Network


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

Hotspot


Dive into the research topics where Sudeh Izadmehr is active.

Publication


Featured researches published by Sudeh Izadmehr.


Science Translational Medicine | 2013

KLF6-SV1 Drives Breast Cancer Metastasis and Is Associated with Poor Survival

Raheleh Hatami; Anieta M. Sieuwerts; Sudeh Izadmehr; Zhong Yao; Rui Fang Qiao; Luena Papa; Maxime P. Look; Marcel Smid; Jessica Ohlssen; Alice C. Levine; Doris Germain; David E. Burstein; Alexander Kirschenbaum; Analisa DiFeo; John A. Foekens; Goutham Narla

The KLF6-SV1 splice variant is associated with poor prognosis in early-stage human breast cancer and drives metastasis through the regulation of an EMT-like program in culture and in vivo. A New TWIST on Breast Cancer Sprawl Suburban sprawl is a complex, multistep process whereby “healthy” land becomes inundated with fast food chains, strip malls, and large parking lots. Yet, what drives sprawl is unclear—Do car-centric residential communities pop up close to businesses, or are the businesses merely opening where the consumers are? Cancer metastasis can be thought of as another type of sprawl, and although we can describe changes associated with metastasis, the drivers are equally unclear. Now, Hatami et al. provide insight into one of the potential drivers of breast cancer metastasis. The authors found that KLF6-SV1, which is a variant of a tumor suppressor gene, was associated with increased metastatic potential and poor survival in breast cancer patients. They then took their studies to the next step, trying to figure out how exactly KLF6-SV1 contributed to metastasis. Overexpressing KLF6-SV1 contributed to an epithelial-to-mesenchymal transition (EMT), which is thought to be important for cancer cells to leave the primary tumor. Indeed, inhibiting KLF6-SV1 returned these cells to a more epithelial (less metastatic) phenotype. Moreover, KLF6-SV1 alters the expression of TWIST1, which regulates EMT. Thus, KLF6-SV1 may be an early driver for metastasis in breast cancer patients. Metastasis is the major cause of cancer mortality. A more thorough understanding of the mechanisms driving this complex multistep process will aid in the identification and characterization of therapeutically targetable genetic drivers of disease progression. We demonstrate that KLF6-SV1, an oncogenic splice variant of the KLF6 tumor suppressor gene, is associated with increased metastatic potential and poor survival in a cohort of 671 lymph node–negative breast cancer patients. KLF6-SV1 overexpression in mammary epithelial cell lines resulted in an epithelial-to-mesenchymal–like transition and drove aggressive multiorgan metastatic disease in multiple in vivo models. Additionally, KLF6-SV1 loss-of-function studies demonstrated reversion to an epithelial and less invasive phenotype. Combined, these findings implicate KLF6-SV1 as a key driver of breast cancer metastasis that distinguishes between indolent and lethal early-stage disease and provides a potential therapeutic target for invasive breast cancer.


Journal of Clinical Investigation | 2012

Targeting the FOXO1/KLF6 axis regulates EGFR signaling and treatment response

Jaya Sangodkar; Neil Dhawan; Heather Melville; Varan J. Singh; Eric Yuan; Huma Q. Rana; Sudeh Izadmehr; Caroline Farrington; Sahar Mazhar; Suzanna Katz; Tara Albano; Pearlann Arnovitz; Rachel Okrent; Michael Ohlmeyer; Matthew D. Galsky; David E. Burstein; David Y. Zhang; Katerina Politi; Analisa DiFeo; Goutham Narla

EGFR activation is both a key molecular driver of disease progression and the target of a broad class of molecular agents designed to treat advanced cancer. Nevertheless, resistance develops through several mechanisms, including activation of AKT signaling. Though much is known about the specific molecular lesions conferring resistance to anti-EGFR-based therapies, additional molecular characterization of the downstream mediators of EGFR signaling may lead to the development of new classes of targeted molecular therapies to treat resistant disease. We identified a transcriptional network involving the tumor suppressors Krüppel-like factor 6 (KLF6) and forkhead box O1 (FOXO1) that negatively regulates activated EGFR signaling in both cell culture and in vivo models. Furthermore, the use of the FDA-approved drug trifluoperazine hydrochloride (TFP), which has been shown to inhibit FOXO1 nuclear export, restored sensitivity to AKT-driven erlotinib resistance through modulation of the KLF6/FOXO1 signaling cascade in both cell culture and xenograft models of lung adenocarcinoma. Combined, these findings define a novel transcriptional network regulating oncogenic EGFR signaling and identify a class of FDA-approved drugs as capable of restoring chemosensitivity to anti-EGFR-based therapy for the treatment of metastatic lung adenocarcinoma.


