Irina Matei
Cornell University
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Irina Matei.
Nature | 2015
Ayuko Hoshino; Bruno Costa-Silva; Tang-Long Shen; Goncalo Rodrigues; Ayako Hashimoto; Milica Tesic Mark; Henrik Molina; Shinji Kohsaka; Angela Di Giannatale; Sophia Ceder; Swarnima Singh; Caitlin Williams; Nadine Soplop; Kunihiro Uryu; Lindsay A. Pharmer; Tari A. King; Linda Bojmar; Alexander E. Davies; Yonathan Ararso; Tuo Zhang; Haiying Zhang; Jonathan M. Hernandez; Joshua Mitchell Weiss; Vanessa D. Dumont-Cole; Kimberly Kramer; Leonard H. Wexler; Aru Narendran; Gary K. Schwartz; John H. Healey; Per Sandström
Ever since Stephen Paget’s 1889 hypothesis, metastatic organotropism has remained one of cancer’s greatest mysteries. Here we demonstrate that exosomes from mouse and human lung-, liver- and brain-tropic tumour cells fuse preferentially with resident cells at their predicted destination, namely lung fibroblasts and epithelial cells, liver Kupffer cells and brain endothelial cells. We show that tumour-derived exosomes uptaken by organ-specific cells prepare the pre-metastatic niche. Treatment with exosomes from lung-tropic models redirected the metastasis of bone-tropic tumour cells. Exosome proteomics revealed distinct integrin expression patterns, in which the exosomal integrins α6β4 and α6β1 were associated with lung metastasis, while exosomal integrin αvβ5 was linked to liver metastasis. Targeting the integrins α6β4 and αvβ5 decreased exosome uptake, as well as lung and liver metastasis, respectively. We demonstrate that exosome integrin uptake by resident cells activates Src phosphorylation and pro-inflammatory S100 gene expression. Finally, our clinical data indicate that exosomal integrins could be used to predict organ-specific metastasis.
Nature Cell Biology | 2013
Cyrus M. Ghajar; Héctor Peinado; Hidetoshi Mori; Irina Matei; Kimberley Evason; Hélène Brazier; Dena Almeida; Antonius Koller; Katherine A. Hajjar; Didier Y. R. Stainier; Emily I. Chen; David Lyden; Mina J. Bissell
In a significant fraction of breast cancer patients, distant metastases emerge after years or even decades of latency. How disseminated tumour cells (DTCs) are kept dormant, and what wakes them up, are fundamental problems in tumour biology. To address these questions, we used metastasis assays in mice and showed that dormant DTCs reside on microvasculature of lung, bone marrow and brain. We then engineered organotypic microvascular niches to determine whether endothelial cells directly influence breast cancer cell (BCC) growth. These models demonstrated that endothelial-derived thrombospondin-1 induces sustained BCC quiescence. This suppressive cue was lost in sprouting neovasculature; time-lapse analysis showed that sprouting vessels not only permit, but accelerate BCC outgrowth. We confirmed this surprising result in dormancy models and in zebrafish, and identified active TGF-β1 and periostin as tumour-promoting factors derived from endothelial tip cells. Our work reveals that stable microvasculature constitutes a dormant niche, whereas sprouting neovasculature sparks micrometastatic outgrowth.
Nature Cell Biology | 2018
Haiying Zhang; Daniela Freitas; Han Sang Kim; Kristina Fabijanic; Zhong Li; Haiyan Chen; Milica Tesic Mark; Henrik Molina; Alberto Martín; Linda Bojmar; Justin Fang; Sham Rampersaud; Ayuko Hoshino; Irina Matei; Candia M. Kenific; Miho Nakajima; Anders Peter Mutvei; Pasquale Sansone; Weston Buehring; Huajuan Wang; Juan Pablo Jimenez; Leona Cohen-Gould; Navid Paknejad; Matthew Brendel; Katia Manova-Todorova; Ana Magalhães; José J.A. Ferreira; Hugo Osório; André M. N. Silva; Ashish Massey
The heterogeneity of exosomal populations has hindered our understanding of their biogenesis, molecular composition, biodistribution and functions. By employing asymmetric flow field-flow fractionation (AF4), we identified two exosome subpopulations (large exosome vesicles, Exo-L, 90–120 nm; small exosome vesicles, Exo-S, 60–80 nm) and discovered an abundant population of non-membranous nanoparticles termed ‘exomeres’ (~35 nm). Exomere proteomic profiling revealed an enrichment in metabolic enzymes and hypoxia, microtubule and coagulation proteins as well as specific pathways, such as glycolysis and mTOR signalling. Exo-S and Exo-L contained proteins involved in endosomal function and secretion pathways, and mitotic spindle and IL-2/STAT5 signalling pathways, respectively. Exo-S, Exo-L and exomeres each had unique N-glycosylation, protein, lipid, DNA and RNA profiles and biophysical properties. These three nanoparticle subsets demonstrated diverse organ biodistribution patterns, suggesting distinct biological functions. This study demonstrates that AF4 can serve as an improved analytical tool for isolating extracellular vesicles and addressing the complexities of heterogeneous nanoparticle subpopulations.Lyden and colleagues use asymmetric flow field-flow fractionation to classify nanoparticles derived from cell lines and human samples, including previously uncharacterized large, Exo-L and small, Exo-S, exosome subsets.
Bone Marrow Transplantation | 2015
Niroshana Anandasabapathy; Gaëlle Breton; Arlene Hurley; Marina Caskey; Christine Trumpfheller; Popi Sarma; James Pring; Maggi Pack; Noreen Buckley; Irina Matei; David Lyden; Jennifer Green; Thomas Hawthorne; Henry Marsh; Michael Yellin; Thomas P. Davis; Tibor Keler; Sarah J. Schlesinger
Fms-like tyrosine kinase-3 ligand (Flt3L) uniquely binds the Flt3 (CD135) receptor expressed on hematopoietic stem cells (HSCs), early progenitor cells, immature thymocytes and steady-state dendritic cells (DCs) and induces their proliferation, differentiation, development and mobilization in the bone marrow, peripheral blood and lymphoid organs. CDX-301 has an identical amino-acid sequence and comparable biological activity to the previously tested rhuFlt3L, which ceased clinical development over a decade ago. This Phase 1 trial assessed the safety, pharmacokinetic, pharmacodynamic and immunologic profile of CDX-301, explored alternate dosing regimens and examined the impact of rhuFlt3L on key immune cell subsets. Thirty healthy volunteers received CDX-301 (1–75 μg/kg/day) over 5–10 days. One event of Grade 3 community-acquired pneumonia occurred. There were no other infections, dose-limiting toxicities or serious adverse events. CDX-301 resulted in effective peripheral expansion of monocytes, hematopoietic stem and progenitor cells and key subsets of myeloid DCs and plasmacytoid DCs, with no clear effect on regulatory T cells. These data from healthy volunteers support the potential for CDX-301, as monotherapy or in combination with other agents, in various indications including allogeneic HSC transplantation and immunotherapy, but the effects of CDX-301 will need to be investigated in each of these patient populations.
Nature Medicine | 2016
Héctor Peinado; Maša Alecˇković; Simon Lavotshkin; Irina Matei; Bruno Costa-Silva; Gema Moreno-Bueno; Marta Hergueta-Redondo; Caitlin Williams; Guillermo García-Santos; Cyrus M. Ghajar; Ayuko Nitadori-Hoshino; Caitlin Hoffman; Karen Badal; Benjamin A. Garcia; Margaret K. Callahan; Jianda Yuan; Vilma R. Martins; Johan Skog; Rosandra N. Kaplan; Mary S. Brady; Jedd D. Wolchok; Paul B. Chapman; Yibin Kang; Jacqueline Bromberg; David Lyden
Corrigendum: Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET
Journal of Clinical Investigation | 2017
Yujie Huang; Prajwal Rajappa; Wenhuo Hu; Caitlin Hoffman; Babacar Cisse; Joon Hyung Kim; Emilie Gorge; Rachel Yanowitch; William Cope; Emma Vartanian; Raymond Xu; Tuo Zhang; David Pisapia; Jenny Xiang; Jason T. Huse; Irina Matei; Héctor Peinado; Jacqueline Bromberg; Eric C. Holland; Bi Sen Ding; Shahin Rafii; David Lyden; Jeffrey P. Greenfield
Tumors are capable of coopting hematopoietic cells to create a suitable microenvironment to support malignant growth. Here, we have demonstrated that upregulation of kinase insert domain receptor (KDR), also known as VEGFR2, in a myeloid cell sublineage is necessary for malignant progression of gliomas in transgenic murine models and is associated with high-grade tumors in patients. KDR expression increased in myeloid cells as myeloid-derived suppressor cells (MDSCs) accumulated, which was associated with the transformation and progression of low-grade fibrillary astrocytoma to high-grade anaplastic gliomas. KDR deficiency in murine BM-derived cells (BMDCs) suppressed the differentiation of myeloid lineages and reduced granulocytic/monocytic populations. The depletion of myeloid-derived KDR compromised its proangiogenic function, which inhibited the angiogenic switch necessary for malignant progression of low-grade to high-grade tumors. We also identified inhibitor of DNA binding protein 2 (ID2) as a key upstream regulator of KDR activation during myeloid differentiation. Deficiency of ID2 in BMDCs led to downregulation of KDR, suppression of proangiogenic myeloid cells, and prevention of low-grade to high-grade transition. Tumor-secreted TGF-&bgr; and granulocyte-macrophage CSF (GM-CSF) enhanced the KDR/ID2 signaling axis in BMDCs. Our results suggest that modulation of KDR/ID2 signaling may restrict tumor-associated myeloid cells and could potentially be a therapeutic strategy for preventing transformation of premalignant gliomas.
Cancer Cell | 2011
Irina Matei; Cyrus M. Ghajar; David Lyden
In the July issue of Nature Medicine, Massagué and colleagues define a biphasic role for the extracellular matrix protein tenascin C as a metastatic niche component in lung colonization by breast cancer cells. These results provide a rationale for designing therapies targeting metastatic progression by disrupting its very foundations.
Nature Medicine | 2012
Héctor Peinado; Maša Alečković; Simon Lavotshkin; Irina Matei; Bruno Costa-Silva; Gema Moreno-Bueno; Marta Hergueta-Redondo; Caitlin Williams; Guillermo García-Santos; Cyrus M. Ghajar; Ayuko Nitadori-Hoshino; Caitlin Hoffman; Karen Badal; Benjamin A. Garcia; Margaret K. Callahan; Jianda Yuan; Vilma R. Martins; Johan Skog; Rosandra N. Kaplan; Mary S. Brady; Jedd D. Wolchok; Paul B. Chapman; Yibin Kang; Jacqueline Bromberg; David Lyden
Nature Cell Biology | 2015
Bruno Costa-Silva; Nicole M. Aiello; Allyson J. Ocean; Swarnima Singh; Haiying Zhang; Basant Kumar Thakur; Annette Becker; Ayuko Hoshino; Milica Tesic Mark; Henrik Molina; Jenny Xiang; Tuo Zhang; Till Theilen; Guillermo García-Santos; Caitlin Williams; Yonathan Ararso; Yujie Huang; Goncalo Rodrigues; Tang-Long Shen; Knut Jørgen Labori; Inger Marie Bowitz Lothe; Elin H. Kure; Jonathan M. Hernandez; Alexandre Doussot; Saya H. Ebbesen; Paul M. Grandgenett; Michael A. Hollingsworth; Maneesh Jain; Kavita Mallya; Surinder K. Batra
Cell Research | 2014
Basant Kumar Thakur; Haiying Zhang; Annette Becker; Irina Matei; Yujie Huang; Bruno Costa-Silva; Yan Zheng; Ayuko Hoshino; Hélène Brazier; Jenny Xiang; Caitlin Williams; Ruth Rodriguez-Barrueco; Jose M. Silva; Weijia Zhang; Stephen Hearn; Olivier Elemento; Navid Paknejad; Katia Manova-Todorova; Karl Welte; Jacqueline Bromberg; Héctor Peinado; David Lyden