Network


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

Hotspot


Dive into the research topics where M. Reza Saadatzadeh is active.

Publication


Featured researches published by M. Reza Saadatzadeh.


Circulation Research | 2009

Robust functional vascular network formation in vivo by cooperation of adipose progenitor and endothelial cells.

Dmitry O. Traktuev; Daniel Prater; Stephanie Merfeld-Clauss; Aravind Raj Sanjeevaiah; M. Reza Saadatzadeh; Michael P. Murphy; Brian H. Johnstone; David A. Ingram; Keith L. March

Rapid induction and maintenance of blood flow through new vascular networks is essential for successfully treating ischemic tissues and maintaining function of engineered neo-organs. We have previously shown that human endothelial progenitor cells (EPCs) form functioning vessels in mice, but these are limited in number and persistence; and also that human adipose stromal cells (ASCs) are multipotent cells with pericytic properties which can stabilize vascular assembly in vitro. In this study, we tested whether ASCs would cooperate with EPCs to coassemble vessels in in vivo implants. Collagen implants containing EPCs, ASCs, or a 4:1 mixture of both were placed subcutaneously into NOD/SCID mice. After a range of time periods, constructs were explanted and evaluated with regard to vascular network assembly and cell fate; and heterotypic cell interactions were explored by targeted molecular perturbations. The density and complexity of vascular networks formed by the synergistic dual-cell system was many-fold higher than found in implants containing either ASCs or EPCs alone. Coimplantation of ASCs and EPCs with either pancreatic islets or adipocytes produced neoorgans populated by these parenchymal cells, as well as by chimeric human vessels conducting flow. This study is the first to demonstrate prompt and consistent assembly of a vascular network by human ASCs and endothelial cells and vascularization by these cells of parenchymal cells in implants. Mixture of these 2 readily available, nontransformed human cell types provides a practical approach to tissue engineering, therapeutic revascularization, and in vivo studies of human vasculogenesis.


Stem Cells | 2007

Clonogenic Endothelial Progenitor Cells Are Sensitive to Oxidative Stress

David A. Ingram; Theresa R. Krier; Laura E. Mead; Colleen McGuire; Daniel Prater; Janak R. Bhavsar; M. Reza Saadatzadeh; Khadijeh Bijangi-Vishehsaraei; Fang Li; Mervin C. Yoder; Laura S. Haneline

Endothelial progenitor cells (EPCs) circulate in the peripheral blood and reside in blood vessel walls. A hierarchy of EPCs exists where progenitors can be discriminated based on their clonogenic potential. EPCs are exposed to oxidative stress during vascular injury as residents of blood vessel walls or as circulating cells homing to sites of neovascularization. Given the links between oxidative injury, endothelial cell dysfunction, and vascular disease, we tested whether EPCs were sensitive to oxidative stress using newly developed clonogenic assays. Strikingly, in contrast to previous reports, we demonstrate that the most proliferative EPCs (high proliferative potential‐endothelial colony‐forming cells and low proliferative potential‐endothelial colony‐forming cells) had decreased clonogenic capacity after oxidant treatment. In addition, EPCs exhibited increased apoptosis and diminished tube‐forming ability in vitro and in vivo in response to oxidative stress, which was directly linked to activation of a redox‐dependent stress‐induced kinase pathway. Thus, this study provides novel insights into the effect of oxidative stress on EPCs. Furthermore, this report outlines a framework for understanding how oxidative injury leads to vascular disease and potentially limits the efficacy of transplantation of EPCs into ischemic tissues enriched for reactive oxygen species and oxidized metabolites.


Circulation | 2012

Experimental Abdominal Aortic Aneurysm Formation Is Mediated by IL-17 and Attenuated by Mesenchymal Stem Cell Treatment

Ashish K. Sharma; Guanyi Lu; Andrea L. Jester; William F. Johnston; Yunge Zhao; Vanessa A. Hajzus; M. Reza Saadatzadeh; Gang Su; Castigliano M. Bhamidipati; Gaurav S. Mehta; Irving L. Kron; Victor E. Laubach; Michael P. Murphy; Gorav Ailawadi; Gilbert R. Upchurch

Background— Abdominal aortic aneurysm (AAA) formation is characterized by inflammation, smooth muscle activation and matrix degradation. This study tests the hypothesis that CD4+ T-cell–produced IL-17 modulates inflammation and smooth muscle cell activation, leading to the pathogenesis of AAA and that human mesenchymal stem cell (MSC) treatment can attenuate IL-17 production and AAA formation. Methods and Results— Human aortic tissue demonstrated a significant increase in IL-17 and IL-23 expression in AAA patients compared with control subjects as analyzed by RT-PCR and ELISA. AAA formation was assessed in C57BL/6 (wild-type; WT), IL-23−/− or IL-17−/− mice using an elastase-perfusion model. Heat-inactivated elastase was used as control. On days 3, 7, and 14 after perfusion, abdominal aorta diameter was measured by video micrometry, and aortic tissue was analyzed for cytokines, cell counts, and IL-17–producing CD4+ T cells. Aortic diameter and cytokine production (MCP-1, RANTES, KC, TNF-&agr;, MIP-1&agr;, and IFN-&ggr;) was significantly attenuated in elastase-perfused IL-17−/− and IL-23−/− mice compared with WT mice on day 14. Cellular infiltration (especially IL-17–producing CD4+ T cells) was significantly attenuated in elastase-perfused IL-17−/− mice compared with WT mice on day 14. Primary aortic smooth muscle cells were significantly activated by elastase or IL-17 treatment. Furthermore, MSC treatment significantly attenuated AAA formation and IL-17 production in elastase-perfused WT mice. Conclusions— These results demonstrate that CD4+ T-cell–produced IL-17 plays a critical role in promoting inflammation during AAA formation and that immunomodulation of IL-17 by MSCs can offer protection against AAA formation.


Blood | 2009

Distinct roles of stress-activated protein kinases in Fanconi anemia type C-deficient hematopoiesis

M. Reza Saadatzadeh; Khadijeh Bijangi-Vishehsaraei; Reuben Kapur; Laura S. Haneline

The underlying molecular mechanisms that promote bone marrow failure in Fanconi anemia are incompletely understood. Evidence suggests that enhanced apoptosis of hematopoietic precursors is a major contributing factor. Previously, enhanced apoptosis of Fanconi anemia type C-deficient (Fancc(-/-)) progenitors was shown to involve aberrant p38 MAPK activation. Given the importance of c-Jun N-terminal kinase (JNK) in the stress response, we tested whether enhanced apoptosis of Fancc(-/-) cells also involved altered JNK activation. In Fancc(-/-) murine embryonic fibroblasts, tumor necrosis factor alpha (TNF-alpha) induced elevated JNK activity. In addition, JNK inhibition protected Fancc(-/-) murine embryonic fibroblasts and c-kit(+) bone marrow cells from TNF-alpha-induced apoptosis. Importantly, hematopoietic progenitor assays demonstrated that JNK inhibition enhanced Fancc(-/-) colony formation in the presence of TNF-alpha. Competitive repopulation assays showed that Fancc(-/-) donor cells cultured with the JNK inhibitor had equivalent levels of donor chimerism compared with Fancc(-/-) donor cells cultured with vehicle control. In contrast, culturing Fancc(-/-) cells with a p38 MAPK inhibitor significantly increased repopulating ability, supporting an integral role of p38 MAPK in maintaining Fancc(-/-) hematopoietic stem cell function. Taken together, these data suggest that p38 MAPK, but not JNK, has a critical role in maintaining the engraftment of Fancc(-/-)-reconstituting cells under conditions of stress.


Journal of Cellular Biochemistry | 2011

Fanconi Anemia D2 Protein Is an Apoptotic Target Mediated by Caspases

Su Jung Park; Brian D. Beck; M. Reza Saadatzadeh; Laura S. Haneline; D. Wade Clapp; Suk Hee Lee

FANCD2, a key factor in the FANC‐BRCA1 pathway is monoubiquitinated and targeted to discrete nuclear foci following DNA damage. Since monoubiquitination of FANCD2 is a crucial indicator for cellular response to DNA damage, we monitored the fate of FANCD2 and its monoubiquitination following DNA damage. Disappearance of FANCD2 protein was induced following DNA damage in a dose‐dependent manner, which correlated with degradation of BRCA1 and poly‐ADP ribose polymerase (PARP), known targets for caspase‐mediated apoptosis. Disappearance of FANCD2 was not affected by a proteasome inhibitor but was blocked by a caspase inhibitor. DNA damage‐induced disappearance of FANCD2 was also observed in cells lacking FANCA, suggesting that disappearance of FANCD2 does not depend on FANC‐BRCA1 pathway and FANCD2 monoubiquitination. In keeping with this, cells treated with TNF‐α, an apoptotic stimulus without causing any DNA damage, also induced disappearance of FANCD2 without monoubiquitination. Together, our data suggest that FANCD2 is a target for caspase‐mediated apoptotic pathway, which may be an early indicator for apoptotic cell death. J. Cell. Biochem. 112: 2383–2391, 2011.


Neurosurgical Focus | 2014

Phosphorylation of NMDA 2B at S1303 in human glioma peritumoral tissue: implications for glioma epileptogenesis

Xiang Gao; Haiyan Wang; Shanbao Cai; M. Reza Saadatzadeh; Helmut Hanenberg; Karen E. Pollok; Aaron A. Cohen-Gadol; Jinhui Chen

OBJECT Peritumoral seizures are an early symptom of a glioma. To gain a better understanding of the molecular mechanism underlying tumor-induced epileptogenesis, the authors studied modulation of the N-methyl-d-aspartate (NMDA) receptor in peritumoral tissue. METHODS To study the possible etiology of peritumoral seizures, NMDA receptor expression, posttranslational modification, and function were analyzed in an orthotopic mouse model of human gliomas and primary patient glioma tissue in which the peritumoral border (tumor-brain interface) was preserved in a tissue block during surgery. RESULTS The authors found that the NMDA receptor containing the 2B subunit (NR2B), a predominantly extrasynaptic receptor, is highly phosphorylated at S1013 in the neurons located in the periglioma area of the mouse brain. NR2B is also highly phosphorylated at S1013 in the neurons located in the peritumoral area from human brain tissue containing a glioma. The phosphorylation of the extrasynaptic NMDA receptor increases its permeability for Ca(2+) influx and subsequently mediates neuronal overexcitation and seizure activity. CONCLUSIONS These data suggest that overexcitation of the extrasynaptic NMDA receptors in the peritumoral neurons may contribute to the development of peritumoral seizures and that the phosphorylated NR2B may be a therapeutic target for blocking primary brain tumor-induced peritumoral seizures.


Cancer Research | 2018

Abstract 3180: Preclinical validation of EZH2 as a therapeutic target in pediatric Ewing's sarcoma

Pankita H. Pandya; Barbara J. Bailey; Adily Elmi; Heather R. Bates; Courtney N. Hemenway; Anthony L. Sinn; Khadijeh Bijangi-Vishehsaraei; M. Reza Saadatzadeh; Harlan E. Shannon; Jixin Ding; Mark S. Marshall; Michael J. Ferguson; Lijun Cheng; Lang Li; Mary E. Murray; Jamie L. Renbarger; Karen E. Pollok

Disease-free survival in relapsed Ewing9s Sarcoma Family of Tumors (ESFT) has not improved over the past 25 years. Current standard-of-care (SOC) agents result in 70% survival in patients with localized ESFT; however, for relapsed patients the survival rates remain between 15-20%. Approximately 85% of ESFTs have the chromosomal translocation t(11;22)(q24;q12) which encodes for the oncogenic EWS/FL1 fusion protein. The EWS/FL1 functions as a potent transcription factor leading to the dysregulated expression of genes that promote and maintain tumorigenesis. A major epigenetic regulator that is a downstream target of EWS/FL1 is the enhancer of Zeste Homolog 2 (EZH2). EZH2 is the catalytic component of the polycomb repressor complex 2 (PRC2). Notably, it is overexpressed in ESFT and maintains tumor oncogenicity by tri-methylating histone 3 lysine 27 (H3K27me3) to modulate gene expression. Genome and transcriptome data obtained by the Pediatric Precision Genomics Program at Riley Hospital for Children at Indiana University Health (IUH) indicate that EZH2 is expressed at high levels in ESFT biopsies. Additionally, other groups have reported that high levels of EZH2 protein in ESFT and other cancers correlate with increased chemoresistance to SOC therapy. We are testing tested the hypothesis that EZH2 contributes to chemoresistance in ESFT by regulating critical growth and survival genes. In addition, we are investigating if pharmacological inhibition of EZH2 will enhance sensitivity to the cytotoxic effects of SOC agents. Pediatric primary and relapsed ESFT cell lines and ESFT xenografts were validated for the EWS/FLI, EZH2, and H3K27me3 signatures. In vitro- and in vivo-pharmacodynamic studies of EZH2 inhibition via tazemetostat were conducted to optimize dosing effect. In ESFT cell lines (TC71, A673, CHLA-9, and CHLA-10), tazemetostat dose-response experiments indicated a significant reduction of H3K27me3 by one day post-treatment which was either sustained or completely blocked by 7-days post-treatment compared to vehicle treated (p Citation Format: Pankita H. Pandya, Barbara Bailey, Adily E. Elmi, Heather R. Bates, Courtney N. Hemenway, Anthony L. Sinn, Khadijeh Bijangi-Vishehsaraei, M. Reza Saadatzadeh, Harlan E. Shannon, Jixin Ding, Mark S. Marshall, Michael J. Ferguson, Lijun Cheng, Lang Li, Mary E. Murray, Jamie L. Renbarger, Karen E. Pollok. Preclinical validation of EZH2 as a therapeutic target in pediatric Ewing9s sarcoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 3180.


Cancer Research | 2014

Abstract 2741: Sensitization of temozolomide-mediated glioblastoma cell death by targeting MDM2: Assessment of PD biomarkers, brain penetration, and efficacy in humanized orthotopic xenograft models

Haiyan Wang; Shanbao Cai; Barbara J. Bailey; Lawrence M. Gelbert; M. Reza Saadatzadeh; Aaron A. Cohen-Gadol; Jann N. Sarkaria; Paul R. Territo; Taxiarchis M. Georgiadis; T. Zachary Gunter; Samy O. Meroueh; Eric C. Long; David R. Jones; Lindsey D. Mayo; Shannon Harlan; Karen E. Pollok

Treatment of glioblastoma multiforme (GBM) continues to be a challenge due to its infiltrative nature, tumor heterogeneity, and lack of therapeutic agents that penetrate the blood-brain barrier (BBB). While the primary tumor is permeable to some degree, the ability of GBM cells to invade areas of parenchyma with an intact BBB indicates development of BBB-penetrable compounds is a necessity. The multifunctional protein MDM2 holds promise as a therapeutic target in a variety of cancers and plays a critical role in controlling cell survival, invasion, and DNA repair. MDM2 antagonists such as nutlin3a and RG7112 are being used to interrogate the impact of modulating MDM2 function in combination with front-line therapy. Our objective was to investigate whether MDM2 antagonists, alone or in combination with temozolomide (TMZ), can augment cell death in orthotopic GBM xenograft models. In vitro data indicate that TMZ and nutlin3a are synergistic in decreasing cell viability in wild type (wt) p53 U87-MG, primary human wt p53 GBM10 and mutant (mt) p53 GBM43 cells. Pharmacodynamic studies demonstrated that the mechanism of action for promoting cell death following exposure to TMZ/nutlin3a was multifactorial. Comet assays indicated that repair of TMZ-mediated DNA damage was significantly delayed in wt and mt p53 GBM cells treated with TMZ/nutlin3a compared to TMZ alone and the base excision repair protein Ape1 was downregulated in cells treated with TMZ/nutlin3a. Pharmacokinetic studies guided development of rational dosing regimens in which 2-3 five-day cycles of TMZ followed by nutlin3a 4 hours later were investigated. In ectopic U87-MG xenografts, nutlin3a sensitized xenografts to TMZ-mediated cell death. Orthotopic studies employing U87-MG, GBM10, and GBM43 tumors are being utilized to determine if nutlin3a levels detectable in the brain via HPLC-MS/MS(API 4000) are sufficient to modulate MDM2 function in the context of TMZ and increase survival. In the TMZ-resistant GBM10 (wtp53, MGMTpos, and PTENnull) orthotopic model, there was a modest increase in median survival from 63 days with TMZ to 73 days with TMZ/nutlin3a. Higher and more sustained brain levels of MDM2 antagonists will likely be necessary to improve survival. The MDM2 antagonist RG7112 has an improved PK profile and structural analysis of RG7112 and nutlin3a via QikProp 3.0 (www.schrodinger.com) indicates that RG7112 also has an improved predicted brain/blood partition coefficient (plog BB) compared to nutlin3a (RG7112 = -0.17; nutlin3a = -0.415). Studies are in progress to assess the brain penetration of RG7112 and its effect on GBM growth in vivo. Taken together, our data suggest that modulation of MDM2 function in the context of cytotoxic therapy has the potential to alter mechanisms involved in DNA repair that can promote cell death and improve survival. Citation Format: Haiyan Wang, Shanbao Cai, Barbara J. Bailey, Lawrence M. Gelbert, M. Reza Saadatzadeh, Aaron A. Cohen-Gadol, Jann N. Sarkaria, Paul Territo, Taxiarchis M. Georgiadis, T. Zachary Gunter, Samy Meroueh, Eric C. Long, David R. Jones, Lindsey D. Mayo, Shannon Harlan, Karen E. Pollok. Sensitization of temozolomide-mediated glioblastoma cell death by targeting MDM2: Assessment of PD biomarkers, brain penetration, and efficacy in humanized orthotopic xenograft models. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 2741. doi:10.1158/1538-7445.AM2014-2741


Experimental Hematology | 2007

Human CD34+AC133+VEGFR-2+ cells are not endothelial progenitor cells but distinct, primitive hematopoietic progenitors

Jamie Case; Laura E. Mead; Waylan K. Bessler; Daniel Prater; Hilary White; M. Reza Saadatzadeh; Janak R. Bhavsar; Mervin C. Yoder; Laura S. Haneline; David A. Ingram


American Journal of Physiology-heart and Circulatory Physiology | 2009

Release of proinflammatory mediators and expression of proinflammatory adhesion molecules by endothelial progenitor cells

Yanmin Zhang; David A. Ingram; Michael P. Murphy; M. Reza Saadatzadeh; Laura E. Mead; Daniel Prater; Jalees Rehman

Collaboration


Dive into the M. Reza Saadatzadeh's collaboration.

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
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge