Network


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

Hotspot


Dive into the research topics where Stasia A. Anderson is active.

Publication


Featured researches published by Stasia A. Anderson.


Stem Cells | 2006

Magnetic Resonance Imaging and Confocal Microscopy Studies of Magnetically Labeled Endothelial Progenitor Cells Trafficking to Sites of Tumor Angiogenesis

Ali S. Arbab; Victor Frenkel; Sunil D. Pandit; Stasia A. Anderson; Gene T. Yocum; Monica Bur; Hanh Khuu; Elizabeth J. Read; Joseph A. Frank

AC133 cells, a subpopulation of CD34+ hematopoietic stem cells, can transform into endothelial cells that may integrate into the neovasculature of tumors or ischemic tissue. Most current imaging modalities do not allow monitoring of early migration and incorporation of endothelial progenitor cells (EPCs) into tumor neovasculature. The goals of this study were to use magnetic resonance imaging (MRI) to track the migration and incorporation of intravenously injected, magnetically labeled EPCs into the blood vessels in a rapidly growing flank tumor model and to determine whether the pattern of EPC incorporation is related to the time of injection or tumor size. Materials and Methods: EPCs labeled with ferumoxide–protamine sulfate (FePro) complexes were injected into mice bearing xenografted glioma, and MRI was obtained at different stages of tumor development and size. Results: Migration and incorporation of labeled EPCs into tumor neovasculature were detected as low signal intensity on MRI at the tumor periphery as early as 3 days after EPC administration in preformed tumors. However, low signal intensities were not observed in tumors implanted at the time of EPC administration until tumor size reached 1 cm at 12 to 14 days. Prussian blue staining showed iron‐positive cells at the sites corresponding to low signal intensity on MRI. Confocal microcopy showed incorporation into the neovasculature, and immunohistochemistry clearly demonstrated the transformation of the administered EPCs into endothelial cells. Conclusion: MRI demonstrated the incorporation of FePro‐labeled human CD34+/AC133+ EPCs into the neovasculature of implanted flank tumors.


Annals of Neurology | 2004

Magnetic resonance imaging of labeled T-cells in a mouse model of multiple sclerosis†

Stasia A. Anderson; Jacqueline Shukaliak‐Quandt; Elaine K. Jordan; Ali S. Arbab; Roland Martin; Henry F. McFarland; Joseph A. Frank

Multiple sclerosis (MS) is a T cell–mediated autoimmune disease with early lesions characterized by mononuclear cellular infiltrate, edema, demyelination, and axonal loss that contribute to the clinical course of the disease. Experimental autoimmune encephalomyelitis (EAE) in the mouse is a valuable model with a similar disease course to relapsing‐remitting MS. The ability to detect the migration of encephalitogenic T cells into the central nervous system in EAE and MS would provide key information on these cells role in the development of lesions observed on magnetic resonance imaging (MRI). T cells were labeled for detection by magnetic resonance imaging using Food and Drug Administration–approved, superparamagnetic iron oxide nanoparticles (Ferumoxides) complexed to poly‐L‐Lysine (FE‐PLL). EAE was induced by adoptive transfer of either labeled or unlabeled T cells. After disease onset, FE‐PLL–labeled T cells were detected in the mouse spinal cord using in vivo and ex vivo cellular MRI. Excellent correlation was seen between MRI‐visible lesions in the spinal cord and histopathology. The results demonstrate that T cells labeled with FE‐PLL can induce EAE disease and can be detected in vivo in the mouse model. The magnetic labeling of cells opens the possibility of monitoring specific cellular phenotypes or pharmacologically or genetically engineered cells by MRI.


Circulation Research | 2009

p53 Improves Aerobic Exercise Capacity and Augments Skeletal Muscle Mitochondrial DNA Content

Joon-Young Park; Ping yuan Wang; Takumi Matsumoto; Ho Joong Sung; Wenzhe Ma; Jeong W. Choi; Stasia A. Anderson; Scot C. Leary; Robert S. Balaban; Ju Gyeong Kang; Paul M. Hwang

Rationale: Exercise capacity is a physiological characteristic associated with protection from both cardiovascular and all-cause mortality. p53 regulates mitochondrial function and its deletion markedly diminishes exercise capacity, but the underlying genetic mechanism orchestrating this is unclear. Understanding the biology of how p53 improves exercise capacity may provide useful insights for improving both cardiovascular as well as general health. Objective: The purpose of this study was to understand the genetic mechanism by which p53 regulates aerobic exercise capacity. Methods and Results: Using a variety of physiological, metabolic, and molecular techniques, we further characterized maximum exercise capacity and the effects of training, measured various nonmitochondrial and mitochondrial determinants of exercise capacity, and examined putative regulators of mitochondrial biogenesis. As p53 did not affect baseline cardiac function or inotropic reserve, we focused on the involvement of skeletal muscle and now report a wider role for p53 in modulating skeletal muscle mitochondrial function. p53 interacts with Mitochondrial Transcription Factor A (TFAM), a nuclear-encoded gene important for mitochondrial DNA (mtDNA) transcription and maintenance, and regulates mtDNA content. The increased mtDNA in p53+/+ compared to p53−/− mice was more marked in aerobic versus glycolytic skeletal muscle groups with no significant changes in cardiac tissue. These in vivo observations were further supported by in vitro studies showing overexpression of p53 in mouse myoblasts increases both TFAM and mtDNA levels whereas depletion of TFAM by shRNA decreases mtDNA content. Conclusions: Our current findings indicate that p53 promotes aerobic metabolism and exercise capacity by using different mitochondrial genes and mechanisms in a tissue-specific manner.


Cytotherapy | 2004

Methods for magnetically labeling stem and other cells for detection by in vivo magnetic resonance imaging

Joseph A. Frank; Stasia A. Anderson; H. Kalsih; Elaine K. Jordan; Bobbi K. Lewis; Gene T. Yocum; Ali S. Arbab

Superparamagnetic iron oxide (SPIO) nanoparticles are being used for intracellular magnetic labeling of stem cells and other cells in order to monitor cell trafficking by magnetic resonance imaging (MRI) as part of cellular-based repair, replacement and treatment strategies. This review focuses on the various methods for magnetic labeling of stem cells and other mammalian cells and on how to translate experimental results from bench to bedside.


Circulation-cardiovascular Imaging | 2010

Late gadolinium-enhancement cardiac magnetic resonance identifies postinfarction myocardial fibrosis and the border zone at the near cellular level in ex vivo rat heart.

Erik B. Schelbert; Li Yueh Hsu; Stasia A. Anderson; Bibhu D Mohanty; Syed M. Karim; Peter Kellman; Anthony H. Aletras; Andrew E. Arai

Background—Using a resolution 1000-fold higher than prior studies, we studied (1) the degree to which late gadolinium-enhancement (LGE) cardiac magnetic resonance tracks fibrosis from chronic myocardial infarction and (2) the relationship between intermediate signal intensity and partial volume averaging at distinct “smooth” infarct borders versus disorganized mixtures of fibrosis and viable cardiomyocytes. Methods and Results—Sprague-Dawley rats underwent myocardial infarction by coronary ligation. Two months later, rats were euthanized 10 minutes after administration of 0.3 mmol/kg intravenous gadolinium. LGE images ex vivo at 7 T with a 3D gradient echo sequence with 50×50×50 &mgr;m voxels were compared with histological sections (Masson trichrome). Planimetered histological and LGE regions of fibrosis correlated well (y=1.01x−0.01; R2=0.96; P<0.001). In addition, LGE images routinely detected clefts of viable cardiomyocytes 2 to 4 cells thick that separated bands of fibrous tissue. Although LGE clearly detected disorganized mixtures of fibrosis and viable cardiomyocytes characterized by intermediate signal intensity voxels, the percentage of apparent intermediate signal intensity myocardium increased significantly (P<0.01) when image resolution was degraded to resemble clinical resolution consistent with significant partial volume averaging. Conclusions—These data provide important validation of LGE at nearly the cellular level for detection of fibrosis after myocardial infarction. Although LGE can detect heterogeneous patches of fibrosis and viable cardiomyocytes as patches of intermediate signal intensity, the percentage of intermediate signal intensity voxels is resolution dependent. Thus, at clinical resolutions, distinguishing the peri-infarct border zone from partial volume averaging with LGE is challenging.


Investigative Radiology | 2006

Gadolinium-fullerenol as a paramagnetic contrast agent for cellular imaging

Stasia A. Anderson; Kristen Kyongae Lee; Joseph A. Frank

Objectives:The objectives of this study were to test cell-labeling methods to achieve intracellular labeling and T1 enhancement of cells on magnetic resonance imaging using a paramagnetic Gd@C82 fullerenol contrast agent, and to determine the effect of labeling on cell viability, metabolism, and differentiation capacity. Materials and Methods:We tested the use of a transfection agent for labeling cells in culture with Gd@C82 fullerenol. Proliferation, viability, and differentiation assays of mesenchymal stem cell (MSC) cultures; light and electron microscopy of MSC and macrophages; and MRI of MSC, macrophage, and HeLa cervical carcinoma cell cultures in vitro and in vivo were performed to evaluate the labeled cells. Results:Protamine sulfate transfection increased cell uptake of Gd@C82 fullerenols. The label was distributed in endosomes in the cytoplasm as shown by electron microscopy. High viability was shown for all cell lines and normal differentiation capacity was shown for MSCs. T1 of labeled MSC at 7 T was reduced 71% compared with unlabeled cells. Conclusions:Cellular labeling with Gd@C82 is feasible and can produce T1-enhanced cells on magnetic resonance imaging. This study suggests that further investigation of Gd fullerenols for tracking studies of viable cells, including stem cells, is warranted.


Cell | 2012

Accumulation of the inner nuclear envelope protein Sun1 is pathogenic in progeric and dystrophic laminopathies.

Chia-Yen Chen; Ya-Hui Chi; Rafidah Mutalif; Matthew F. Starost; Timothy G. Myers; Stasia A. Anderson; Colin L. Stewart; Kuan-Teh Jeang

Human LMNA gene mutations result in laminopathies that include Emery-Dreifuss muscular dystrophy (AD-EDMD) and Hutchinson-Gilford progeria, the premature aging syndrome (HGPS). The Lmna null (Lmna(-/-)) and progeroid LmnaΔ9 mutant mice are models for AD-EDMD and HGPS, respectively. Both animals develop severe tissue pathologies with abbreviated life spans. Like HGPS cells, Lmna(-/-) and LmnaΔ9 fibroblasts have typically misshapen nuclei. Unexpectedly, Lmna(-/-) or LmnaΔ9 mice that are also deficient for the inner nuclear membrane protein Sun1 show markedly reduced tissue pathologies and enhanced longevity. Concordantly, reduction of SUN1 overaccumulation in LMNA mutant fibroblasts and in cells derived from HGPS patients corrected nuclear defects and cellular senescence. Collectively, these findings implicate Sun1 protein accumulation as a common pathogenic event in Lmna(-/-), LmnaΔ9, and HGPS disorders.


Magnetic Resonance in Medicine | 2007

Viral capsids as MRI contrast agents.

Lars O. Liepold; Stasia A. Anderson; Deborah A. Willits; Luke Oltrogge; Joseph A. Frank; Trevor Douglas; Mark J. Young

Viral capsids have the potential for combined cell/tissue targeting, drug delivery, and imaging. Described here is the development of a viral capsid as an efficient and potentially relevant MRI contrast agent. Two approaches are outlined to fuse high affinity Gd3+ chelating moieties to the surface of the cowpea chlorotic mottle virus (CCMV) capsid. In the first approach, a metal binding peptide has been genetically engineered into the subunit of CCMV. In a second approach gadolinium‐tetraazacyclododecane tetraacetic acid (GdDOTA) was attached to CCMV by reactions with endogenous lysine residues on the surface of the viral capsid. T1 and T2 ionic relaxivity rates for the genetic fusion particle were R1 = 210 and R2 = 402 mM−1s−1 (R2 at 56 MHz) and for CCMV functionalized with GdDOTA were R1 = 46 and R2 = 142 mM−1s−1 at 61 MHz. The relaxivities per intact capsid for the genetic fusion were R1 = 36,120 and R2 = 69,144 mM−1s−1 (R2 at 56 MHz) and for the GdDOTA CCMV construct were R1 = 2,806 and R2 = 8,662 mM−1s−1 at 61 MHz. The combination of high relaxivity, stable Gd3+ binding, and large Gd3+ payloads indicates the potential of viral capsids as high‐performance contrast agents. Magn Reson Med 58:871–879, 2007.


Journal of Clinical Investigation | 2009

A selective EP4 PGE2 receptor agonist alleviates disease in a new mouse model of X-linked nephrogenic diabetes insipidus.

Jian Hua Li; Chung-Lin Chou; Bo Li; Oksana Gavrilova; Christoph Eisner; Jurgen Schnermann; Stasia A. Anderson; Chu-Xia Deng; Mark A. Knepper; Jürgen Wess

X-linked nephrogenic diabetes insipidus (XNDI) is a severe kidney disease caused by inactivating mutations in the V2 vasopressin receptor (V2R) gene that result in the loss of renal urine-concentrating ability. At present,no specific pharmacological therapy has been developed for XNDI, primarily due to the lack of suitable animal models. To develop what we believe to be the first viable animal model of XNDI, we generated mice in which the V2R gene could be conditionally deleted during adulthood by administration of 4-OH-tamoxifen.Radioligand-binding studies confirmed the lack of V2R-binding sites in kidneys following 4-OH-tamoxifen treatment, and further analysis indicated that upon V2R deletion, adult mice displayed all characteristic symptoms of XNDI, including polyuria, polydipsia, and resistance to the antidiuretic actions of vasopressin. Gene expression analysis suggested that activation of renal EP4 PGE2 receptors might compensate for the lack of renal V2R activity in XNDI mice. Strikingly, both acute and chronic treatment of the mutant mice with a selective EP4 receptor agonist greatly reduced all major manifestations of XNDI, including changes in renal morphology.These physiological improvements were most likely due to a direct action on EP4 receptors expressed on collecting duct cells. These findings illustrate the usefulness of the newly generated V2R mutant mice for elucidating and testing new strategies for the potential treatment of humans with XNDI.


The FASEB Journal | 2008

Detection of migration of locally implanted AC133+ stem cells by cellular magnetic resonance imaging with histological findings

Ali S. Arbab; Branislava Janic; Robert A. Knight; Stasia A. Anderson; Edyta Pawelczyk; Ali M. Rad; Elizabeth J. Read; Sunil D. Pandit; Joseph A. Frank

This study investigated the factors responsible for migration and homing of magnetically labeled AC133+ cells at the sites of active angiogenesis in tumor. AC133+ cells labeled with ferumoxide‐protamine sulfate were mixed with either rat glioma or human melanoma cells and implanted in flank of nude mice. An MRI of the tumors including surrounding tissues was performed. Tumor sections were stained for Prussian blue (PB), platelet‐derived growth factor (PDGF), hypoxia‐inducible factor‐1α (HIF‐1α), stromal cell derived factor‐1 (SDF‐1), matrix metalloproteinase‐2 (MMP‐2), vascular endothelial growth factor (VEGF), and endothelial markers. Fresh snap‐frozen strips from the central and peripheral parts of the tumor were collected for Western blotting. MRIs demonstrated hypointense regions at the periphery of the tumors where the PB+/AC133+ cells were positive for endothelial cells markers. At the sites of PB+/AC133+ cells, both HIF‐1α and SDF‐1 were strongly positive and PDGF and MMP‐2 showed generalized expression in the tumor and surrounding tissues. There was no significant association of PB+/AC133+ cell localization and VEGF expression in tumor cells. Western blot demonstrated strong expression of the SDF‐1, MMP‐2, and PDGF at the peripheral parts of the tumors. HIF‐1α was expressed at both the periphery and central parts of the tumor. This work demonstrates that magnetically labeled cells can be used as probes for MRI and histological identification of administered cells.—Arbab, A. S., Janic, B., Knight, R. A., Anderson, S. A., Pawelczyk, E., Rad, A. M., Read, E. J., Pandit, S. D., Frank, J. A. Detection of migration of locally implanted AC133+ stem cells by cellular magnetic resonance imaging with histological findings. FASEB J. 22, 3234–3246 (2008)

Collaboration


Dive into the Stasia A. Anderson's collaboration.

Top Co-Authors

Avatar

Joseph A. Frank

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Ali S. Arbab

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar

Andrew E. Arai

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Li-Yueh Hsu

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Cecilia W. Lo

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

Rajeev Samtani

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Elaine K. Jordan

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Elizabeth J. Read

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Gene T. Yocum

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge