Network


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

Hotspot


Dive into the research topics where Maksim Sinyuk is active.

Publication


Featured researches published by Maksim Sinyuk.


Cancer Research | 2015

Cancer Stem Cells: Targeting the Roots of Cancer, Seeds of Metastasis, and Sources of Therapy Resistance

Valery Adorno-Cruz; Golam Kibria; Xia Liu; Mary R. Doherty; Damian J. Junk; Dongyin Guan; Christopher G. Hubert; Monica Venere; Erin E. Mulkearns-Hubert; Maksim Sinyuk; Alvaro G. Alvarado; Arnold I. Caplan; Jeremy N. Rich; Stanton L. Gerson; Justin D. Lathia; Huiping Liu

With the goal to remove the roots of cancer, eliminate metastatic seeds, and overcome therapy resistance, the 2014 inaugural International Cancer Stem Cell (CSC) Conference at Cleveland, OH, convened together over 320 investigators, including 55 invited world-class speakers, 25 short oral presenters, and 100 poster presenters, to gain an in-depth understanding of CSCs and explore therapeutic opportunities targeting CSCs. The meeting enabled intriguing discussions on several topics including: genetics and epigenetics; cancer origin and evolution; microenvironment and exosomes; metabolism and inflammation; metastasis and therapy resistance; single cell and heterogeneity; plasticity and reprogramming; as well as other new concepts. Reports of clinical trials targeting CSCs emphasized the urgent need for strategically designing combinational CSC-targeting therapies against cancer.


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


Neuro-oncology | 2016

The intersection of cancer, cancer stem cells, and the immune system: therapeutic opportunities.

Daniel J. Silver; Maksim Sinyuk; Michael A. Vogelbaum; Manmeet S. Ahluwalia; Justin D. Lathia

During brain neoplasia, malignant cells subjugate the immune system to provide an environment that favors tumor growth. These mechanisms capitalize on tumor-promoting functions of various immune cell types and typically result in suppression of tumor immune rejection. Immunotherapy efforts are underway to disrupt these mechanisms and turn the immune system against developing tumors. While many of these therapies are already in early-stage clinical trials, understanding how these therapies impact various tumor cell populations, including self-renewing cancer stem cells, may help to predict their efficacy and clarify their mechanisms of action. Moreover, interrogating the biology of glioma cell, cancer stem cell, and immune cell interactions may provide additional therapeutic targets to leverage against disease progression. In this review, we begin by highlighting a series of investigations into immune cell-mediated tumor promotion that do not parse the tumor into stem and non-stem components. We then take a closer look at the immune-suppressive mechanisms derived specifically from cancer stem cell interactions with the immune system and end with an update on immunotherapy and cancer stem cell-directed clinical trials in glioblastoma.


PLOS ONE | 2012

Context Dependent Role of the CD36 - Thrombospondin - Histidine-Rich Glycoprotein Axis in Tumor Angiogenesis and Growth

James S. Hale; Meizhang Li; Maksim Sinyuk; Willi Jahnen-Dechent; Justin D. Lathia; Roy L. Silverstein

The angiogenic switch is a promising therapeutic target in cancer. Work by our laboratory and others has described an important endogenous anti-angiogenic pathway mediated by interactions of CD36, a receptor on microvascular endothelial cells, with proteins containing thrombospondin (TSP) type I repeat domains (TSR). Recent studies revealed that circulating Histidine Rich Glycoprotein (HRG) inhibits the anti-angiogenic potential of the CD36-TSR pathway by functioning as a decoy receptor that binds and sequesters TSR proteins. As tumors of different origin display variable expression profiles of numerous targets, we hypothesized that the TSP-CD36-HRG axis regulates vascularization and growth in the tumor microenvironment in a context, or tumor type, dependent manner. Growth of Lewis Lung Carcinoma (LL2) and B16F1 Melanoma tumor cell implants in syngeneic wild type (WT), hrg, or cd36 null mice were used as a model to interrogate this signaling axis. LL2 tumor volumes were greater in cd36 null mice and smaller in hrg null mice compared to WT. Immunofluorescent staining showed increased vascularity in cd36 null vs. WT and WT vs. hrg null mice. No differences in tumor growth or vascularity were observed with B16F1 implants, consistent with lack of expression of TSP-1 in B16F1 cells. When TSR expression was induced in B16F1 cells by cDNA transfection, tumor growth and vascularity were similar to that seen with LL2 cells. These data show a role for CD36-mediated anti-angiogenic activity in the tumor microenvironment when TSR proteins are available and demonstrate that HRG modulates this activity. Further, they suggest a mechanism by which tumor microenvironments may regulate sensitivity to TSR containing proteins.


Neuro-oncology | 2016

Insulin-mediated signaling promotes proliferation and survival of glioblastoma through Akt activation

Yuanying Gong; Yufang Ma; Maksim Sinyuk; Sudan Loganathan; Reid C. Thompson; Jann N. Sarkaria; Wenbiao Chen; Justin D. Lathia; Bret C. Mobley; Stephen W. Clark; Jialiang Wang

BACKGROUND Metabolic complications such as obesity, hyperglycemia, and type 2 diabetes are associated with poor outcomes in patients with glioblastoma. To control peritumoral edema, use of chronic high-dose steroids in glioblastoma patients is common, which can result in de novo diabetic symptoms. These metabolic complications may affect tumors via profound mechanisms, including activation of insulin receptor (InsR) and the related insulin-like growth factor 1 receptor (IGF1R) in malignant cells. METHODS In the present study, we assessed expression of InsR in glioblastoma surgical specimens and glioblastoma response to insulin at physiologically relevant concentrations. We further determined whether genetic or pharmacological targeting of InsR affected oncogenic functions of glioblastoma in vitro and in vivo. RESULTS We showed that InsR was commonly expressed in glioblastoma surgical specimens and xenograft tumor lines, with mitogenic isoform-A predominating. Insulin at physiologically relevant concentrations promoted glioblastoma cell growth and survival, potentially via Akt activation. Depletion of InsR impaired cellular functions and repressed orthotopic tumor growth. The absence of InsR compromised downstream Akt activity, but yet stimulated IGF1R expression. Targeting both InsR and IGF1R with dual kinase inhibitors resulted in effective blockade of downstream signaling, loss of cell viability, and repression of xenograft tumor growth. CONCLUSIONS Taken together, our work suggests that glioblastoma is sensitive to the mitogenic functions of insulin, thus significant insulin exposure imposes risks to glioblastoma patients. Additionally, dual inhibition of InsR and IGF1R exhibits promise for treating glioblastoma.


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.

Collaboration


Dive into the Maksim Sinyuk's collaboration.

Top Co-Authors

Avatar
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
Researchain Logo
Decentralizing Knowledge