substrates. We have also constructed a set of plasmids for constructing repair templates to generate fluorescent protein::AID fusions through CRISPR/Cas9-mediated genome editing. These plasmids are compatible with commonly used genome editing approaches in the C. elegans community (Gibson or SapTrap assembly of plasmid repair templates or PCR-derived linear repair templates). Together these reagents will complement existing TIR1 strains and facilitate rapid and high-throughput fluorescent protein::AID tagging of genes. This battery of new TIR1-expressing strains and modular, efficient cloning vectors serves as a platform for straightforward assembly of CRISPR/Cas9 repair templates for conditional protein depletion.
- Published
- 2020
30. Auxin-mediated Protein Degradation in
- Author
-
Michael A Q, Martinez and David Q, Matus
- Subjects
food and beverages ,Article - Abstract
The auxin-inducible degron (AID) technology was recently adapted for use in the nematode Caenorhabditis elegans. Rapid degradation of C. elegans proteins tagged with an AID is mediated by a plant-specific F-box protein, transport inhibitor response 1 (TIR1), and occurs only in the presence of the phytohormone auxin. The first iteration of this technology elicited protein degradation in C. elegans through a naturally occurring form of auxin, indole-3-acetic acid (IAA). Here, we present a protocol that uses 1-naphthaleneacetic acid, potassium salt (K-NAA), an indole-free synthetic auxin analog. At equal concentration, K-NAA is as effective as IAA in standard nematode growth media (NGM). K-NAA is also effective in physiological buffer (M9), allowing for high-throughput experimentation. The main advantages of K-NAA are twofold: first, its photostability prevents light-induced compound degradation during storage and the production of toxic indole-derivatives during fluorescence microscopy of live cells; and second, its water solubility eliminates the need of using ethanol to dissolve the auxin compound, a solvent that may confound C. elegans lifespan and behavioral assays. In this protocol, we describe our method of degrading C. elegans proteins using K-NAA on solid and in liquid media, as well as our method of analyzing protein degradation.
- Published
- 2020
31. Expanding the Caenorhabditis elegans auxin-inducible degron system toolkit with internal expression and degradation controls and improved modular constructs for CRISPR/Cas9-mediated genome editing
- Author
-
James Matthew Ragle, Raquel Martinez-Mendez, Max T. Levenson, Ryan Doonan, Wan Zhang, Sydney S. Sirota, David J. Reiner, Brittany R Davidson, Bob Goldstein, Nicholas J. Palmisano, Jordan D. Ward, Hannah N. Saeger, Tam Duong, David Q. Matus, Daniel J. Dickinson, Michael A. Q. Martinez, Jonathan D. Hibshman, Guinevere Ashley, and Taylor N. Medwig-Kinney
- Subjects
Gibson assembly ,Plasmid ,biology ,Genome editing ,Cas9 ,Cloning vector ,CRISPR ,Computational biology ,Degron ,biology.organism_classification ,Caenorhabditis elegans - Abstract
The auxin-inducible degron (AID) system has emerged as a powerful tool to conditionally deplete proteins in a range of organisms and cell-types. Here, we describe a toolkit to augment the use of the AID system inCaenorhabditis elegans. We have generated a set of single-copy, tissue-specific (germline, intestine, neuron, muscle, hypodermis, seam cell, anchor cell) and pan-somaticTIR1-expressing strains carrying an equimolar co-expressed blue fluorescent reporter to enable use of both red and green channels in experiments. We have also constructed a set of plasmids to generate fluorescent protein::AID fusions through CRISPR/Cas9-mediated genome editing. These templates can be produced through frequently used cloning systems (Gibson assembly or SapTrap) or through ribonucleoprotein complex-mediated insertion of PCR-derived, linear repair templates. We have generated a set of sgRNA plasmids carrying modifications shown to boost editing efficiency, targeting standardized transgene insertion sites on chromosomes I and II. Together these reagents should complement existingTIR1strains and facilitate rapid and high-throughput fluorescent protein::AID* tagging of factors of interest. This battery of new TIR1-expressing strains and modular, efficient cloning vectors serves as a platform for facile assembly of CRISPR/Cas9 repair templates for conditional protein depletion.
- Published
- 2020
- Full Text
- View/download PDF
32. Auxin-mediated Protein Degradation in Caenorhabditis elegans
- Author
-
David Q. Matus and Michael A. Q. Martinez
- Subjects
chemistry.chemical_classification ,biology ,Chemistry ,Strategy and Management ,Mechanical Engineering ,Metals and Alloys ,Morphogenesis ,food and beverages ,Protein degradation ,biology.organism_classification ,Transport inhibitor ,Industrial and Manufacturing Engineering ,Cell biology ,Auxin ,Fluorescence microscope ,Degron ,Developmental biology ,Caenorhabditis elegans - Abstract
The auxin-inducible degron (AID) technology was recently adapted for use in the nematode Caenorhabditis elegans. Rapid degradation of C. elegans proteins tagged with an AID is mediated by a plant-specific F-box protein, transport inhibitor response 1 (TIR1), and occurs only in the presence of the phytohormone auxin. The first iteration of this technology elicited protein degradation in C. elegans through a naturally occurring form of auxin, indole-3-acetic acid (IAA). Here, we present a protocol that uses 1-naphthaleneacetic acid, potassium salt (K-NAA), an indole-free synthetic auxin analog. At equal concentration, K-NAA is as effective as IAA in standard nematode growth media (NGM). K-NAA is also effective in physiological buffer (M9), allowing for high-throughput experimentation. The main advantages of K-NAA are twofold: first, its photostability prevents light-induced compound degradation during storage and the production of toxic indole-derivatives during fluorescence microscopy of live cells; and second, its water solubility eliminates the need of using ethanol to dissolve the auxin compound, a solvent that may confound C. elegans lifespan and behavioral assays. In this protocol, we describe our method of degrading C. elegans proteins using K-NAA on solid and in liquid media, as well as our method of analyzing protein degradation.
- Published
- 2020
- Full Text
- View/download PDF
33. Genomic organization and expression demonstrate spatial and temporal Hox gene colinearity in the lophotrochozoan Capitella sp. I.
- Author
-
Andreas C Fröbius, David Q Matus, and Elaine C Seaver
- Subjects
Medicine ,Science - Abstract
Hox genes define regional identities along the anterior-posterior axis in many animals. In a number of species, Hox genes are clustered in the genome, and the relative order of genes corresponds with position of expression in the body. Previous Hox gene studies in lophotrochozoans have reported expression for only a subset of the Hox gene complement and/or lack detailed genomic organization information, limiting interpretations of spatial and temporal colinearity in this diverse animal clade. We studied expression and genomic organization of the single Hox gene complement in the segmented polychaete annelid Capitella sp. I. Total genome searches identified 11 Hox genes in Capitella, representing 11 distinct paralog groups thought to represent the ancestral lophotrochozoan complement. At least 8 of the 11 Capitella Hox genes are genomically linked in a single cluster, have the same transcriptional orientation, and lack interspersed non-Hox genes. Studying their expression by situ hybridization, we find that the 11 Capitella Hox genes generally exhibit spatial and temporal colinearity. With the exception of CapI-Post1, Capitella Hox genes are all expressed in broad ectodermal domains during larval development, consistent with providing positional information along the anterior-posterior axis. The anterior genes CapI-lab, CapI-pb, and CapI-Hox3 initiate expression prior to the appearance of segments, while more posterior genes appear at or soon after segments appear. Many of the Capitella Hox genes have either an anterior or posterior expression boundary coinciding with the thoracic-abdomen transition, a major body tagma boundary. Following metamorphosis, several expression patterns change, including appearance of distinct posterior boundaries and restriction to the central nervous system. Capitella Hox genes have maintained a clustered organization, are expressed in the canonical anterior-posterior order found in other metazoans, and exhibit spatial and temporal colinearity, reflecting Hox gene characteristics that likely existed in the protostome-deuterostome ancestor.
- Published
- 2008
- Full Text
- View/download PDF
34. Pre-bilaterian origins of the Hox cluster and the Hox code: evidence from the sea anemone, Nematostella vectensis.
- Author
-
Joseph F Ryan, Maureen E Mazza, Kevin Pang, David Q Matus, Andreas D Baxevanis, Mark Q Martindale, and John R Finnerty
- Subjects
Medicine ,Science - Abstract
Hox genes were critical to many morphological innovations of bilaterian animals. However, early Hox evolution remains obscure. Phylogenetic, developmental, and genomic analyses on the cnidarian sea anemone Nematostella vectensis challenge recent claims that the Hox code is a bilaterian invention and that no "true" Hox genes exist in the phylum Cnidaria.Phylogenetic analyses of 18 Hox-related genes from Nematostella identify putative Hox1, Hox2, and Hox9+ genes. Statistical comparisons among competing hypotheses bolster these findings, including an explicit consideration of the gene losses implied by alternate topologies. In situ hybridization studies of 20 Hox-related genes reveal that multiple Hox genes are expressed in distinct regions along the primary body axis, supporting the existence of a pre-bilaterian Hox code. Additionally, several Hox genes are expressed in nested domains along the secondary body axis, suggesting a role in "dorsoventral" patterning.A cluster of anterior and posterior Hox genes, as well as ParaHox cluster of genes evolved prior to the cnidarian-bilaterian split. There is evidence to suggest that these clusters were formed from a series of tandem gene duplication events and played a role in patterning both the primary and secondary body axes in a bilaterally symmetrical common ancestor. Cnidarians and bilaterians shared a common ancestor some 570 to 700 million years ago, and as such, are derived from a common body plan. Our work reveals several conserved genetic components that are found in both of these diverse lineages. This finding is consistent with the hypothesis that a set of developmental rules established in the common ancestor of cnidarians and bilaterians is still at work today.
- Published
- 2007
- Full Text
- View/download PDF
35. Breaking down barriers: the evolution of cell invasion
- Author
-
Taylor N Medwig and David Q. Matus
- Subjects
Gene Editing ,0301 basic medicine ,Cell invasion ,Cell signaling ,Mechanism (biology) ,Ecology ,Cell ,Cell Communication ,Biological evolution ,Biology ,Biological Evolution ,Basement Membrane ,Article ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,Invasive phenotype ,Genome editing ,Evolutionary biology ,Genetics ,medicine ,Animals ,Caenorhabditis elegans ,Developmental Biology - Abstract
Cell invasion is a specialized cell behavior that likely co-evolved with the emergence of basement membranes in metazoans, as a mechanism to break down the barriers that separate tissues. A variety of conserved and lineage-specific biological processes that occur during development and homeostasis rely on cell invasive behavior. Recent innovations in genome editing and live-cell imaging have shed some light on the programs that mediate acquisition of an invasive phenotype; however, comparative approaches among species are necessary to understand how this cell behavior evolved. Here, we discuss the contexts of cell invasion, highlighting both established and emerging model systems, and underscore gaps in our understanding of the evolution of this key cellular behavior.
- Published
- 2017
- Full Text
- View/download PDF
36. Visualizing the metazoan proliferation-terminal differentiation decisionin vivo
- Author
-
Abraham Q. Kohrman, Maria D. Sallee, Mingwei Min, Ariel M. Pani, Wan Zhang, David Q. Matus, Nicholas Weeks, Taylor N. Medwig-Kinney, Michael A. Q. Martinez, Michalis Barkoulas, Nicholas J. Palmisano, Jayson J. Smith, Nuri Kim, Ononnah B. Ahmed, Qinyun Zhao, Benjamin L. Martin, Simeiyun Liu, Sabrina L. Spencer, Robert D Morabito, Rebecca C. Adikes, and Jessica L. Feldman
- Subjects
0303 health sciences ,Cell growth ,Kinase ,Cellular differentiation ,030302 biochemistry & molecular biology ,Cell ,Biology ,biology.organism_classification ,Cell biology ,03 medical and health sciences ,medicine.anatomical_structure ,Cytoplasm ,Cyclin-dependent kinase ,medicine ,biology.protein ,Phosphorylation ,Zebrafish ,030304 developmental biology - Abstract
SummaryCell proliferation and terminal differentiation are intimately coordinated during metazoan development. Here, we adapt a cyclin-dependent kinase (CDK) sensor to uncouple these cell cycle-associated events live inC. elegansand zebrafish. The CDK sensor consists of a fluorescently tagged CDK substrate that steadily translocates from the nucleus to the cytoplasm in response to increasing CDK activity and consequent sensor phosphorylation. We show that the CDK sensor can distinguish cycling cells in G1 from terminally differentiated cells in G0, revealing a commitment point and a cryptic stochasticity in an otherwise invariantC. eleganscell lineage. We also derive a predictive model of future proliferation behavior inC. elegansand zebrafish based on a snapshot of CDK activity in newly born cells. Thus, we introduce a live-cell imaging tool to facilitatein vivostudies of cell cycle control in a wide-range of developmental contexts.
- Published
- 2019
- Full Text
- View/download PDF
37. Rapid Degradation of
- Author
-
Michael A Q, Martinez, Brian A, Kinney, Taylor N, Medwig-Kinney, Guinevere, Ashley, James M, Ragle, Londen, Johnson, Joseph, Aguilera, Christopher M, Hammell, Jordan D, Ward, and David Q, Matus
- Subjects
Indoleacetic Acids ,AID system ,Ubiquitin-Protein Ligases ,SCF complex ,Microfluidics ,Investigations ,NHR-25 ,Naphthaleneacetic Acids ,C . elegans ,Larva ,Proteolysis ,synthetic auxin ,Animals ,Single-Cell Analysis ,Caenorhabditis elegans ,Caenorhabditis elegans Proteins - Abstract
As developmental biologists in the age of genome editing, we now have access to an ever-increasing array of tools to manipulate endogenous gene expression. The auxin-inducible degradation system allows for spatial and temporal control of protein degradation via a hormone-inducible Arabidopsis F-box protein, transport inhibitor response 1 (TIR1). In the presence of auxin, TIR1 serves as a substrate-recognition component of the E3 ubiquitin ligase complex SKP1-CUL1-F-box (SCF), ubiquitinating auxin-inducible degron (AID)-tagged proteins for proteasomal degradation. Here, we optimize the Caenorhabditis elegans AID system by utilizing 1-naphthaleneacetic acid (NAA), an indole-free synthetic analog of the natural auxin indole-3-acetic acid (IAA). We take advantage of the photostability of NAA to demonstrate via quantitative high-resolution microscopy that rapid degradation of target proteins can be detected in single cells within 30 min of exposure. Additionally, we show that NAA works robustly in both standard growth media and physiological buffer. We also demonstrate that K-NAA, the water-soluble, potassium salt of NAA, can be combined with microfluidics for targeted protein degradation in C. elegans larvae. We provide insight into how the AID system functions in C. elegans by determining that TIR1 depends on C. elegans SKR-1/2, CUL-1, and RBX-1 to degrade target proteins. Finally, we present highly penetrant defects from NAA-mediated degradation of the FTZ-F1 nuclear hormone receptor, NHR-25, during C. elegans uterine-vulval development. Together, this work improves our use and understanding of the AID system for dissecting gene function at the single-cell level during C. elegans development.
- Published
- 2019
38. Rapid degradation of C. elegans proteins at single-cell resolution with a synthetic auxin
- Author
-
Taylor N. Medwig-Kinney, Christopher M. Hammell, Londen C. Johnson, Brian A. Kinney, Jordan D. Ward, Joseph Aguilera, David Q. Matus, Guinevere Ashley, Michael A. Q. Martinez, and James Matthew Ragle
- Subjects
chemistry.chemical_classification ,0303 health sciences ,biology ,Protein degradation ,biology.organism_classification ,Transport inhibitor ,Ubiquitin ligase ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,chemistry ,Nuclear receptor ,Auxin ,Arabidopsis ,biology.protein ,Degron ,030217 neurology & neurosurgery ,Caenorhabditis elegans ,030304 developmental biology - Abstract
As developmental biologists in the age of genome editing, we now have access to an ever-increasing array of tools to manipulate endogenous gene expression. The auxin-inducible degradation system, allows for spatial and temporal control of protein degradation, functioning through the activity of a hormone-inducible Arabidopsis F-box protein, transport inhibitor response 1 (TIR1). In the presence of auxin, TIR1 serves as a substrate recognition component of the E3 ubiquitin ligase complex SKP1-CUL1-F-box (SCF), ubiquitinating auxin-inducible degron (AID)-tagged proteins for proteasomal degradation. Here, we optimize the Caenorhabditis elegans AID method, utilizing 1-naphthaleneacetic acid (NAA), an indole-free synthetic analog of the natural auxin indole-3-acetic acid (IAA). We take advantage of the photostability of NAA to demonstrate via quantitative high-resolution microscopy that rapid degradation of target proteins can be detected in single cells within 30 minutes of exposure. Additionally, we show that NAA works robustly in both standard growth media and physiological buffer. We also demonstrate that K-NAA, the water-soluble, potassium salt of NAA, can be combined with microfluidics for targeted protein degradation in C. elegans larvae. We provide insight into how the AID system functions in C. elegans by determining that TIR1 interacts with C. elegans SKR-1/2, CUL-1, and RBX-1 to degrade target proteins. Finally, we present highly penetrant defects from NAA-mediated degradation of the Ftz-F1 nuclear hormone receptor, NHR-25, during C. elegans uterine-vulval development. Together, this work provides a conceptual improvement to the AID system for dissecting gene function at the single-cell level during C. elegans development.
- Published
- 2019
- Full Text
- View/download PDF
39. A developmental gene regulatory network for invasive differentiation of the C. elegans anchor cell
- Author
-
Taylor N. Medwig-Kinney, Jayson J. Smith, Nicholas J. Palmisano, Sujata Tank, Wan Zhang, and David Q. Matus
- Subjects
0303 health sciences ,Cell ,Gene regulatory network ,Endogeny ,Biology ,Epithelium ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Genome editing ,RNA interference ,medicine ,Transcription factor ,030217 neurology & neurosurgery ,030304 developmental biology ,Positive feedback - Abstract
Cellular invasion is a key part of development, immunity, and disease. Using thein vivomodel ofC. elegansanchor cell invasion, we characterize the gene regulatory network that promotes invasive differentiation. The anchor cell is initially specified in a stochastic cell fate decision mediated by Notch signaling. Previous research has identified four conserved transcription factors,fos-1a(Fos),egl-43(EVI1/MEL),hlh-2(E/Daughterless) andnhr-67(NR2E1/TLX), that mediate anchor cell specification and/or invasive differentiation. Connections between these transcription factors and the underlying cell biology that they regulate is poorly understood. Here, using genome editing and RNA interference, we examine transcription factor interactions prior to and after anchor cell specification. During invasion we identify thategl-43,hlh-2, andnhr-67function together in a type I coherent feed-forward loop with positive feedback. Conversely, prior to specification, these transcription factors function independent of one another to regulate LIN-12 (Notch) activity. Together, these results demonstrate that, although the same transcription factors can function in fate specification and differentiated cell behavior, a gene regulatory network can be rapidly re-wired to reinforce a post-mitotic, pro-invasive state.SUMMARY STATEMENTBasement membrane invasion by theC. elegansanchor cell is coordinated by a dynamic gene regulatory network encompassing cell cycle dependent and independent sub-circuits.
- Published
- 2019
- Full Text
- View/download PDF
40. Adaptive F-Actin Polymerization and Localized ATP Production Drive Basement Membrane Invasion in the Absence of MMPs
- Author
-
Julie Plastino, Rodrigo Cáceres, Adam J. Schindler, Eric Hastie, Yue Jiang, Laura C. Kelley, Qiuyi Chi, David R. Sherwood, David Q. Matus, Duke University [Durham], Laboratoire Physico-Chimie Curie [Institut Curie] (PCC), Institut Curie [Paris]-Institut de Chimie du CNRS (INC)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), MOLTECH-Anjou, Université d'Angers (UA)-Institut de Chimie du CNRS (INC)-Centre National de la Recherche Scientifique (CNRS), and Sorbonne Université (SU)
- Subjects
matrix metalloproteinase ,[SDV]Life Sciences [q-bio] ,Cell ,Nerve Tissue Proteins ,Mitochondrion ,Matrix metalloproteinase ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Polymerization ,Cell membrane ,actin dynamics ,03 medical and health sciences ,Adenosine Triphosphate ,0302 clinical medicine ,Invasion ,Cell Movement ,Live cell imaging ,medicine ,Animals ,Caenorhabditis elegans ,Caenorhabditis elegans Proteins ,Molecular Biology ,Actin ,030304 developmental biology ,Basement membrane ,0303 health sciences ,ATP transport ,Cell Membrane ,Gene Expression Regulation, Developmental ,Cell Biology ,live imaging ,basement membrane ,Actins ,Matrix Metalloproteinases ,Cell biology ,mitochondria ,Actin Cytoskeleton ,medicine.anatomical_structure ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
International audience; Matrix metalloproteinases (MMPs) are associated with decreased patient prognosis, but have failed as anti-invasive drug targets despite promoting cancer cell invasion. Through time-lapse imaging, optical highlighting, and combined genetic removal of the five MMPs expressed during anchor cell (AC) invasion in C. elegans, we find that MMPs hasten invasion by degrading basement membrane (BM). Though irregular and delayed, AC invasion persists in MMP-animals via adaptive enrichment of the Arp2/3 complex at the invasive cell membrane, which drives formation of an F-actin-rich protrusion that physically breaches and displaces BM. Using a largescale RNAi synergistic screen and a genetically encoded ATP FRET sensor, we discover that mitochondria enrich within the protrusion and provide localized ATP that fuels F-actin network growth. Thus, without MMPs an invasive cell can alter its BM breaching tactics, suggesting that targeting adaptive mechanisms will be necessary to mitigate BM invasion in human pathologies.
- Published
- 2019
- Full Text
- View/download PDF
41. A developmental gene regulatory network for C. elegans anchor cell invasion
- Author
-
Wan Zhang, Sujata Tank, Nicholas J. Palmisano, Jayson J. Smith, David Q. Matus, and Taylor N. Medwig-Kinney
- Subjects
Cellular differentiation ,Green Fluorescent Proteins ,Cell ,Notch signaling pathway ,Gene regulatory network ,Receptors, Cytoplasmic and Nuclear ,Cell fate determination ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Cell Movement ,RNA interference ,Basic Helix-Loop-Helix Transcription Factors ,medicine ,Animals ,Protein Isoforms ,Cell Lineage ,Gene Regulatory Networks ,Caenorhabditis elegans ,Caenorhabditis elegans Proteins ,Molecular Biology ,Transcription factor ,030304 developmental biology ,0303 health sciences ,Receptors, Notch ,Cell Cycle ,Uterus ,Gene Expression Regulation, Developmental ,biology.organism_classification ,Cell biology ,medicine.anatomical_structure ,Female ,RNA Interference ,030217 neurology & neurosurgery ,Research Article ,Protein Binding ,Signal Transduction ,Transcription Factors ,Developmental Biology - Abstract
Cellular invasion is a key part of development, immunity, and disease. Using the in vivo model of C. elegans anchor cell invasion, we characterize the gene regulatory network that promotes cell invasion. The anchor cell is initially specified in a stochastic cell fate decision mediated by Notch signaling. Previous research has identified four conserved transcription factors, fos-1a (Fos), egl-43 (EVI1/MEL), hlh-2 (E/Daughterless) and nhr-67 (NR2E1/TLX), that mediate anchor cell specification and/or invasive behavior. Connections between these transcription factors and the underlying cell biology that they regulate are poorly understood. Here, using genome editing and RNA interference, we examine transcription factor interactions before and after anchor cell specification. Initially, these transcription factors function independently of one another to regulate LIN-12 (Notch) activity. Following anchor cell specification, egl-43, hlh-2, and nhr-67, function largely parallel to fos-1 in a type I coherent feed-forward loop with positive feedback to promote invasion. Together, these results demonstrate that the same transcription factors can function in cell fate specification and differentiated cell behavior, and that a gene regulatory network can be rapidly assembled to reinforce a post-mitotic, pro-invasive state.
- Published
- 2019
- Full Text
- View/download PDF
42. Observing the cell in its native state: Imaging subcellular dynamics in multicellular organisms
- Author
-
Abraham Q. Kohrman, Kishore R. Mosaliganti, Kai Wang, Srigokul Upadhyayula, Hanako Yashiro, David Q. Matus, Elliot M. Meyerowitz, Yuan Ruan, Tom W. Hiscock, Brian Cunniff, Dirk Hockemeyer, David G. Drubin, Minoru Koyama, Eric Betzig, Zach M. Collins, Ryan Forster, Ved P. Singh, Daniel E. Milkie, Ian A. Swinburne, Daphné Dambournet, Benjamin L. Martin, Tsung-Li Liu, Sean G. Megason, Steffen Scholpp, Tom Kirchhausen, Jamien Shea, and Taylor N Medwig
- Subjects
0301 basic medicine ,General Science & Technology ,1.1 Normal biological development and functioning ,Cell ,Mitosis ,Bioengineering ,Lattice light-sheet microscopy ,Endocytosis ,Eye ,Article ,Imaging ,03 medical and health sciences ,0302 clinical medicine ,Imaging, Three-Dimensional ,Single-cell analysis ,Underpinning research ,Cell Movement ,Organelle ,Genetics ,medicine ,Animals ,Humans ,Zebrafish ,Organelles ,Microscopy ,Multidisciplinary ,biology ,biology.organism_classification ,Phenotype ,Cell biology ,Multicellular organism ,030104 developmental biology ,medicine.anatomical_structure ,Three-Dimensional ,Cancer cell ,Biomedical Imaging ,Generic health relevance ,Single-Cell Analysis ,030217 neurology & neurosurgery - Abstract
True physiological imaging of subcellular dynamics requires studying cells within their parent organisms, where all the environmental cues that drive gene expression, and hence the phenotypes we actually observe, are present. A complete understanding also requires volumetric imaging of the cell and its surroundings at high spatiotemporal resolution without inducing undue stress on either. We combined lattice light sheet microscopy with two-channel adaptive optics to achieve, across large multicellular volumes, noninvasive aberration-free imaging of subcellular processes, including endocytosis, organelle remodeling during mitosis, and the migration of axons, immune cells, and metastatic cancer cells in vivo. The technology reveals the phenotypic diversity within cells across different organisms and developmental stages, and may offer insights into how cells harness their intrinsic variability to adapt to different physiological environments.One Sentence SummaryCombining lattice light sheet microscopy with adaptive optics enables high speed, high resolution in vivo 3D imaging of dynamic processes inside cells under physiological conditions within their parent organisms.
- Published
- 2018
43. Invasive Cell Fate Requires G1 Cell-Cycle Arrest and Histone Deacetylase-Mediated Changes in Gene Expression
- Author
-
Abraham Q. Kohrman, Adam J. Schindler, Michalis Barkoulas, David R. Sherwood, David Q. Matus, Wan Zhang, Lauren L. Lohmer, Qiuyi Chi, and Laura C. Kelley
- Subjects
Podosome ,Cellular differentiation ,Receptors, Cytoplasmic and Nuclear ,Cell fate determination ,Biology ,Histone Deacetylases ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Animals ,Neoplasm Invasiveness ,Caenorhabditis elegans ,Caenorhabditis elegans Proteins ,Molecular Biology ,Mitosis ,Transcription factor ,Cyclin-Dependent Kinase Inhibitor Proteins ,030304 developmental biology ,0303 health sciences ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Cell Biology ,G1 Phase Cell Cycle Checkpoints ,Actins ,Cell biology ,030220 oncology & carcinogenesis ,Podosomes ,Invadopodia ,Histone deacetylase ,Developmental Biology ,Cyclin-dependent kinase inhibitor protein - Abstract
SummaryDespite critical roles in development and cancer, the mechanisms that specify invasive cellular behavior are poorly understood. Through a screen of transcription factors in Caenorhabditis elegans, we identified G1 cell-cycle arrest as a precisely regulated requirement of the anchor cell (AC) invasion program. We show that the nuclear receptor nhr-67/tlx directs the AC into G1 arrest in part through regulation of the cyclin-dependent kinase inhibitor cki-1. Loss of nhr-67 resulted in non-invasive, mitotic ACs that failed to express matrix metalloproteinases or actin regulators and lack invadopodia, F-actin-rich membrane protrusions that facilitate invasion. We further show that G1 arrest is necessary for the histone deacetylase HDA-1, a key regulator of differentiation, to promote pro-invasive gene expression and invadopodia formation. Together, these results suggest that invasive cell fate requires G1 arrest and that strategies targeting both G1-arrested and actively cycling cells may be needed to halt metastatic cancer.
- Published
- 2015
- Full Text
- View/download PDF
44. The significance and scope of evolutionary developmental biology: a vision for the 21st century
- Author
-
Armin P. Moczek, Chi Hua Chiu, Thomas J. Sanger, Ehab Abouheif, Deirdre C. Lyons, Pamela K. Diggle, Angelika Stollewerk, Cassandra G. Extavour, Cristina C. Ledón-Rettig, Patricia J. Wittkopp, Scott F. Gilbert, C. Sarah Cohen, David Q. Matus, Federico D. Brown, Mario Vallejo-Marín, Anthony W. De Tomaso, Ian Dworkin, Siegfried Roth, Chelsea D. Specht, Joel Smith, Alan C. Love, Brian K. Hall, and Karen E. Sears
- Subjects
Cognitive science ,Scope (project management) ,Process (engineering) ,4. Education ,Ecology (disciplines) ,Gene regulatory network ,Environmental ethics ,Biology ,Science education ,Situated ,Evolutionary developmental biology ,Biological sciences ,Ecology, Evolution, Behavior and Systematics ,Developmental Biology - Abstract
Evolutionary developmental biology (evo-devo) has undergone dramatic transformations since its emergence as a distinct discipline. This paper aims to highlight the scope, power, and future promise of evo-devo to transform and unify diverse aspects of biology. We articulate key questions at the core of eleven biological disciplines-from Evolution, Development, Paleontology, and Neurobiology to Cellular and Molecular Biology, Quantitative Genetics, Human Diseases, Ecology, Agriculture and Science Education, and lastly, Evolutionary Developmental Biology itself-and discuss why evo-devo is uniquely situated to substantially improve our ability to find meaningful answers to these fundamental questions. We posit that the tools, concepts, and ways of thinking developed by evo-devo have profound potential to advance, integrate, and unify biological sciences as well as inform policy decisions and illuminate science education. We look to the next generation of evolutionary developmental biologists to help shape this process as we confront the scientific challenges of the 21st century.
- Published
- 2015
- Full Text
- View/download PDF
45. Divide or Conquer: Cell cycle regulation of invasive behavior
- Author
-
Abraham Q. Kohrman and David Q. Matus
- Subjects
0301 basic medicine ,Cell cycle checkpoint ,Embryonic Development ,Disease ,Biology ,Models, Biological ,Article ,Basement Membrane ,Metastasis ,03 medical and health sciences ,Neoplasms ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,Basement membrane ,Cell growth ,Cell Cycle ,Cancer ,Cell Biology ,Cell cycle ,medicine.disease ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Cancer cell - Abstract
Cell invasion through the basement membrane (BM) occurs during normal embryonic development and is a fundamental feature of cancer metastasis. The underlying cellular and genetic machinery required for invasion has been difficult to identify, due to a lack of adequate in vivo models to accurately examine invasion in single cells at subcellular resolution. Recent evidence has documented a functional link between cell cycle arrest and invasive activity. While cancer progression is traditionally thought of as a disease of uncontrolled cell proliferation, cancer cell dissemination, a critical aspect of metastasis, may require a switch from a proliferative to an invasive state. In this work, we review evidence that BM invasion requires cell cycle arrest and discuss the implications of this concept with regard to limiting the lethality associated with cancer metastasis.
- Published
- 2016
46. Imaging multicellular specimens with real-time optimized tiling light-sheet selective plane illumination microscopy
- Author
-
Liang Gao, Qinyi Fu, Benjamin L. Martin, and David Q. Matus
- Subjects
0301 basic medicine ,Embryo, Nonmammalian ,Science ,General Physics and Astronomy ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Imaging, Three-Dimensional ,Optics ,Live cell imaging ,Microscopy ,Animals ,Caenorhabditis elegans ,Zebrafish ,Multidisciplinary ,Extramural ,Plane (geometry) ,business.industry ,General Chemistry ,Multicellular organism ,030104 developmental biology ,Temporal resolution ,Zebrafish embryo ,business - Abstract
Despite the progress made in selective plane illumination microscopy, high-resolution 3D live imaging of multicellular specimens remains challenging. Tiling light-sheet selective plane illumination microscopy (TLS-SPIM) with real-time light-sheet optimization was developed to respond to the challenge. It improves the 3D imaging ability of SPIM in resolving complex structures and optimizes SPIM live imaging performance by using a real-time adjustable tiling light sheet and creating a flexible compromise between spatial and temporal resolution. We demonstrate the 3D live imaging ability of TLS-SPIM by imaging cellular and subcellular behaviours in live C. elegans and zebrafish embryos, and show how TLS-SPIM can facilitate cell biology research in multicellular specimens by studying left-right symmetry breaking behaviour of C. elegans embryos., Selective plane illumination microscopy (SPIM) is capable of high-resolution, high-speed 3D imaging of single cells, but application to multicellular samples is challenging. Here the authors develop tiling light sheet SPIM to image large multicellular specimens in 3D with subcellular resolution.
- Published
- 2016
- Full Text
- View/download PDF
47. Developmental Mechanisms Controlling Cell Fate, Evolution of
- Author
-
David Q. Matus, Deirdre C. Lyons, and Mansi Srivastava
- Subjects
Multicellular organism ,Mesoderm ,Cell type ,medicine.anatomical_structure ,Ecology ,Evolutionary biology ,Cellular differentiation ,Embryogenesis ,medicine ,Gene regulatory network ,Stem cell ,Biology ,Cell fate determination - Abstract
The specification of individual cells into differentiated cell types is a hallmark of multicellular organisms and driving force underlying body-plan diversity. The molecular underpinnings of cell fate acquisition during embryogenesis, adult homeostasis and following injury have been the subject of intense focus for many years. Advances in the ability to perform functional perturbations paired with ease of generating genomic data in a wide variety of taxa now allows biologists to address cell fate specification throughout the Metazoa. We discuss here three different aspects of cell fate specification: the formation of nematode equivalence groups, the evolution of a novel cell type, the echinoderm skeletogenic mesoderm, and the regulation of adult cell fate specification strategies.
- Published
- 2016
- Full Text
- View/download PDF
48. An expression screen for RhoGEF genes involved in C. elegans gonadogenesis
- Author
-
David R. Sherwood, David Q. Matus, and Joshua W. Ziel
- Subjects
Genetics ,RHOA ,Cell fusion ,biology ,Morphogenesis ,CDC42 ,GTPase ,biology.organism_classification ,Article ,Cell polarity ,biology.protein ,Molecular Biology ,Gene ,Caenorhabditis elegans ,Developmental Biology - Abstract
The gonad in Caenorhabditis elegans is an important model system for understanding complex morphogenetic processes including cellular movement, cell fusion, cell invasion and cell polarity during development. One class of signaling proteins known to be critical for the cellular events underlying morphogenesis is the Rho family GTPases, particularly RhoA, Rac and Cdc42. In C. elegans orthologues of these genes have been shown to be important for gonad development. In our current study we have extended those findings by examining the patterns of 5'cis-regulatory element (5'CRE) activity associated with nineteen putative guanine nucleotide exchange factors (GEFs) encoded by the C. elegans genome predicted to activate Rho family GTPases. Here we identify 13 RhoGEF genes that are expressed during gonadogenesis and characterize the cells in which their 5'CREs are active. These data provide the basis for designing experiments to examine Rho GTPase activation during morphogenetic processes central to normal gonad development.
- Published
- 2009
- Full Text
- View/download PDF
49. Anatomy and development of the nervous system ofNematostella vectensis, an anthozoan cnidarian
- Author
-
Mark Q. Martindale, Daniel S. Rokhsar, Heather Marlow, Mansi Srivastava, and David Q. Matus
- Subjects
Nervous system ,food.ingredient ,Nerve net ,Cellular differentiation ,Molecular Sequence Data ,Nematostella ,Nervous System ,Polymerase Chain Reaction ,Cellular and Molecular Neuroscience ,food ,Developmental Neuroscience ,medicine ,Animals ,Neural Cell Adhesion Molecules ,Neurons ,biology ,Stem Cells ,Starlet sea anemone ,Cell Differentiation ,Anatomy ,Anthozoa ,biology.organism_classification ,Immunohistochemistry ,Gastrulation ,medicine.anatomical_structure ,Homeobox ,Lernaean Hydra - Abstract
Nematostella vectensis, an anthozoan cnidarian, whose genome has been sequenced and is suitable for developmental and ecological studies, has a complex neural morphology that is modified during development from the larval to adult form. N. vectensis' nervous system is a diffuse nerve net with both ectodermal sensory and effector cells and endodermal multipolar ganglion cells. This nerve net consists of several distinct neural territories along the oral-aboral axis including the pharyngeal and oral nerve rings, and the larval apical tuft. These neuralized regions correspond to expression of conserved bilaterian neural developmental regulatory genes including homeodomain transcription factors and NCAMs. Early neurons and stem cell populations identified with NvMsi, NvELAV, and NvGCM, indicate that neural differentiation occurs throughout the animal and initiates prior to the conclusion of gastrulation. Neural specification in N. vectensis appears to occur through an independent mechanism from that in the classical cnidarian model Hydra.
- Published
- 2009
- Full Text
- View/download PDF
50. Broad phylogenomic sampling improves resolution of the animal tree of life
- Author
-
Gonzalo Giribet, Matthias Obst, Andreas Schmidt-Rhaesa, Akiko Okusu, Ward C. Wheeler, Martin V. Sørensen, Andreas Hejnol, Elaine C. Seaver, Mark Q. Martindale, William E. Browne, Greg W. Rouse, Gregory D. Edgecombe, Stephen A. Smith, David Q. Matus, Steven H. D. Haddock, Kevin Pang, Casey W. Dunn, and Reinhardt Møbjerg Kristensen
- Subjects
Lophotrochozoa ,Zoology ,Sensitivity and Specificity ,Evolution, Molecular ,Databases, Genetic ,Animals ,Humans ,Spiralia ,Phoronid ,Phylogeny ,Panarthropoda ,Gene Library ,Expressed Sequence Tags ,Multidisciplinary ,biology ,Computational Biology ,Reproducibility of Results ,Bayes Theorem ,Classification ,biology.organism_classification ,Markov Chains ,Eumetazoa ,Sister group ,Sample Size ,Ecdysozoa ,Platyzoa - Abstract
Long-held ideas regarding the evolutionary relationships among animals have recently been upended by sometimes controversial hypotheses based largely on insights from molecular data. These new hypotheses include a clade of moulting animals (Ecdysozoa) and the close relationship of the lophophorates to molluscs and annelids (Lophotrochozoa). Many relationships remain disputed, including those that are required to polarize key features of character evolution, and support for deep nodes is often low. Phylogenomic approaches, which use data from many genes, have shown promise for resolving deep animal relationships, but are hindered by a lack of data from many important groups. Here we report a total of 39.9 Mb of expressed sequence tags from 29 animals belonging to 21 phyla, including 11 phyla previously lacking genomic or expressed-sequence-tag data. Analysed in combination with existing sequences, our data reinforce several previously identified clades that split deeply in the animal tree (including Protostomia, Ecdysozoa and Lophotrochozoa), unambiguously resolve multiple long-standing issues for which there was strong conflicting support in earlier studies with less data (such as velvet worms rather than tardigrades as the sister group of arthropods), and provide molecular support for the monophyly of molluscs, a group long recognized by morphologists. In addition, we find strong support for several new hypotheses. These include a clade that unites annelids (including sipunculans and echiurans) with nemerteans, phoronids and brachiopods, molluscs as sister to that assemblage, and the placement of ctenophores as the earliest diverging extant multicellular animals. A single origin of spiral cleavage (with subsequent losses) is inferred from well-supported nodes. Many relationships between a stable subset of taxa find strong support, and a diminishing number of lineages remain recalcitrant to placement on the tree.
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.