Network


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

Hotspot


Dive into the research topics where Reut Avni is active.

Publication


Featured researches published by Reut Avni.


NMR in Biomedicine | 2011

Hypoxic stress and cancer: Imaging the axis of evil in tumor metastasis

Reut Avni; Batya Cohen; Michal Neeman

Tumors emerge as a result of the sequential acquisition of genetic, epigenetic and somatic alterations promoting cell proliferation and survival. The maintenance and expansion of tumor cells rely on their ability to adapt to changes in their microenvironment, together with the acquisition of the ability to remodel their surroundings. Tumor cells interact with two types of interconnected microenvironments: the metabolic cell autonomous microenvironment and the nonautonomous cellular–molecular microenvironment comprising interactions between tumor cells and the surrounding stroma. Hypoxia is a central player in cancer progression, affecting not only tumor cell autonomous functions, such as cell division and invasion, resistance to therapy and genetic instability, but also nonautonomous processes, such as angiogenesis, lymphangiogenesis and inflammation, all contributing to metastasis. Closely related microenvironmental stressors affecting cancer progression include, in addition to hypoxia, elevated interstitial pressure and oxidative stress. Noninvasive imaging offers multiple means to monitor the tumor microenvironment and its consequences, and can thus assist in the understanding of the biological basis of hypoxia and microenvironmental stress in cancer progression, and in the development of strategies to monitor therapies targeted at stress‐induced tumor progression. Copyright


Proceedings of the National Academy of Sciences of the United States of America | 2014

Major mouse placental compartments revealed by diffusion-weighted MRI, contrast-enhanced MRI, and fluorescence imaging.

Eddy Solomon; Reut Avni; Ron Hadas; Tal Raz; Joel R. Garbow; Peter Bendel; Lucio Frydman; Michal Neeman

Significance Fluid motion measurements can provide valuable insight regarding the structure and function of developing placentas. This study presents to our knowledge the first MRI characterization of multicompartmental diffusion and incoherent flow in pregnant mice at different gestation stages, made possible by methods herein introduced for the single-scan acquisition of diffusion-encoded images in the challenging environment associated with in vivo embryonic studies. These methods were combined with a customized contrast agent to reveal a freely diffusive maternal blood pool, a strongly perfused fetal blood flow, and an intermediate behavior for the trophoblastic labyrinth cell layer. Structural features associated with these dynamics were corroborated with ex vivo fluorescence microscopy and are discussed within the context of the anatomical structure of developing mouse placentas. Mammalian models, and mouse studies in particular, play a central role in our understanding of placental development. Magnetic resonance imaging (MRI) could be a valuable tool to further these studies, providing both structural and functional information. As fluid dynamics throughout the placenta are driven by a variety of flow and diffusion processes, diffusion-weighted MRI could enhance our understanding of the exchange properties of maternal and fetal blood pools—and thereby of placental function. These studies, however, have so far been hindered by the small sizes, the unavoidable motions, and the challenging air/water/fat heterogeneities, associated with mouse placental environments. The present study demonstrates that emerging methods based on the spatiotemporal encoding (SPEN) of the MRI information can robustly overcome these obstacles. Using SPEN MRI in combination with albumin-based contrast agents, we analyzed the diffusion behavior of developing placentas in a cohort of mice. These studies successfully discriminated the maternal from the fetal blood flows; the two orders of magnitude differences measured in these fluids’ apparent diffusion coefficients suggest a nearly free diffusion behavior for the former and a strong flow-based component for the latter. An intermediate behavior was observed by these methods for a third compartment that, based on maternal albumin endocytosis, was associated with trophoblastic cells in the interphase labyrinth. Structural features associated with these dynamic measurements were consistent with independent intravital and ex vivo fluorescence microscopy studies and are discussed within the context of the anatomy of developing mouse placentas.


Magnetic Resonance in Medicine | 2012

Unique in utero identification of fetuses in multifetal mouse pregnancies by placental bidirectional arterial spin labeling MRI

Reut Avni; Tal Raz; Inbal E. Biton; Vyacheslav Kalchenko; Joel R. Garbow; Michal Neeman

Noninvasive imaging is a critical part of the study of developing embryos/fetuses, particularly in the context of alterations of gene expression in genetically modified animals. However, in litter‐bearing animals, such as mice, the inability to accurately identify individual embryo/fetus in utero is a major obstacle to longitudinal, noninvasive in vivo studies. Arterial spin labeling MRI was adopted here to determine the fetal order along the uterine horns in vivo, based on the specific pattern of dual arterial blood supply within the mouse uterine horns. Blood enters the mouse uterus cranially through the ovarian artery and caudally through the uterine artery. Saturation slices were alternately placed on the maternal heart or on the bifurcation point of the common iliac artery, thereby saturating either downward inflow via the ovarian arteries or upward inflow via the uterine arteries, respectively. Saturation maps provided a unique signature with highly significant correlation between the direction‐dependent magnetization transfer and the position of the fetuses/placentas along the uterine horns. The bidirectional arterial spin labeling‐MRI method reported here opens possibilities to determine and pursue phenotypic alterations in fetuses and placentas in longitudinal studies of transgenic and knockout mice models, and for studying defects in placental vascular architecture. Magn Reson Med, 2012.


Placenta | 2015

Functional MRI of the placenta – From rodents to humans

Reut Avni; Michal Neeman; Joel R. Garbow

The placenta performs a wide range of physiological functions; insufficiencies in these functions may result in a variety of severe prenatal and postnatal syndromes with long-term negative impacts on human adult health. Recent advances in magnetic resonance imaging (MRI) studies of placental function, in both animal models and humans, have contributed significantly to our understanding of placental structure, blood flow, oxygenation status, and metabolic profile, and have provided important insights into pregnancy complications.


Contrast Media & Molecular Imaging | 2010

Novel MRI and fluorescent probes responsive to the Factor XIII transglutaminase activity

Lorenzo Tei; Galit Mazooz; Yael Shellef; Reut Avni; Katrien Vandoorne; Alessandro Barge; Vyacheslav Kalchenko; Mark W. Dewhirst; Linda Chaabane; Luigi Miragoli; Dario Livio Longo; Michal Neeman; Silvio Aime

Transglutaminases, including factor XIII and tissue transglutaminase, participate in multiple extracellular processes associated with remodeling of the extracellular matrix during wound repair, blood clotting, tumor progression and fibrosis of ischemic injuries. The aim of this work was to evaluate a novel substrate analog for transglutaminase optimized by molecular modeling calculations (DCCP16), which can serve for molecular imaging of transglutaminase activity by magnetic resonance imaging and by near-infrared imaging. Experimental data showed covalent binding of Gd-DCCP16 and DCCP16-IRIS Blue to human clots, to basement membrane components and to casein in purified systems as well as in three-dimensional multicellular spheroids. In vivo, DCCP16 showed enhancement with a prolonged retention in clots and tumors, demonstrating the ability to detect both factor XIII and tissue transglutaminase mediated covalent binding of the contrast material.


PLOS ONE | 2012

The hemodynamic basis for positional- and inter-fetal dependent effects in dual arterial supply of mouse pregnancies.

Tal Raz; Reut Avni; Yoseph Addadi; Yoni Cohen; Ariel J. Jaffa; Brian A. Hemmings; Joel R. Garbow; Michal Neeman

In mammalian pregnancy, maternal cardiovascular adaptations must match the requirements of the growing fetus(es), and respond to physiologic and pathologic conditions. Such adaptations are particularly demanding for mammals bearing large-litter pregnancies, with their inherent conflict between the interests of each individual fetus and the welfare of the entire progeny. The mouse is the most common animal model used to study development and genetics, as well as pregnancy-related diseases. Previous studies suggested that in mice, maternal blood flow to the placentas occurs via a single arterial uterine loop generated by arterial-arterial anastomosis of the uterine artery to the uterine branch of the ovarian artery, resulting in counter bi-directional blood flow. However, we provide here experimental evidence that each placenta is actually supplied by two distinct arterial inputs stemming from the uterine artery and from the uterine branch of the ovarian artery, with position-dependent contribution of flow from each source. Moreover, we report significant positional- and inter-fetal dependent alteration of placental perfusion, which were detected by in vivo MRI and fluorescence imaging. Maternal blood flow to the placentas was dependent on litter size and was attenuated for placentas located centrally along the uterine horn. Distinctive apposing, inter-fetal hemodynamic effects of either reduced or elevated maternal blood flow, were measured for placenta of normal fetuses that are positioned adjacent to either pathological, or to hypovascular Akt1-deficient placentas, respectively. The results reported here underscore the critical importance of confounding local and systemic in utero effects on phenotype presentation, in general and in the setting of genetically modified mice. The unique robustness and plasticity of the uterine vasculature architecture, as reported in this study, can explain the ability to accommodate varying litter sizes, sustain large-litter pregnancies and overcome pathologic challenges. Remarkably, the dual arterial supply is evolutionary conserved in mammals bearing a single offspring, including primates.


Radiology | 2016

MR imaging–derived oxygen-hemoglobin dissociation curves and fetal-placental oxygen-hemoglobin affinities

Reut Avni; Ofra Golani; Ayelet Akselrod-Ballin; Yonni Cohen; Inbal E. Biton; Joel R. Garbow; Michael Neeman

The authors of this study present a noninvasive approach for obtaining MR imaging–based oxygen-hemoglobin dissociation curves and for deriving oxygen tension values at which hemoglobin is 50% saturated and maps for the placenta and fetus in pregnant mice.


Fertility and Sterility | 2014

In search of signaling pathways critical for ovarian graft reception: Akt1 is essential for long-term survival of ovarian grafts

Yoni Cohen; Hagit Dafni; Reut Avni; Tal Raz; Inbal E. Biton; Brian A. Hemmings; Michal Neeman

OBJECTIVE To explore the role of Akt1, a principle modulator of angiogenesis, in ovarian graft reception and to investigate whether Akt1 deficiency can alter ovarian graft reception. DESIGN Experimental mouse model. SETTING Research institute. ANIMAL(S) Donors: Akt1 knockout (Akt1(-/-)) and wild types (Akt1(+/+)) mice. Recipients: CD-1 nude immune deficient female mice. INTERVENTION(S) Ovaries from Akt1(-/-) and Akt1(+/+) mice transplanted in the biceps femoris muscle of immunocompromised CD-1 mice, and ovarian graft viability, perfusion, and revascularization explored in vivo by magnetic resonance imaging (MRI). MAIN OUTCOME MEASURE(S) Vascular density and permeability of newly formed graft blood vessels quantified by dynamic contrast-enhanced MRI 7, 14, 30, and 60 days after grafting as indicators for angiogenesis and reestablishment of blood perfusion. RESULT(S) The Akt1(-/-) ovarian grafts showed a gradual decrease in angiogenic response with time after transplantation, ultimately leading to complete or near-complete graft destruction coinciding with massive follicular loss. Sixty days after transplantation, the mean blood volume fraction (fBV) and vessel permeability (PS) were statistically significantly lower in Akt1(-/-) transplants compared with Akt1(+/+). CONCLUSION(S) Akt1 is essential for ovarian graft reception. However, surprisingly the impact of Akt1 deficiency was most profound not in the early stages of angiogenesis but rather in long-term survival of the graft.


Scientific Reports | 2016

Multimodal Correlative Preclinical Whole Body Imaging and Segmentation

Ayelet Akselrod-Ballin; Hagit Dafni; Yoseph Addadi; Inbal E. Biton; Reut Avni; Yafit Brenner; Michal Neeman

Segmentation of anatomical structures and particularly abdominal organs is a fundamental problem for quantitative image analysis in preclinical research. This paper presents a novel approach for whole body segmentation of small animals in a multimodal setting of MR, CT and optical imaging. The algorithm integrates multiple imaging sequences into a machine learning framework, which generates supervoxels by an efficient hierarchical agglomerative strategy and utilizes multiple SVM-kNN classifiers each constrained by a heatmap prior region to compose the segmentation. We demonstrate results showing segmentation of mice images into several structures including the heart, lungs, liver, kidneys, stomach, vena cava, bladder, tumor, and skeleton structures. Experimental validation on a large set of mice and organs, indicated that our system outperforms alternative state of the art approaches. The system proposed can be generalized to various tissues and imaging modalities to produce automatic atlas-free segmentation, thereby enabling a wide range of applications in preclinical studies of small animal imaging.


Biology of Reproduction | 2016

Genetic and Pharmacological Modulation of Akt1 for Improving Ovarian Graft Revascularization in a Mouse Model

Yoni Cohen; Hagit Dafni; Reut Avni; Liat Fellus; Filip Bochner; Ron Rotkopf; Tal Raz; Laura E. Benjamin; Kenneth Walsh; Michal Neeman

ABSTRACT Ovarian tissue cryopreservation and transplantation is one of a few available treatments for fertility preservation in women diagnosed with cancer. Rapid revascularization is essential for reducing hypoxic damage after grafting and protecting the primordial follicles reserve. Using a mouse model of heterotopic ovarian graft transplantation, we have delineated the role of endothelial Akt1 expression using longitudinal magnetic resonance imaging follow-up to quantify angiogenic response. Endothelial Akt1 activation in ovarian grafts promoted angiogenesis to support the graft during posttransplantation hypoxic period. Similarly, simvastatin therapy activated Akt1 at the transplantation site and improved the revascularization and vascular support of ovarian grafts. These results serve as an important first step toward pharmacological intervention to improve revascularization of ovarian grafts and restoration of fertility in cancer survivors. The pro-angiogenic effects reported here may extend beyond improving ovarian graft reception in fertility preservation and could potentially be used for different organ or tissue transplantation.

Collaboration


Dive into the Reut Avni's collaboration.

Top Co-Authors

Avatar

Michal Neeman

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Tal Raz

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Joel R. Garbow

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Hagit Dafni

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Yoni Cohen

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Inbal E. Biton

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Liat Fellus

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Filip Bochner

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Lucio Frydman

Weizmann Institute of Science

View shared research outputs
Researchain Logo
Decentralizing Knowledge