Network


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

Hotspot


Dive into the research topics where Gregory R. Wojtkiewicz is active.

Publication


Featured researches published by Gregory R. Wojtkiewicz.


Science | 2014

Vascular and neurogenic rejuvenation of the aging mouse brain by young systemic factors

Lida Katsimpardi; Nadia K. Litterman; Pamela A. Schein; Christine M. Miller; Francesco Loffredo; Gregory R. Wojtkiewicz; John W. Chen; Richard T. Lee; Amy J. Wagers; Lee L. Rubin

Help the Aged Muscle function declines with age, as does neurogenesis in certain brain regions. Two teams analyzed the effects of heterochronic parabiosis in mice. Sinha et al. (p. 649) found that when an aged mouse shares a circulatory system with a youthful mouse, the aged mouse sees improved muscle function, and Katsimpardi et al. (p. 630) observed increased generation of olfactory neurons. In both cases, Growth Differentiation Factor 11 appeared to be one of the key components of the young blood. A circulating growth factor promotes youthful muscles and brains in aged mice. In the adult central nervous system, the vasculature of the neurogenic niche regulates neural stem cell behavior by providing circulating and secreted factors. Age-related decline of neurogenesis and cognitive function is associated with reduced blood flow and decreased numbers of neural stem cells. Therefore, restoring the functionality of the niche should counteract some of the negative effects of aging. We show that factors found in young blood induce vascular remodeling, culminating in increased neurogenesis and improved olfactory discrimination in aging mice. Further, we show that GDF11 alone can improve the cerebral vasculature and enhance neurogenesis. The identification of factors that slow the age-dependent deterioration of the neurogenic niche in mice may constitute the basis for new methods of treating age-related neurodegenerative and neurovascular diseases.


PLOS ONE | 2013

Measuring Myeloperoxidase Activity in Biological Samples

Benjamin Pulli; Muhammad Ali; Reza Forghani; Stefan Schob; Kevin Li Chun Hsieh; Gregory R. Wojtkiewicz; Jenny Linnoila; John W. Chen

Background Enzymatic activity measurements of the highly oxidative enzyme myeloperoxidase (MPO), which is implicated in many diseases, are widely used in the literature, but often suffer from nonspecificity and lack of uniformity. Thus, validation and standardization are needed to establish a robust method that is highly specific, sensitive, and reproducible for assaying MPO activity in biological samples. Principal findings We found conflicting results between in vivo molecular MR imaging of MPO, which measures extracellular activity, and commonly used in vitro MPO activity assays. Thus, we established and validated a protocol to obtain extra- and intracellular MPO from murine organs. To validate the MPO activity assays, three different classes of MPO activity assays were used in spike and recovery experiments. However, these assay methods yielded inconsistent results, likely because of interfering substances and other peroxidases present in tissue extracts. To circumvent this, we first captured MPO with an antibody. The MPO activity of the resultant samples was assessed by ADHP and validated against samples from MPO-knockout mice in murine disease models of multiple sclerosis, steatohepatitis, and myocardial infarction. We found the measurements performed using this protocol to be highly specific and reproducible, and when performed using ADHP, to be highly sensitive over a broad range. In addition, we found that intracellular MPO activity correlated well with tissue neutrophil content, and can be used as a marker to assess neutrophil infiltration in the tissue. Conclusion We validated a highly specific and sensitive assay protocol that should be used as the standard method for all MPO activity assays in biological samples. We also established a method to obtain extra- and intracellular MPO from murine organs. Extracellular MPO activity gives an estimate of the oxidative stress in inflammatory diseases, while intracellular MPO activity correlates well with tissue neutrophil content. A detailed step-by-step protocol is provided.


Nano Letters | 2012

Tracking mesenchymal stem cells with iron oxide nanoparticle loaded poly(lactide-co-glycolide) microparticles.

Chenjie Xu; David Miranda-Nieves; James A. Ankrum; Mads Emil Matthiesen; Joseph A. Phillips; Isaac Roes; Gregory R. Wojtkiewicz; Vikram Juneja; Jens Roat Kultima; Weian Zhao; Praveen Kumar Vemula; Charles P. Lin; Matthias Nahrendorf; Jeffrey M. Karp

Monitoring the location, distribution and long-term engraftment of administered cells is critical for demonstrating the success of a cell therapy. Among available imaging-based cell tracking tools, magnetic resonance imaging (MRI) is advantageous due to its noninvasiveness, deep penetration, and high spatial resolution. While tracking cells in preclinical models via internalized MRI contrast agents (iron oxide nanoparticles, IO-NPs) is a widely used method, IO-NPs suffer from low iron content per particle, low uptake in nonphagocytotic cell types (e.g., mesenchymal stem cells, MSCs), weak negative contrast, and decreased MRI signal due to cell proliferation and cellular exocytosis. Herein, we demonstrate that internalization of IO-NP (10 nm) loaded biodegradable poly(lactide-co-glycolide) microparticles (IO/PLGA-MPs, 0.4-3 μm) in MSCs enhances MR parameters such as the r(2) relaxivity (5-fold), residence time inside the cells (3-fold) and R(2) signal (2-fold) compared to IO-NPs alone. Intriguingly, in vitro and in vivo experiments demonstrate that internalization of IO/PLGA-MPs in MSCs does not compromise inherent cell properties such as viability, proliferation, migration and their ability to home to sites of inflammation.


Science Translational Medicine | 2015

Predicting therapeutic nanomedicine efficacy using a companion magnetic resonance imaging nanoparticle

Miles A. Miller; Suresh Gadde; Christina Pfirschke; Camilla Engblom; Melissa M. Sprachman; Rainer H. Kohler; Katherine S. Yang; Ashley M. Laughney; Gregory R. Wojtkiewicz; Nazila Kamaly; Sushma Bhonagiri; Mikael J. Pittet; Omid C. Farokhzad; Ralph Weissleder

Magnetic nanoparticles predict the efficacy of drug-loaded polymeric nanoparticles in vivo, helping select for tumors more responsive to nanomedicine. Particle prediction One particle, it seems, can predict the behavior of another. Thankfully, this is not the beginning of a lesson on quantum physics; instead, it is the basis for potentially designing targeted clinical trials in nanomedicine, by knowing if a tumor is likely to respond to a particular therapeutic nanoparticle. Miller et al. hypothesized that if a tumor readily takes up magnetic nanoparticles (MNP), it will also accumulate other nanoparticles carrying a deadly payload. The authors injected MNPs and a fluorescent version of the therapeutic nanoparticles into mice and followed their biodistribution using imaging. Both types of nanoparticles had similar pharmacokinetics and uptake in tumor-associated host cells owing to the enhanced permeability and retention effect. In mice with human tumors, Miller and colleagues found that the tumors with high MNP uptake were significantly more responsive than those with medium or low uptake to nanoparticles delivering chemotherapeutics. Thus, MNPs can be used as companion imaging agents during nanomedicine trials to predict the therapeutic effect of their nanosized counterparts. Therapeutic nanoparticles (TNPs) have shown heterogeneous responses in human clinical trials, raising questions of whether imaging should be used to identify patients with a higher likelihood of NP accumulation and thus therapeutic response. Despite extensive debate about the enhanced permeability and retention (EPR) effect in tumors, it is increasingly clear that EPR is extremely variable; yet, little experimental data exist to predict the clinical utility of EPR and its influence on TNP efficacy. We hypothesized that a 30-nm magnetic NP (MNP) in clinical use could predict colocalization of TNPs by magnetic resonance imaging (MRI). To this end, we performed single-cell resolution imaging of fluorescently labeled MNPs and TNPs and studied their intratumoral distribution in mice. MNPs circulated in the tumor microvasculature and demonstrated sustained uptake into cells of the tumor microenvironment within minutes. MNPs could predictably demonstrate areas of colocalization for a model TNP, poly(d,l-lactic-co-glycolic acid)-b-polyethylene glycol (PLGA-PEG), within the tumor microenvironment with >85% accuracy and circulating within the microvasculature with >95% accuracy, despite their markedly different sizes and compositions. Computational analysis of NP transport enabled predictive modeling of TNP distribution based on imaging data and identified key parameters governing intratumoral NP accumulation and macrophage uptake. Finally, MRI accurately predicted initial treatment response and drug accumulation in a preclinical efficacy study using a paclitaxel-encapsulated NP in tumor-bearing mice. These approaches yield valuable insight into the in vivo kinetics of NP distribution and suggest that clinically relevant imaging modalities and agents can be used to select patients with high EPR for treatment with TNPs.


Journal of Immunology | 2006

Oligomerization of CXCL10 Is Necessary for Endothelial Cell Presentation and In Vivo Activity

Gabriele S. V. Campanella; Jan Grimm; Lindsay A. Manice; Richard A. Colvin; Benjamin D. Medoff; Gregory R. Wojtkiewicz; Ralph Weissleder; Andrew D. Luster

The chemokine IFN-γ-inducible protein of 10 kDa (IP-10; CXCL10) plays an important role in the recruitment of activated T lymphocytes into sites of inflammation by interacting with the G protein-coupled receptor CXCR3. IP-10, like other chemokines, forms oligomers, the role of which has not yet been explored. In this study, we used a monomeric IP-10 mutant to elucidate the functional significance of oligomerization. Although monomeric IP-10 had reduced binding affinity for CXCR3 and heparin, it was able to induce in vitro chemotaxis of activated T cells with the same efficacy as wild-type IP-10. However, monomeric IP-10 was unable to induce recruitment of activated CD8+ T cells into the airways of mice after intratracheal instillation. Use of a different IP-10 mutant demonstrated that this inability was due to lack of oligomerization rather than reduced CXCR3 or heparin binding. Molecular imaging demonstrated that both wild-type and monomeric IP-10 were retained in the lung after intratracheal instillation. However, in vitro binding assays indicated that wild-type, but not monomeric, IP-10 was retained on endothelial cells and could induce transendothelial chemotaxis of activated T cells. We therefore propose that oligomerization of IP-10 is required for presentation on endothelial cells and subsequent transendothelial migration, an essential step for lymphocyte recruitment in vivo.


Nature Immunology | 2012

Early window of diabetes determinism in NOD mice, dependent on the complement receptor CRIg, identified by noninvasive imaging

Wenxian Fu; Gregory R. Wojtkiewicz; Ralph Weissleder; Christophe Benoist; Diane Mathis

All juvenile mice of the nonobese diabetic (NOD) strain develop insulitis, but there is considerable variation in their progression to diabetes. Here we used a strategy based on magnetic resonance imaging (MRI) of magnetic nanoparticles to noninvasively visualize local effects of pancreatic-islet inflammation to predict the onset of diabetes in NOD mice. MRI signals acquired during a narrow early time window allowed us to sort mice into groups that would progress to clinical disease or not and to estimate the time to diabetes development. We exploited this approach to identify previously unknown molecular and cellular elements correlated with disease protection, including the complement receptor of the immunoglobulin superfamily (CRIg), which marked a subset of macrophages associated with diabetes resistance. Administration of a fusion of CRIg and the Fc portion of immunoglobulin resulted in lower MRI signals and diabetes incidence. In addition to identifying regulators of disease progression, we show here that diabetes is set at an early age in NOD mice.


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

In vivo imaging of T cell delivery to tumors after adoptive transfer therapy

Mikael J. Pittet; Jan Grimm; Cedric R. Berger; Takahiko Tamura; Gregory R. Wojtkiewicz; Matthias Nahrendorf; Pedro Romero; Filip K. Swirski; Ralph Weissleder

Adoptive transfer therapy of in vitro-expanded tumor-specific cytolytic T lymphocytes (CTLs) can mediate objective cancer regression in patients. Yet, technical limitations hamper precise monitoring of posttherapy T cell responses. Here we show in a mouse model that fused single photon emission computed tomography and x-ray computed tomography allows quantitative whole-body imaging of 111In-oxine-labeled CTLs at tumor sites. Assessment of CTL localization is rapid, noninvasive, three-dimensional, and can be repeated for longitudinal analyses. We compared the effects of lymphodepletion before adoptive transfer on CTL recruitment and report that combined treatment increased intratumoral delivery of CTLs and improved antitumor efficacy. Because 111In-oxine is a Food and Drug Administration-approved clinical agent, and human SPECT-CT systems are available, this approach should be clinically translatable, insofar as it may assess the efficacy of immunization procedures in individual patients and lead to development of more effective therapies.


Journal of Experimental Medicine | 2015

Macrophages retain hematopoietic stem cells in the spleen via VCAM-1

Partha Dutta; Friedrich Felix Hoyer; Lubov S. Grigoryeva; Hendrik B. Sager; Florian Leuschner; Gabriel Courties; Anna Borodovsky; Tatiana Novobrantseva; Vera M. Ruda; Kevin Fitzgerald; Yoshiko Iwamoto; Gregory R. Wojtkiewicz; Yuan Sun; Nicolas Da Silva; Peter Libby; Daniel G. Anderson; Filip K. Swirski; Ralph Weissleder; Matthias Nahrendorf

Dutta et al. show that targeting VACM-1 expression in splenic macrophages impairs extramedullary hematopoiesis, thus reducing inflammation in mouse ischemic heart and atherosclerotic plaques.


Circulation | 2015

Targeting Interleukin-1β Reduces Leukocyte Production After Acute Myocardial Infarction

Hendrik B. Sager; Timo Heidt; Maarten Hulsmans; Partha Dutta; Gabriel Courties; Matthew Sebas; Gregory R. Wojtkiewicz; Benoit Tricot; Yoshiko Iwamoto; Yuan Sun; Ralph Weissleder; Peter Libby; Filip K. Swirski; Matthias Nahrendorf

Background— Myocardial infarction (MI) is an ischemic wound that recruits millions of leukocytes. MI-associated blood leukocytosis correlates inversely with patient survival, yet the signals driving heightened leukocyte production after MI remain incompletely understood. Methods and Results— With the use of parabiosis surgery, this study shows that soluble danger signals, among them interleukin-1&bgr;, increase bone marrow hematopoietic stem cell proliferation after MI. Data obtained in bone marrow reconstitution experiments reveal that interleukin-1&bgr; enhances hematopoietic stem cell proliferation by both direct actions on hematopoietic cells and through modulation of the bone marrow’s hematopoietic microenvironment. An antibody that neutralizes interleukin-1&bgr; suppresses these effects. Anti-interleukin-1&bgr; treatment dampens the post-MI increase in hematopoietic stem cell proliferation. Consequently, decreased leukocyte numbers in the blood and infarct reduce inflammation and diminish post-MI heart failure in ApoE–/– mice with atherosclerosis. Conclusions— The presented insight into post-MI bone marrow activation identifies a mechanistic target for muting inflammation in the ischemically damaged heart.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Noninvasive mapping of pancreatic inflammation in recent-onset type-1 diabetes patients

Jason L. Gaglia; Mukesh G. Harisinghani; Iman Aganj; Gregory R. Wojtkiewicz; Sandeep Hedgire; Christophe Benoist; Diane Mathis; Ralph Weissleder

Significance We describe a readily exportable method for noninvasive imaging of the pancreatic inflammation underlying type-1 diabetes (T1D), based on MRI of the clinically approved magnetic nanoparticle ferumoxytol. This approach, which reflects nanoparticle uptake by macrophages in the inflamed pancreatic lesion, has been validated rigorously in mouse T1D models. Methodological advances reported here include extensive optimization of image acquisition and improved MRI registration and visualization technologies. A proof-of-principle study revealed a clear difference in whole-pancreas nanoparticle accumulation in patients with recent-onset T1D versus healthy controls and pronounced intra- and interpancreatic signal heterogeneity in patients. Noninvasive generation of 3D, high-resolution maps of pancreatic inflammation should prove invaluable in assessing T1D progression and as an indicator of response to therapy. The inability to visualize the initiation and progression of type-1 diabetes (T1D) noninvasively in humans is a major research and clinical stumbling block. We describe an advanced, exportable method for imaging the pancreatic inflammation underlying T1D, based on MRI of the clinically approved magnetic nanoparticle (MNP) ferumoxytol. The MNP-MRI approach, which reflects nanoparticle uptake by macrophages in the inflamed pancreatic lesion, has been validated extensively in mouse models of T1D and in a pilot human study. The methodological advances reported here were enabled by extensive optimization of image acquisition at 3T, as well as by the development of improved MRI registration and visualization technologies. A proof-of-principle study on patients recently diagnosed with T1D versus healthy controls yielded two major findings: First, there was a clear difference in whole-pancreas nanoparticle accumulation in patients and controls; second, the patients with T1D exhibited pronounced inter- and intrapancreatic heterogeneity in signal intensity. The ability to generate noninvasive, 3D, high-resolution maps of pancreatic inflammation in autoimmune diabetes should prove invaluable in assessing disease initiation and progression and as an indicator of response to emerging therapies.

Collaboration


Dive into the Gregory R. Wojtkiewicz'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
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge