Network


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

Hotspot


Dive into the research topics where Joan M. Greve is active.

Publication


Featured researches published by Joan M. Greve.


Cancer Cell | 2010

A Hierarchy of Self-Renewing Tumor-Initiating Cell Types in Glioblastoma

Ruihuan Chen; Merry Nishimura; Stephanie M. Bumbaca; Samir Kharbanda; William F. Forrest; Ian Kasman; Joan M. Greve; Robert Soriano; Laurie L. Gilmour; Celina Sanchez Rivers; Zora Modrusan; Serban Nacu; Steve Guerrero; Kyle A. Edgar; Jeffrey Wallin; Katrin Lamszus; Manfred Westphal; Susanne Heim; C. David James; Scott R. VandenBerg; Joseph F. Costello; Scott Moorefield; Cynthia Cowdrey; Michael D. Prados; Heidi S. Phillips

The neural stem cell marker CD133 is reported to identify cells within glioblastoma (GBM) that can initiate neurosphere growth and tumor formation; however, instances of CD133(-) cells exhibiting similar properties have also been reported. Here, we show that some PTEN-deficient GBM tumors produce a series of CD133(+) and CD133(-) self-renewing tumor-initiating cell types and provide evidence that these cell types constitute a lineage hierarchy. Our results show that the capacities for self-renewal and tumor initiation in GBM need not be restricted to a uniform population of stemlike cells, but can be shared by a lineage of self-renewing cell types expressing a range of markers of forebrain lineage.


Proceedings of the National Academy of Sciences of the United States of America | 2007

Long-term monitoring of transplanted human neural stem cells in developmental and pathological contexts with MRI

Raphael Guzman; Nobuko Uchida; Tonya Bliss; Dongping He; Karen K. Christopherson; David Stellwagen; Alexandra Capela; Joan M. Greve; Robert C. Malenka; Michael E. Moseley; Theo D. Palmer; Gary K. Steinberg

Noninvasive monitoring of stem cells, using high-resolution molecular imaging, will be instrumental to improve clinical neural transplantation strategies. We show that labeling of human central nervous system stem cells grown as neurospheres with magnetic nanoparticles does not adversely affect survival, migration, and differentiation or alter neuronal electrophysiological characteristics. Using MRI, we show that human central nervous system stem cells transplanted either to the neonatal, the adult, or the injured rodent brain respond to cues characteristic for the ambient microenvironment resulting in distinct migration patterns. Nanoparticle-labeled human central nervous system stem cells survive long-term and differentiate in a site-specific manner identical to that seen for transplants of unlabeled cells. We also demonstrate the impact of graft location on cell migration and describe magnetic resonance characteristics of graft cell death and subsequent clearance. Knowledge of migration patterns and implementation of noninvasive stem cell tracking might help to improve the design of future clinical neural stem cell transplantation.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Monovalent antibody design and mechanism of action of onartuzumab, a MET antagonist with anti-tumor activity as a therapeutic agent

Mark Merchant; Xiaolei Ma; Henry R. Maun; Zhong Zheng; Jing Peng; Mally Romero; Arthur Huang; Nai Ying Yang; Merry Nishimura; Joan M. Greve; Lydia Santell; Yu-Wen Zhang; Yanli Su; Dafna Kaufman; Karen Billeci; Elaine Mai; Barbara Moffat; Amy Lim; Eileen T. Duenas; Heidi S. Phillips; Hong Xiang; Judy Young; George F. Vande Woude; Mark S. Dennis; Dorothea Reilly; Ralph Schwall; Melissa A. Starovasnik; Robert A. Lazarus; Daniel G. Yansura

Significance Therapeutic antibodies have revolutionized the treatment of human disease. Despite these advances, antibody bivalency limits their utility against some targets. Here, we describe the development of a one-armed (monovalent) antibody, onartuzumab, targeting the receptor tyrosine kinase MET. While initial screening of bivalent antibodies produced agonists of MET, engineering them into monovalent antibodies produced antagonists instead. We explain the structural basis of the mechanism of action with the crystal structure of onartuzumab antigen-binding fragment in complex with MET and HGF-β. These discoveries have led to an additional antibody-based therapeutic option and shed light on the underpinnings of HGF/MET signaling. Binding of hepatocyte growth factor (HGF) to the receptor tyrosine kinase MET is implicated in the malignant process of multiple cancers, making disruption of this interaction a promising therapeutic strategy. However, targeting MET with bivalent antibodies can mimic HGF agonism via receptor dimerization. To address this limitation, we have developed onartuzumab, an Escherichia coli-derived, humanized, and affinity-matured monovalent monoclonal antibody against MET, generated using the knob-into-hole technology that enables the antibody to engage the receptor in a one-to-one fashion. Onartuzumab potently inhibits HGF binding and receptor phosphorylation and signaling and has antibody-like pharmacokinetics and antitumor activity. Biochemical data and a crystal structure of a ternary complex of onartuzumab antigen-binding fragment bound to a MET extracellular domain fragment, consisting of the MET Sema domain fused to the adjacent Plexins, Semaphorins, Integrins domain (MET Sema-PSI), and the HGF β-chain demonstrate that onartuzumab acts specifically by blocking HGF α-chain (but not β-chain) binding to MET. These data suggest a likely binding site of the HGF α-chain on MET, which when dimerized leads to MET signaling. Onartuzumab, therefore, represents the founding member of a class of therapeutic monovalent antibodies that overcomes limitations of antibody bivalency for targets impacted by antibody crosslinking.


Cell Death & Differentiation | 2016

RIPK3 deficiency or catalytically inactive RIPK1 provides greater benefit than MLKL deficiency in mouse models of inflammation and tissue injury

Kim Newton; D L Dugger; A Maltzman; Joan M. Greve; Maj Hedehus; Baby Martin-McNulty; Rick Carano; Tim C. Cao; N. van Bruggen; Lisa J. Bernstein; Wyne P. Lee; Xiumin Wu; J DeVoss; Juan Zhang; S Jeet; I Peng; B S McKenzie; M Roose-Girma; P Caplazi; Lauri Diehl; Joshua D. Webster; Domagoj Vucic

Necroptosis is a caspase-independent form of cell death that is triggered by activation of the receptor interacting serine/threonine kinase 3 (RIPK3) and phosphorylation of its pseudokinase substrate mixed lineage kinase-like (MLKL), which then translocates to membranes and promotes cell lysis. Activation of RIPK3 is regulated by the kinase RIPK1. Here we analyze the contribution of RIPK1, RIPK3, or MLKL to several mouse disease models. Loss of RIPK3 had no effect on lipopolysaccharide-induced sepsis, dextran sodium sulfate-induced colitis, cerulein-induced pancreatitis, hypoxia-induced cerebral edema, or the major cerebral artery occlusion stroke model. However, kidney ischemia–reperfusion injury, myocardial infarction, and systemic inflammation associated with A20 deficiency or high-dose tumor necrosis factor (TNF) were ameliorated by RIPK3 deficiency. Catalytically inactive RIPK1 was also beneficial in the kidney ischemia–reperfusion injury model, the high-dose TNF model, and in A20−/− mice. Interestingly, MLKL deficiency offered less protection in the kidney ischemia–reperfusion injury model and no benefit in A20−/− mice, consistent with necroptosis-independent functions for RIPK1 and RIPK3. Combined loss of RIPK3 (or MLKL) and caspase-8 largely prevented the cytokine storm, hypothermia, and morbidity induced by TNF, suggesting that the triggering event in this model is a combination of apoptosis and necroptosis. Tissue-specific RIPK3 deletion identified intestinal epithelial cells as the major target organ. Together these data emphasize that MLKL deficiency rather than RIPK1 inactivation or RIPK3 deficiency must be examined to implicate a role for necroptosis in disease.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2011

Influences of Aortic Motion and Curvature on Vessel Expansion in Murine Experimental Aneurysms

Craig J. Goergen; Junya Azuma; Kyla N. Barr; Lars Magdefessel; Dara Y. Kallop; Alvin Gogineni; Amarjeet K. Grewall; Robby M. Weimer; Andrew J. Connolly; Ronald L. Dalman; Charles A. Taylor; Philip S. Tsao; Joan M. Greve

Objective—To quantitatively compare aortic curvature and motion with resulting aneurysm location, direction of expansion, and pathophysiological features in experimental abdominal aortic aneurysms (AAAs). Methods and Results—MRI was performed at 4.7 T with the following parameters: (1) 3D acquisition for vessel geometry and (2) 2D cardiac-gated acquisition to quantify luminal motion. Male 24-week-old mice were imaged before and after AAA formation induced by angiotensin II (AngII)–filled osmotic pump implantation or infusion of elastase. AngII-induced AAAs formed near the location of maximum abdominal aortic curvature, and the leftward direction of expansion was correlated with the direction of suprarenal aortic motion. Elastase-induced AAAs formed in a region of low vessel curvature and had no repeatable direction of expansion. AngII significantly increased mean blood pressure (22.7 mm Hg, P<0.05), whereas both models showed a significant 2-fold decrease in aortic cyclic strain (P<0.05). Differences in patterns of elastin degradation and localization of fluorescent signal from protease-activated probes were also observed. Conclusion—The direction of AngII aneurysm expansion correlated with the direction of motion, medial elastin dissection, and adventitial remodeling. Anterior infrarenal aortic motion correlated with medial elastin degradation in elastase-induced aneurysms. Results from both models suggest a relationship between aneurysm pathological features and aortic geometry and motion.


Osteoarthritis and Cartilage | 2008

Meniscectomy alters the dynamic deformational behavior and cumulative strain of tibial articular cartilage in knee joints subjected to cyclic loads

Y. Song; Joan M. Greve; Dennis R. Carter; Nicholas J. Giori

OBJECTIVE Meniscectomy-induced osteoarthritis may be mechanically based. We asked how meniscectomy alters time-dependent deformation of physiologically loaded articular cartilage. We hypothesized that meniscectomy alters nominal strain in tibial articular cartilage, and that meniscectomy affects cartilage thickness recovery following cessation of loading. METHODS A cyclic load simulating normal gait was applied to four sheep knees. A custom device was used to obtain MR images of cartilage at 4.7T during cyclic loading. Articular cartilage thickness and nominal strain were measured every 2.5 min during 1h of cyclic loading, and during 2.5h after cessation of loading. RESULTS Following meniscectomy the loaded joints rapidly developed high strain centrally and minimal strain peripherally. Maximum nominal strains after 1h of loading were about 55% in the intact knees and 72% in the meniscectomized knees. Nominal strains in the peripheral tibial cartilage were significantly reduced in the meniscectomized knees. Strain recovery was markedly prolonged in the meniscectomized knees. CONCLUSIONS With meniscectomy, tibial articular cartilage in the central load bearing region remains chronically deformed and dehydrated, even after cessation of loading. Post-meniscectomy osteoarthritis may be initiated in this region by direct damage to the cartilage matrix, or by altering the hydration of the tissue. In peripheral regions, reduced loading and strain may facilitate subchondral vascular invasion, and endochondral ossification. This is consistent with the central fibrillation and peripheral osteophyte formation seen in post-meniscectomy osteoarthritis.


Clinical Cancer Research | 2012

Targeting the PI3K Pathway in the Brain - Efficacy of a PI3K Inhibitor Optimized to Cross the Blood-Brain Barrier

Laurent Salphati; Timothy P. Heffron; Bruno Alicke; Merry Nishimura; Kai H. Barck; Richard A. D. Carano; Jonathan Cheong; Kyle A. Edgar; Joan M. Greve; Samir Kharbanda; Hartmut Koeppen; Shari Lau; Leslie Lee; Jodie Pang; Emile Plise; Jenny L. Pokorny; Hani Bou Reslan; Jann N. Sarkaria; Jeffrey Wallin; Xiaolin Zhang; Stephen E. Gould; Alan G. Olivero; Heidi S. Phillips

Purpose: Glioblastoma (GBM), the most common primary brain tumor in adults, presents a high frequency of alteration in the PI3K pathway. Our objectives were to identify a dual PI3K/mTOR inhibitor optimized to cross the blood–brain barrier (BBB) and characterize its brain penetration, pathway modulation in the brain and efficacy in orthotopic xenograft models of GBM. Experimental Design: Physicochemical properties of PI3K inhibitors were optimized using in silico tools, leading to the identification of GNE-317. This compound was tested in cells overexpressing P-glycoprotein (P-gp) or breast cancer resistance protein (BCRP). Following administration to mice, GNE-317 plasma and brain concentrations were determined, and phosphorylated biomarkers (pAkt, p4EBP1, and pS6) were measured to assess PI3K pathway suppression in the brain. GNE-317 efficacy was evaluated in the U87, GS2, and GBM10 orthotopic models of GBM. Results: GNE-317 was identified as having physicochemical properties predictive of low efflux by P-gp and BCRP. Studies in transfected MDCK cells showed that GNE-317 was not a substrate of either transporter. GNE-317 markedly inhibited the PI3K pathway in mouse brain, causing 40% to 90% suppression of the pAkt and pS6 signals up to 6-hour postdose. GNE-317 was efficacious in the U87, GS2, and GBM10 orthotopic models, achieving tumor growth inhibition of 90% and 50%, and survival benefit, respectively. Conclusions: These results indicated that specific optimization of PI3K inhibitors to cross the BBB led to potent suppression of the PI3K pathway in healthy brain. The efficacy of GNE-317 in 3 intracranial models of GBM suggested that this compound could be effective in the treatment of GBM. Clin Cancer Res; 18(22); 6239–48. ©2012 AACR.


