Network


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

Hotspot


Dive into the research topics where G. V. Shivashankar is active.

Publication


Featured researches published by G. V. Shivashankar.


FEBS Letters | 2014

YAP/TAZ as mechanosensors and mechanotransducers in regulating organ size and tumor growth

Boon Chuan Low; Catherine Pan; G. V. Shivashankar; Alexander D. Bershadsky; Marius Sudol; Michael P. Sheetz

Organ size is controlled by the concerted action of biochemical and physical processes. Although mechanical forces are known to regulate cell and tissue behavior, as well as organogenesis, the precise molecular events that integrate mechanical and biochemical signals to control these processes are not fully known. The recently delineated Hippo‐tumor suppressor network and its two nuclear effectors, YAP and TAZ, shed light on these mechanisms. YAP and TAZ are proto‐oncogene proteins that respond to complex physical milieu represented by the rigidity of the extracellular matrix, cell geometry, cell density, cell polarity and the status of the actin cytoskeleton. Here, we review the current knowledge of how YAP and TAZ function as mechanosensors and mechanotransducers. We also suggest that by deciphering the mechanical and biochemical signals controlling YAP/TAZ function, we will gain insights into new strategies for cancer treatment and organ regeneration.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Cell geometric constraints induce modular gene-expression patterns via redistribution of HDAC3 regulated by actomyosin contractility

Nikhil Jain; Iyer Kv; Abhishek Kumar; G. V. Shivashankar

Physical forces in the form of substrate rigidity or geometrical constraints have been shown to alter gene expression profile and differentiation programs. However, the underlying mechanism of gene regulation by these mechanical cues is largely unknown. In this work, we use micropatterned substrates to alter cellular geometry (shape, aspect ratio, and size) and study the nuclear mechanotransduction to regulate gene expression. Genome-wide transcriptome analysis revealed cell geometry-dependent alterations in actin-related gene expression. Increase in cell size reinforced expression of matrix-related genes, whereas reduced cell-substrate contact resulted in up-regulation of genes involved in cellular homeostasis. We also show that large-scale changes in gene-expression profile mapped onto differential modulation of nuclear morphology, actomyosin contractility and histone acetylation. Interestingly, cytoplasmic-to-nuclear redistribution of histone deacetylase 3 modulated histone acetylation in an actomyosin-dependent manner. In addition, we show that geometric constraints altered the nuclear fraction of myocardin-related transcription factor. These fractions exhibited hindered diffusion time scale within the nucleus, correlated with enhanced serum-response element promoter activity. Furthermore, nuclear accumulation of myocardin-related transcription factor also modulated NF-κB activity. Taken together, our work provides modularity in switching gene-expression patterns by cell geometric constraints via actomyosin contractility.


Biophysical Journal | 2012

Mechanical Activation of Cells Induces Chromatin Remodeling Preceding MKL Nuclear Transport

K. Venkatesan Iyer; Stephanie Pulford; Alex Mogilner; G. V. Shivashankar

For cells to adapt to different tissues and changes in tissue mechanics, they must be able to respond to mechanical cues by changing their gene expression patterns. Biochemical signaling pathways for these responses have been elucidated, and recent evidence points to the involvement of force-induced deformation of the nucleus. However, it is still unclear how physical cues received at the plasma membrane (PM) spatiotemporally integrate to the functional chromatin organization of the cell nucleus. To investigate this issue, we applied mechanical forces through magnetic particles adhered to the PM of single cells and mapped the accompanying changes in actin polymerization, nuclear morphology, chromatin remodeling, and nuclear transport of soluble signaling intermediates using high-resolution fluorescence anisotropy imaging. Using this approach, we show the timescales associated with force-induced polymerization of actin and changes in the F/G actin ratio resulting in nuclear translocation of the G-actin-associated transcriptional cofactor, megakaryoblastic acute leukemia factor-1 (MKL). Further, this method of measuring nuclear organization at high spatiotemporal resolution with simultaneous force application revealed the physical propagation of forces to the nucleus, resulting in changes to chromatin organization, followed by nuclear deformation. We also describe a quantitative model that incorporates active stresses and chemical kinetics to evaluate the observed timescales. Our work suggests that mechanical activation of cells is accompanied by distinct timescales involved in the reorganization of actin and chromatin assembly, followed by translocation of transcription cofactors from the cytoplasm to the nucleus.


Biophysical Journal | 2009

Spatio-temporal plasticity in chromatin organization in mouse cell differentiation and during Drosophila embryogenesis.

Dipanjan Bhattacharya; Shefali Talwar; Aprotim Mazumder; G. V. Shivashankar

Cellular differentiation and developmental programs require changing patterns of gene expression. Recent experiments have revealed that chromatin organization is highly dynamic within living cells, suggesting possible mechanisms to alter gene expression programs, yet the physical basis of this organization is unclear. In this article, we contrast the differences in the dynamic organization of nuclear architecture between undifferentiated mouse embryonic stem cells and terminally differentiated primary mouse embryonic fibroblasts. Live-cell confocal tracking of nuclear lamina evidences highly flexible nuclear architecture within embryonic stem cells as compared to primary mouse embryonic fibroblasts. These cells also exhibit significant changes in histone and heterochromatin binding proteins correlated with their distinct epigenetic signatures as quantified by immunofluorescence analysis. Further, we follow histone dynamics during the development of the Drosophila melanogaster embryo, which gives an insight into spatio-temporal evolution of chromatin plasticity in an organismal context. Core histone dynamics visualized by fluorescence recovery after photobleaching, fluorescence correlation spectroscopy, and fluorescence anisotropy within the developing embryo, revealed an intriguing transition from plastic to frozen chromatin assembly synchronous with cellular differentiation. In the embryo, core histone proteins are highly mobile before cellularization, actively exchanging with the pool in the yolk. This hyperdynamic mobility decreases as cellularization and differentiation programs set in. These findings reveal a direct correlation between the dynamic transitions in chromatin assembly with the onset of cellular differentiation and developmental programs.


Biomaterials | 2014

The regulation of dynamic mechanical coupling between actin cytoskeleton and nucleus by matrix geometry

Qingsen Li; Abhishek Kumar; Ekta Makhija; G. V. Shivashankar

Cells sense their physical microenvironment and transduce these signals through actin-nuclear links to regulate nuclear functions including gene expression. However, the spatio-temporal coupling between perinuclear actin and nucleus and their functional importance are still unclear. Using micropatterned substrates to control cell geometry, we show that perinuclear actin organization at the apical plane remodels from mesh-like structure to stress fibers. The formation of these apical stress fibers (ASFs) correlated with significant reduction in nuclear height and was found to exert an active compressive load on the nucleus via direct contact with mature focal adhesion sites. Interestingly, the dynamic nature of ASFs was found to transduce forces to chromatin assembly. In addition, geometric perturbations or using pharmacological drugs to inhibit actomyosin contractility of ASFs resulted in nuclear instability. Taken together, our work provides direct evidence of physical links between the nucleus and focal adhesion sites via ASFs, which modulate nuclear homeostatic balance and internal chromatin structure. We suggest that such direct links may underlie nuclear mechanotransduction to regulate genomic programs.


Biophysical Journal | 2008

Dynamics of Chromatin Decondensation Reveals the Structural Integrity of a Mechanically Prestressed Nucleus

Aprotim Mazumder; T. Roopa; Aakash Basu; L. Mahadevan; G. V. Shivashankar

Genome organization within the cell nucleus is a result of chromatin condensation achieved by histone tail-tail interactions and other nuclear proteins that counter the outward entropic pressure of the polymeric DNA. We probed the entropic swelling of chromatin driven by enzymatic disruption of these interactions in isolated mammalian cell nuclei. The large-scale decondensation of chromatin and the eventual rupture of the nuclear membrane and lamin network due to this entropic pressure were observed by fluorescence imaging. This swelling was accompanied by nuclear softening, an effect that we quantified by measuring the fluctuations of an optically trapped bead adhered onto the nucleus. We also measured the pressure at which the nuclear scaffold ruptured using an atomic force microscope cantilever. A simple theory based on a balance of forces in a swelling porous gel quantitatively explains the diffusive dynamics of swelling. Our experiments on decondensation of chromatin in nuclei suggest that its compaction is a critical parameter in controlling nuclear stability.


Journal of the Royal Society Interface | 2010

Emergence of a prestressed eukaryotic nucleus during cellular differentiation and development

Aprotim Mazumder; G. V. Shivashankar

Nuclear shape and size are emerging as mechanistic regulators of genome function. Yet, the coupling between chromatin assembly and various nuclear and cytoplasmic scaffolds is poorly understood. The present work explores the structural organization of a prestressed nucleus in a variety of cellular systems ranging from cells in culture to those in an organism. A combination of laser ablation and cellular perturbations was used to decipher the dynamic nature of the nucleo-cytoplasmic contacts. In primary mouse embryonic fibroblasts, ablation of heterochromatin nodes caused an anisotropic shrinkage of the nucleus. Depolymerization of actin and microtubules, and inhibition of myosin motors, resulted in the differential stresses that these cytoplasmic systems exert on the nucleus. The onset of nuclear prestress was then mapped in two contexts—first, in the differentiation of embryonic stem cells, where signatures of prestress appeared with differentiation; second, at an organism level, where nuclear or cytoplasmic laser ablations of cells in the early Drosophila embryo induced a collapse of the nucleus only after cellularization. We thus show that the interplay of physical connections bridging the nucleus with the cytoplasm governs the size and shape of a prestressed eukaryotic nucleus.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Multivariate biophysical markers predictive of mesenchymal stromal cell multipotency

Wong Cheng J. Lee; Hui Shi; Zhiyong Poon; Lin Myint Nyan; Tanwi Kaushik; G. V. Shivashankar; Jerry Chan; Chwee Teck Lim; Jongyoon Han; Krystyn J. Van Vliet

Significance We identify a set of unique biophysical markers of multipotent mesenchymal stromal cell populations. Multivariate biophysical analysis of cells from 10 adult and fetal bone marrow donors shows that distinct subpopulations exist within supposed mesenchymal stem cell populations that are otherwise indistinguishable by accepted stem cell marker surface antigens. We find that although no single biophysical parameter is wholly predictive of stem cell multipotency, three of these together—cell diameter, cell mechanical stiffness, and nuclear membrane fluctuations—distinguish multipotent stem cell from osteochondral progenitor subpopulations. Together, these results (along with the corresponding statistical correlations) show that a minimal set of biophysical markers can be used to identify, isolate, and predict the function of stem and progenitor cells within mixed cell populations. The capacity to produce therapeutically relevant quantities of multipotent mesenchymal stromal cells (MSCs) via in vitro culture is a common prerequisite for stem cell-based therapies. Although culture expanded MSCs are widely studied and considered for therapeutic applications, it has remained challenging to identify a unique set of characteristics that enables robust identification and isolation of the multipotent stem cells. New means to describe and separate this rare cell type and its downstream progenitor cells within heterogeneous cell populations will contribute significantly to basic biological understanding and can potentially improve efficacy of stem and progenitor cell-based therapies. Here, we use multivariate biophysical analysis of culture-expanded, bone marrow-derived MSCs, correlating these quantitative measures with biomolecular markers and in vitro and in vivo functionality. We find that, although no single biophysical property robustly predicts stem cell multipotency, there exists a unique and minimal set of three biophysical markers that together are predictive of multipotent subpopulations, in vitro and in vivo. Subpopulations of culture-expanded stromal cells from both adult and fetal bone marrow that exhibit sufficiently small cell diameter, low cell stiffness, and high nuclear membrane fluctuations are highly clonogenic and also exhibit gene, protein, and functional signatures of multipotency. Further, we show that high-throughput inertial microfluidics enables efficient sorting of committed osteoprogenitor cells, as distinct from these mesenchymal stem cells, in adult bone marrow. Together, these results demonstrate novel methods and markers of stemness that facilitate physical isolation, study, and therapeutic use of culture-expanded, stromal cell subpopulations.


Journal of Molecular Biology | 2015

Cytoskeletal Control of Nuclear Morphology and Chromatin Organization

Nisha M. Ramdas; G. V. Shivashankar

The nucleus is sculpted toward various morphologies during cellular differentiation and development. Alterations in nuclear shape often result in changes to chromatin organization and genome function. This is thought to be reflective of its role as a cellular mechanotransducer. Recent evidence has highlighted the importance of cytoskeletal organization in defining how nuclear morphology regulates chromatin dynamics. However, the mechanisms underlying cytoskeletal control of chromatin remodeling are not well understood. We demonstrate here the differential influence of perinuclear actin- and microtubule-driven assemblies on nuclear architecture using pharmacological inhibitors and targeted RNA interference knockdown of cytoskeleton components in Drosophila cells. We find evidence that the loss of perinuclear actin assembly results in basolateral enhancement of microtubule organization and this is reflected functionally by enhanced nuclear dynamics. Cytoskeleton reorganization leads to nuclear lamina deformation that influences heterochromatin localization and core histone protein mobility. We also show that modulations in actin-microtubule assembly result in differential gene expression patterns. Taken together, we suggest that perinuclear actin and basolateral microtubule organization exerts mechanical control on nuclear morphology and chromatin dynamics.


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

Nuclear deformability and telomere dynamics are regulated by cell geometric constraints

Ekta Makhija; Doorgesh Sharma Jokhun; G. V. Shivashankar

Significance Physical properties of the cell nucleus are important for various cellular functions. However, the role of cell geometry and active cytoskeletal forces in regulating nuclear dynamics and chromatin dynamics is not well understood. Our results show cells with reduced matrix constraints have short actomyosin structures. These dynamic structures together with lower lamin A/C levels, resulting in softer nuclei, may provide the driving force for nuclear fluctuations. Furthermore, we observed increased dynamics of heterochromatin and telomere structures under such reduced cell–matrix interactions. We conclude that extracellular matrix signals alter cytoskeletal organization and lamin A/C expression levels, which together lead to nuclear and chromatin dynamics. These results highlight the importance of matrix constraints in regulating gene expression and maintaining genome integrity. Forces generated by the cytoskeleton can be transmitted to the nucleus and chromatin via physical links on the nuclear envelope and the lamin meshwork. Although the role of these active forces in modulating prestressed nuclear morphology has been well studied, the effect on nuclear and chromatin dynamics remains to be explored. To understand the regulation of nuclear deformability by these active forces, we created different cytoskeletal states in mouse fibroblasts using micropatterned substrates. We observed that constrained and isotropic cells, which lack long actin stress fibers, have more deformable nuclei than elongated and polarized cells. This nuclear deformability altered in response to actin, myosin, formin perturbations, or a transcriptional down-regulation of lamin A/C levels in the constrained and isotropic geometry. Furthermore, to probe the effect of active cytoskeletal forces on chromatin dynamics, we tracked the spatiotemporal dynamics of heterochromatin foci and telomeres. We observed increased dynamics and decreased correlation of the heterochromatin foci and telomere trajectories in constrained and isotropic cell geometry. The observed enhanced dynamics upon treatment with actin depolymerizing reagents in elongated and polarized geometry were regained once the reagent was washed off, suggesting an inherent structural memory in chromatin organization. We conclude that active forces from the cytoskeleton and rigidity from lamin A/C nucleoskeleton can together regulate nuclear and chromatin dynamics. Because chromatin remodeling is a necessary step in transcription control and its memory, genome integrity, and cellular deformability during migration, our results highlight the importance of cell geometric constraints as critical regulators in cell behavior.

Collaboration


Dive into the G. V. Shivashankar's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Abhishek Kumar

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar

Yejun Wang

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar

Doorgesh Sharma Jokhun

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar

K. Venkatesan Iyer

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar

Shovamayee Maharana

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar

Shefali Talwar

National Centre for Biological Sciences

View shared research outputs
Top Co-Authors

Avatar

Caroline Uhler

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Feroz M. Hameed

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge