Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Cell Rep ; 42(5): 112485, 2023 05 30.
Article in English | MEDLINE | ID: mdl-37149866

ABSTRACT

Neurovascular abnormalities in mouse models of 16p11.2 deletion autism syndrome are reminiscent of alterations reported in murine models of glucose transporter deficiency, including reduced brain angiogenesis and behavioral alterations. Yet, whether cerebrovascular alterations in 16p11.2df/+ mice affect brain metabolism is unknown. Here, we report that anesthetized 16p11.2df/+ mice display elevated brain glucose uptake, a phenomenon recapitulated in mice with endothelial-specific 16p11.2 haplodeficiency. Awake 16p11.2df/+ mice display attenuated relative fluctuations of extracellular brain glucose following systemic glucose administration. Targeted metabolomics on cerebral cortex extracts reveals enhanced metabolic responses to systemic glucose in 16p11.2df/+ mice that also display reduced mitochondria number in brain endothelial cells. This is not associated with changes in mitochondria fusion or fission proteins, but 16p11.2df/+ brain endothelial cells lack the splice variant NT-PGC-1α, suggesting defective mitochondrial biogenesis. We propose that altered brain metabolism in 16p11.2df/+ mice is compensatory to endothelial dysfunction, shedding light on previously unknown adaptative responses.


Subject(s)
Endothelial Cells , Haploinsufficiency , Mice , Animals , Endothelial Cells/metabolism , Organelle Biogenesis , Chromosome Deletion , Brain
2.
Ultrasound Med Biol ; 47(11): 3291-3300, 2021 11.
Article in English | MEDLINE | ID: mdl-34373135

ABSTRACT

Right ventricular (RV) strain measurements from ultrasound via speckle-tracking techniques are being used more frequently as a non-invasive diagnostic tool for a variety of cardiopulmonary pathologies. However, despite the clinical utility of ultrasound RV strain measurements, quantification of RV strain in rodents remains difficult owing to unique image artifacts and non-standardized methodologies. We demonstrate here a simple approach for measuring RV strain in both mice and rats using high-frequency ultrasound and automated speckle tracking. Our results show estimated peak RV free-wall longitudinal strain values (mean ± standard error of the mean) in mice (n = 15) and rats (n = 5) of, respectively, -10.38% ± 0.4% and -4.85% ± 0.42%. We further estimated the 2-D Green-Lagrange strain within the RV free wall, with longitudinal components estimated at -5.7% ± 0.48% in mice and -2.1% ± 0.28% in rats. These methods and data may provide a foundation for future work aimed at evaluating murine RV strain levels in different disease models.


Subject(s)
Heart Ventricles , Ventricular Dysfunction, Right , Animals , Echocardiography , Heart , Heart Ventricles/diagnostic imaging , Mice , Rats , Ultrasonography , Ventricular Dysfunction, Right/diagnostic imaging
3.
Nat Neurosci ; 23(9): 1090-1101, 2020 09.
Article in English | MEDLINE | ID: mdl-32661394

ABSTRACT

While the neuronal underpinnings of autism spectrum disorder (ASD) are being unraveled, vascular contributions to ASD remain elusive. Here, we investigated postnatal cerebrovascular development in the 16p11.2df/+ mouse model of 16p11.2 deletion ASD syndrome. We discover that 16p11.2 hemizygosity leads to male-specific, endothelium-dependent structural and functional neurovascular abnormalities. In 16p11.2df/+ mice, endothelial dysfunction results in impaired cerebral angiogenesis at postnatal day 14, and in altered neurovascular coupling and cerebrovascular reactivity at postnatal day 50. Moreover, we show that there is defective angiogenesis in primary 16p11.2df/+ mouse brain endothelial cells and in induced-pluripotent-stem-cell-derived endothelial cells from human carriers of the 16p11.2 deletion. Finally, we find that mice with an endothelium-specific 16p11.2 deletion (16p11.2ΔEC) partially recapitulate some of the behavioral changes seen in 16p11.2 syndrome, specifically hyperactivity and impaired motor learning. By showing that developmental 16p11.2 haploinsufficiency from endothelial cells results in neurovascular and behavioral changes in adults, our results point to a potential role for endothelial impairment in ASD.


Subject(s)
Autism Spectrum Disorder/physiopathology , Endothelial Cells/pathology , Neurovascular Coupling/physiology , Animals , Autistic Disorder , Cerebrovascular Circulation/physiology , Chromosome Deletion , Chromosome Disorders , Chromosomes, Human, Pair 16 , Disease Models, Animal , Endothelial Cells/metabolism , Female , Intellectual Disability , Male , Mice , Neovascularization, Physiologic/genetics
4.
Ultrasound Med Biol ; 46(2): 359-368, 2020 02.
Article in English | MEDLINE | ID: mdl-31708270

ABSTRACT

Gas vesicles (GVs) are nanosized structures (45-800 nm) and have been reported to produce non-linear contrast signals, making them an attractive agent for molecular targeting of tumors. One barrier to their use for pre-clinical oncology studies is rapid uptake into the reticuloendothelial system (RES) and consequent rapid decrease in contrast signal after infusion ends and low signal on reperfusion after a bubble burst sequence. The purpose of this study was to examine suppression of the RES and surface modification of GVs to prolong contrast circulation in tumors for ultrasound imaging. Ultrasound imaging to measure dynamics of contrast signal intensity in tumor models was carried out using a 21-MHz high-frequency array transducer with the Vevo 2100 ultrasound system. The non-linear contrast signal from intravenously injected GVs compared with peak enhancement was measured during contrast wash-out and on reperfusion after a contrast burst sequence. Disrupting the RES by saturating the macrophage population or chemically inhibiting the Kupffer cell population with gadolinium or Intralipid preserves 62%-88% of GVs' contrast enhancement relative to peak during the wash-out phase and 32%-56% on reperfusion compared with 38% and 14%, respectively, for no disruption of the RES, indicating longer circulation of GVs in the tumor. Additionally, coating the GVs with 2-, 5- or 10-kDa polyethylene glycol (PEG) chains resulted in >70% contrast signal retention in the tumors during wash-out and, for 5- or 10-kDa PEG chains, a return to >45% of peak contrast signal on reperfusion. These findings indicate that GVs can be used as a contrast agent for tumor imaging and that disruption of the RES improved recirculation and maintained contrast enhancement caused by GVs in the tumors.


Subject(s)
Contrast Media/administration & dosage , Contrast Media/pharmacokinetics , Gases , Microbubbles , Mononuclear Phagocyte System/metabolism , Neoplasms/diagnostic imaging , Cell Line, Tumor , Humans , Neoplasms/pathology , Polyethylene Glycols , Ultrasonography/methods
5.
Mol Imaging Biol ; 20(2): 230-239, 2018 04.
Article in English | MEDLINE | ID: mdl-28956265

ABSTRACT

PURPOSE: Contrast-enhanced ultrasound plays an expanding role in oncology, but its applicability to molecular imaging is hindered by a lack of nanoscale contrast agents that can reach targets outside the vasculature. Gas vesicles (GVs)-a unique class of gas-filled protein nanostructures-have recently been introduced as a promising new class of ultrasound contrast agents that can potentially access the extravascular space and be modified for molecular targeting. The purpose of the present study is to determine the quantitative biodistribution of GVs, which is critical for their development as imaging agents. PROCEDURES: We use a novel bioorthogonal radiolabeling strategy to prepare technetium-99m-radiolabeled ([99mTc])GVs in high radiochemical purity. We use single photon emission computed tomography (SPECT) and tissue counting to quantitatively assess GV biodistribution in mice. RESULTS: Twenty minutes following administration to mice, the SPECT biodistribution shows that 84 % of [99mTc]GVs are taken up by the reticuloendothelial system (RES) and 13 % are found in the gall bladder and duodenum. Quantitative tissue counting shows that the uptake (mean ± SEM % of injected dose/organ) is 0.6 ± 0.2 for the gall bladder, 46.2 ± 3.1 for the liver, 1.91 ± 0.16 for the lungs, and 1.3 ± 0.3 for the spleen. Fluorescence imaging confirmed the presence of GVs in RES. CONCLUSIONS: These results provide essential information for the development of GVs as targeted nanoscale imaging agents for ultrasound.


Subject(s)
Acoustics , Nanostructures/chemistry , Proteins/chemistry , Radiopharmaceuticals/chemistry , Animals , Female , Fluorescence , Imaging, Three-Dimensional , Liver/diagnostic imaging , Mice , Spleen/diagnostic imaging , Technetium/chemistry , Tissue Distribution , Tomography, Emission-Computed, Single-Photon
6.
Nat Protoc ; 12(10): 2050-2080, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28880278

ABSTRACT

Gas vesicles (GVs) are a unique class of gas-filled protein nanostructures that are detectable at subnanomolar concentrations and whose physical properties allow them to serve as highly sensitive imaging agents for ultrasound and MRI. Here we provide a protocol for isolating GVs from native and heterologous host organisms, functionalizing these nanostructures with moieties for targeting and fluorescence, characterizing their biophysical properties and imaging them using ultrasound and MRI. GVs can be isolated from natural cyanobacterial and haloarchaeal host organisms or from Escherichia coli expressing a heterologous GV gene cluster and purified using buoyancy-assisted techniques. They can then be modified by replacing surface-bound proteins with engineered, heterologously expressed variants or through chemical conjugation, resulting in altered mechanical, surface and targeting properties. Pressurized absorbance spectroscopy is used to characterize their mechanical properties, whereas dynamic light scattering (DLS)and transmission electron microscopy (TEM) are used to determine nanoparticle size and morphology, respectively. GVs can then be imaged with ultrasound in vitro and in vivo using pulse sequences optimized for their detection versus background. They can also be imaged with hyperpolarized xenon MRI using chemical exchange saturation transfer between GV-bound and dissolved xenon-a technique currently implemented in vitro. Taking 3-8 d to prepare, these genetically encodable nanostructures enable multimodal, noninvasive biological imaging with high sensitivity and potential for molecular targeting.


Subject(s)
Contrast Media/chemistry , Magnetic Resonance Imaging/methods , Nanostructures/chemistry , Ultrasonography/methods , Escherichia coli , Microscopy, Electron, Transmission
7.
PLoS One ; 12(5): e0176958, 2017.
Article in English | MEDLINE | ID: mdl-28472168

ABSTRACT

Prostate specific membrane antigen (PSMA) targeted microbubbles (MBs) were developed using bioorthogonal chemistry. Streptavidin-labeled MBs were treated with a biotinylated tetrazine (MBTz) and targeted to PSMA expressing cells using trans-cyclooctene (TCO)-functionalized anti-PSMA antibodies (TCO-anti-PSMA). The extent of MB binding to PSMA positive cells for two different targeting strategies was determined using an in vitro flow chamber. The initial approach involved pretargeting, where TCO-anti-PSMA was first incubated with PSMA expressing cells and followed by MBTz, which subsequently showed a 2.8 fold increase in the number of bound MBs compared to experiments performed in the absence of TCO-anti-PSMA. Using direct targeting, where TCO-anti-PSMA was linked to MBTz prior to initiation of the assay, a 5-fold increase in binding compared to controls was observed. The direct targeting approach was subsequently evaluated in vivo using a human xenograft tumor model and two different PSMA-targeting antibodies. The US signal enhancements observed were 1.6- and 5.9-fold greater than that for non-targeted MBs. The lead construct was also evaluated in a head-to-head study using mice bearing both PSMA positive or negative tumors in separate limbs. The human PSMA expressing tumors exhibited a 2-fold higher US signal compared to those tumors deficient in human PSMA. The results demonstrate both the feasibility of preparing PSMA-targeted MBs and the benefits of using bioorthogonal chemistry to create targeted US probes.


Subject(s)
Antigens, Surface/metabolism , Glutamate Carboxypeptidase II/metabolism , Microbubbles , Prostatic Neoplasms/immunology , Ultrasonics , Animals , Antibodies/immunology , Antigens, Surface/immunology , Glutamate Carboxypeptidase II/immunology , Heterografts , Humans , Male , Mice , Mice, Nude , Prostatic Neoplasms/pathology
8.
Ultrasound Med Biol ; 43(5): 1016-1030, 2017 05.
Article in English | MEDLINE | ID: mdl-28258771

ABSTRACT

Gas vesicles (GVs) are a new and unique class of biologically derived ultrasound contrast agents with sub-micron size whose acoustic properties have not been fully elucidated. In this study, we investigated the acoustic collapse pressure and behavior of Halobacterium salinarum gas vesicles at transmit center frequencies ranging from 12.5 to 27.5 MHz. The acoustic collapse pressure was found to be above 550 kPa at all frequencies, nine-fold higher than the critical pressure observed under hydrostatic conditions. We illustrate that gas vesicles behave non-linearly when exposed to ultrasound at incident pressure ranging from 160 kPa to the collapse pressure and generate second harmonic amplitudes of -2 to -6 dB below the fundamental in media with viscosities ranging from 0.89 to 8 mPa·s. Simulations performed using a Rayleigh-Plesset-type model accounting for buckling and a dynamic finite-element analysis suggest that buckling is the mechanism behind the generation of harmonics. We found good agreement between the level of second harmonic relative to the fundamental measured at 20 MHz and the Rayleigh-Plesset model predictions. Finite-element simulations extended these findings to a non-spherical geometry, confirmed that the acoustic buckling pressure corresponds to the critical pressure under hydrostatic conditions and support the hypothesis of limited gas flow across the GV shell during the compression phase in the frequency range investigated. From simulations, estimates of GV bandwidth-limited scattering indicate that a single GV has a scattering cross section comparable to that of a red blood cell. These findings will inform the development of GV-based contrast agents and pulse sequences to optimize their detection with ultrasound.


Subject(s)
Contrast Media , Halobacterium salinarum , Ultrasonic Waves , Computer Simulation , Microbubbles , Models, Theoretical , Pressure
9.
Ultrasound Med Biol ; 43(2): 531-540, 2017 02.
Article in English | MEDLINE | ID: mdl-27727022

ABSTRACT

Phase-shift perfluorocarbon droplets have been investigated for over 20 years as pre-clinical ultrasound contrast agents with distinctive advantages in imaging and therapy. A number of formulation strategies exist, each with inherent advantages and limitations. In this note, we demonstrate a unique opportunity: that phase-shift droplets can be generated directly from commercially available microbubbles. This may facilitate pre-clinical and translational development by reducing the in-house synthesis expertise and resources required to generate high concentration droplet emulsions. Proof-of-principle in vitro and in vivo is given using droplets created from Definity and MicroMarker. The results demonstrate the role of perfluorocarbon choice in the trade-off between thermal stability and vaporization threshold, and suggest that commercial microbubbles with decafluorobutane cores may be ideal for this approach.


Subject(s)
Contrast Media/chemistry , Fluorocarbons/chemistry , Image Enhancement/methods , Microbubbles , Ultrasonography/methods , Animals , Liver/diagnostic imaging , Mice , Mice, Inbred C3H , Models, Animal , Phantoms, Imaging
10.
Cancer Res ; 76(15): 4493-503, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27325647

ABSTRACT

VEGF pathway-targeting antiangiogenic drugs, such as bevacizumab, when combined with chemotherapy have changed clinical practice for the treatment of a broad spectrum of human cancers. However, adaptive resistance often develops, and one major mechanism is elevated tumor hypoxia and upregulated hypoxia-inducible factor-1α (HIF1α) caused by antiangiogenic treatment. Reduced tumor vessel numbers and function following antiangiogenic therapy may also affect intratumoral delivery of concurrently administered chemotherapy. Nonetheless, combining chemotherapy and bevacizumab can lead to improved response rates, progression-free survival, and sometimes, overall survival, the extent of which can partly depend on the chemotherapy backbone. A rational, complementing chemotherapy partner for combination with bevacizumab would not only reduce HIF1α to overcome hypoxia-induced resistance, but also improve tumor perfusion to maintain intratumoral drug delivery. Here, we evaluated bevacizumab and CRLX101, an investigational nanoparticle-drug conjugate containing camptothecin, in preclinical mouse models of orthotopic primary triple-negative breast tumor xenografts, including a patient-derived xenograft. We also evaluated long-term efficacy of CRLX101 and bevacizumab to treat postsurgical, advanced metastatic breast cancer in mice. CRLX101 alone and combined with bevacizumab was highly efficacious, leading to complete tumor regressions, reduced metastasis, and greatly extended survival of mice with metastatic disease. Moreover, CRLX101 led to improved tumor perfusion and reduced hypoxia, as measured by contrast-enhanced ultrasound and photoacoustic imaging. CRLX101 durably suppressed HIF1α, thus potentially counteracting undesirable effects of elevated tumor hypoxia caused by bevacizumab. Our preclinical results show pairing a potent cytotoxic nanoparticle chemotherapeutic that complements and improves concurrent antiangiogenic therapy may be a promising treatment strategy for metastatic breast cancer. Cancer Res; 76(15); 4493-503. ©2016 AACR.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Bevacizumab/therapeutic use , Camptothecin/therapeutic use , Cyclodextrins/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/pharmacology , Animals , Bevacizumab/administration & dosage , Bevacizumab/pharmacology , Camptothecin/administration & dosage , Camptothecin/pharmacology , Cell Line, Tumor , Cyclodextrins/administration & dosage , Cyclodextrins/pharmacology , Female , Humans , Mice , Mice, SCID , Nanoparticles , Triple Negative Breast Neoplasms/pathology
11.
Cancer Res ; 76(15): 4320-31, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27325651

ABSTRACT

Alterations in tumor perfusion and microenvironment have been shown to be associated with aggressive cancer phenotypes, raising the need for noninvasive methods of tracking these changes. Dynamic contrast-enhanced ultrasound (DCEUS) and photoacoustic (PA) imaging serve as promising candidates-one has the ability to measure tissue perfusion, whereas the other can be used to monitor tissue oxygenation and hemoglobin concentration. In this study, we investigated the relationship between the different functional parameters measured with DCEUS and PA imaging, using two morphologically different hind-limb tumor models and drug-induced alterations in an orthotopic breast tumor model. Imaging results showed some correlation between perfusion and oxygen saturation maps and the ability to sensitively monitor antivascular treatment. In addition, DCEUS measurements revealed different vascular densities in the core of specific tumors compared with their rims. Noncorrelated perfusion and hemoglobin concentration measurements facilitated discrimination between blood lakes and necrotic areas. Taken together, our results illustrate the utility of a combined contrast-enhanced ultrasound method with photoacoustic imaging to visualize blood flow patterns in tumors. Cancer Res; 76(15); 4320-31. ©2016 AACR.


Subject(s)
Neoplasms/diagnostic imaging , Photoacoustic Techniques/methods , Ultrasonography/methods , Animals , Contrast Media , Humans , Mice , Xenograft Model Antitumor Assays
12.
J Natl Cancer Inst ; 108(8)2016 08.
Article in English | MEDLINE | ID: mdl-27059374

ABSTRACT

BACKGROUND: The anti-angiogenic Sorafenib is the only approved systemic therapy for advanced hepatocellular carcinoma (HCC). However, acquired resistance limits its efficacy. An emerging theory to explain intrinsic resistance to other anti-angiogenic drugs is 'vessel co-option,' ie, the ability of tumors to hijack the existing vasculature in organs such as the lungs or liver, thus limiting the need for sprouting angiogenesis. Vessel co-option has not been evaluated as a potential mechanism for acquired resistance to anti-angiogenic agents. METHODS: To study sorafenib resistance mechanisms, we used an orthotopic human HCC model (n = 4-11 per group), where tumor cells are tagged with a secreted protein biomarker to monitor disease burden and response to therapy. Histopathology, vessel perfusion assessed by contrast-enhanced ultrasound, and miRNA sequencing and quantitative real-time polymerase chain reaction were used to monitor changes in tumor biology. RESULTS: While sorafenib initially inhibited angiogenesis and stabilized tumor growth, no angiogenic 'rebound' effect was observed during development of resistance unless therapy was stopped. Instead, resistant tumors became more locally infiltrative, which facilitated extensive incorporation of liver parenchyma and the co-option of liver-associated vessels. Up to 75% (±10.9%) of total vessels were provided by vessel co-option in resistant tumors relative to 23.3% (±10.3%) in untreated controls. miRNA sequencing implicated pro-invasive signaling and epithelial-to-mesenchymal-like transition during resistance development while functional imaging further supported a shift from angiogenesis to vessel co-option. CONCLUSIONS: This is the first documentation of vessel co-option as a mechanism of acquired resistance to anti-angiogenic therapy and could have important implications including the potential therapeutic benefits of targeting vessel co-option in conjunction with vascular endothelial growth factor receptor signaling.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/blood supply , Drug Resistance, Neoplasm , Liver Neoplasms/blood supply , Liver/blood supply , Neovascularization, Pathologic/metabolism , Niacinamide/analogs & derivatives , Phenylurea Compounds/therapeutic use , Actins/metabolism , Animals , Antigens, CD34/metabolism , Blood Vessels/diagnostic imaging , Blood Vessels/pathology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Contrast Media , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition/genetics , Homeodomain Proteins/genetics , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Mice , Mice, SCID , MicroRNAs/analysis , Neoplasm Invasiveness , Neoplasm Transplantation , Neovascularization, Pathologic/diagnostic imaging , Neovascularization, Pathologic/prevention & control , Niacinamide/therapeutic use , Osteopontin/blood , Repressor Proteins/genetics , Sequence Analysis, RNA , Signal Transduction/genetics , Sorafenib , Ultrasonography , Up-Regulation , Vascular Endothelial Growth Factor A/blood , Vimentin/genetics , Zinc Finger E-box Binding Homeobox 2 , Zinc Finger E-box-Binding Homeobox 1/genetics
13.
IEEE Trans Biomed Eng ; 62(8): 1969-80, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25730824

ABSTRACT

BACKGROUND: Speckle noise is an inherent characteristic of dynamic contrast-enhanced ultrasound (DCEUS) movies and ultrasound images in general. Speckle noise considerably reduces the quality of these images and limits their clinical use. Currently, temporal compounding and maximum intensity persistence (MIP) are among the most widely accepted processing methods enabling the visualization of vasculature using DCEUS. GOAL: A different approach has been used in this study, in order to improve the noise removal, while enabling the investigation of CEUS dynamics. METHODS: A multiplicative model for the formation of DCEUS speckled images is adopted and the log-transformed cines are processed. A preprocessing step was performed, locally removing low value outliers. Due to the fast-changing spatial distribution of microbubbles inside the vasculature, the noise in consecutive DCEUS frames is independent, facilitating its removal by temporal denoising. Noise reduction is efficiently achieved by wavelet denoising, in which the signal's wavelet coefficients are thresholded and small-value noise-related coefficients are discarded. The main advantage of using wavelet denoising in the present context is its ability to estimate ultrasound contrast agents' (UCA) concentration over time adaptively, without assuming a model or predefining the signal's degree of smoothness. The performance of wavelet denoising was compared against MIP, temporal compounding, and Log-normal model fitting. RESULTS: Phantom experiments showed improved SNR, using wavelet denoising over a wide range of UCA concentrations (MicroMarker, 0.001-1%). In the in vivo tests, improved noise removal was achieved, reflected by a significantly lower coefficient of variation in homogeneous vascular regions (p < 0.01).


Subject(s)
Contrast Media/chemistry , Image Processing, Computer-Assisted/methods , Ultrasonography/methods , Animals , Female , Mice , Mice, Nude , Microbubbles , Models, Theoretical , Phantoms, Imaging , Reproducibility of Results , Wavelet Analysis
14.
Nat Nanotechnol ; 9(4): 311-6, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24633522

ABSTRACT

Ultrasound is among the most widely used non-invasive imaging modalities in biomedicine, but plays a surprisingly small role in molecular imaging due to a lack of suitable molecular reporters on the nanoscale. Here, we introduce a new class of reporters for ultrasound based on genetically encoded gas nanostructures from microorganisms, including bacteria and archaea. Gas vesicles are gas-filled protein-shelled compartments with typical widths of 45-250 nm and lengths of 100-600 nm that exclude water and are permeable to gas. We show that gas vesicles produce stable ultrasound contrast that is readily detected in vitro and in vivo, that their genetically encoded physical properties enable multiple modes of imaging, and that contrast enhancement through aggregation permits their use as molecular biosensors.


Subject(s)
Biosensing Techniques/methods , Contrast Media/chemistry , Gases/chemistry , Nanoparticles/chemistry , Ultrasonography/methods
15.
Angiogenesis ; 17(3): 641-59, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24510304

ABSTRACT

Chronic intestinal inflammation is associated with pathological angiogenesis that further amplifies the inflammatory response. Vascular endothelial growth factor (VEGF), is a major angiogenic cytokine that has been implicated in chronic colitis and inflammatory bowel diseases. Endoglin (CD105), a transforming growth factor-ß superfamily co-receptor expressed on endothelial and some myeloid cells, is a modulator of angiogenesis involved in wound healing and potentially in resolution of inflammation. We showed previously that Endoglin heterozygous (Eng (+/-)) mice subjected to dextran sodium sulfate developed severe colitis, abnormal colonic vessels and high tissue VEGF. We therefore tested in the current study if treatment with a monoclonal antibody to VEGF could ameliorate chronic colitis in Eng (+/-) mice. Tissue inflammation and microvessel density (MVD) were quantified on histological slides. Colonic wall thickness, microvascular hemodynamics and targeted MAdCAM-1(+) inflamed vessels were assessed in vivo by ultrasound. Mediators of angiogenesis and inflammation were measured by Milliplex and ELISA assays. Colitic Eng (+/-) mice showed an increase in intestinal inflammation, MVD, colonic wall thickness, microvascular hemodynamics and the number of MAdCAM-1(+) microvessels relative to colitic Eng (+/+) mice; these parameters were all attenuated by anti-VEGF treatment. Of all factors up-regulated in the inflamed gut, granulocyte colony-stimulating factor (G-CSF) and amphiregulin were further increased in colitic Eng (+/-) versus Eng (+/+) mice. Anti-VEGF therapy decreased tissue VEGF and inflammation-induced endoglin, IL-1ß and G-CSF in colitic Eng (+/-) mice. Our results suggest that endoglin modulates intestinal angiogenic and inflammatory responses in colitis. Furthermore, contrast-enhanced ultrasound provides an excellent non-invasive imaging modality to monitor gut angiogenesis, inflammation and responses to anti-angiogenic treatment.


Subject(s)
Colitis/drug therapy , Inflammation/drug therapy , Intestines/pathology , Intracellular Signaling Peptides and Proteins/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Colitis/pathology , Colon/drug effects , Colon/pathology , Endoglin , Female , Granulocyte Colony-Stimulating Factor/metabolism , Hemodynamics/drug effects , Heterozygote , Inflammation/physiopathology , Inflammation Mediators/metabolism , Interleukin-1beta/metabolism , Intestines/drug effects , Intestines/physiopathology , Intracellular Signaling Peptides and Proteins/deficiency , Male , Mice, Inbred C57BL , Microvessels/drug effects , Microvessels/pathology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism , Vascular Endothelial Growth Factor A/metabolism
16.
Angiogenesis ; 17(1): 129-46, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24061911

ABSTRACT

Hereditary hemorrhagic telangiectasia (HHT) is a vascular dysplasia associated with dysregulated angiogenesis and arteriovascular malformations. The disease is caused by mutations in endoglin (ENG; HHT1) or activin receptor-like kinase 1 (ALK1; HHT2) genes, coding for transforming growth factor ß (TGF-ß) superfamily receptors. Vascular endothelial growth factor (VEGF) has been implicated in HHT and beneficial effects of anti-VEGF treatment were recently reported in HHT patients. To investigate the systemic angiogenic phenotype of Endoglin and Alk1 mutant mice and their response to anti-VEGF therapy, we assessed microvessel density (MVD) in multiple organs after treatment with an antibody to mouse VEGF or vehicle. Lungs were the only organ showing an angiogenic defect, with reduced peripheral MVD and secondary right ventricular hypertrophy (RVH), yet distinctly associated with a fourfold increase in thrombospondin-1 (TSP-1) in Eng (+/-) versus a rise in angiopoietin-2 (Ang-2) in Alk1 (+/-) mice. Anti-VEGF treatment did reduce lung VEGF levels but interestingly, led to an increase in peripheral pulmonary MVD and attenuation of RVH; it also normalized TSP-1 and Ang-2 expression. Hepatic MVD, unaffected in mutant mice, was reduced by anti-VEGF therapy in heterozygous and wild type mice, indicating a liver-specific effect of treatment. Contrast-enhanced micro-ultrasound demonstrated a reduction in hepatic microvascular perfusion after anti-VEGF treatment only in Eng (+/-) mice. Our findings indicate that the mechanisms responsible for the angiogenic imbalance and the response to anti-VEGF therapy differ between Eng and Alk1 heterozygous mice and raise the need for systemic monitoring of anti-angiogenic therapy effects in HHT patients.


Subject(s)
Activin Receptors, Type I/metabolism , Antibodies, Monoclonal/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Liver , Lung , Neovascularization, Pathologic/drug therapy , Telangiectasia, Hereditary Hemorrhagic/drug therapy , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Activin Receptors, Type I/genetics , Activin Receptors, Type II , Animals , Endoglin , Heterozygote , Intracellular Signaling Peptides and Proteins/genetics , Liver/blood supply , Liver/metabolism , Liver/pathology , Lung/blood supply , Lung/metabolism , Lung/pathology , Mice , Mice, Mutant Strains , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Ribonuclease, Pancreatic/genetics , Ribonuclease, Pancreatic/metabolism , Telangiectasia, Hereditary Hemorrhagic/genetics , Telangiectasia, Hereditary Hemorrhagic/metabolism , Telangiectasia, Hereditary Hemorrhagic/pathology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
17.
Article in English | MEDLINE | ID: mdl-23661123

ABSTRACT

Photoacoustic (PA) imaging for biomedical applications has been under development for many years. Based on the many advances over the past decade, a new photoacoustic imaging system has been integrated into a micro-ultrasound platform for co-registered PA-ultrasound (US) imaging. The design and implementation of the new scanner is described and its performance quantified. Beamforming techniques and signal processing are described, in conjunction with in vivo PA images of normal subcutaneous mouse tissue and selected tumor models. In particular, the use of the system to estimate the spatial distribution of oxygen saturation (sO2) in blood and co-registered with B-mode images of the surrounding anatomy are investigated. The system was validated in vivo against a complementary technique for measuring partial pressure of oxygen in blood (pO2). The pO2 estimates were converted to sO2 values based on a standard dissociation curve found in the literature. Preliminary studies of oxygenation effects were performed in a mouse model of breast cancer (MDA-MB-231) in which control mice were compared with mice treated with a targeted antiangiogenic agent over a 3 d period. Treated mice exhibited a >90% decrease in blood volume, an 85% reduction in blood wash-in rate, and a 60% decrease in relative tissue oxygenation.


Subject(s)
Microtechnology/instrumentation , Photoacoustic Techniques/methods , Ultrasonography/methods , Animals , Antineoplastic Agents/therapeutic use , Female , Fiber Optic Technology , Mammary Glands, Animal/blood supply , Mammary Glands, Animal/diagnostic imaging , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/diagnostic imaging , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Mice , Oxygen/blood , Oxyhemoglobins/analysis , Photoacoustic Techniques/instrumentation , Regional Blood Flow/physiology , Ultrasonography/instrumentation
18.
Mol Cancer Ther ; 11(3): 680-9, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22188817

ABSTRACT

Metronomic chemotherapy refers to the close, regular administration of conventional chemotherapy drugs at relatively low, minimally toxic doses, with no prolonged break periods; it is now showing encouraging results in various phase II clinical trials and is currently undergoing phase III trial evaluation. It is thought to cause antitumor effects primarily by antiangiogenic mechanisms, both locally by targeting endothelial cells of the tumor neovasculature and systemically by effects on bone marrow-derived cells, including circulating endothelial progenitor cells (CEP). Previous studies have shown reduction of CEPs by metronomic administration of a number of different chemotherapeutic drugs, including vinblastine, cyclophosphamide, paclitaxel, topotecan, and tegafur plus uracil (UFT). However in addition to, or even instead of, antiangiogenic effects, metronomic chemotherapy may cause suppression of tumor growth by other mechanisms such as stimulating cytotoxic T-cell responses or by direct antitumor effects. Here we report results evaluating the properties of metronomic administration of an oral prodrug of gemcitabine LY2334737 in nontumor-bearing mice and in preclinical models of human ovarian (SKOV3-13) and breast cancer (LM2-4) xenografts. Through daily gavage (at 6 mg/kg/d), the schedules tested were devoid of toxicity and caused antitumor effects; however, a suppressive effect on CEPs was not detected. Unexpectedly, metronomic LY2334737 administration caused increased blood flow in luciferase-tagged LM2-4 tumor xenografts, and this effect, readily measured using contrast micro-ultrasound, coincided with a relative increase in tumor bioluminescence. These results highlight the possibility of significant antitumor effects mediated by metronomic administration of some chemotherapy drugs without a concomitant inhibition of systemic angiogenesis.


Subject(s)
Deoxycytidine/analogs & derivatives , Neoplasms/drug therapy , Prodrugs/pharmacology , Administration, Oral , Animals , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/pharmacology , Blood Vessels/drug effects , Blood Vessels/metabolism , Blood Vessels/pathology , Cell Line, Tumor , Deoxycytidine/administration & dosage , Deoxycytidine/pharmacology , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Humans , Mice , Mice, Inbred BALB C , Mice, SCID , Neoplasms/blood supply , Neoplasms/pathology , Neovascularization, Pathologic/prevention & control , Prodrugs/administration & dosage , Tumor Burden/drug effects , Xenograft Model Antitumor Assays , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL
...