Journal of Endovascular Therapy | 2007

Increased Anterior Abdominal Aortic Wall Motion: Possible Role in Aneurysm Pathogenesis and Design of Endovascular Devices

Craig J. Goergen; Bonnie L. Johnson; Joan M. Greve; Charles A. Taylor; Christopher K. Zarins

PURPOSE To determine whether variations in aortic wall motion exist in mammalian species other than humans and to consider the potential implications of such variations. METHODS M-mode ultrasound was used to measure abdominal aortic wall motion in 4 animal species [mice (n=10), rats (n=8), rabbits (n=7), and pigs (n=5)], and humans (n=6). Anterior wall displacement, posterior wall displacement, and diastolic diameter were measured. The ratio of displacement to diameter and cyclic strain were calculated. RESULTS Body mass varied from 24.1+/-2.4 g (mouse) to 61.8+/-13.4 kg (human); aortic diameter varied from 0.53+/-0.07 mm (mouse) to 1.2+/-1 mm (human). Anterior wall displacement was 2.5 to 4.0 times greater than posterior among the species studied. The ratios of wall displacement to diastolic diameter were similar for the anterior (range 9.40%-11.80%) and posterior (range 2.49%-3.91%) walls among species. The ratio of anterior to posterior displacement (range 2.47-4.03) and aortic wall circumferential cyclic strain (range 12.1%-15.7%) were also similar. An allometric scaling exponent was experimentally derived relating anterior wall (0.377+/-0.032, R2=0.94) and posterior wall (0.378+/-0.037, R2=0.93) displacement to body mass. CONCLUSION Abdominal aortic wall dynamics are similar in animals and humans regardless of aortic size, wih more anterior than posterior wall motion. Wall displacement increases linearly with diameter, but allometrically with body mass. These data suggest increased dynamic strain of the anterior wall. Increased strain, corresponding to increased elastin fatigue, may help explain why human abdominal aortic aneurysms initially develop anteriorly. Aortic wall motion should be considered when developing endovascular devices, since asymmetric motion may affect device migration, fixation, and sealing.


Journal of Magnetic Resonance Imaging | 2010

In Vivo Quantification of Murine Aortic Cyclic Strain, Motion, and Curvature: Implications for Abdominal Aortic Aneurysm Growth

Craig J. Goergen; Kyla N. Barr; Diem T. Huynh; Jeffrey Eastham-Anderson; Gilwoo Choi; Maj Hedehus; Ronald L. Dalman; Andrew J. Connolly; Charles A. Taylor; Philip S. Tsao; Joan M. Greve

To develop methods to quantify cyclic strain, motion, and curvature of the murine abdominal aorta in vivo.


The Journal of Nuclear Medicine | 2009

PET of Glial Metabolism Using 2-18F-Fluoroacetate

Jan Marik; Annie Ogasawara; Baby Martin-McNulty; Jed Ross; Judith E. Flores; Herman S. Gill; Jeff N. Tinianow; Alexander N. Vanderbilt; Merry Nishimura; Franklin Peale; Cinthia V. Pastuskovas; Joan M. Greve; Nicholas van Bruggen; Simon Williams

Imaging of the glial activation that occurs in response to central nervous system trauma and inflammation could become a powerful technique for the assessment of several neuropathologies. The selective uptake and metabolism of 2-18F-fluoroacetate (18F-FAC) in glia may represent an attractive strategy for imaging glial metabolism. Methods: We have evaluated the use of 18F-FAC as a specific PET tracer of glial cell metabolism in rodent models of glioblastoma, stroke, and ischemia–hypoxia. Results: Enhanced uptake of 18F-FAC was observed (6.98 ± 0.43 percentage injected dose per gram [%ID/g]; tumor-to-normal ratio, 1.40) in orthotopic U87 xenografts, compared with healthy brain tissue. The lesion extent determined by 18F-FAC PET correlated with that determined by MRI (R2 = 0.934, P = 0.007). After transient middle cerebral artery occlusion in the rat brain, elevated uptake of 18F-FAC (1.00 ± 0.03 %ID/g; lesion-to-normal ratio, 1.90) depicted the ischemic territory and correlated with infarct volumes as determined by 2,3,5-triphenyltetrazolium chloride staining (R2 = 0.692, P = 0.010) and with the presence of activated astrocytes detected by anti–glial fibrillary acidic protein. Ischemia–hypoxia, induced by permanent ligation of the common carotid artery with transient hypoxia, resulted in persistent elevation of 18F-FAC uptake within 30 min of the induction of hypoxia. Conclusion: Our data support the further evaluation of 18F-FAC PET for the assessment of glial cell metabolism associated with neuroinflammation.

Collaboration


Dive into the Joan M. Greve'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
Researchain Logo
Decentralizing Knowledge