Network


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

Hotspot


Dive into the research topics where Joel A. Spencer is active.

Publication


Featured researches published by Joel A. Spencer.


Cell | 2014

Circulating Tumor Cell Clusters Are Oligoclonal Precursors of Breast Cancer Metastasis

Nicola Aceto; Aditya Bardia; David T. Miyamoto; Maria C. Donaldson; Ben S. Wittner; Joel A. Spencer; Min Yu; Adam Pely; Amanda Engstrom; Huili Zhu; Brian W. Brannigan; Ravi Kapur; Shannon L. Stott; Toshi Shioda; Sridhar Ramaswamy; David T. Ting; Charles P. Lin; Mehmet Toner; Daniel A. Haber; Shyamala Maheswaran

Circulating tumor cell clusters (CTC clusters) are present in the blood of patients with cancer but their contribution to metastasis is not well defined. Using mouse models with tagged mammary tumors, we demonstrate that CTC clusters arise from oligoclonal tumor cell groupings and not from intravascular aggregation events. Although rare in the circulation compared with single CTCs, CTC clusters have 23- to 50-fold increased metastatic potential. In patients with breast cancer, single-cell resolution RNA sequencing of CTC clusters and single CTCs, matched within individual blood samples, identifies the cell junction component plakoglobin as highly differentially expressed. In mouse models, knockdown of plakoglobin abrogates CTC cluster formation and suppresses lung metastases. In breast cancer patients, both abundance of CTC clusters and high tumor plakoglobin levels denote adverse outcomes. Thus, CTC clusters are derived from multicellular groupings of primary tumor cells held together through plakoglobin-dependent intercellular adhesion, and though rare, they greatly contribute to the metastatic spread of cancer.


Nature | 2014

Direct measurement of local oxygen concentration in the bone marrow of live animals

Joel A. Spencer; Francesca Ferraro; Emmanuel Roussakis; Alyssa Klein; Juwell Wu; Judith Runnels; Walid Zaher; Luke J. Mortensen; Clemens Alt; Raphaël Turcotte; Rushdia Z. Yusuf; Daniel Côté; Sergei A. Vinogradov; David T. Scadden; Charles P. Lin

Characterization of how the microenvironment, or niche, regulates stem cell activity is central to understanding stem cell biology and to developing strategies for the therapeutic manipulation of stem cells. Low oxygen tension (hypoxia) is commonly thought to be a shared niche characteristic in maintaining quiescence in multiple stem cell types. However, support for the existence of a hypoxic niche has largely come from indirect evidence such as proteomic analysis, expression of hypoxia inducible factor-1α (Hif-1α) and related genes, and staining with surrogate hypoxic markers (for example, pimonidazole). Here we perform direct in vivo measurements of local oxygen tension (pO2) in the bone marrow of live mice. Using two-photon phosphorescence lifetime microscopy, we determined the absolute pO2 of the bone marrow to be quite low (<32 mm Hg) despite very high vascular density. We further uncovered heterogeneities in local pO2, with the lowest pO2 (∼9.9 mm Hg, or 1.3%) found in deeper peri-sinusoidal regions. The endosteal region, by contrast, is less hypoxic as it is perfused with small arteries that are often positive for the marker nestin. These pO2 values change markedly after radiation and chemotherapy, pointing to the role of stress in altering the stem cell metabolic microenvironment.


Science Translational Medicine | 2011

Diabetes Impairs Hematopoietic Stem Cell Mobilization by Altering Niche Function

Francesca Ferraro; Stefania Lymperi; Simón Méndez-Ferrer; Borja Saez; Joel A. Spencer; Beow Y. Yeap; Elena Masselli; Gallia Graiani; Lucia Prezioso; Elisa Lodi Rizzini; Marcellina Mangoni; Vittorio Rizzoli; Stephen M. Sykes; Charles P. Lin; Paul S. Frenette; Federico Quaini; David T. Scadden

Impaired mobilization of hematopoietic stem cells in diabetic mice is due to sympathetic nervous system dysregulation of CXCL12 distribution. Boosting Stem Cell Mobilization Transplantation of hematopoietic stem cells (HSCs) from the bone marrow is a successful approach for treating blood diseases and certain cancers. Usually, the patient’s own (autologous) HSCs are used for transplant, but in some patients, their HSCs cannot be mobilized in sufficient numbers using the growth factor G-CSF (granulocyte colony-stimulating factor) to enable a successful transplant. In a new study, Ferraro and colleagues set out to discover the causes of this poor HSC mobilization. The investigators discovered by analyzing data from a number of bone marrow transplant patients that patients with diabetes showed poorer mobilization of HSCs in response to G-CSF than did those patients who did not have diabetes. The authors then confirmed in mouse models of type 1 and type 2 diabetes that HSCs were poorly mobilized from the bone marrow in response to G-CSF in these mice but not healthy control animals. The authors discovered that there was a defect in the bone marrow microenvironment of the diabetic mice rather than a problem with the HSCs themselves. Specifically, in diabetic (but not control) mice, the researchers observed mislocalization of HSCs in the bone marrow and an increase in the number of perivascular sympathetic nerve fibers in the niche with a concomitant inability of bone marrow mesenchymal stem cells to down-modulate production of the chemokine CXCL12 (a molecule known to mediate HSC localization). Finally, the authors were able to overcome the defect in HSC mobilization using a clinically approved drug called AMD3100 that interrupts the interaction of CXCL12 with its receptor CXCR4. The authors suggest that AMD3100 could be used to boost HSC mobilization in diabetic patients who require a bone marrow transplant. Success with transplantation of autologous hematopoietic stem and progenitor cells (HSPCs) in patients depends on adequate collection of these cells after mobilization from the bone marrow niche by the cytokine granulocyte colony-stimulating factor (G-CSF). However, some patients fail to achieve sufficient HSPC mobilization. Retrospective analysis of bone marrow transplant patient records revealed that diabetes correlated with poor mobilization of CD34+ HSPCs. In mouse models of type 1 and type 2 diabetes (streptozotocin-induced and db/db mice, respectively), we found impaired egress of murine HSPCs from the bone marrow after G-CSF treatment. Furthermore, HSPCs were aberrantly localized in the marrow niche of the diabetic mice, and abnormalities in the number and function of sympathetic nerve termini were associated with this mislocalization. Aberrant responses to β-adrenergic stimulation of the bone marrow included an inability of marrow mesenchymal stem cells expressing the marker nestin to down-modulate the chemokine CXCL12 in response to G-CSF treatment (mesenchymal stem cells are reported to be critical for HSPC mobilization). The HSPC mobilization defect was rescued by direct pharmacological inhibition of the interaction of CXCL12 with its receptor CXCR4 using the drug AMD3100. These data suggest that there are diabetes-induced changes in bone marrow physiology and microanatomy and point to a potential intervention to overcome poor HSPC mobilization in diabetic patients.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Engineered vascularized bone grafts

Olga Tsigkou; Irina Pomerantseva; Joel A. Spencer; Patricia A. Redondo; Alison Hart; Elisabeth O'Doherty; Yunfeng Lin; Claudia C. Friedrich; Laurence Daheron; Charles P. Lin; Cathryn A. Sundback; Joseph Vacanti; Craig M. Neville

Clinical protocols utilize bone marrow to seed synthetic and decellularized allogeneic bone grafts for enhancement of scaffold remodeling and fusion. Marrow-derived cytokines induce host neovascularization at the graft surface, but hypoxic conditions cause cell death at the core. Addition of cellular components that generate an extensive primitive plexus-like vascular network that would perfuse the entire scaffold upon anastomosis could potentially yield significantly higher-quality grafts. We used a mouse model to develop a two-stage protocol for generating vascularized bone grafts using mesenchymal stem cells (hMSCs) from human bone marrow and umbilical cord-derived endothelial cells. The endothelial cells formed tube-like structures and subsequently networks throughout the bone scaffold 4–7 days after implantation. hMSCs were essential for stable vasculature both in vitro and in vivo; however, contrary to expectations, vasculature derived from hMSCs briefly cultured in medium designed to maintain a proliferative, nondifferentiated state was more extensive and stable than that with hMSCs with a TGF-β-induced smooth muscle cell phenotype. Anastomosis occurred by day 11, with most hMSCs associating closely with the network. Although initially immature and highly permeable, at 4 weeks the network was mature. Initiation of scaffold mineralization had also occurred by this period. Some human-derived vessels were still present at 5 months, but the majority of the graft vasculature had been functionally remodeled with host cells. In conclusion, clinically relevant progenitor sources for pericytes and endothelial cells can serve to generate highly functional microvascular networks for tissue engineered bone grafts.


