4.7 Article

A developmental gene regulatory network for C. elegans anchor cell invasion

Journal

DEVELOPMENT
Volume 147, Issue 1, Pages -

Publisher

COMPANY BIOLOGISTS LTD
DOI: 10.1242/dev.185850

Keywords

EGL-43; FOS-1; HLH-2; NHR-67; Gene regulatory network; Cell invasion; Cell cycle arrest

Funding

  1. National Institutes of Health (NIH) National Cancer Institute [5R00CA154870-05]
  2. National Institute of General Medical Sciences (NIGMS) [1R01GM121597-01]
  3. Damon Runyon Cancer Research Foundation [DRR-47-17]
  4. NIH Eunice Kennedy Shriver National Institute of Child Health and Human Development [F31HD100091-01]
  5. NIGMS [3R01GM121597-02S1]
  6. American Cancer Society [132969-PF-18-226-01-CSM]

Ask authors/readers for more resources

Cellular invasion is a key part of development, immunity and disease. Using an in vivo model of Caenorhabditis 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-1 (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.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

Article Genetics & Heredity

The SWI/SNF chromatin remodeling assemblies BAF and PBAF differentially regulate cell cycle exit and cellular invasion in vivo

Jayson J. Smith, Yutong Q. Xiao, Nithin Parsan, Taylor C. Medwig-Kinney, Michael A. Q. Martinez, Frances E. Q. A. Moore, Nicholas Palmisano, Abraham Kohrman, Mana Chandhok Delos Reyes, Rebecca Q. Adikes, Simeiyun P. Liu, Sydney Bracht, Wan P. Zhang, Kailong Wen, Paschalis P. Kratsios, David Matus

Summary: The SWI/SNF complex plays a critical role in regulating cellular invasion, with BAF and PBAF assemblies contributing to invasion via different mechanisms. Research demonstrates that the SWI/SNF complex functions in a dose-dependent manner to control invasion, and provides insights into how it can regulate switching from invasive to proliferative fates. This study highlights the importance of appropriate regulation of the SWI/SNF complex in combating inappropriate cellular invasion.

PLOS GENETICS (2022)

Article Genetics & Heredity

An engineered, orthogonal auxin analog/AtTIR1(F79G) pairing improves both specificity and efficacy of the auxin degradation system in Caenorhabditis elegans

Kelly Hills-Muckey, Michael A. Q. Martinez, Natalia Stec, Shilpa Hebbar, Joanne Saldanha, Taylor N. Medwig-Kinney, Frances E. Q. Moore, Maria Ivanova, Ana Morao, J. D. Ward, Eric G. Moss, Sevinc Ercan, Anna Y. Zinovyeva, David Q. Matus, Christopher M. Hammell

Summary: The auxin-inducible degradation system in C. elegans allows for spatial and temporal control of protein degradation. In this study, the authors developed a mutant (At)TIR1(F79G) allele that reduces ligand-independent degradation of multiple AID*-tagged proteins, providing improved specificity and efficacy for the AID system.

GENETICS (2022)

Review Biochemistry & Molecular Biology

CDK activity sensors: genetically encoded ratiometric biosensors for live analysis of the cell cycle

Michael A. Q. Martinez, David Q. Matus

Summary: CDK sensors are genetically encoded fluorescent biosensors that change location upon CDK activation, facilitating investigations of the cell cycle. CDK activity is regulated by its association with cyclins, which is crucial for stem cell maintenance and cancer cell dormancy. The evolutionary conservation of CDKs allows for rapid development of CDK activity sensors in various organisms. These sensors enable visualization of the moment of cell-cycle commitment, providing insights into proliferation-quiescence decision and the regulation of development, aging, and cancer.

BIOCHEMICAL SOCIETY TRANSACTIONS (2022)

Article Multidisciplinary Sciences

Maintenance of neurotransmitter identity by Hox proteins through a homeostatic mechanism

Weidong Feng, Honorine Destain, Jayson J. Smith, Paschalis Kratsios

Summary: To maintain neuronal function, neuronal cells need to synthesize and release specific neurotransmitters. In this study, the authors found that Hox proteins are crucial for this ability. They discovered that LIN-39, MAB-5, and UNC-3 work together to ensure continuous and robust expression of genes related to cholinergic identity in motor neurons. This study reveals a noncanonical role for Hox proteins in controlling neurotransmitter identity during post-embryonic life.

NATURE COMMUNICATIONS (2022)

Article Cell Biology

A proliferative to invasive switch is mediated by srGAP1 downregulation through the activation of TGF-(32 signaling

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, Jose Javier Bravo-Cordero

Summary: In this study, srGAP1 is identified as a regulator of the proliferative-to-invasive switch in breast cancer cells. srGAP1low cells display a motile and invasive phenotype, facilitating the extravasation of cancer cells from blood vessels and attenuating tumor growth. Interestingly, a population of srGAP1low cells remain as solitary disseminated tumor cells in the lungs.

CELL REPORTS (2022)

Article Cell Biology

A light sheet fluorescence microscopy protocol for Caenorhabditis elegans larvae and adults

Jayson J. Smith, Isabel W. Kenny, Carsten Wolff, Rachel Cray, Abhishek Kumar, David R. Sherwood, David Q. Matus

Summary: Light sheet fluorescence microscopy (LSFM) has advantages of fast acquisition and reduced photobleaching and phototoxicity, making it a preferred method for live imaging. However, there is currently no generalized LSFM mounting protocol for live imaging of post-embryonic stages of C. elegans. In this study, a simple mounting and immobilization protocol using a refractive-index matched UV-curable hydrogel within FEP tubes was developed for efficient and reliable imaging of larval and adult C. elegans stages.

FRONTIERS IN CELL AND DEVELOPMENTAL BIOLOGY (2022)

Article Biochemistry & Molecular Biology

A membrane reticulum, the centriculum, affects centrosome size and function in Caenorhabditis elegans

Richa Maheshwari, Mohammad M. Rahman, Seth Drey, Megan Onyundo, Gunar Fabig, Michael A. Q. Martinez, David Q. Matus, Thomas Mueller-Reichert, Orna Cohen-Fix

Summary: In this study, using volume electron microscopy, researchers discovered that centrosomes in the Caenorhabditis elegans early embryo are surrounded by a three-dimensional membrane reticulum called the centriculum. The formation of the centriculum depends on the presence of centrosomes and microtubules. Moreover, the centriculum affects the function of centrosomes.

CURRENT BIOLOGY (2023)

Article Biology

Reevaluating the relationship between EGL-43 (EVI1) and LIN-12 (Notch) during C. elegans anchor cell invasion

Michael A. Q. Martinez, Angelina A. Mullarkey, Callista Yee, Chris Z. Zhao, Wan Zhang, Kang Shen, David Q. Matus

Summary: Development of the Caenorhabditis elegans reproductive tract is regulated by the anchor cell (AC). Transcription factors EGL-43, HLH-2, and NHR-67 play crucial roles in the regulation of AC invasion. Recent research has shown a mechanistic connection between EGL-43 loss and AC cell-cycle entry, suggesting that Notch signaling has mitogenic effects in the invasive AC. Using a novel co-expression system called AIDHB, researchers discovered that LIN-12 is not required for AC proliferation following loss of EGL-43.

BIOLOGY OPEN (2022)

Article Developmental Biology

The Caenorhabditis elegans anchor cell transcriptome: ribosome biogenesis drives cell invasion through basement membrane

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, David R. Sherwood

Summary: Cell invasion through basement membrane (BM) barriers is crucial in development, immune function, and cancer progression. The AC transcriptome of Caenorhabditis elegans anchor cell invasion provides insights into the mechanisms underlying BM transmigration, revealing the roles of ribosome biogenesis and endomembrane expansion. The identification of new invasion regulators, such as TCTP, and the enrichment of ribosomal proteins contribute to our understanding of the invasion process.

DEVELOPMENT (2023)

Article Cell Biology

Architecture of the cortical actomyosin network driving apical constriction in C. elegans

Pu Zhang, Taylor N. Medwig-Kinney, Bob Goldstein

Summary: Zhang, Medwig-Kinney, and Goldstein show that the medioapical actomyosin network driving apical constriction in C. elegans gastrulation has a diffuse organization with a mixed-polarity actin filament network. This differs from the sarcomere-like architecture observed in the Drosophila ventral furrow. They further demonstrate that C. elegans endodermal precursor cells apically constrict using a mixed-polarity actin filament network and with myosin and myosin activator distributed throughout the network. These results suggest that diverse actomyosin architectures are used in animal cells to accomplish apical constriction.

JOURNAL OF CELL BIOLOGY (2023)

Article Multidisciplinary Sciences

Loss of the E3 ubiquitin ligases UBR-5 or HECD-1 restores Caenorhabditis elegans development in the absence of SWI/SNF function

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

Summary: SWI/SNF complexes are important regulators of gene expression and development. This study identifies a specific mutation in the SWI/SNF subunit snfc-5 that prevents embryonic lethality in nematodes with a loss-of-function mutation in the subunit swsn-1. The study also reveals a potential therapeutic target for developmental disorders caused by mutations in SWI/SNF subunits.

PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA (2023)

No Data Available