Nature | 2017

Blocking FSH induces thermogenic adipose tissue and reduces body fat

Peng Liu; Yaoting Ji; Tony Yuen; Elizabeth Rendina-Ruedy; Victoria E. DeMambro; Samarth Dhawan; Wahid Abu-Amer; Sudeh Izadmehr; Bin Zhou; Andrew C. Shin; Rauf Latif; Priyanthan Thangeswaran; Animesh Gupta; Jianhua Li; Valeria Shnayder; Samuel T. Robinson; Yue Eric Yu; Xingjian Zhang; Feiran Yang; Ping Lu; Yu Zhou; Ling-Ling Zhu; Douglas J. Oberlin; Terry F. Davies; Michaela R. Reagan; Aaron Brown; T. Rajendra Kumar; Solomon Epstein; Jameel Iqbal; Narayan G. Avadhani

Menopause is associated with bone loss and enhanced visceral adiposity. A polyclonal antibody that targets the β-subunit of the pituitary hormone follicle-stimulating hormone (Fsh) increases bone mass in mice. Here, we report that this antibody sharply reduces adipose tissue in wild-type mice, phenocopying genetic haploinsufficiency for the Fsh receptor gene Fshr. The antibody also causes profound beiging, increases cellular mitochondrial density, activates brown adipose tissue and enhances thermogenesis. These actions result from the specific binding of the antibody to the β-subunit of Fsh to block its action. Our studies uncover opportunities for simultaneously treating obesity and osteoporosis.


Cancer Research | 2014

Inducible Nitric Oxide Synthase Drives mTOR Pathway Activation and Proliferation of Human Melanoma by Reversible Nitrosylation of TSC2

Esther Lopez-Rivera; Padmini Jayaraman; Falguni Parikh; Michael A. Davies; Suhendan Ekmekcioglu; Sudeh Izadmehr; Denái R. Milton; Jerry E. Chipuk; Elizabeth A. Grimm; Yeriel Estrada; Julio A. Aguirre-Ghiso; Andrew G. Sikora

Melanoma is one of the cancers of fastest-rising incidence in the world. Inducible nitric oxide synthase (iNOS) is overexpressed in melanoma and other cancers, and previous data suggest that iNOS and nitric oxide (NO) drive survival and proliferation of human melanoma cells. However, specific mechanisms through which this occurs are poorly defined. One candidate is the PI3K-AKT-mTOR pathway, which plays a major role in proliferation, angiogenesis, and metastasis of melanoma and other cancers. We used the chick embryo chorioallantoic membrane (CAM) assay to test the hypothesis that melanoma growth is regulated by iNOS-dependent mTOR pathway activation. Both pharmacologic inhibition and siRNA-mediated gene silencing of iNOS suppressed melanoma proliferation and in vivo growth on the CAM in human melanoma models. This was associated with strong downregulation of mTOR pathway activation by Western blot analysis of p-mTOR, p70 ribosomal S6 kinase (p-P70S6K), p-S6RP, and p-4EBP1. iNOS expression and NO were associated with reversible nitrosylation of tuberous sclerosis complex (TSC) 2, and inhibited dimerization of TSC2 with its inhibitory partner TSC1, enhancing GTPase activity of its target Ras homolog enriched in brain (Rheb), a critical activator of mTOR signaling. Immunohistochemical analysis of tumor specimens from stage III melanoma patients showed a significant correlation between iNOS expression levels and expression of the mTOR pathway members. Exogenously supplied NO was also sufficient to reverse the mTOR pathway inhibition by the B-Raf inhibitor vemurafenib. In summary, covalent modification of TSC2 by iNOS-derived NO is associated with impaired TSC2/TSC1 dimerization, mTOR pathway activation, and proliferation of human melanoma. This model is consistent with the known association of iNOS overexpression and poor prognosis in melanoma and other cancers.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Repurposing of bisphosphonates for the prevention and therapy of nonsmall cell lung and breast cancer

Agnes Stachnik; Tony Yuen; Jameel Iqbal; Miriam Sgobba; Yogesh K. Gupta; Ping Lu; Graziana Colaianni; Yaoting Ji; Ling Ling Zhu; Se Min Kim; Jianhua Li; Peng Liu; Sudeh Izadmehr; Jaya Sangodkar; Thomas Scherer; Matthew D. Galsky; Jorge Gomez; Solomon Epstein; Christoph Buettner; Zhuan Bian; Alberta Zallone; Aneel K. Aggarwal; Shozeb Haider; Maria I. New; Li Sun; Goutham Narla; Mone Zaidi

Significance Small molecules to target oncogenic signaling cascades in cancer have achieved success in molecularly defined patient subsets. The path to approval is often protracted and plagued with failures. Repositioning Food and Drug Administration-approved drugs with known side effects has become a major focus. Bisphosphonates are a commonly prescribed therapy for osteoporosis and skeletal metastases. The drugs have also been associated with reduced tumor burden in some patients, but the mechanism is unknown. Here we provide evidence that bisphosphonates inhibit the human EGFR (HER) receptor tyrosine kinase, including the commonly mutated forms that drive nonsmall cell lung cancer, as well as a resistance mutation. This new mechanism lays the basis for the future use of bisphosphonates for the prevention and therapy of HER family-driven cancers. A variety of human cancers, including nonsmall cell lung (NSCLC), breast, and colon cancers, are driven by the human epidermal growth factor receptor (HER) family of receptor tyrosine kinases. Having shown that bisphosphonates, a class of drugs used widely for the therapy of osteoporosis and metastatic bone disease, reduce cancer cell viability by targeting HER1, we explored their potential utility in the prevention and therapy of HER-driven cancers. We show that bisphosphonates inhibit colony formation by HER1ΔE746-A750-driven HCC827 NSCLCs and HER1wt-expressing MB231 triple negative breast cancers, but not by HERlow-SW620 colon cancers. In parallel, oral gavage with bisphosphonates of mice xenografted with HCC827 or MB231 cells led to a significant reduction in tumor volume in both treatment and prevention protocols. This result was not seen with mice harboring HERlow SW620 xenografts. We next explored whether bisphosphonates can serve as adjunctive therapies to tyrosine kinase inhibitors (TKIs), namely gefitinib and erlotinib, and whether the drugs can target TKI-resistant NSCLCs. In silico docking, together with molecular dynamics and anisotropic network modeling, showed that bisphosphonates bind to TKIs within the HER1 kinase domain. As predicted from this combinatorial binding, bisphosphonates enhanced the effects of TKIs in reducing cell viability and driving tumor regression in mice. Impressively, the drugs also overcame erlotinib resistance acquired through the gatekeeper mutation T790M, thus offering an option for TKI-resistant NSCLCs. We suggest that bisphosphonates can potentially be repurposed for the prevention and adjunctive therapy of HER1-driven cancers.


Journal of Clinical Investigation | 2017

Activation of tumor suppressor protein PP2A inhibits KRAS-driven tumor growth

Jaya Sangodkar; Abbey Perl; Rita Tohme; Janna Kiselar; David Kastrinsky; Nilesh Zaware; Sudeh Izadmehr; Sahar Mazhar; Danica Wiredja; Caitlin M. O’Connor; Divya Hoon; Neil Dhawan; Daniela Schlatzer; Shen Yao; Daniel Leonard; Alain C. Borczuk; Giridharan Gokulrangan; Lifu Wang; Elena Svenson; Caroline C. Farrington; Eric Yuan; Rita A. Avelar; Agnes Stachnik; Blake Smith; Vickram Gidwani; Heather M. Giannini; Daniel McQuaid; Kimberly McClinch; Zhizhi Wang; Alice C. Levine

Targeted cancer therapies, which act on specific cancer-associated molecular targets, are predominantly inhibitors of oncogenic kinases. While these drugs have achieved some clinical success, the inactivation of kinase signaling via stimulation of endogenous phosphatases has received minimal attention as an alternative targeted approach. Here, we have demonstrated that activation of the tumor suppressor protein phosphatase 2A (PP2A), a negative regulator of multiple oncogenic signaling proteins, is a promising therapeutic approach for the treatment of cancers. Our group previously developed a series of orally bioavailable small molecule activators of PP2A, termed SMAPs. We now report that SMAP treatment inhibited the growth of KRAS-mutant lung cancers in mouse xenografts and transgenic models. Mechanistically, we found that SMAPs act by binding to the PP2A A&agr; scaffold subunit to drive conformational changes in PP2A. These results show that PP2A can be activated in cancer cells to inhibit proliferation. Our strategy of reactivating endogenous PP2A may be applicable to the treatment of other diseases and represents an advancement toward the development of small molecule activators of tumor suppressor proteins.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Bisphosphonates inactivate human EGFRs to exert antitumor actions

Tony Yuen; Agnes Stachnik; Jameel Iqbal; Miriam Sgobba; Yogesh K. Gupta; Ping Lu; Graziana Colaianni; Yaoting Ji; Ling Ling Zhu; Se Min Kim; Jianhua Li; Peng Liu; Sudeh Izadmehr; Jaya Sangodkar; Jack Bailey; Yathin Latif; Solomon Epstein; Terry F. Davies; Zhuan Bian; Alberta Zallone; Aneel K. Aggarwal; Shozeb Haider; Maria I. New; Li Sun; Goutham Narla; Mone Zaidi

Significance For over three decades, bisphosphonates have been used for the therapy of osteoporosis and skeletal metastasis. Here we show that this class of drugs reduces the viability of tumor cells that are driven by the human epidermal growth factor receptor (HER) family of receptor tyrosine kinases. We also show that bisphosphonates directly bind to and inhibit HER kinases. Because bisphosphonates are inexpensive and readily available worldwide, our findings may have important healthcare implications by offering an affordable and multiuse alternative or adjunct to current therapies for HER-driven malignancy. Bisphosphonates are the most commonly prescribed medicines for osteoporosis and skeletal metastases. The drugs have also been shown to reduce cancer progression, but only in certain patient subgroups, suggesting that there is a molecular entity that mediates bisphosphonate action on tumor cells. Using connectivity mapping, we identified human epidermal growth factor receptors (human EGFR or HER) as a potential new molecular entity for bisphosphonate action. Protein thermal shift and cell-free kinase assays, together with computational modeling, demonstrated that N-containing bisphosphonates directly bind to the kinase domain of HER1/2 to cause a global reduction in downstream signaling. By doing so, the drugs kill lung, breast, and colon cancer cells that are driven by activating mutations or overexpression of HER1. Knocking down HER isoforms thus abrogates cell killing by bisphosphonates, establishing complete HER dependence and ruling out a significant role for other receptor tyrosine kinases or the enzyme farnesyl pyrophosphate synthase. Consistent with this finding, colon cancer cells expressing low levels of HER do not respond to bisphosphonates. The results suggest that bisphosphonates can potentially be repurposed for the prevention and therapy of HER family-driven cancers.


Cell Death and Disease | 2012

Sensitization to the mitochondrial pathway of apoptosis augments melanoma tumor cell responses to conventional chemotherapeutic regimens

R A Anvekar; James J. Asciolla; Esther Lopez-Rivera; Konstantinos V. Floros; Sudeh Izadmehr; Rana Elkholi; G Belbin; Andrew G. Sikora; Jerry E. Chipuk

Metastatic malignant melanoma is highly resistant to chemotherapy, and the average survival rate is under 1 year. The only FDA-approved conventional chemotherapy (i.e., dacarbazine) targets melanoma tumor cells by inducing a form of cell death referred to as apoptosis. However, dacarbazine exhibits a response rate of ∼5%, and combination chemotherapies consisting of cisplatin, vinblastine, and dacarbazine often offer little clinical advantage over dacarbazine alone. Apoptosis is governed by the BCL-2 family of proteins, which is comprised of anti-apoptotic and pro-apoptotic members. To determine if the anti-apoptotic BCL-2 repertoire established the cell death threshold and chemoresistance in melanoma, a novel treatment strategy was designed to inhibit the anti-apoptotic BCL-2 members with ABT-737. Using various melanoma model systems, we determined the affects of ABT-737 on sensitivity to dacarbazine-based regimens. Strikingly, ABT-737 re-sensitized melanoma cell lines to common chemotherapeutics leading to marked BIM-mediated apoptosis. Cellular features of the ABT-737 combination treatments were loss of proliferation, mitochondrial fragmentation, nuclear condensation, phosphatidylserine exposure, and decreased clonogenic survival. We also observed significant anti-tumor activity in an in vivo melanoma model system. Our data indicate that ABT-737 may be a beneficial adjuvant therapy to improve melanoma response rates when conventional chemotherapy is the only option.


Bioorganic & Medicinal Chemistry | 2015

Reengineered tricyclic anti-cancer agents.

David Kastrinsky; Jaya Sangodkar; Nilesh Zaware; Sudeh Izadmehr; Neil Dhawan; Goutham Narla; Michael Ohlmeyer

The phenothiazine and dibenzazepine tricyclics are potent neurotropic drugs with a documented but underutilized anti-cancer side effect. Reengineering these agents (TFP, CPZ, CIP) by replacing the basic amine with a neutral polar functional group (e.g., RTC-1, RTC-2) abrogated their CNS effects as demonstrated by in vitro pharmacological assays and in vivo behavioral models. Further optimization generated several phenothiazines and dibenzazepines with improved anti-cancer potency, exemplified by RTC-5. This new lead demonstrated efficacy against a xenograft model of an EGFR driven cancer without the neurotropic effects exhibited by the parent molecules. Its effects were attributed to concomitant negative regulation of PI3K-AKT and RAS-ERK signaling.


Journal of Clinical Investigation | 2018

Factor XII and uPAR upregulate neutrophil functions to influence wound healing

Evi X. Stavrou; Chao Fang; Kara L. Bane; Andy T. Long; Clément Naudin; Erdem Kucukal; Agharnan Gandhi; Adina Brett-Morris; Michele M. Mumaw; Sudeh Izadmehr; Alona Merkulova; Cindy C. Reynolds; Omar Alhalabi; Lalitha Nayak; Wen Mei Yu; Cheng Kui Qu; Howard Meyerson; George R. Dubyak; Umut A. Gurkan; Marvin T. Nieman; Anirban Sen Gupta; Thomas Renné; Alvin H. Schmaier

Coagulation factor XII (FXII) deficiency is associated with decreased neutrophil migration, but the mechanisms remain uncharacterized. Here, we examine how FXII contributes to the inflammatory response. In 2 models of sterile inflammation, FXII-deficient mice (F12–/–) had fewer neutrophils recruited than WT mice. We discovered that neutrophils produced a pool of FXII that is functionally distinct from hepatic-derived FXII and contributes to neutrophil trafficking at sites of inflammation. FXII signals in neutrophils through urokinase plasminogen activator receptor–mediated (uPAR-mediated) Akt2 phosphorylation at S474 (pAktS474). Downstream of pAkt2S474, FXII stimulation of neutrophils upregulated surface expression of &agr;M&bgr;2 integrin, increased intracellular calcium, and promoted extracellular DNA release. The sum of these activities contributed to neutrophil cell adhesion, migration, and release of neutrophil extracellular traps in a process called NETosis. Decreased neutrophil signaling in F12–/– mice resulted in less inflammation and faster wound healing. Targeting hepatic F12 with siRNA did not affect neutrophil migration, whereas WT BM transplanted into F12–/– hosts was sufficient to correct the neutrophil migration defect in F12–/– mice and restore wound inflammation. Importantly, these activities were a zymogen FXII function and independent of FXIIa and contact activation, highlighting that FXII has a sophisticated role in vivo that has not been previously appreciated.

Collaboration


Dive into the Sudeh Izadmehr's collaboration.

Top Co-Authors

Avatar

Goutham Narla

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Jaya Sangodkar

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Michael Ohlmeyer

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

David Kastrinsky

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Alice C. Levine

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Analisa DiFeo

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Shen Yao

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Alexander Kirschenbaum

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Daniela Schlatzer

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Peng Liu

Icahn School of Medicine at Mount Sinai

View shared research outputs
Researchain Logo
Decentralizing Knowledge