Nature | 2016

Distinct bone marrow blood vessels differentially regulate haematopoiesis.

Tomer Itkin; Shiri Gur-Cohen; Joel A. Spencer; Amir Schajnovitz; Saravana K. Ramasamy; Anjali P. Kusumbe; Guy Ledergor; Yookyung Jung; Idan Milo; Michael G. Poulos; Alexander Kalinkovich; Aya Ludin; Orit Kollet; Guy Shakhar; Jason M. Butler; Shahin Rafii; Ralf H. Adams; David T. Scadden; Charles P. Lin; Tsvee Lapidot

Bone marrow endothelial cells (BMECs) form a network of blood vessels that regulate both leukocyte trafficking and haematopoietic stem and progenitor cell (HSPC) maintenance. However, it is not clear how BMECs balance these dual roles, and whether these events occur at the same vascular site. We found that mammalian bone marrow stem cell maintenance and leukocyte trafficking are regulated by distinct blood vessel types with different permeability properties. Less permeable arterial blood vessels maintain haematopoietic stem cells in a low reactive oxygen species (ROS) state, whereas the more permeable sinusoids promote HSPC activation and are the exclusive site for immature and mature leukocyte trafficking to and from the bone marrow. A functional consequence of high permeability of blood vessels is that exposure to blood plasma increases bone marrow HSPC ROS levels, augmenting their migration and differentiation, while compromising their long-term repopulation and survival. These findings may have relevance for clinical haematopoietic stem cell transplantation and mobilization protocols.


Blood | 2011

Engineered cell homing

Debanjan Sarkar; Joel A. Spencer; Joseph A. Phillips; Weian Zhao; Sebastian Schafer; Dawn P. Spelke; Luke J. Mortensen; Juan P. Ruiz; Praveen Kumar Vemula; Rukmani Sridharan; Sriram Kumar; Rohit Karnik; Charles P. Lin; Jeffrey M. Karp

One of the greatest challenges in cell therapy is to minimally invasively deliver a large quantity of viable cells to a tissue of interest with high engraftment efficiency. Low and inefficient homing of systemically delivered mesenchymal stem cells (MSCs), for example, is thought to be a major limitation of existing MSC-based therapeutic approaches, caused predominantly by inadequate expression of cell surface adhesion receptors. Using a platform approach that preserves the MSC phenotype and does not require genetic manipulation, we modified the surface of MSCs with a nanometer-scale polymer construct containing sialyl Lewis(x) (sLe(x)) that is found on the surface of leukocytes and mediates cell rolling within inflamed tissue. The sLe(x) engineered MSCs exhibited a robust rolling response on inflamed endothelium in vivo and homed to inflamed tissue with higher efficiency compared with native MSCs. The modular approach described herein offers a simple method to potentially target any cell type to specific tissues via the circulation.


IEEE Journal of Selected Topics in Quantum Electronics | 2008

In Vivo Cell Tracking With Video Rate Multimodality Laser Scanning Microscopy

Israel Veilleux; Joel A. Spencer; David P. Biss; Daniel Côté; Charles P. Lin

Studies of biological processes, such as disease progression and response to therapy, call for live imaging methods that allow continuous observation without terminating the study subject for histological tissue processing. Among all current imaging modalities, optical microscopy is the only method capable of probing live tissue with cellular and subcellular resolution. We present a video-rate (30 frames/s), multimodality imaging system that is designed specifically for live animal imaging and cell tracking. In vivo depth-sectioned, high-resolution images are obtained using confocal and nonlinear optical techniques that extract structural, functional, and molecular information by combining multiple contrast mechanisms, including back scattering, fluorescence (from single- and two-photon excitation), second harmonic generation, and coherent anti-Stokes Raman scattering. Simultaneous use of up to three modalities is possible and eliminates the need for coregistration, especially on large-scale images. A real-time movement correction algorithm was developed to extend integration times in cases where the image needs to be stabilized against subject movement. Finally, imaging of fast moving leukocytes in blood vessels is made possible with a modification that permits operation at 120 frames/s over a smaller area. Sample imagery obtained in vivo with the microscope is presented to illustrate the capabilities.


Nature Medicine | 2006

G-CSF induces E-selectin ligand expression on human myeloid cells.

Nilesh M. Dagia; Samah Zeineb Gadhoum; Christine A Knoblauch; Joel A. Spencer; Parisa Zamiri; Charles P. Lin; Robert Sackstein

Clinical use of G-CSF can result in vascular and inflammatory complications. To investigate the molecular basis of these effects, we analyzed the adherence of G-CSF–mobilized human peripheral blood leukocytes (ML) to inflamed (TNF-α–stimulated) vascular endothelium. Studies using parallel plate assays under physiologic flow conditions and intravital microscopy in a mouse inflammation model each showed that ML take part in heightened adhesive interactions with endothelium compared to unmobilized (native) blood leukocytes, mediated by markedly increased E-selectin receptor-ligand interactions. Biochemical studies showed that ML express the potent E-selectin ligand HCELL (ref. 8) and another, previously unrecognized ∼65-kDa E-selectin ligand, and possess enhanced levels of transcripts encoding glycosyltransferases (ST3GalIV, FucT-IV and FucT-VII) conferring glycan modifications associated with E-selectin ligand activity. Enzymatic treatments and physiologic binding assays showed that HCELL and the ∼65-kDa E-selectin ligand contribute prominently to the observed G-CSF–induced myeloid cell adhesion to inflamed endothelium. Treatment of normal human bone marrow cells with a pharmacokinetically relevant concentration of G-CSF in vitro resulted in increased expression of these two molecules, coincident with increased transcripts encoding pertinent glycosyltransferases and heightened E-selectin binding. These findings provide direct evidence for a role of G-CSF in the induction of E-selectin ligands on myeloid cells, thus providing mechanistic insight into the pathobiology of G-CSF complications.


Analytical Chemistry | 2014

Two-Photon Antenna-Core Oxygen Probe with Enhanced Performance

Emmanuel Roussakis; Joel A. Spencer; Charles P. Lin; Sergei A. Vinogradov

Recent development of two-photon phosphorescence lifetime microscopy (2PLM) of oxygen enabled first noninvasive high-resolution measurements of tissue oxygenation in vivo in 3D, providing valuable physiological information. The so far developed two-photon-enhanced phosphorescent probes comprise antenna-core constructs, in which two-photon absorbing chromophores (antenna) capture and channel excitation energy to a phosphorescent core (metalloporphyrin) via intramolecular excitation energy transfer (EET). These probes allowed demonstration of the methods’ potential; however, they suffer from a number of limitations, such as partial loss of emissivity to competing triplet state deactivation pathways (e.g., electron transfer) and suboptimal sensitivity to oxygen, thereby limiting spatial and temporal resolution of the method. Here we present a new probe, PtTCHP-C307, designed to overcome these limitations. The key improvements include significant increase in the phosphorescence quantum yield, higher efficiency of the antenna-core energy transfer, minimized quenching of the phosphorescence by electron transfer and increased signal dynamic range. For the same excitation flux, the new probe is able to produce up to 6-fold higher signal output than previously reported molecules. Performance of PtTCHP-C307 was demonstrated in vivo in pO2 measurements through the intact mouse skull into the bone marrow, where all blood cells are made from hematopoietic stem cells.


Molecular Imaging | 2006

Imaging molecular expression on vascular endothelial cells by in vivo immunofluorescence microscopy.

Judith Runnels; Parisa Zamiri; Joel A. Spencer; Isreal Veilleux; Xunbin Wei; Alexei Bogdanov; Charles P. Lin

Molecular expression on the vascular endothelium is critical in regulating the interaction of circulating cells with the blood vessel wall. Leukocytes as well as circulating cancer cells gain entry into tissue by interacting with adhesion molecules on the endothelial cells (EC). Molecular targets on the EC are increasingly being explored for tissue-specific delivery of therapeutic and imaging agents. Here we use in vivo immunofluorescence microscopy to visualize the endothelial molecular expression in the vasculature of live animals. High-resolution images are obtained by optical sectioning through the intact skin using in vivo confocal and multiphoton microscopy after in situ labeling of EC surface markers with fluorescent antibodies. Other vascular beds such as the bone marrow and ocular blood vessels can be imaged with little or no tissue manipulation. Live imaging is particularly useful for following the dynamic expression of inducible molecules such as E-selectin during an inflammatory response.

Collaboration


Dive into the Joel A. Spencer'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

Robert Sackstein

Brigham and Women's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge