Network


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

Hotspot


Dive into the research topics where Balint Otvos is active.

Publication


Featured researches published by Balint Otvos.


Stem Cells | 2014

Cancer Stem Cell-Specific Scavenger Receptor CD36 Drives Glioblastoma Progression

James S. Hale; Balint Otvos; Maksim Sinyuk; Alvaro G. Alvarado; Masahiro Hitomi; Kevin Stoltz; Qiulian Wu; William A. Flavahan; Bruce S. Levison; Mette L. Johansen; David Schmitt; Janna M. Neltner; Ping Huang; Bin Ren; Andrew E. Sloan; Roy L. Silverstein; Candece L. Gladson; Joseph A. DiDonato; J. Mark Brown; Thomas M. McIntyre; Stanley L. Hazen; Craig Horbinski; Jeremy N. Rich; Justin D. Lathia

Glioblastoma (GBM) contains a self‐renewing, tumorigenic cancer stem cell (CSC) population which contributes to tumor propagation and therapeutic resistance. While the tumor microenvironment is essential to CSC self‐renewal, the mechanisms by which CSCs sense and respond to microenvironmental conditions are poorly understood. Scavenger receptors are a broad class of membrane receptors well characterized on immune cells and instrumental in sensing apoptotic cellular debris and modified lipids. Here, we provide evidence that CSCs selectively use the scavenger receptor CD36 to promote their maintenance using patient‐derived CSCs and in vivo xenograft models. CD36 expression was observed in GBM cells in addition to previously described cell types including endothelial cells, macrophages, and microglia. CD36 was enriched in CSCs and was able to functionally distinguish self‐renewing cells. CD36 was coexpressed with integrin alpha 6 and CD133, previously described CSC markers, and CD36 reduction resulted in concomitant loss of integrin alpha 6 expression, self‐renewal, and tumor initiation capacity. We confirmed oxidized phospholipids, ligands of CD36, were present in GBM and found that the proliferation of CSCs, but not non‐CSCs, increased with exposure to oxidized low‐density lipoprotein. CD36 was an informative biomarker of malignancy and negatively correlated to patient prognosis. These results provide a paradigm for CSCs to thrive by the selective enhanced expression of scavenger receptors, providing survival, and metabolic advantages. Stem Cells 2014;32:1746–1758


Cell Reports | 2015

Differential Connexin Function Enhances Self-Renewal in Glioblastoma

Masahiro Hitomi; Loic P. Deleyrolle; Erin E. Mulkearns-Hubert; Awad Jarrar; Meizhang Li; Maksim Sinyuk; Balint Otvos; Sylvain Brunet; William A. Flavahan; Christopher G. Hubert; Winston Goan; James S. Hale; Alvaro G. Alvarado; Ao Zhang; Mark Rohaus; Muna Oli; Vinata Vedam-Mai; Jeff M. Fortin; Hunter S. Futch; Benjamin Griffith; Qiulian Wu; Chun hong Xia; Xiaohua Gong; Manmeet S. Ahluwalia; Jeremy N. Rich; Brent A. Reynolds; Justin D. Lathia

SUMMARY The coordination of complex tumor processes requires cells to rapidly modify their phenotype and is achieved by direct cell-cell communication through gap junction channels composed of connexins. Previous reports have suggested that gap junctions are tumor suppressive based on connexin43 (Cx43), but this does not take into account differences in connexin-mediated ion selectivity and intercellular communication rate that drive gap junction diversity. We find that glioblastoma cancer stem cells (CSCs) possess functional gap junctions that can be targeted using clinically relevant compounds to reduce self-renewal and tumor growth. Our analysis reveals that CSCs express Cx46, while Cx43 is predominantly expressed in non-CSCs. During differentiation, Cx46 is reduced, while Cx43 is increased, and targeting Cx46 compromises CSC maintenance. The difference between Cx46 and Cx43 is reflected in elevated cell-cell communication and reduced resting membrane potential in CSCs. Our data demonstrate a pro-tumorigenic role for gap junctions that is dependent on connexin expression.


Stem Cells | 2015

Development of a Fluorescent Reporter System to Delineate Cancer Stem Cells in Triple-Negative Breast Cancer.

Praveena S. Thiagarajan; Masahiro Hitomi; James S. Hale; Alvaro G. Alvarado; Balint Otvos; Maksim Sinyuk; Kevin Stoltz; Andrew Wiechert; Erin E. Mulkearns-Hubert; Awad Jarrar; Qiao Zheng; Dustin Thomas; Thomas T. Egelhoff; Jeremy N. Rich; Huiping Liu; Justin D. Lathia; Ofer Reizes

Advanced cancers display cellular heterogeneity driven by self‐renewing, tumorigenic cancer stem cells (CSCs). The use of cell lines to model CSCs is challenging due to the difficulty of identifying and isolating cell populations that possess differences in self‐renewal and tumor initiation. To overcome these barriers in triple‐negative breast cancer (TNBC), we developed a CSC system using a green fluorescent protein (GFP) reporter for the promoter of the well‐established pluripotency gene NANOG. NANOG‐GFP+ cells gave rise to both GFP+ and GFP− cells, and GFP+ cells possessed increased levels of the embryonic stem cell transcription factors NANOG, SOX2, and OCT4 and elevated self‐renewal and tumor initiation capacities. GFP+ cells also expressed mesenchymal markers and demonstrated increased invasion. Compared with the well‐established CSC markers CD24−/CD44+, CD49f, and aldehyde dehydrogenase (ALDH) activity, our NANOG‐GFP reporter system demonstrated increased enrichment for CSCs. To explore the utility of this system as a screening platform, we performed a flow cytometry screen that confirmed increased CSC marker expression in the GFP+ population and identified new cell surface markers elevated in TNBC CSCs, including junctional adhesion molecule‐A (JAM‐A). JAM‐A was highly expressed in GFP+ cells and patient‐derived xenograft ALDH+ CSCs compared with the GFP− and ALDH− cells, respectively. Depletion of JAM‐A compromised self‐renewal, whereas JAM‐A overexpression induced self‐renewal in GFP− cells. Our data indicate that we have defined and developed a robust system to monitor differences between CSCs and non‐CSCs in TNBC that can be used to identify CSC‐specific targets for the development of future therapeutic strategies. Stem Cells. Stem Cells 2015;33:2114–2125


Stem Cells | 2016

Cancer Stem Cell-Secreted Macrophage Migration Inhibitory Factor Stimulates Myeloid Derived Suppressor Cell Function and Facilitates Glioblastoma Immune Evasion.

Balint Otvos; Daniel J. Silver; Erin E. Mulkearns-Hubert; Alvaro G. Alvarado; Soumya M. Turaga; Mia D. Sørensen; Patricia Rayman; William A. Flavahan; James S. Hale; Kevin Stoltz; Maksim Sinyuk; Qiulian Wu; Awad Jarrar; Sung Hak Kim; Paul L. Fox; Ichiro Nakano; Jeremy N. Rich; Richard M. Ransohoff; James Finke; Bjarne Winther Kristensen; Michael A. Vogelbaum; Justin D. Lathia

Shifting the balance away from tumor‐mediated immune suppression toward tumor immune rejection is the conceptual foundation for a variety of immunotherapy efforts currently being tested. These efforts largely focus on activating antitumor immune responses but are confounded by multiple immune cell populations, including myeloid‐derived suppressor cells (MDSCs), which serve to suppress immune system function. We have identified immune‐suppressive MDSCs in the brains of GBM patients and found that they were in close proximity to self‐renewing cancer stem cells (CSCs). MDSCs were selectively depleted using 5‐flurouracil (5‐FU) in a low‐dose administration paradigm, which resulted in prolonged survival in a syngeneic mouse model of glioma. In coculture studies, patient‐derived CSCs but not nonstem tumor cells selectively drove MDSC‐mediated immune suppression. A cytokine screen revealed that CSCs secreted multiple factors that promoted this activity, including macrophage migration inhibitory factor (MIF), which was produced at high levels by CSCs. Addition of MIF increased production of the immune‐suppressive enzyme arginase‐1 in MDSCs in a CXCR2‐dependent manner, whereas blocking MIF reduced arginase‐1 production. Similarly to 5‐FU, targeting tumor‐derived MIF conferred a survival advantage to tumor‐bearing animals and increased the cytotoxic T cell response within the tumor. Importantly, tumor cell proliferation, survival, and self‐renewal were not impacted by MIF reduction, demonstrating that MIF is primarily an indirect promoter of GBM progression, working to suppress immune rejection by activating and protecting immune suppressive MDSCs within the GBM tumor microenvironment. Stem Cells 2016;34:2026–2039


Neurosurgery | 2015

Tumor progression in patients receiving adjuvant whole-brain radiotherapy vs localized radiotherapy after surgical resection of brain metastases.

Jason K. Hsieh; Paul Elson; Balint Otvos; Jonathan A. Rose; Christopher Loftus; Gazanfar Rahmathulla; Lilyana Angelov; Gene H. Barnett; Robert J. Weil; Michael A. Vogelbaum

BACKGROUND Surgery followed by adjuvant radiotherapy is a well-established treatment paradigm for brain metastases. OBJECTIVE To examine the effect of postsurgical whole-brain radiotherapy (WBRT) or localized radiotherapy (LRT), including stereotactic radiosurgery and intraoperative radiotherapy, on the rate of recurrence both local and distal to the resection site in the treatment of brain metastases. METHODS We retrospectively identified patients who underwent surgery for brain metastasis at the Cleveland Clinic between 2004 and 2012. Institutional review board-approved chart review was conducted, and patients who had radiation before surgery, who had nonmetastatic lesions, or who lacked postadjuvant imaging were excluded. RESULTS The final analysis included 212 patients. One hundred fifty-six patients received WBRT, 37 received stereotactic radiosurgery only, and 19 received intraoperative radiotherapy. One hundred forty-six patients were deceased, of whom 60 (41%) died with no evidence of recurrence. Competing risks methodology was used to test the association between adjuvant modality and progression. Multivariable analysis revealed no significant difference in the rate of recurrence at the resection site (hazard ratio [HR] 1.46, P = .26) or of unresected, radiotherapy-treated lesions (HR 1.70, P = .41) for LRT vs WBRT. Patients treated with LRT had an increased hazard of the development of new lesions (HR 2.41, P < .001) and leptomeningeal disease (HR 2.45, P = .04). Median survival was 16.5 months and was not significantly different between groups. CONCLUSION LRT as adjuvant treatment to surgical resection of brain metastases is associated with an increased rate of development of new distant metastases and leptomeningeal disease compared with WBRT, but not with recurrence at the resection site or of unresected lesions treated with radiation.


Neuro-oncology | 2016

Coordination of self-renewal in glioblastoma by integration of adhesion and microRNA signaling

Alvaro G. Alvarado; Soumya M. Turaga; Pratheesh Sathyan; Erin E. Mulkearns-Hubert; Balint Otvos; Daniel J. Silver; James S. Hale; William A. Flavahan; Pascal O. Zinn; Maksim Sinyuk; Meizhang Li; Maheedhara R. Guda; Kiran Kumar Velpula; Andrew J. Tsung; Ichiro Nakano; Michael A. Vogelbaum; Sadhan Majumder; Jeremy N. Rich; Justin D. Lathia

BACKGROUND Cancer stem cells (CSCs) provide an additional layer of complexity for tumor models and targets for therapeutic development. The balance between CSC self-renewal and differentiation is driven by niche components including adhesion, which is a hallmark of stemness. While studies have demonstrated that the reduction of adhesion molecules, such as integrins and junctional adhesion molecule-A (JAM-A), decreases CSC maintenance. The molecular circuitry underlying these interactions has yet to be resolved. METHODS MicroRNA screening predicted that microRNA-145 (miR-145) would bind to JAM-A. JAM-A overexpression in CSCs was evaluated both in vitro (proliferation and self-renewal) and in vivo (intracranial tumor initiation). miR-145 introduction into CSCs was similarly assessed in vitro. Additionally, The Cancer Genome Atlas dataset was evaluated for expression levels of miR-145 and overall survival of the different molecular groups. RESULTS Using patient-derived glioblastoma CSCs, we confirmed that JAM-A is suppressed by miR-145. CSCs expressed low levels of miR-145, and its introduction decreased self-renewal through reductions in AKT signaling and stem cell marker (SOX2, OCT4, and NANOG) expression; JAM-A overexpression rescued these effects. These findings were predictive of patient survival, with a JAM-A/miR-145 signature robustly predicting poor patient prognosis. CONCLUSIONS Our results link CSC-specific niche signaling to a microRNA regulatory network that is altered in glioblastoma and can be targeted to attenuate CSC self-renewal.


Neuro-oncology | 2015

Development of a Sox2 reporter system modeling cellular heterogeneity in glioma

Kevin Stoltz; Maksim Sinyuk; James S. Hale; Qiulian Wu; Balint Otvos; Kiera Walker; Amit Vasanji; Jeremy N. Rich; Anita B. Hjelmeland; Justin D. Lathia

BACKGROUND Malignant gliomas are complex systems containing a number of factors that drive tumor initiation and progression, including genetic aberrations that lead to extensive cellular heterogeneity within the neoplastic compartment. Mouse models recapitulate these genetic aberrations, but readily observable heterogeneity remains challenging. METHODS To interrogate cellular heterogeneity in mouse glioma models, we utilized a replication-competent avian sarcoma-leukosis virus long terminal repeat with splice acceptor/tumor virus A (RCAS-tva) system to generate spontaneous mouse gliomas that contained a Sox2-enhanced green fluorescent protein (EGFP) reporter. Glial fibrillary acidic protein-tva mice were crossed with Sox2-EGFP mice, and tumors were initiated that contained a subpopulation of Sox2-EGFP-high cells enriched for tumor-initiating cell properties such as self-renewal, multilineage differentiation potential, and perivascular localization. RESULTS Following implantation into recipient mice, Sox2-EGFP-high cells generated tumors containing Sox2-EGFP-high and Sox2-EGFP-low cells. Kinomic analysis of Sox2-EGFP-high cells revealed activation of known glioma signaling pathways that are strongly correlated with patient survival including platelet-derived growth factor receptor beta, phosphoinositide-3 kinase, and vascular endothelial growth factor. Our functional analysis identified active feline sarcoma (Fes) signaling in Sox2-EGFP-high cells. Fes negatively correlated with glioma patient survival and was coexpressed with Sox2-positive cells in glioma xenografts and primary patient-derived tissue. CONCLUSIONS Our RCAS-tva/Sox2-EGFP model will empower closer examination of cellular heterogeneity and will be useful for identifying novel glioma pathways as well as testing preclinical treatment efficacy.


International Journal of Surgery Case Reports | 2016

Delayed presentation of an arteriovenous malformation after cerebellar hemangioblastoma resection—Case report

E. Emily Bennett; Balint Otvos; Varun R. Kshettry; Jorge Gonzalez-Martinez

Highlights • Hemangioblastoma rarely occur in coexistence either temporally or spatially with the development of an AVM. We present a case of delayed AVM following hemangioblastoma resection.• Recent research supports both hemangioblastoma and AVM are of embryologic origin but require later genetic alterations to develop into symptomatic lesions.• It is unclear in our case if the AVM was present at the time of the initial hemangioblastoma resection or developed de novo after tumor resection.• More research is needed to elucidate the rare intermixture of these lesions.


Neuro-oncology | 2014

SC-11DIFFERENTIAL CONNEXIN FUNCTION ENHANCES SELF-RENEWAL IN GLIOBLASTOMA

Masahiro Hitomi; Loic P. Deleyrolle; Maksim Sinyuk; Balint Otvos; Sylvain Brunet; Awad Jarrar; William A. Flavahan; Winston Goan; Erin E. Mulkearns-Hubert; Ao Zhang; Mark Rohaus; Muna Oli; Vinata Vedam-Mai; Jeff M. Fortin; Hunter S. Futch; Qiulian Wu; Manmeet S. Ahluwalia; Jeremy N. Rich; Brent A. Reynolds; Justin D. Lathia

The coordination of complex tumor processes requires cells to rapidly modify their phenotypes using direct cell-cell communication through gap junction channels composed of connexins. Previous reports suggest that gap junctions are tumor suppressors based on connexin 43 (Cx43), but this hypothesis fails to consider the differences in connexin-mediated intercellular communication rate and ion selectivity that drive gap junction diversity. Using patient-derived specimens, we screened connexin proteins and found that glioblastoma cancer stem cells (CSCs) expressed Cx46, while Cx43 was predominantly expressed in non-CSCs. Targeting Cx46 compromised CSC proliferation, self-renewal, and tumor initiation. Consistent with the divergent physiological nature of intercellular communication and ion selectivity between Cx46 and Cx43, CSCs with higher Cx46 had an elevated intercellular communication rate and were more depolarized than non-CSCs. The difference in connexin subtype was responsible for these phenotypic differences; Cx46 knockdown reduced the communication rate of CSCs, and Cx43 knockdown increased the depolarization of non-CSCs. The differences between the two connexins were reflected in GBM patient survival: Cx46 expression correlated with poor prognosis, while Cx43 expression was not informative. Ongoing studies are identifying differentially transported signaling molecules that are responsible for CSC maintenance based on connexin subunits. As clinically relevant gap junction inhibitors including 1-Octanol are being tested for other neurological disorders (essential tremor), we evaluated if these inhibitors were effective in glioblastoma. We confirmed that CSCs possessed functional gap junctions and that inhibitors reduced communication. These inhibitors potently inhibited proliferation and self-renewal of CSCs compared with non-CSCs and neural progenitor cells. In established xenograft tumors, gap junction inhibitors suppressed tumor growth and had an additive effect when combined with Temozolomide. Taken together, our data demonstrate a pro-tumorigenic role of gap junctions that is dependent on connexin subunit expression and provide a rationale for gap junction targeting in glioblastoma.


Journal for ImmunoTherapy of Cancer | 2013

Interrogating the interactions between myeloid derived suppressor cells and cancer stem cells in glioblastoma

Balint Otvos; James H. Finke; Michael A. Vogelbaum; Justin D. Lathia

Cellular and molecular regulation of the immune system is exquisitely controlled in the brain and is disrupted in neoplasia. Despite accumulation of immune cells in the tumor microenvironment, glioblastoma (GBM) growth persists while the mechanisms suppressing immune function remain largely unknown. Myeloid derived suppressor cells (MDSCs) are a heterogeneous class of immune cells responsible for immunomodulation through the suppression of cytotoxic T-cells. These populations are elevated in the peripheral blood of GBM patients, but their roles within the GBM microenvironment are uncharacterized. Through immunofluorescence analysis, we identified MDSCs within human GBM specimens, suggesting an immunosuppressive phenotype marked by arginase 1 (Arg1) staining. We have also observed co-localization between MDSCs and GBM cancer stem cells (CSCs) in both human tissues and mouse xenografts, leading to the hypothesis that CSCs recruit MDSCs to the tumor microenvironment, promote their survival, and that MDSCs are responsible for the immune-evasive properties of CSCs (Fig. ​(Fig.1A).1A). Intracranial (IC) injections of CSCs into mice have led to GBM formation, expansion of the Arg1-producing MDSCs within the marrow of GBM-bearing mice, and increased levels of MDSCs within the GBM microenvironment (Fig. 1B-E). CSC conditioned media promoted decreased apoptosis and increased Arg1 production of murine bone marrow derived MDSCs. Our results suggest a critical role by which GBM is able to promote immunosuppression through the recruitment of MDSCs and a novel paradigm of immunomodulation by CSCs in GBM. Figure 1 A. MDSCs detected in GBM xenografts by labeling with Sox2, CD244.1, and Arg1. Yellow arrows indicate CSCs; white arrows indicate MDSCs. B. H&E staining of IC xenografts. C. GBM-bearing mice have increased MDSCs by flow cytometry plots (boxed area). ...

Collaboration


Dive into the Balint Otvos's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jeremy N. Rich

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge