Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Minna Roh-Johnson is active.

Publication


Featured researches published by Minna Roh-Johnson.


Developmental Biology | 2010

Apical constriction: A cell shape change that can drive morphogenesis

Jacob M. Sawyer; Jessica R. Harrell; Gidi Shemer; Jessica Sullivan-Brown; Minna Roh-Johnson; Bob Goldstein

Biologists have long recognized that dramatic bending of a cell sheet may be driven by even modest shrinking of the apical sides of cells. Cell shape changes and tissue movements like these are at the core of many of the morphogenetic movements that shape animal form during development, driving processes such as gastrulation, tube formation, and neurulation. The mechanisms of such cell shape changes must integrate developmental patterning information in order to spatially and temporally control force production-issues that touch on fundamental aspects of both cell and developmental biology and on birth defects research. How does developmental patterning regulate force-producing mechanisms, and what roles do such mechanisms play in development? Work on apical constriction from multiple systems including Drosophila, Caenorhabditis elegans, sea urchin, Xenopus, chick, and mouse has begun to illuminate these issues. Here, we review this effort to explore the diversity of mechanisms of apical constriction, the diversity of roles that apical constriction plays in development, and the common themes that emerge from comparing systems.


Science | 2012

Triggering a Cell Shape Change by Exploiting Preexisting Actomyosin Contractions

Minna Roh-Johnson; Gidi Shemer; Christopher D. Higgins; Joseph H. McClellan; Adam D. Werts; U. Serdar Tulu; Liang Gao; Eric Betzig; Daniel P. Kiehart; Bob Goldstein

A Time and a Place The onset of morphogenetic cell shape changes is thought to be triggered by initiation of actomyosin contractions. Roh-Johnson et al. (p. 1232, published online 9 February; see the Perspective by Razzell and Martin) have now discovered in both Caenorhabditis elegans and Drosophila embryos that the actomyosin contractions driving morphogenesis run constitutively, only being engaged to trigger cell shape changes at a specific time during development. Morphogenesis in developing worms and flies harnesses ongoing cortical motility. Apical constriction changes cell shapes, driving critical morphogenetic events, including gastrulation in diverse organisms and neural tube closure in vertebrates. Apical constriction is thought to be triggered by contraction of apical actomyosin networks. We found that apical actomyosin contractions began before cell shape changes in both Caenorhabitis elegans and Drosophila. In C. elegans, actomyosin networks were initially dynamic, contracting and generating cortical tension without substantial shrinking of apical surfaces. Apical cell-cell contact zones and actomyosin only later moved increasingly in concert, with no detectable change in actomyosin dynamics or cortical tension. Thus, apical constriction appears to be triggered not by a change in cortical tension, but by dynamic linking of apical cell-cell contact zones to an already contractile apical cortex.


Oncogene | 2014

Macrophage contact induces RhoA GTPase signaling to trigger tumor cell intravasation

Minna Roh-Johnson; Jose Javier Bravo-Cordero; Antonia Patsialou; Ved P. Sharma; Peng Guo; Huiping Liu; Louis Hodgson; John Condeelis

Most cancer patients die as a result of metastasis, thus it is important to understand the molecular mechanisms of dissemination, including intra- and extravasation. Although the mechanisms of extravasation have been vastly studied in vitro and in vivo, the process of intravasation is still unclear. Furthermore, how cells in the tumor microenvironment facilitate tumor cell intravasation is still unknown. Using high-resolution imaging, we found that macrophages enhance tumor cell intravasation upon physical contact. Macrophage and tumor cell contact induce RhoA activity in tumor cells, triggering the formation of actin-rich degradative protrusions called invadopodia, enabling tumor cells to degrade and break through matrix barriers during tumor cell transendothelial migration. Interestingly, we show that macrophage-induced invadopodium formation and tumor cell intravasation also occur in patient-derived tumor cells and in vivo models, revealing a conserved mechanism of tumor cell intravasation. Our results illustrate a novel heterotypic cell contact-mediated signaling role for RhoA, as well as yield mechanistic insight into the ability of cells within the tumor microenvironment to facilitate steps of the metastatic cascade.


Oncogene | 2014

Autocrine HBEGF expression promotes breast cancer intravasation, metastasis and macrophage-independent invasion in vivo

Zhen Ni Zhou; Ved P. Sharma; Bt Beaty; Minna Roh-Johnson; Ea Peterson; N. van Rooijen; Pa Kenny; H. S. Wiley; John Condeelis; Jeffrey E. Segall

Increased expression of HBEGF in estrogen receptor-negative breast tumors is correlated with enhanced metastasis to distant organ sites and more rapid disease recurrence upon removal of the primary tumor. Our previous work has demonstrated a paracrine loop between breast cancer cells and macrophages in which the tumor cells are capable of stimulating macrophages through the secretion of colony-stimulating factor-1 while the tumor-associated macrophages (TAMs), in turn, aid in tumor cell invasion by secreting epidermal growth factor. To determine how the autocrine expression of epidermal growth factor receptor (EGFR) ligands by carcinoma cells would affect this paracrine loop mechanism, and in particular whether tumor cell invasion depends on spatial ligand gradients generated by TAMs, we generated cell lines with increased HBEGF expression. We found that autocrine HBEGF expression enhanced in vivo intravasation and metastasis and resulted in a novel phenomenon in which macrophages were no longer required for in vivo invasion of breast cancer cells. In vitro studies revealed that expression of HBEGF enhanced invadopodium formation, thus providing a mechanism for cell autonomous invasion. The increased invadopodium formation was directly dependent on EGFR signaling, as demonstrated by a rapid decrease in invadopodia upon inhibition of autocrine HBEGF/EGFR signaling as well as inhibition of signaling downstream of EGFR activation. HBEGF expression also resulted in enhanced invadopodium function via upregulation of matrix metalloprotease 2 (MMP2) and MMP9 expression levels. We conclude that high levels of HBEGF expression can short-circuit the tumor cell/macrophage paracrine invasion loop, resulting in enhanced tumor invasion that is independent of macrophage signaling.


Journal of Cell Science | 2013

Spatial regulation of RhoC activity defines protrusion formation in migrating cells

Jose Javier Bravo-Cordero; Ved P. Sharma; Minna Roh-Johnson; Xiaoming Chen; Robert J. Eddy; John Condeelis; Louis Hodgson

Summary Protrusion formation is the first step that precedes cell movement of motile cells. Spatial control of actin polymerization is necessary to achieve directional protrusion during cell migration. Here we show that the spatial coordinators p190RhoGEF and p190RhoGAP regulate actin polymerization during leading edge protrusions by regulating the actin barbed end distribution and amplitude. The distribution of RhoC activity and proper balance of cofilin activation achieved by p190RhoGEF and p190RhoGAP determines the direction of final protrusive activity. These findings provide a new insight into the dynamic plasticity in the amplitude and distribution of barbed ends, which can be modulated by fine-tuning RhoC activity by upstream GEFs and GAPs for directed cell motility.


Journal of Cell Science | 2009

In vivo roles for Arp2/3 in cortical actin organization during C. elegans gastrulation

Minna Roh-Johnson; Bob Goldstein

The Arp2/3 complex is important for morphogenesis in various developmental systems, but specific in vivo roles for this complex in cells that move during morphogenesis are not well understood. We have examined cellular roles for Arp2/3 in the Caenorhabditis elegans embryo. In C. elegans, the first morphogenetic movement, gastrulation, is initiated by the internalization of two endodermal precursor cells. These cells undergo a myosin-dependent apical constriction, pulling a ring of six neighboring cells into a gap left behind on the ventral surface of the embryo. In agreement with a previous report, we found that in Arp2/3-depleted C. elegans embryos, membrane blebs form and the endodermal precursor cells fail to fully internalize. We show that these cells are normal with respect to several key requirements for gastrulation: cell cycle timing, cell fate, apicobasal cell polarity and apical accumulation and activation of myosin-II. To further understand the function of Arp2/3 in gastrulation, we examined F-actin dynamics in wild-type embryos. We found that three of the six neighboring cells extend short, dynamic F-actin-rich processes at their apical borders with the internalizing cells. These processes failed to form in embryos that were depleted of Arp2/3 or the apical protein PAR-3. Our results identify an in vivo role for Arp2/3 in the formation of subcellular structures during morphogenesis. The results also suggest a new layer to the model of C. elegans gastrulation: in addition to apical constriction, internalization of the endoderm might involve dynamic Arp2/3-dependent F-actin-rich extensions on one side of a ring of cells.


Scientific Reports | 2016

Macrophage-dependent tumor cell transendothelial migration is mediated by Notch1/Mena INV -initiated invadopodium formation

Jeanine Pignatelli; Jose Javier Bravo-Cordero; Minna Roh-Johnson; Saumil J. Gandhi; Yarong Wang; Xiaoming Chen; Robert J. Eddy; Alice Xue; Robert H. Singer; Louis Hodgson; Maja H. Oktay; John Condeelis

The process of intravasation involving transendothelial migration is a key step in metastatic spread. How the triple cell complex composed of a macrophage, Mena over-expressing tumor cell and endothelial cell, called the tumor microenvironment of metastasis (TMEM), facilitates tumor cell transendothelial migration is not completely understood. Previous work has shown that the physical contact between a macrophage and tumor cell results in the formation of invadopodia, actin-rich matrix degrading protrusions, important for tumor cell invasion and transendothelial migration and tumor cell dissemination. Herein, we show that the macrophage-induced invadopodium is formed through a Notch1/MenaINV signaling pathway in the tumor cell upon macrophage contact. This heterotypic tumor cell – macrophage interaction results in the upregulation of MenaINV through the activation of MENA transcription. Notch1 and MenaINV expression are required for tumor cell transendothelial migration, a necessary step during intravasation. Inhibition of the Notch signaling pathway blocked macrophage-induced invadopodium formation in vitro and the dissemination of tumor cells from the primary tumor in vivo. Our findings indicate a novel role for Notch1 signaling in the regulation of MenaINV expression and transendothelial migration and provide mechanistic information essential to the use of therapeutic inhibitors of metastasis.


Development | 2011

Dynamic localization of C. elegans TPR-GoLoco proteins mediates mitotic spindle orientation by extrinsic signaling

Adam D. Werts; Minna Roh-Johnson; Bob Goldstein

Cell divisions are sometimes oriented by extrinsic signals, by mechanisms that are poorly understood. Proteins containing TPR and GoLoco-domains (C. elegans GPR-1/2, Drosophila Pins, vertebrate LGN and AGS3) are candidates for mediating mitotic spindle orientation by extrinsic signals, but the mechanisms by which TPR-GoLoco proteins may localize in response to extrinsic cues are not well defined. The C. elegans TPR-GoLoco protein pair GPR-1/2 is enriched at a site of contact between two cells – the endomesodermal precursor EMS and the germline precursor P2 – and both cells align their divisions toward this shared cell-cell contact. To determine whether GPR-1/2 is enriched at this site within both cells, we generated mosaic embryos with GPR-1/2 bearing a different fluorescent tag in different cells. We were surprised to find that GPR-1/2 distribution is symmetric in EMS, where GPR-1/2 had been proposed to function as an asymmetric cue for spindle orientation. Instead, GPR-1/2 is asymmetrically distributed only in P2. We demonstrate a role for normal GPR-1/2 localization in P2 division orientation. We show that MES-1/Src signaling plays an instructive role in P2 for asymmetric GPR-1/2 localization and normal spindle orientation. We ruled out a model in which signaling localizes GPR-1/2 by locally inhibiting LET-99, a GPR-1/2 antagonist. Instead, asymmetric GPR-1/2 distribution is established by destabilization at one cell contact, diffusion, and trapping at another cell contact. Once the mitotic spindle of P2 is oriented normally, microtubule-dependent removal of GPR-1/2 prevented excess accumulation, in an apparent negative-feedback loop. These results highlight the role of dynamic TPR-GoLoco protein localization as a key mediator of mitotic spindle alignment in response to instructive, external cues.


Proceedings of the National Academy of Sciences of the United States of America | 2016

MYC-nick promotes cell migration by inducing fascin expression and Cdc42 activation.

Sarah Anderson; Kumud R. Poudel; Minna Roh-Johnson; Thomas Brabletz; Ming Yu; Nofit Borenstein-Auerbach; William N. Grady; Jihong Bai; Cecilia B. Moens; Robert N. Eisenman; Maralice Conacci-Sorrell

Significance The MYC family of transcription factors is deregulated in a broad range of cancers and drives the expression of genes that mediate biomass accumulation and promote cell proliferation and tumor initiation. We find that MYC can also trigger tumor cell migration and metastasis independently of its transcriptional activity, via its conversion to MYC-nick, a truncated form of MYC localized in the cytoplasm. MYC-nick promotes reorganization of the actin cytoskeleton by inducing expression of the actin-bundling protein fascin and by activating the Rho GTPase Cdc42, both of which lead to formation of filopodia, cellular structures known to drive cell migration. Our work links the repurposing of the MYC transcription factor to altered cytoskeletal structure and tumor cell metastatic behavior. MYC-nick is a cytoplasmic, transcriptionally inactive member of the MYC oncoprotein family, generated by a proteolytic cleavage of full-length MYC. MYC-nick promotes migration and survival of cells in response to chemotherapeutic agents or withdrawal of glucose. Here we report that MYC-nick is abundant in colonic and intestinal tumors derived from mouse models with mutations in the Wnt, TGF-β, and PI3K pathways. Moreover, MYC-nick is elevated in colon cancer cells deleted for FBWX7, which encodes the major E3 ligase of full-length MYC frequently mutated in colorectal cancers. MYC-nick promotes the migration of colon cancer cells assayed in 3D cultures or grown as xenografts in a zebrafish metastasis model. MYC-nick accelerates migration by activating the Rho GTPase Cdc42 and inducing fascin expression. MYC-nick, fascin, and Cdc42 are frequently up-regulated in cells present at the invasive front of human colorectal tumors, suggesting a coordinated role for these proteins in tumor migration.


Archive | 2010

Roles for Actin Dynamics in Cell Movements During Development

Minna Roh-Johnson; Jessica Sullivan-Brown; Bob Goldstein

Actin-dependent cellular movements and rearrangements are crucial for development. Studies in vitro have contributed much to the knowledge of actin biology. However, interesting environmental influences common in developing systems can differentially regulate actin dynamics and organization. In this chapter, we highlight several selected examples of directed cell migration during morphogenesis, in which actin dynamics have been observed directly in live-imaging studies. We discuss similarities and differences between collective cell and single cell migration during development, and we compare what has been learned from in vivo studies in developmental systems with in vitro studies of single cells.

Collaboration


Dive into the Minna Roh-Johnson's collaboration.

Top Co-Authors

Avatar

John Condeelis

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Bob Goldstein

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Jose Javier Bravo-Cordero

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Louis Hodgson

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Maja H. Oktay

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Cecilia B. Moens

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Jeanine Pignatelli

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Kumud R. Poudel

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Ved P. Sharma

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Adam D. Werts

University of North Carolina at Chapel Hill

View shared research outputs
Researchain Logo
Decentralizing Knowledge