Network


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

Hotspot


Dive into the research topics where Afsar Barlas is active.

Publication


Featured researches published by Afsar Barlas.


Cell | 2009

Nuclear CDKs Drive Smad Transcriptional Activation and Turnover in BMP and TGF-β Pathways

Claudio Alarcón; Alexia-Ileana Zaromytidou; Qiaoran Xi; Sheng Gao; Jianzhong Yu; Sho Fujisawa; Afsar Barlas; Alexandria N. Miller; Katia Manova-Todorova; Maria J. Macias; Gopal P. Sapkota; Duojia Pan; Joan Massagué

TGF-beta and BMP receptor kinases activate Smad transcription factors by C-terminal phosphorylation. We have identified a subsequent agonist-induced phosphorylation that plays a central dual role in Smad transcriptional activation and turnover. As receptor-activated Smads form transcriptional complexes, they are phosphorylated at an interdomain linker region by CDK8 and CDK9, which are components of transcriptional mediator and elongation complexes. These phosphorylations promote Smad transcriptional action, which in the case of Smad1 is mediated by the recruitment of YAP to the phosphorylated linker sites. An effector of the highly conserved Hippo organ size control pathway, YAP supports Smad1-dependent transcription and is required for BMP suppression of neural differentiation of mouse embryonic stem cells. The phosphorylated linker is ultimately recognized by specific ubiquitin ligases, leading to proteasome-mediated turnover of activated Smad proteins. Thus, nuclear CDK8/9 drive a cycle of Smad utilization and disposal that is an integral part of canonical BMP and TGF-beta pathways.


Cell | 2011

A Poised Chromatin Platform for TGF-β Access to Master Regulators

Qiaoran Xi; Zhanxin Wang; Alexia Ileana Zaromytidou; Xiang H.-F. Zhang; Lai Fong Chow-Tsang; Jing X. Liu; Hyesoo Kim; Afsar Barlas; Katia Manova-Todorova; Vesa Kaartinen; Lorenz Studer; Willie Mark; Dinshaw J. Patel; Joan Massagué

Specific chromatin marks keep master regulators of differentiation silent yet poised for activation by extracellular signals. We report that nodal TGF-β signals use the poised histone mark H3K9me3 to trigger differentiation of mammalian embryonic stem cells. Nodal receptors induce the formation of companion Smad4-Smad2/3 and TRIM33-Smad2/3 complexes. The PHD-Bromo cassette of TRIM33 facilitates binding of TRIM33-Smad2/3 to H3K9me3 and H3K18ac on the promoters of mesendoderm regulators Gsc and Mixl1. The crystal structure of this cassette, bound to histone H3 peptides, illustrates that PHD recognizes K9me3, and Bromo binds an adjacent K18ac. The interaction between TRIM33-Smad2/3 and H3K9me3 displaces the chromatin-compacting factor HP1γ, making nodal response elements accessible to Smad4-Smad2/3 for Pol II recruitment. In turn, Smad4 increases K18 acetylation to augment TRIM33-Smad2/3 binding. Thus, nodal effectors use the H3K9me3 mark as a platform to switch master regulators of stem cell differentiation from the poised to the active state.


PLOS ONE | 2012

Tunneling Nanotubes Provide a Unique Conduit for Intercellular Transfer of Cellular Contents in Human Malignant Pleural Mesothelioma

Emil Lou; Sho Fujisawa; Alexei Morozov; Afsar Barlas; Yevgeniy Romin; Yildirim Dogan; Sepideh Gholami; Andre L. Moreira; Katia Manova-Todorova; Malcolm A. S. Moore

Tunneling nanotubes are long, non-adherent F-actin-based cytoplasmic extensions which connect proximal or distant cells and facilitate intercellular transfer. The identification of nanotubes has been limited to cell lines, and their role in cancer remains unclear. We detected tunneling nanotubes in mesothelioma cell lines and primary human mesothelioma cells. Using a low serum, hyperglycemic, acidic growth medium, we stimulated nanotube formation and bidirectional transfer of vesicles, proteins, and mitochondria between cells. Notably, nanotubes developed between malignant cells or between normal mesothelial cells, but not between malignant and normal cells. Immunofluorescent staining revealed their actin-based assembly and structure. Metformin and an mTor inhibitor, Everolimus, effectively suppressed nanotube formation. Confocal microscopy with 3-dimensional reconstructions of sectioned surgical specimens demonstrated for the first time the presence of nanotubes in human mesothelioma and lung adenocarcinoma tumor specimens. We provide the first evidence of tunneling nanotubes in human primary tumors and cancer cells and propose that these structures play an important role in cancer cell pathogenesis and invasion.


Endocrinology | 2012

GLP-1 Receptor Agonists and the Thyroid: C-Cell Effects in Mice Are Mediated via the GLP-1 Receptor and not Associated with RET Activation

Lars Wichmann Madsen; Jeffrey A. Knauf; Carsten F. Gotfredsen; Andrew M. Pilling; Ingrid Sjögren; Søren Andersen; Lene Nonboe Andersen; Anne Sietske de Boer; Katia Manova; Afsar Barlas; Sushil Vundavalli; Niels C. Berg Nyborg; Lotte Bjerre Knudsen; Anne Marie Moelck; James A. Fagin

Liraglutide and exenatide are glucagon-like peptide receptor (GLP-1R) agonists used in the treatment of type 2 diabetes. Both molecules have been associated with the development of thyroid C-cell tumors after lifetime exposure in rodents. Previously, it has been reported that these tumors are preceded by increased plasma calcitonin and C-cell hyperplasia. We can now document that the murine C-cell effects are mediated via GLP-1R. Thus, 13 wk of continuous exposure to GLP-1R agonists was associated with marked increases in plasma calcitonin and in the incidence of C-cell hyperplasia in wild-type mice. In contrast, similar effects were not seen in GLP-1R knockout mice. Human C-cell cancer is often caused by activating mutations in the rearranged-during-transfection (RET) protooncogene. We developed an immunohistochemical method to assess RET activation in tissues. Liraglutide dosing to mice was not found to activate RET. Further evaluation of the signaling pathways demonstrated that liraglutide increased ribosomal S6, but not MAPK kinase, phosphorylation. These observations are consistent with effects of GLP-1R agonists on rodent C cells being mediated via mammalian target of rapamycin activation in a RET- and MAPK-independent manner.


Communicative & Integrative Biology | 2012

Tunneling Nanotubes: A new paradigm for studying intercellular communication and therapeutics in cancer.

Emil Lou; Sho Fujisawa; Afsar Barlas; Yevgeniy Romin; Katia Manova-Todorova; Malcolm A. S. Moore; Subbaya Subramanian

Tunneling nanotubes are actin-based cytoplasmic extensions that function as intercellular channels in a wide variety of cell types.There is a renewed and keen interest in the examination of modes of intercellular communication in cells of all types, especially in the field of cancer biology. Tunneling nanotubes –which in the literature have also been referred to as “membrane nanotubes,” “’intercellular’ or ‘epithelial’ bridges,” or “cytoplasmic extensions” – are under active investigation for their role in facilitating direct intercellular communication. These structures have not, until recently, been scrutinized as a unique and previously unrecognized form of direct cell-to-cell transmission of cellular cargo in the context of human cancer. Our recent study of tunneling nanotubes in human malignant pleural mesothelioma and lung adenocarcinomas demonstrated efficient transfer of cellular contents, including proteins, Golgi vesicles, and mitochondria, between cells derived from several well-established cancer cell lines. Further, we provided effective demonstration that such nanotubes can form between primary malignant cells from human patients. For the first time, we also demonstrated the in vivo relevance of these structures in humans, having effectively imaged nanotubes in intact solid tumors from patients. Here we provide further analysis and discussion on our findings, and offer a prospective ‘road map’ for studying tunneling nanotubes in the context of human cancer. We hope that further understanding of the mechanisms, methods of transfer, and particularly the role of nanotubes in tumor-stromal cross-talk will lead to identification of new selective targets for cancer therapeutics.


Experimental Cell Research | 2014

Tumor exosomes induce tunneling nanotubes in lipid raft-enriched regions of human mesothelioma cells.

Venugopal Thayanithy; Victor Babatunde; Elizabeth L. Dickson; Phillip Y.-P. Wong; Sanghoon Oh; Xu Ke; Afsar Barlas; Sho Fujisawa; Yevgeniy Romin; Andre L. Moreira; Robert J. Downey; Clifford J. Steer; Subbaya Subramanian; Katia Manova-Todorova; Malcolm A. S. Moore; Emil Lou

Tunneling nanotubes (TnTs) are long, non-adherent, actin-based cellular extensions that act as conduits for transport of cellular cargo between connected cells. The mechanisms of nanotube formation and the effects of the tumor microenvironment and cellular signals on TnT formation are unknown. In the present study, we explored exosomes as potential mediators of TnT formation in mesothelioma and the potential relationship of lipid rafts to TnT formation. Mesothelioma cells co-cultured with exogenous mesothelioma-derived exosomes formed more TnTs than cells cultured without exosomes within 24-48 h; and this effect was most prominent in media conditions (low-serum, hyperglycemic medium) that support TnT formation (1.3-1.9-fold difference). Fluorescence and electron microscopy confirmed the purity of isolated exosomes and revealed that they localized predominantly at the base of and within TnTs, in addition to the extracellular environment. Time-lapse microscopic imaging demonstrated uptake of tumor exosomes by TnTs, which facilitated intercellular transfer of these exosomes between connected cells. Mesothelioma cells connected via TnTs were also significantly enriched for lipid rafts at nearly a 2-fold higher number compared with cells not connected by TnTs. Our findings provide supportive evidence of exosomes as potential chemotactic stimuli for TnT formation, and also lipid raft formation as a potential biomarker for TnT-forming cells.


Molecular Cell | 2013

The Mre11 Complex Suppresses Oncogene-Driven Breast Tumorigenesis and Metastasis

Gaorav P. Gupta; Katelynd Vanness; Afsar Barlas; Katia Manova-Todorova; Yong H. Wen; John H.J. Petrini

The DNA damage response (DDR) is activated by oncogenic stress, but the mechanisms by which this occurs, and the particular DDR functions that constitute barriers to tumorigenesis, remain unclear. We established a mouse model of sporadic oncogene-driven breast tumorigenesis in a series of mutant mouse strains with specific DDR deficiencies to reveal a role for the Mre11 complex in the response to oncogene activation. We demonstrate that an Mre11-mediated DDR restrains mammary hyperplasia by effecting an oncogene-induced G2 arrest. Impairment of Mre11 complex functions promotes the progression of mammary hyperplasias into invasive and metastatic breast cancers, which are often associated with secondary inactivation of the Ink4a-Arf (CDKN2a) locus. These findings provide insight into the mechanism of DDR engagement by activated oncogenes and highlight genetic interactions between the DDR and Ink4a-Arf pathways in suppression of oncogene-driven tumorigenesis and metastasis.


Radiology | 2016

Colorectal Cancer Liver Metastases: Biopsy of the Ablation Zone and Margins Can Be Used to Predict Oncologic Outcome

Vlasios S. Sotirchos; Lydia M. Petrovic; Mithat Gonen; David S. Klimstra; Richard K. G. Do; Elena N. Petre; Alessandra R. Garcia; Afsar Barlas; Joseph P. Erinjeri; Karen T. Brown; Anne M. Covey; William Alago; Lynn A. Brody; Ronald P. DeMatteo; Nancy E. Kemeny; Stephen B. Solomon; Katia Manova-Todorova; Constantinos T. Sofocleous

Purpose To establish the prognostic value of biopsy of the central and marginal ablation zones for time to local tumor progression (LTP) after radiofrequency (RF) ablation of colorectal cancer liver metastasis (CLM). Materials and Methods A total of 47 patients with 67 CLMs were enrolled in this prospective institutional review board-approved and HIPAA-compliant study between November 2009 and August 2012. Mean tumor size was 2.1 cm (range, 0.6-4.3 cm). Biopsy of the center and margin of the ablation zone was performed immediately after RF ablation (mean number of biopsy samples per ablation zone, 1.9) and was evaluated for the presence of viable tumor cells. Samples containing tumor cells at morphologic evaluation were further interrogated with immunohistochemistry and were classified as either positive, viable tumor (V) or negative, necrotic (N). Minimal ablation margin size was evaluated in the first postablation CT study performed 4-8 weeks after ablation. Variables were evaluated as predictors of time to LTP with the competing-risks model (uni- and multivariate analyses). Results Technical effectiveness was evident in 66 of 67 (98%) ablated lesions on the first contrast material-enhanced CT images at 4-8-week follow-up. The cumulative incidence of LTP at 12-month follow-up was 22% (95% confidence interval [CI]: 12, 32). Samples from 16 (24%) of 67 ablation zones were classified as viable tumor. At univariate analysis, tumor size, minimal margin size, and biopsy results were significant in predicting LTP. When these variables were subsequently entered in a multivariate model, margin size of less than 5 mm (P < .001; hazard ratio [HR], 6.7) and positive biopsy results (P = .008; HR, 3.4) were significant. LTP within 12 months after RF ablation was noted in 3% (95% CI: 0, 9) of necrotic CLMs with margins of at least 5 mm. Conclusion Biopsy proof of complete tumor ablation and minimal ablation margins of at least 5 mm are independent predictors of LTP and yield the best oncologic outcomes. (©) RSNA, 2016.


Scientific Reports | 2015

Machine-based method for multiplex in situ molecular characterization of tissues by immunofluorescence detection

Dmitry Yarilin; Ke Xu; Mesruh Turkekul; Ning Fan; Yevgeniy Romin; Sho Fijisawa; Afsar Barlas; Katia Manova-Todorova

Immunofluorescent staining is an informative tool that is widely used in basic research. Automation of immunostaining improves reproducibility and quality of the results. Up to now, use of automation in immunofluorescent staining was mostly limited to one marker. Here we present tyramide signal amplification based method of multiple marker immunofluorescent detection, including detection of antibodies, raised in the same species, in tissue sections and cultured cells. This method can be beneficial for both basic and clinical research.


Nature Communications | 2016

Deconvoluting hepatic processing of carbon nanotubes.

Simone Alidori; Robert L. Bowman; Dmitry Yarilin; Yevgeniy Romin; Afsar Barlas; J. Justin Mulvey; Sho Fujisawa; Ke Xu; Alessandro Ruggiero; Vladimir Riabov; Daniel L. J. Thorek; Hans David S. Ulmert; Elliott J. Brea; Katja Behling; Julia Kzhyshkowska; Katia Manova-Todorova; David A. Scheinberg; Michael R. McDevitt

Single-wall carbon nanotubes present unique opportunities for drug delivery, but have not advanced into the clinic. Differential nanotube accretion and clearance from critical organs have been observed, but the mechanism not fully elucidated. The liver has a complex cellular composition that regulates a range of metabolic functions and coincidently accumulates most particulate drugs. Here we provide the unexpected details of hepatic processing of covalently functionalized nanotubes including receptor-mediated endocytosis, cellular trafficking and biliary elimination. Ammonium-functionalized fibrillar nanocarbon is found to preferentially localize in the fenestrated sinusoidal endothelium of the liver but not resident macrophages. Stabilin receptors mediate the endocytic clearance of nanotubes. Biocompatibility is evidenced by the absence of cell death and no immune cell infiltration. Towards clinical application of this platform, nanotubes were evaluated for the first time in non-human primates. The pharmacologic profile in cynomolgus monkeys is equivalent to what was reported in mice and suggests that nanotubes should behave similarly in humans.

Collaboration


Dive into the Afsar Barlas's collaboration.

Top Co-Authors

Avatar

Katia Manova-Todorova

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Sho Fujisawa

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Mesruh Turkekul

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ning Fan

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Dmitry Yarilin

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Yevgeniy Romin

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ke Xu

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Emil Lou

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar

Katia Manova

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Malcolm A. S. Moore

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge