Network


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

Hotspot


Dive into the research topics where Srikanth Talluri is active.

Publication


Featured researches published by Srikanth Talluri.


Cell Cycle | 2012

Regulation of transcription and chromatin structure by pRB: here, there and everywhere.

Srikanth Talluri; Frederick A. Dick

Commitment to divide is one of the most crucial steps in the mammalian cell division cycle. It is critical for tissue and organismal homeostasis, and consequently is highly regulated. The vast majority of cancers evade proliferative control, further emphasizing the importance of the commitment step in cell cycle regulation. The Retinoblastoma (RB) tumor suppressor pathway regulates this decision-making step. Since being the subject of Knudson’s ‘two hit hypothesis’, there has been considerable interest in understanding pRB’s role in cancer. It is best known for repressing E2F dependent transcription of cell cycle genes. However, pRB’s role in controlling chromatin structure is expanding and bringing it into new regulatory paradigms. In this review we discuss pRB function through protein-protein interactions, at the level of transcriptional regulation of individual promoters and in organizing higher order chromatin domains.


Molecular and Cellular Biology | 2010

A G1 checkpoint mediated by the retinoblastoma protein that is dispensable in terminal differentiation but essential for senescence

Srikanth Talluri; Christian E. Isaac; Mohammad E. Ahmad; Shauna A. Henley; Sarah M. Francis; Alison L. Martens; Rod Bremner; Frederick A. Dick

ABSTRACT Terminally differentiated cell types are needed to live and function in a postmitotic state for a lifetime. Cellular senescence is another type of permanent arrest that blocks the proliferation of cells in response to genotoxic stress. Here we show that the retinoblastoma protein (pRB) uses a mechanism to block DNA replication in senescence that is distinct from its role in permanent cell cycle exit associated with terminal differentiation. Our work demonstrates that a subtle mutation in pRB that cripples its ability to interact with chromatin regulators impairs heterochromatinization and repression of E2F-responsive promoters during senescence. In contrast, terminally differentiated nerve and muscle cells bearing the same mutation fully exit the cell cycle and block E2F-responsive gene expression by a different mechanism. Remarkably, this reveals that pRB recruits chromatin regulators primarily to engage a stress-responsive G1 arrest program.


Cancer Discovery | 2014

Haploinsufficiency of an RB–E2F1–Condensin II Complex Leads to Aberrant Replication and Aneuploidy

Courtney H. Coschi; Charles A. Ishak; David Gallo; Aren Marshall; Srikanth Talluri; Jianxin Wang; Matthew J. Cecchini; Alison L. Martens; Vanessa Percy; Ian Welch; Paul C. Boutros; Grant W. Brown; Frederick A. Dick

UNLABELLED Genome instability is a characteristic of malignant cells; however, evidence for its contribution to tumorigenesis has been enigmatic. In this study, we demonstrate that the retinoblastoma protein, E2F1, and Condensin II localize to discrete genomic locations including major satellite repeats at pericentromeres. In the absence of this complex, aberrant replication ensues followed by defective chromosome segregation in mitosis. Surprisingly, loss of even one copy of the retinoblastoma gene reduced recruitment of Condensin II to pericentromeres and caused this phenotype. Using cancer genome data and gene-targeted mice, we demonstrate that mutation of one copy of RB1 is associated with chromosome copy-number variation in cancer. Our study connects DNA replication and chromosome structure defects with aneuploidy through a dosage-sensitive complex at pericentromeric repeats. SIGNIFICANCE Genome instability is inherent to most cancers and is the basis for selective killing of cancer cells by genotoxic therapeutics. In this report, we demonstrate that instability can be caused by loss of a single allele of the retinoblastoma gene that prevents proper replication and condensation of pericentromeric chromosomal regions, leading to elevated levels of aneuploidy in cancer.


Molecular and Cellular Biology | 2014

A retinoblastoma allele that is mutated at its common E2F interaction site inhibits cell proliferation in gene targeted mice

Matthew J. Cecchini; Michael J. Thwaites; Srikanth Talluri; James I. S. MacDonald; Daniel T. Passos; Jean-Leon Chong; Paul G. Cantalupo; P. M. Stafford; Maria Teresa Sáenz-Robles; Sarah M. Francis; James M. Pipas; Gustavo Leone; Ian Welch; Frederick A. Dick

ABSTRACT The retinoblastoma protein (pRB) is best known for regulating cell proliferation through E2F transcription factors. In this report, we investigate the properties of a targeted mutation that disrupts pRB interactions with the transactivation domain of E2Fs. Mice that carry this mutation endogenously (Rb1ΔG) are defective for pRB-dependent repression of E2F target genes. Except for an accelerated entry into S phase in response to serum stimulation, cell cycle regulation in Rb1ΔG/ΔG mouse embryonic fibroblasts (MEFs) strongly resembles that of the wild type. In a serum deprivation-induced cell cycle exit, Rb1ΔG/ΔG MEFs display a magnitude of E2F target gene derepression similar to that of Rb1−/− cells, even though Rb1ΔG/ΔG cells exit the cell cycle normally. Interestingly, cell cycle arrest in Rb1ΔG/ΔG MEFs is responsive to p16 expression and gamma irradiation, indicating that alternate mechanisms can be activated in G1 to arrest proliferation. Some Rb1ΔG/ΔG mice die neonatally with a muscle degeneration phenotype, while the others live a normal life span with no evidence of spontaneous tumor formation. Most tissues appear histologically normal while being accompanied by derepression of pRB-regulated E2F targets. This suggests that non-E2F-, pRB-dependent pathways may have a more relevant role in proliferative control than previously identified.


Molecular and Cellular Biology | 2014

Loss of the mammalian DREAM complex deregulates chondrocyte proliferation.

Chantal Forristal; Shauna A. Henley; James I. S. MacDonald; Jason R Bush; Carley Ort; Daniel T. Passos; Srikanth Talluri; Charles A. Ishak; Michael J. Thwaites; Chris J.D. Norley; Larisa Litovchick; James A. DeCaprio; Gabriel E. DiMattia; David W. Holdsworth; Frank Beier; Frederick A. Dick

ABSTRACT Mammalian DREAM is a conserved protein complex that functions in cellular quiescence. DREAM contains an E2F, a retinoblastoma (RB)-family protein, and the MuvB core (LIN9, LIN37, LIN52, LIN54, and RBBP4). In mammals, MuvB can alternatively bind to BMYB to form a complex that promotes mitotic gene expression. Because BMYB-MuvB is essential for proliferation, loss-of-function approaches to study MuvB have generated limited insight into DREAM function. Here, we report a gene-targeted mouse model that is uniquely deficient for DREAM complex assembly. We have targeted p107 (Rbl1) to prevent MuvB binding and combined it with deficiency for p130 (Rbl2). Our data demonstrate that cells from these mice preferentially assemble BMYB-MuvB complexes and fail to repress transcription. DREAM-deficient mice show defects in endochondral bone formation and die shortly after birth. Micro-computed tomography and histology demonstrate that in the absence of DREAM, chondrocytes fail to arrest proliferation. Since DREAM requires DYRK1A (dual-specificity tyrosine phosphorylation-regulated protein kinase 1A) phosphorylation of LIN52 for assembly, we utilized an embryonic bone culture system and pharmacologic inhibition of (DYRK) kinase to demonstrate a similar defect in endochondral bone growth. This reveals that assembly of mammalian DREAM is required to induce cell cycle exit in chondrocytes.


Cell Cycle | 2014

The retinoblastoma protein and PML collaborate to organize heterochromatin and silence E2F-responsive genes during senescence

Srikanth Talluri; Frederick A. Dick

Cellular senescence is characterized by silencing of genes involved in DNA replication and cell cycle progression. Stable repression is crucial for preventing inappropriate DNA synthesis and the maintenance of a prolonged senescent state. Many of these genes are targets for E2F transcription factors. The pRB pathway plays a major role in senescence by directly repressing E2Fs and also by regulating chromatin at the promoters of E2F target genes using its LXCXE cleft-dependent interactions. In this study, we sought to investigate the mechanisms by which pRB stably silences E2F target gene transcription during cellular senescence. We report that in mouse embryonic fibroblasts, endogenous promyelocytic leukemia protein (PML) associates with E2F target genes in a pRB LXCXE-dependent manner during HrasV12-induced senescence. Furthermore, using a PML-IV-induced senescence model, we show that the pRB LXCXE binding cleft is essential for PML association with gene promoters, silencing of E2F target genes, and stable cell cycle exit. Binding assays show that pRB can interact with PML specifically during senescence, suggesting that signaling events in senescence regulate assembly of PML and pRB to establish heterochromatin and create a permanent cell cycle arrest.


PLOS ONE | 2013

Mutation of the LXCXE binding cleft of pRb facilitates transformation by ras in vitro but does not promote tumorigenesis in vivo.

Srikanth Talluri; Sarah M. Francis; Frederick A. Dick

Background The Retinoblastoma protein (pRB) is a key tumor suppressor that is functionally inactivated in most cancers. pRB regulates the cell division cycle and cell cycle exit through protein–protein interactions mediated by its multiple binding interfaces. The LXCXE binding cleft region of pRB mediates interactions with cellular proteins that have chromatin regulatory functions. Chromatin regulation mediated by pRB is required for a stress responsive cell cycle arrest, including oncogene induced senescence. The in vivo role of chromatin regulation by pRB during senescence, and its relevance to cancer is not clear. Methodology/Principal Findings Using gene-targeted mice, uniquely defective for pRB mediated chromatin regulation, we investigated its role during transformation and tumor progression in response to activation of oncogenic ras. We report that the pRB∆L mutation confers susceptibility to escape from HrasV12 induced senescence and allows transformation in vitro, although these cells possess high levels of DNA damage. Intriguingly, LSL-Kras, Rb1 ∆L/∆L mice show delayed lung tumor formation compared to controls. This is likely due to the increased apoptosis seen in the early hyperplastic lesions shortly following ras activation that inhibits tumor progression. Furthermore, DMBA treatment to induce sporadic ras mutations in other tissues also failed to reveal greater susceptibility to cancer in Rb1 ∆L/∆L mice. Conclusions/Significance Our data suggests that chromatin regulation by pRB can function to limit proliferation, but its loss fails to contribute to cancer susceptibility in ras driven tumor models because of elevated levels of DNA damage and apoptosis.


Scientific Reports | 2017

Isolation of Circulating Plasma Cells in Multiple Myeloma Using CD138 Antibody-Based Capture in a Microfluidic Device

Mohammad A. Qasaimeh; Yichao C. Wu; Suman Bose; Anoop Menachery; Srikanth Talluri; Gabriel Gonzalez; Mariateresa Fulciniti; Jeffrey M. Karp; Rao Prabhala; Rohit Karnik

The necessity for bone marrow aspiration and the lack of highly sensitive assays to detect residual disease present challenges for effective management of multiple myeloma (MM), a plasma cell cancer. We show that a microfluidic cell capture based on CD138 antigen, which is highly expressed on plasma cells, permits quantitation of rare circulating plasma cells (CPCs) in blood and subsequent fluorescence-based assays. The microfluidic device is based on a herringbone channel design, and exhibits an estimated cell capture efficiency of ~40–70%, permitting detection of <10 CPCs/mL using 1-mL sample volumes, which is difficult using existing techniques. In bone marrow samples, the microfluidic-based plasma cell counts exhibited excellent correlation with flow cytometry analysis. In peripheral blood samples, the device detected a baseline of 2–5 CD138+ cells/mL in healthy donor blood, with significantly higher numbers in blood samples of MM patients in remission (20–24 CD138+ cells/mL), and yet higher numbers in MM patients exhibiting disease (45–184 CD138+ cells/mL). Analysis of CPCs isolated using the device was consistent with serum immunoglobulin assays that are commonly used in MM diagnostics. These results indicate the potential of CD138-based microfluidic CPC capture as a useful ‘liquid biopsy’ that may complement or partially replace bone marrow aspiration.


Medical Physics | 2015

Technical Note: Immunohistochemical evaluation of mouse brain irradiation targeting accuracy with 3D-printed immobilization device.

Niloufar Zarghami; Michael D. Jensen; Srikanth Talluri; Paula J. Foster; Ann F. Chambers; Frederick A. Dick; Eugene Wong

PURPOSE Small animal immobilization devices facilitate positioning of animals for reproducible imaging and accurate focal radiation therapy. In this study, the authors demonstrate the use of three-dimensional (3D) printing technology to fabricate a custom-designed mouse head restraint. The authors evaluate the accuracy of this device for the purpose of mouse brain irradiation. METHODS A mouse head holder was designed for a microCT couch using cad software and printed in an acrylic based material. Ten mice received half-brain radiation while positioned in the 3D-printed head holder. Animal placement was achieved using on-board image guidance and computerized asymmetric collimators. To evaluate the precision of beam localization for half-brain irradiation, mice were sacrificed approximately 30 min after treatment and brain sections were stained for γ-H2AX, a marker for DNA breaks. The distance and angle of the γ-H2AX radiation beam border to longitudinal fissure were measured on histological samples. Animals were monitored for any possible trauma from the device. RESULTS Visualization of the radiation beam on ex vivo brain sections with γ-H2AX immunohistochemical staining showed a sharp radiation field within the tissue. Measurements showed a mean irradiation targeting error of 0.14±0.09 mm (standard deviation). Rotation between the beam axis and mouse head was 1.2°±1.0° (standard deviation). The immobilization device was easily adjusted to accommodate different sizes of mice. No signs of trauma to the mice were observed from the use of tooth block and ear bars. CONCLUSIONS The authors designed and built a novel 3D-printed mouse head holder with many desired features for accurate and reproducible radiation targeting. The 3D printing technology was found to be practical and economical for producing a small animal imaging and radiation restraint device and allows for customization for study specific needs.


Cell Division | 2017

Multiple molecular interactions redundantly contribute to RB-mediated cell cycle control

Michael J. Thwaites; Matthew J. Cecchini; Srikanth Talluri; Daniel T. Passos; Jasmyne Carnevale; Frederick A. Dick

BackgroundThe G1-S phase transition is critical to maintaining proliferative control and preventing carcinogenesis. The retinoblastoma tumor suppressor is a key regulator of this step in the cell cycle.ResultsHere we use a structure–function approach to evaluate the contributions of multiple protein interaction surfaces on pRB towards cell cycle regulation. SAOS2 cell cycle arrest assays showed that disruption of three separate binding surfaces were necessary to inhibit pRB-mediated cell cycle control. Surprisingly, mutation of some interaction surfaces had no effect on their own. Rather, they only contributed to cell cycle arrest in the absence of other pRB dependent arrest functions. Specifically, our data shows that pRB–E2F interactions are competitive with pRB–CDH1 interactions, implying that interchangeable growth arrest functions underlie pRB’s ability to block proliferation. Additionally, disruption of similar cell cycle control mechanisms in genetically modified mutant mice results in ectopic DNA synthesis in the liver.ConclusionsOur work demonstrates that pRB utilizes a network of mechanisms to prevent cell cycle entry. This has important implications for the use of new CDK4/6 inhibitors that aim to activate this proliferative control network.

Collaboration


Dive into the Srikanth Talluri's collaboration.

Top Co-Authors

Avatar

Frederick A. Dick

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Daniel T. Passos

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Matthew J. Cecchini

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Michael J. Thwaites

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Sarah M. Francis

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Alison L. Martens

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Charles A. Ishak

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Ian Welch

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

James I. S. MacDonald

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Shauna A. Henley

University of Western Ontario

View shared research outputs
Researchain Logo
Decentralizing Knowledge