36 results on '"Cooper, Itzik"'
Search Results
2. BBB opening by low pulsed electric fields, depicted by delayed-contrast MRI, enables efficient delivery of therapeutic doxorubicin doses into mice brains.
- Author
-
Cooper, Itzik, Last, David, Ravid, Orly, Rand, Daniel, Matsree, Erez, Omesi, Liora, Shemesh, Chen, Liberman, Meir, Zach, Leor, Furman, Orit, Daniels, Dianne, Liraz-Zaltsman, Sigal, Mardor, Yael, and Sharabi, Shirley
- Subjects
ELECTRIC fields ,BRAIN tumors ,CONTRAST-enhanced magnetic resonance imaging ,DRUG delivery systems ,TUMOR growth ,DOXORUBICIN - Abstract
Background: Pharmacological treatment of CNS diseases is limited due to the presence of the blood-brain barrier (BBB). Recent years showed significant advancement in the field of CNS drug delivery enablers, with technologies such as MR-guided focused ultrasound reaching clinical trials. This have inspired researchers in the field to invent novel brain barriers opening (BBo) technologies that are required to be simple, fast, safe and efficient. One such technology, recently developed by us, is BDF (Barrier Disrupting Fields), based on low pulsed electric fields (L-PEFs) for opening the BBB in a controlled, safe, reversible and non-invasive manner. Here, we conducted an in vivo study to show that BDF is a feasible technology for delivering Doxorubicin (Doxo) into mice brain. Means for depicting BBBo levels were developed and applied for monitoring the treatment and predicting response. Overall, the goals of the presented study were to demonstrate the feasibility for delivering therapeutic Doxo doses into naïve and tumor-bearing mice brains and applying delayed–contrast MRI (DCM) for monitoring the levels of BBBo. Methods: L-PEFs were applied using plate electrodes placed on the intact skull of naïve mice. L-PEFs/Sham mice were scanned immediately after the procedure by DCM ("MRI experiment"), or injected with Doxo and Trypan blue followed by delayed (4 h) perfusion and brain extraction ("Doxo experiment"). Doxo concentrations were measured in brain samples using confocal microscopy and compared to IC
50 of Doxo in glioma cell lines in vitro. In order to map BBBo extent throughout the brain, pixel by pixel MR image analysis was performed using the DCM data. Finally, the efficacy of L-PEFs in combination with Doxo was tested in nude mice bearing intracranial human glioma tumors. Results: Significant amount of Doxo was found in cortical regions of all L-PEFs-treated mice brains (0.50 ± 0.06 µg Doxo/gr brain) while in Sham brains, Doxo concentrations were below or on the verge of detection limit (0.03 ± 0.02 µg Doxo/gr brain). This concentration was x97 higher than IC50 of Doxo calculated in gl261 mouse glioma cells and x8 higher than IC50 of Doxo calculated in U87 human glioma cells. DCM analysis revealed significant BBBo levels in the cortical regions of L-PEFs-treated mice; the average volume of BBBo in the L-PEFs-treated mice was x29 higher than in the Sham group. The calculated BBBo levels dropped exponentially as a function of BBBo threshold, similarly to the electric fields distribution in the brain. Finally, combining non-invasive L-PEFs with Doxo significantly decreased brain tumors growth rates in nude mice. Conclusions: Our results demonstrate significant BBBo levels induced by extra-cranial L-PEFs, enabling efficient delivery of therapeutic Doxo doses into the brain and reducing tumor growth. As BBBo was undetectable by standard contrast-enhanced MRI, DCM was applied to generate maps depicting the BBBo levels throughout the brain. These findings suggest that BDF is a promising technology for efficient drug delivery into the brain with important implications for future treatment of brain cancer and additional CNS diseases. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
3. Definition of the contribution of an Osteopontin-producing CD11c+ microglial subset to Alzheimer's disease.
- Author
-
Yiguo Qiu, Xianli Shen, Ravid, Orly, Atrakchi, Dana, Rand, Daniel, Wight, Andrew E., Hye-Jung Kim, Liraz-Zaltsman, Sigal, Cooper, Itzik, Beeri, Michal Schnaider, and Cantor, Harvey
- Subjects
ALZHEIMER'S disease ,MICROGLIA ,NEURONS ,PUBLIC health ,OSTEOPONTIN - Abstract
Alzheimer's disease (AD) is the most common form of incurable dementia and represents a critical public health issue as the world's population ages. Although microglial dysregulation is a cardinal feature of AD, the extensive heterogeneity of these immunological cells in the brain has impeded our understanding of their contribution to this disease. Here, we identify a pathogenic microglial subset which expresses the CD11c surface marker as the sole producer of Osteopontin (OPN) in the 5XFAD mouse model of AD. OPN production divides Disease-Associated Microglia (DAM) into two functionally distinct subsets, i.e., a protective CD11c
+ OPN- subset that robustly ingests amyloid β (Aβ) in a noninflammatory fashion and a pathogenic CD11c+ OPN+ subset that produces proinflammatory cytokines and fails to ingest significant amounts of Aβ. Genetic ablation of OPN or administration of monoclonal anti-OPN antibody to 5XFAD mice reduces proinflammatory microglia, plaque formation, and numbers of dystrophic neurites and results in improved cognitive function. Analysis of brain tissue from AD patients indicates that levels of OPN-producing CD11c+ microglia correlate strongly with the degree of cognitive deficit and AD neuropathology. These findings define an OPN-dependent pathway to disease driven by a distinct microglial subset, and identify OPN as a novel therapeutic target for potentially effective immunotherapy to treat AD. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
4. How Alpha Linolenic Acid May Sustain Blood–Brain Barrier Integrity and Boost Brain Resilience against Alzheimer's Disease.
- Author
-
Leikin-Frenkel, Alicia, Schnaider Beeri, Michal, and Cooper, Itzik
- Abstract
Cognitive decline, the primary clinical phenotype of Alzheimer's disease (AD), is currently attributed mainly to amyloid and tau protein deposits. However, a growing body of evidence is converging on brain lipids, and blood–brain barrier (BBB) dysfunction, as crucial players involved in AD development. The critical role of lipids metabolism in the brain and its vascular barrier, and its constant modifications particularly throughout AD development, warrants investigation of brain lipid metabolism as a high value therapeutic target. Yet, there is limited knowledge on the biochemical and structural roles of lipids in BBB functionality in AD. Within this framework, we hypothesize that the ApoE4 genotype, strongly linked to AD risk and progression, may be related to altered fatty acids composition in the BBB. Interestingly, alpha linolenic acid (ALA), the precursor of the majoritarian brain component docosahexaenoic acid (DHA), emerges as a potential novel brain savior, acting via BBB functional improvements, and this may be primarily relevant to ApoE4 carriers. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
5. Cytoskeleton Elements Contribute to Prion Peptide-Induced Endothelial Barrier Breakdown in a Blood–Brain Barrier In Vitro System.
- Author
-
Cooper, Itzik, Cohen-Kashi Malina, Katayun, Levin, Yishai, Gabashvili, Alexandra, Mohar, Boaz, Cagnotto, Alfredo, Salmona, Mario, and Teichberg, Vivian I.
- Subjects
BLOOD-brain barrier ,PRIONS ,LIQUID chromatography-mass spectrometry ,CYTOSKELETON ,PEPTIDES ,ENDOTHELIAL cells - Abstract
The mechanisms involved in the interaction of PrP 106-126, a peptide corresponding to the prion protein amyloidogenic region, with the blood–brain barrier (BBB) were studied. PrP 106-126 treatment that was previously shown to impair BBB function, reduced cAMP levels in cultured brain endothelial cells, increased nitric oxide (NO) levels, and changed the activation mode of the small GTPases Rac1 (inactivation) and RhoA (activation). The latter are well established regulators of endothelial barrier properties that act via cytoskeletal elements. Indeed, liquid chromatography-mass spectrometry (LC-MS)-based proteomic profiling study revealed extensive changes in expression of cytoskeleton-related proteins. These results shed light on the nature of the interaction between the prion peptide PrP 106-126 and the BBB and emphasize the importance of the cytoskeleton in endothelium response to prion- induced stress. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
6. Engagement of vascular early response genes typifies mild cognitive impairment.
- Author
-
Katsel, Pavel, Fam, Peter, Tan, Weilun, Khan, Sonia, Gama‐Sosa, Miguel, De Gasperi, Rita, Roussos, Panos, Robinson, Ari, Cooper, Itzik, Schnaider‐Beeri, Michal, and Haroutunian, Vahram
- Published
- 2022
- Full Text
- View/download PDF
7. The Israel National Brain Repository.The establishment and uniqueness of the first brain bank in Israel.
- Author
-
Manzali, Sigalit, Ravona‐Springer, Ramit, Heymann, Anthony, Greenbaum, Lior, Cooper, Itzik, Gardner, Raquel C., Liraz‐Zaltsman, Sigal, and Beeri, Michal S
- Published
- 2023
- Full Text
- View/download PDF
8. Naphthoquinone–Dopamine Hybrids Inhibit α‐Synuclein Aggregation, Disrupt Preformed Fibrils, and Attenuate Aggregate‐Induced Toxicity.
- Author
-
Paul, Ashim, Huber, Adi, Rand, Daniel, Gosselet, Fabien, Cooper, Itzik, Gazit, Ehud, and Segal, Daniel
- Subjects
DOPAMINE ,AMYLOID beta-protein ,SMALL molecules ,PARKINSON'S disease ,AMYLOID ,BLOOD-brain barrier ,NEURODEGENERATION - Abstract
Accumulation and aggregation of the intrinsically disordered protein α‐synuclein (α‐Syn) into amyloid fibrils are hallmarks of a series of heterogeneous neurodegenerative disorders, known as synucleinopathies and most notably Parkinson's disease (PD). The crucial role of α‐Syn aggregation in PD makes it an attractive target for the development of disease‐modifying therapeutics that would inhibit α‐Syn aggregation or disrupt its preformed fibrillar assemblies. To this end, we have designed and synthesized two naphthoquinone–dopamine‐based hybrid small molecules, NQDA and Cl‐NQDA, and demonstrated their ability to inhibit in vitro amyloid formation by α‐Syn using ThT assay, CD, TEM, and Congo red birefringence. Moreover, these hybrid molecules efficiently disassembled preformed fibrils of α‐Syn into nontoxic species, as evident from LUV leakage assay. NQDA and Cl‐NQDA were found to have low cytotoxicity and they attenuated the toxicity induced by α‐Syn towards SH‐SY5Y neuroblastoma cells. NQDA was found to efficiently cross an in vitro human blood–brain barrier model. These naphthoquinone–dopamine based derivatives can be an attractive scaffold for therapeutic design towards PD. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
9. Caspase-1 has a critical role in blood-brain barrier injury and its inhibition contributes to multifaceted repair.
- Author
-
Israelov, Hila, Ravid, Orly, Atrakchi, Dana, Rand, Daniel, Elhaik, Shirin, Bresler, Yael, Twitto-Greenberg, Rachel, Omesi, Liora, Liraz-Zaltsman, Sigal, Gosselet, Fabien, Schnaider Beeri, Michal, and Cooper, Itzik
- Subjects
BLOOD-brain barrier ,NLRP3 protein ,INFLAMMATION ,ENCEPHALITIS ,CENTRAL nervous system ,ENDOTHELIAL cells - Abstract
Background: Excessive inflammation might activate and injure the blood-brain barrier (BBB), a common feature of many central nervous system (CNS) disorders. We previously developed an in vitro BBB injury model in which the organophosphate paraoxon (PX) affects the BBB endothelium by attenuating junctional protein expression leading to weakened barrier integrity. The objective of this study was to investigate the inflammatory cellular response at the BBB to elucidate critical pathways that might lead to effective treatment in CNS pathologies in which the BBB is compromised. We hypothesized that caspase-1, a core component of the inflammasome complex, might have important role in BBB function since accumulating evidence indicates its involvement in brain inflammation and pathophysiology.Methods: An in vitro human BBB model was employed to investigate BBB functions related to inflammation, primarily adhesion and transmigration of peripheral blood mononuclear cells (PBMCs). Caspase-1 pathway was studied by measurements of its activation state and its role in PBMCs adhesion, transmigration, and BBB permeability were investigated using the specific caspase-1 inhibitor, VX-765. Expression level of adhesion and junctional molecules and the secretion of pro-inflammatory cytokines were measured in vitro and in vivo at the BBB endothelium after exposure to PX. The potential repair effect of blocking caspase-1 and downstream molecules was evaluated by immunocytochemistry, ELISA, and Nanostring technology.Results: PX affected the BBB in vitro by elevating the expression of the adhesion molecules E-selectin and ICAM-1 leading to increased adhesion of PBMCs to endothelial monolayer, followed by elevated transendothelial-migration which was ICAM-1 and LFA-1 dependent. Blocking caspase-8 and 9 rescued the viability of the endothelial cells but not the elevated transmigration of PBMCs. Inhibition of caspase-1, on the other hand, robustly restored all of barrier insults tested including PBMCs adhesion and transmigration, permeability, and VE-cadherin protein levels. The in vitro inflammatory response induced by PX and the role of caspase-1 in BBB injury were corroborated in vivo in isolated blood vessels from hippocampi of mice exposed to PX and treated with VX-765.Conclusions: These results shed light on the important role of caspase-1 in BBB insult in general and specifically in the inflamed endothelium, and suggest therapeutic potential for various CNS disorders, by targeting caspase-1 in the injured BBB. [ABSTRACT FROM AUTHOR]- Published
- 2020
- Full Text
- View/download PDF
10. Purpurin modulates Tau-derived VQIVYK fibrillization and ameliorates Alzheimer's disease-like symptoms in animal model.
- Author
-
Viswanathan, Guru Krishnakumar, Shwartz, Dana, Losev, Yelena, Arad, Elad, Shemesh, Chen, Pichinuk, Edward, Engel, Hamutal, Raveh, Avi, Jelinek, Raz, Cooper, Itzik, Gosselet, Fabien, Gazit, Ehud, and Segal, Daniel
- Subjects
TAU proteins ,SURFACE plasmon resonance ,ISOTHERMAL titration calorimetry ,NEUROFIBRILLARY tangles ,ANTHRAQUINONE derivatives ,ANIMAL models in research - Abstract
Neurofibrillary tangles of the Tau protein and plaques of the amyloid β peptide are hallmarks of Alzheimer's disease (AD), which is characterized by the conversion of monomeric proteins/peptides into misfolded β-sheet rich fibrils. Halting the fibrillation process and disrupting the existing aggregates are key challenges for AD drug development. Previously, we performed in vitro high-throughput screening for the identification of potent inhibitors of Tau aggregation using a proxy model, a highly aggregation-prone hexapeptide fragment
306 VQIVYK311 (termed PHF6) derived from Tau. Here we have characterized a hit molecule from that screen as a modulator of Tau aggregation using in vitro, in silico, and in vivo techniques. This molecule, an anthraquinone derivative named Purpurin, inhibited ~ 50% of PHF6 fibrillization in vitro at equimolar concentration and disassembled pre-formed PHF6 fibrils. In silico studies showed that Purpurin interacted with key residues of PHF6, which are responsible for maintaining its β-sheets conformation. Isothermal titration calorimetry and surface plasmon resonance experiments with PHF6 and full-length Tau (FL-Tau), respectively, indicated that Purpurin interacted with PHF6 predominantly via hydrophobic contacts and displayed a dose-dependent complexation with FL-Tau. Purpurin was non-toxic when fed to Drosophila and it significantly ameliorated the AD-related neurotoxic symptoms of transgenic flies expressing WT-FL human Tau (hTau) plausibly by inhibiting Tau accumulation and reducing Tau phosphorylation. Purpurin also reduced hTau accumulation in cell culture overexpressing hTau. Importantly, Purpurin efficiently crossed an in vitro human blood–brain barrier model. Our findings suggest that Purpurin could be a potential lead molecule for AD therapeutics. [ABSTRACT FROM AUTHOR]- Published
- 2020
- Full Text
- View/download PDF
11. The application of point source electroporation and chemotherapy for the treatment of glioma: a randomized controlled rat study.
- Author
-
Sharabi, Shirley, Guez, David, Daniels, Dianne, Cooper, Itzik, Atrakchi, Dana, Liraz-Zaltsman, Sigal, Last, David, and Mardor, Yael
- Subjects
ELECTROPORATION ,CANCER chemotherapy ,GLIOMA treatment ,RANDOMIZED controlled trials ,METHOTREXATE ,CISPLATIN ,RAT physiology - Abstract
The prognosis of Glioblastoma Multiforme patients is poor despite aggressive therapy. Reasons include poor chemotherapy penetration across the blood-brain barrier and tumor infiltration into surrounding tissues. Here we studied the effects of combined point-source electroporation (EP) and systemic chemotherapy in glioma-bearing rats. 128 rats were studied. Treatment groups were administered systemic Cisplatin/Methotrexate before EP (either 90 or 180 pulses). Control groups were treated by EP, chemotherapy, or no treatment. Tumor volumes were determined by MRI. Tumors growth rates of the EP + Methotrexate group (1.02 ± 0.77) were significantly lower (p < 0.01) than the control (5.2 ± 1.0) 1-week post treatment. No significant difference was found compared to Methotrexate (1.7 ± 0.5). Objective response rates (ORR) were 40% and 57% for the Methotrexate and EP + Methotrexate groups respectively. Tumor growth rates and ORR of the EP + Cisplatin groups (90 pulses 0.98 ± 0.2, 57%, 180 pulses 1.2 ± 0.1, 33%) were significantly smaller than the control (6.4 ± 1.0, p < 0.01, p < 0.02, 0%) and Cisplatin (3.9 ± 1.0, p < 0.04, p < 0.05, 13%) groups. No significant differences were found between the control groups. Increased survival was found in the EP + Cisplatin group, Χ
2 = 7.54, p < 0.006 (Log Rank). Point-source EP with systemic chemotherapy is a rapid, minimal-invasive treatment that was found to induce significant antineoplastic effects in a rat glioma model. [ABSTRACT FROM AUTHOR]- Published
- 2020
- Full Text
- View/download PDF
12. Transient blood–brain barrier disruption is induced by low pulsed electrical fields in vitro: an analysis of permeability and trans-endothelial electric resistivity.
- Author
-
Sharabi, Shirley, Bresler, Yael, Ravid, Orly, Shemesh, Chen, Atrakchi, Dana, Schnaider-Beeri, Michal, Gosselet, Fabien, Dehouck, Lucie, Last, David, Guez, David, Daniels, Dianne, Mardor, Yael, and Cooper, Itzik
- Subjects
ELECTRICAL resistivity ,BLOOD-brain barrier ,PERMEABILITY ,ELECTRIC impedance ,PARKINSON'S disease ,BRAIN diseases ,ELECTRIC fields ,SOIL permeability - Abstract
The blood–brain barrier (BBB) is limiting transcellular and paracellular movement of molecules and cells, controls molecular traffic, and keeps out toxins. However, this protective function is the major hurdle for treating brain diseases such as brain tumors, Parkinson's disease, Alzheimer's disease, etc. It was previously demonstrated that high pulsed electrical fields (PEFs) can disrupt the BBB by inducing electroporation (EP) which increases the permeability of the transcellular route. Our goal was to study the effects of low PEFs, well below the threshold of EP on the integrity and function of the BBB. Ten low voltage pulses (5–100 V) were applied to a human in vitro BBB model. Changes in permeability to small molecules (NaF) were studied as well as changes in impedance spectrum and trans-endothelial electric resistivity. Viability and EP were evaluated by Presto-Blue and endogenous Lactate dehydrogenase release assays. The effect on tight junction and adherent junction protein was also studied. The results of low voltage experiments were compared to high voltage experiments (200–1400 V). A significant increase in permeability was found at voltages as low as 10 V despite EP only occurring from 100 V. The changes in permeability as a function of applied voltage were fitted to an inverse-exponential function, suggesting a plateau effect. Staining of VE-cadherin showed specific changes in protein expression. The results indicate that low PEFs can transiently disrupt the BBB by affecting the paracellular route, although the mechanism remains unclear. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
13. Blood-Brain Barrier Cellular Responses Toward Organophosphates: Natural Compensatory Processes and Exogenous Interventions to Rescue Barrier Properties.
- Author
-
Ravid, Orly, Elhaik Goldman, Shirin, Macheto, David, Bresler, Yael, De Oliveira, Raquel Ines, Liraz-Zaltsman, Sigal, Gosselet, Fabien, Dehouck, Lucie, Beeri, Michal Schnaider, and Cooper, Itzik
- Abstract
Organophosphorus compounds (OPs) are highly toxic chemicals widely used as pesticides (e.g., paraoxon (PX)- the active metabolite of the insecticide parathion) and as chemical warfare nerve agents. Blood-brain barrier (BBB) leakage has been shown in rodents exposed to PX, which is an organophosphate oxon. In this study, we investigated the cellular mechanisms involved in BBB reaction after acute exposure to PX in an established in vitro BBB system made of stem-cell derived, human brain-like endothelial cells (BLECs) together with brain pericytes that closely mimic the in vivo BBB. Our results show that PX directly affects the BBB in vitro both at toxic and non-toxic concentrations by attenuating tight junctional (TJ) protein expression and that only above a certain threshold the paracellular barrier integrity is compromised. Below this threshold, BLECs exhibit a morphological coping mechanism in which they enlarge their cell area thus preventing the formation of meaningful intercellular gaps and maintaining barrier integrity. Importantly, we demonstrate that reversal of the apoptotic cell death induced by PX, by a pan-caspase-inhibitor ZVAD-FMK (ZVAD) can reduce PX-induced cell death and elevate cell area but do not prevent the induced BBB permeability, implying that TJ complex functionality is hindered. This is corroborated by formation of ROS at all toxic concentrations of PX and which are even higher with ZVAD. We suggest that while lower levels of ROS can induce compensating mechanisms, higher PX-induced oxidative stress levels interfere with barrier integrity. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
14. High‐fat diet protects the blood–brain barrier in an Alzheimer's disease mouse model.
- Author
-
Elhaik Goldman, Shirin, Goez, David, Last, David, Naor, Sharone, Liraz Zaltsman, Sigal, Sharvit‐Ginon, Inbal, Atrakchi‐Baranes, Dana, Shemesh, Chen, Twitto‐Greenberg, Rachel, Tsach, Shoval, Lotan, Roni, Leikin‐Frenkel, Alicia, Shish, Aviv, Mardor, Yael, Schnaider Beeri, Michal, and Cooper, Itzik
- Subjects
HIGH-fat diet ,BLOOD-brain barrier ,ALZHEIMER'S disease risk factors ,LIPID metabolism ,INSULIN receptors - Abstract
Abstract: Type 2 diabetes (T2D) is associated with increased risk of Alzheimer's disease (AD). There is evidence for impaired blood–brain barrier (BBB) in both diseases, but its role in the interplay between them is not clear. Here, we investigated the effects of high‐fat diet (HFD), a model for T2D, on the Tg2576 mouse model of AD, in regard to BBB function. We showed that HFD mice had higher weight, more insulin resistance, and higher serum HDL cholesterol levels, primarily in Tg2576 mice, which also had higher brain lipids content. In terms of behavior, Tg2576 HFD mice were less active and more anxious, but had better learning in the Morris Water Maze compared to Tg2576 on regular diet. HFD had no effect on the level of amyloid beta 1–42 in the cortex of Tg2576 mice, but increased the transcription level of insulin receptor in the hippocampus. Tg2576 mice on regular diet demonstrated more BBB disruption at 8 and 12 months accompanied by larger lateral ventricles volume in contrast to Tg2576 HFD mice, whose BBB leakage and ventricular volume were similar to wild‐type (WT) mice. Our results suggest that in AD, HFD may promote better cognitive function through improvements of BBB function and of brain atrophy but not of amyloid beta levels. Lipid metabolism in the CNS and peripheral tissues and brain insulin signaling may underlie this protection. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
15. Haptoglobin 1-1 Genotype Modulates the Association of Glycemic Control With Hippocampal Volume in Elderly Individuals With Type 2 Diabetes.
- Author
-
Livny, Abigail, Ravona-Springer, Ramit, Heymann, Anthony, Priess, Rachel, Kushnir, Tammar, Tsarfaty, Galia, Rabinov, Leeron, Moran, Reut, Tik, Niv, Moshier, Erin, Cooper, Itzik, Greenbaum, Lior, Silverman, Jeremy, Levy, Andrew, Sano, Mary, Bendlin, Barbara B., Buchman, Aron S., and Schnaider-Beeri, Michal
- Subjects
GLYCEMIC control ,COGNITIVE ability ,TYPE 2 diabetes ,DISEASES in older people ,HIPPOCAMPUS (Brain) - Abstract
Recent evidence suggests that glycemic control is associated with cognitive function in older patients with type 2 diabetes who are carriers of the haptoglobin (Hp) 1-1 genotype compared with noncarriers. We assessed whether poor glycemic control in Hp 1-1 carriers is more strongly associated with smaller hippocampal volume than in noncarriers. Hippocampal volume was generated from high-resolution structural T1 MRI obtained for 224 participants (28 Hp 1-1 carriers [12.5%] and 196 noncarriers [87.5%]) from the Israel Diabetes and Cognitive Decline (IDCD) study, who had a mean (SD) number of years in the Maccabi Healthcare Services (MHS) registry of 8.35 (2.63) and a mean (SD) HbA1c level of 6.66 (0.73)% [49 mmol/mol]. A stronger negative association between right hippocampal volume and HbA1c was found in patients with the Hp 1-1 genotype, with a 0.032-mL decrease in right hippocampal volume per 14% increase in HbA1c (P = 0.0007) versus a 0.009-mL decrease in Hp 1-1 noncarriers (P = 0.047), after adjusting for total intracranial volume, age, sex, follow-up years in the registry, and cardiovascular factor (interaction, P = 0.025). This indicates that 29.66% of the total variance in right hippocampal volume is explained by HbA1c levels among Hp 1-1 carriers and that 3.22% is explained by HbA1c levels among Hp 1-1 noncarriers. Our results suggest that the hippocampus of Hp 1-1 carriers may be more vulnerable to the insults of poor glycemic control. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
16. Conjugation of Methotrexate-Amino Derivatives to Macromolecules through Carboxylate Moieties Is Superior Over Conventional Linkage to Amino Residues: Chemical, Cell-Free and In Vitro Characterizations.
- Author
-
Cooper, Itzik, Fridkin, Mati, and Shechter, Yoram
- Subjects
METHOTREXATE ,CARBOXYLATES ,SCISSION (Chemistry) ,GLUTAMIC acid ,PRECIPITATION (Chemistry) ,CONJUGATED polymers - Abstract
In this study, we examined the possibility of introducing methotrexate (MTX) to the carboxylate rather than to the ε-amino side chains of proteins. We found that MTX—amino compounds covalently linked to the carboxylate moieties of macromolecules, undergo unusual peptide-bond cleavage, with the release of the MTX amino derivatives from the conjugates. This event takes place at an accelerated rate under acidic conditions, and at a slower rate at physiological pH values. The glutamate portion of MTX is responsible for this behavior, with little or no contribution of the p-aminobenzoate-pteridine ring that is linked to the α-amino side chain of the glutamate. Carboxylate-linked Fmoc-Glu-γ-CONH-(CH
2 )6 -NH2 undergoes hydrolysis in a nearly indistinguishable fashion. A free α carboxylate moiety is essential for this effect. Carboxylate linked Fmoc-glutamic-amide-γ-CONH-(CH2 )6 -NH2 undergoes no hydrolysis under acidic conditions. Based on these findings, we engineered a cysteine specific MTX containing reagent. Its linkage to bovine serum albumin (BSA) yielded a conjugate with profound antiproliferative efficacy in a MTX-sensitive glioma cell line. In conclusion, carboxylate linked MTX-amino derivatives in particular, and carboxylate linked R-α-GLU-γ amino compounds in general are equipped with‘built-in chemical machinery’ that releases them under mild acidic conditions. [ABSTRACT FROM AUTHOR]- Published
- 2016
- Full Text
- View/download PDF
17. Long-term Variability in Glycemic Control Is Associated With White Matter Hyperintensities in APOE4 Genotype Carriers With Type 2 Diabetes.
- Author
-
Livny, Abigail, Ravona-Springer, Ramit, Heymann, Anthony, Priess, Rachel, Kushnir, Tammar, Tsarfaty, Galia, Rabinov, Leeron, Moran, Reut, Hoffman, Hadass, Cooper, Itzik, Greenbaum, Lior, Silverman, Jeremy, Sano, Mary, Johnson, Sterling C., Bendlin, Barbara B., Beeri, Michal Schnaider, and Schnaider Beeri, Michal
- Subjects
GLYCEMIC control ,LEUKOENCEPHALOPATHIES ,TYPE 2 diabetes ,APOLIPOPROTEINS ,GENOTYPES ,BRAIN diseases ,ALLELES ,ANTHROPOMETRY ,BLOOD sugar ,BRAIN ,CARDIOVASCULAR diseases ,GLYCOSYLATED hemoglobin ,MAGNETIC resonance imaging ,RESEARCH funding ,GENETIC carriers - Abstract
Objective: We assessed whether the apolipoprotein ε4 (APOE4) genotype affects the relationship of variability in long-term glycemic control (measured by HbA1c SD of multiple measurements) with white matter hyperintensities (WMHs) in elderly patients with type 2 diabetes (T2D).Research Design and Methods: WMH volume was generated from structural T1 and fluid-attenuated inversion recovery MRI in each subject. The analysis included 124 subjects; 27 (21.8%) had one or more APOE4 alleles.Results: HbA1c variability was associated with significantly higher WMH in APOE4 carriers (r = 0.47, P = 0.03), controlling for age, sex, mean HbA1c, number of follow-up years, and a composite of cardiovascular risk factors, but not in noncarriers (r = -0.04, P = 0.71; P for interaction = 0.050).Conclusions: The results suggest that the APOE4 genotype affects the relationship of long-term glycemic control with WMH load so that APOE4 carriers may be more vulnerable to the insults of poor control. [ABSTRACT FROM AUTHOR]- Published
- 2016
- Full Text
- View/download PDF
18. High dietary advanced glycation end products are associated with poorer spatial learning and accelerated Aβ deposition in an Alzheimer mouse model.
- Author
-
Lubitz, Irit, Ricny, Jan, Atrakchi‐Baranes, Dana, Shemesh, Chen, Kravitz, Efrat, Liraz‐Zaltsman, Sigal, Maksin‐Matveev, Anna, Cooper, Itzik, Leibowitz, Avshalom, Uribarri, Jaime, Schmeidler, James, Cai, Weijing, Kristofikova, Zdena, Ripova, Daniela, LeRoith, Derek, and Schnaider‐Beeri, Michal
- Subjects
ALZHEIMER'S disease treatment ,ADVANCED glycation end-products ,FOOD irradiation ,HIPPOCAMPUS (Brain) ,NEURODEGENERATION ,LABORATORY mice - Abstract
There is growing evidence of the involvement of advanced glycation end products ( AGEs) in the pathogenesis of neurodegenerative processes including Alzheimer's disease ( AD) and their function as a seed for the aggregation of Aβ, a hallmark feature of AD. AGEs are formed endogenously and exogenously during heating and irradiation of foods. We here examined the effect of a diet high in AGEs in the context of an irradiated diet on memory, insoluble Aβ
42 , AGEs levels in hippocampus, on expression of the receptor for AGEs ( RAGE), and on oxidative stress in the vasculature. We found that AD-like model mice on high- AGE diet due to irradiation had significantly poorer memory, higher hippocampal levels of insoluble Aβ42 and AGEs as well as higher levels of oxidative stress on vascular walls, compared to littermates fed an isocaloric diet. These differences were not due to weight gain. The data were further supported by the overexpression of RAGE, which binds to Aβ42 and regulates its transport across the blood-brain barrier, suggesting a mediating pathway. Because exposure to AGEs can be diminished, these insights provide an important simple noninvasive potential therapeutic strategy for alleviating a major lifestyle-linked disease epidemic. [ABSTRACT FROM AUTHOR]- Published
- 2016
- Full Text
- View/download PDF
19. Normal human CD4 helper T cells express Kv1.1 voltage-gated K channels, and selective Kv1.1 block in T cells induces by itself robust TNFα production and secretion and activation of the NFκB non-canonical pathway.
- Author
-
Fellerhoff-Losch, Barbara, Korol, Sergiy, Ganor, Yonatan, Gu, Songhai, Cooper, Itzik, Eilam, Raya, Besser, Michal, Goldfinger, Meidan, Chowers, Yehuda, Wank, Rudolf, Birnir, Bryndis, and Levite, Mia
- Subjects
CD4 antigen ,T cells ,VOLTAGE-gated ion channels ,TUMOR necrosis factors ,IMMUNE system ,CANCER immunotherapy ,CANCER patients ,NEURAL development - Abstract
TNFα is a very potent and pleiotropic pro-inflammatory cytokine, essential to the immune system for eradicating cancer and microorganisms, and to the nervous system, for brain development and ongoing function. Yet, excess and/or chronic TNFα secretion causes massive tissue damage in autoimmune, inflammatory and neurological diseases and injuries. Therefore, many patients with autoimmune/inflammatory diseases receive anti-TNFα medications. TNFα is secreted primarily by CD4 T cells, macrophages, monocytes, neutrophils and NK cells, mainly after immune stimulation. Yet, the cause for the pathologically high and chronic TNFα secretion is unknown. Can blocking of a particular ion channel in T cells induce by itself TNFα secretion? Such phenomenon was never revealed or even hypothesized. In this interdisciplinary study we discovered that: (1) normal human T cells express Kv1.1 voltage-gated potassium channel mRNA, and the Kv1.1 membrane-anchored protein channel; (2) Kv1.1 is expressed in most CD4CD3 helper T cells (mean CD4CD3Kv1.1 T cells of 7 healthy subjects: 53.09 ± 22.17 %), but not in CD8CD3 cytotoxic T cells (mean CD8CD3Kv1.1 T cells: 4.12 ± 3.04 %); (3) electrophysiological whole-cell recordings in normal human T cells revealed Kv currents; (4) Dendrotoxin-K (DTX-K), a highly selective Kv1.1 blocker derived from snake toxin, increases the rate of rise and decay of Kv currents in both resting and activated T cells, without affecting the peak current; (5) DTX-K by itself induces robust TNFα production and secretion by normal human T cells, without elevating IFNγ, IL-4 and IL-10; (6) intact Ca channels are required for DTX-induced TNFα secretion; (7) selective anti-Kv1.1 antibodies also induce by themselves TNFα secretion; (8) DTX-K activates NFκB in normal human T cells via the unique non-canonical-pathway; (9) injection of Kv1.1-blocked human T cells to SCID mice, causes recruitment of resident mouse cells into the liver, alike reported after TNFα injection into the brain. Based on our discoveries we speculate that abnormally blocked Kv1.1 in T cells (and other immune cells?), due to either anti-Kv1.1 autoimmune antibodies, or Kv1.1-blocking toxins alike DTX-K, or Kv1.1-blocking genetic mutations, may be responsible for the chronic/excessive TNFα in autoimmune/inflammatory diseases. Independently, we also hypothesize that selective block of Kv1.1 in CD4 T cells of patients with cancer or chronic infectious diseases could be therapeutic, since it may: a. augment beneficial secretion and delivery of TNFα to the disease-affected sites; b. induce recruitment and extravasation of curative immune cells and factors; c. improve accessibility of drugs to the brain and few peripheral organs thanks to TNFα-induced increased permeability of organ's barriers. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
20. Combined Treatment of an Amyotrophic Lateral Sclerosis Rat Model with Recombinant GOT1 and Oxaloacetic Acid: A Novel Neuroprotective Treatment.
- Author
-
Ruban, angela, Cohen-Kashi Malina, Katayun, Cooper, Itzik, Graubardt, Nadine, Babakin, Leonid, Jona, Ghil, and Teichberg, Vivian I.
- Subjects
AMYOTROPHIC lateral sclerosis treatment ,NEUROPROTECTIVE agents ,OXALACETATE ,AMINOTRANSFERASES ,RECOMBINANT proteins ,GLUTAMIC acid ,BLOOD-brain barrier ,LABORATORY rats ,THERAPEUTICS - Abstract
Background/Aim: The sporadic form of the disease affects the majority of amyotrophic lateral sclerosis (ALS) patients. The role of glutamate (Glu) excitotoxicity in ALS has been extensively documented and remains one of the prominent hypotheses of ALS pathogenesis. In light of this evidence, the availability of a method to remove excess Glu from brain and spinal cord extracellular fluids without the need to deliver drugs across the blood-brain barrier and with minimal or no adverse effects may provide a major therapeutic asset, which is the primary aim of this study. Methods: The therapeutic efficacy of the combined treatment with recombinant Glu-oxaloacetate-transaminase (rGOT) and its co-factor oxaloacetic acid (OxAc) has been tested in an animal model of sporadic ALS. Results: We found that OxAc/rGOT treatment provides significant neuroprotection to spinal cord motor neurons. It also slows down the development of motor weakness and prolongs survival. Conclusion: In this study we bring evidence that the administration of Glu scavengers to rats with sporadic ALS inhibited the massive death of spinal cord motor neurons, slowed the onset of motor weakness and prolonged survival. This treatment may be of high clinical significance for the future treatment of chronic neurodegenerative diseases. © 2015 S. Karger AG, Basel [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
21. Combined local blood-brain barrier opening and systemic methotrexate for the treatment of brain tumors.
- Author
-
Cooper, Itzik, Last, David, Guez, David, Sharabi, Shirley, Elhaik Goldman, Shirin, Lubitz, Irit, Daniels, Dianne, Salomon, Sharona, Tamar, Gregory, Tamir, Tzur, Mardor, Ronni, Fridkin, Mati, Shechter, Yoram, and Mardor, Yael
- Subjects
BRAIN tumor treatment ,BLOOD-brain barrier ,GLIOBLASTOMA multiforme ,METHOTREXATE ,SERUM albumin ,PATIENTS ,PROGNOSIS - Abstract
Despite aggressive therapy, existing treatments offer poor prognosis for glioblastoma multiforme patients, in part due to poor penetration of most drugs across the blood-brain barrier (BBB). We propose a minimal-invasive combined treatment approach consisting of local BBB disruption in the tumor in parallel to systemic drug administration. Local BBB disruption is obtained by convection-enhanced delivery of a novel BBB disruption agent, enabling efficient/targeted delivery of the systemically administered drug by the tumors own vasculature. Various human serum albumin (HSA) analogs were synthesized and screened for BBB disruption efficacy in custom in vitro systems. The candidate analogs were then delivered into naïve rat brains by convection-enhanced delivery and screened for maximal BBB disruption and minimal brain toxicity. These studies found a noncationized/neutralized analog, ethylamine (EA)-HSA, to be the optimal BBB-opening agent. Immunocytochemical studies suggested that BBB disruption by EA-HSA may be explained by alterations in occludin expression. Finally, an efficacy study in rats bearing intracranial gliomas was performed. The rats were treated by convection-enhanced delivery of EA-HSA in parallel to systemic administration of Methotrexate, showing significant antineoplastic effects of the combined approached reflected in suppressed tumor growth and significantly (~x3) prolonged survival. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
22. Claudin-5 Expression in In Vitro Models of the Blood–Brain Barrier.
- Author
-
Cooper, Itzik, Cohen-Kashi-Malina, Katayun, and Teichberg, Vivian I.
- Published
- 2011
- Full Text
- View/download PDF
23. MRI-based Treatment Response Assessment Maps, Trams, for Depicting Short/long-term Subtle BBB Disruption in Traumatic Brain Injury.
- Author
-
Mardor, Yael, Last, David, Guez, David, Shohami, Ester, Shemesh, Chen, Cooper, Itzik, Daniels, Dianne, Sharabi, Shirley, and Liraz Zaltsman, Sigal
- Published
- 2022
- Full Text
- View/download PDF
24. Non-invasive Low Pulsed Electric Fields Induce Transient BBB Disruption, Depicted by the Trams, Enabling Efficient Delivery of Biologics into the Brain.
- Author
-
Mardor, Yael, Last, David, Bresler, yael, Daniels, Dianne, Cooper, Itzik, and Sharabi, Shirley
- Published
- 2022
- Full Text
- View/download PDF
25. Blood glutamate scavengers prolong the survival of rats and mice with brain-implanted gliomas.
- Author
-
Ruban, Angela, Berkutzki, Tamara, Cooper, Itzik, Mohar, Boaz, and Teichberg, Vivian
- Published
- 2012
- Full Text
- View/download PDF
26. Mechanisms of glutamate efflux at the blood-brain barrier: involvement of glial cells.
- Author
-
Cohen-Kashi-Malina, Katayun, Cooper, Itzik, and Teichberg, Vivian I
- Subjects
BLOOD-brain barrier ,GLUTAMIC acid ,BRAIN damage ,NEUROGLIA ,NEUROTOXIC agents ,DRUG synergism ,NEUROLOGICAL disorders ,HOMEOSTASIS - Abstract
At high concentrations, glutamate (Glu) exerts potent neurotoxic properties, leading to irreversible brain damages found in numerous neurological disorders. The accepted notion that Glu homeostasis in brain interstitial fluid is maintained primarily through the activity of Glu transporters present on glial cells does not take into account the possible contribution of endothelial cells constituting the blood-brain barrier (BBB) to this process. Here, we present evidence for the presence of the Glu transporters, excitatory amino-acid transporters (EAATs) 1 to 3, in porcine brain endothelial cells (PBECs) and show their participation in Glu uptake into PBECs. Moreover, transport of Glu across three in vitro models of the BBB is investigated for the first time, and evidence for Glu transport across the BBB in both directions is presented. Our results provide evidence that the BBB can function in the efflux mode to selectively remove Glu, via specific transporters, from the abluminal side (brain) into the luminal compartment (blood). Furthermore, we found that glial cells lining the BBB have an active role in the efflux process by taking up Glu and releasing it, through hemichannels, anion channels, and possibly the reversal of its EAATs, in close proximity to ECs, which in turn take up Glu and release it to the blood. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
27. Interactions of the prion peptide (PrP 106-126) with brain capillary endothelial cells: coordinated cell killing and remodeling of intercellular junctions.
- Author
-
Cooper, Itzik, Malina, Katayun Cohen-Kashi, Cagnotto, Alfredo, Bazzoni, Gianfranco, Salmona, Mario, and Teichberg, Vivian I.
- Subjects
PEPTIDES ,BLOOD-brain barrier ,PRIONS ,TIGHT junctions ,CELL death ,THERAPEUTIC use of immunoglobulins ,PHYSIOLOGY - Abstract
We studied here the interactions of PrP 106-126, a peptide corresponding to the prion protein (PrP) amyloidogenic region, with a blood-brain barrier in vitro model consisting of confluent porcine brain endothelial cells (PBEC). PrP 106-126 interacted selectively with PBEC via their luminal side, and caused cumulative cell death, as shown by lactate dehydrogenase release, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide reduction, Caspase 3 induction and direct cell counting. In addition, PrP 106-126, but not its corresponding scrambled peptide, produced a 50% reduction of the transendothelial electrical resistance, while the PBEC maintained confluency. This process was accompanied by a 23% increase of PBEC average size and the selective disappearance from the cell borders of the junction proteins occludin, claudin-5 and VE-cadherin (but not ZO-1), as evaluated by immunostaining. These results fit with a mechanism by which PrP 106-126 initiates a coordinated cell killing process ultimately causing the remaining cells to undergo a coordinated remodeling of the intercellular junctions and an expansion of their cell territory. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
28. Human T-leukemia and T-lymphoma express glutamate receptor AMPA GluR3, and the neurotransmitter glutamate elevates the cancer-related matrix-metalloproteinases inducer CD147/EMMPRIN, MMP-9 secretion and engraftment of T-leukemia in vivo.
- Author
-
Ganor, Yonatan, Grinberg, Igor, Reis, Arbel, Cooper, Itzik, Goldstein, Ronald S., and Levite, Mia
- Subjects
ADULT T-cell leukemia ,LYMPHOMAS ,T cells ,GLUTAMIC acid ,CHEMOTAXIS ,METALLOPROTEINASES ,MEDICAL research ,CANCER - Abstract
Glutamate is the major excitatory neurotransmitter of the nervous system. We previously found that glutamate activates normal human T-cells, inducing their adhesion and chemotaxis, via its glutamate receptors of the alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) subtype 3 (GluR3) expressed in these cells. Here, we discovered that human T-leukemia (Jurkat) and cutaneous sezary T-lymphoma (HuT-78) cells also express high levels of GluR3. Furthermore, glutamate (10 nM) elevates CD147/EMMPRIN, a cancer-associated matrix metalloproteinases (MMPs) inducer, promoting spread of many tumors. Glutamate-induced CD147 elevation in both cancerous and normal human T-cells was mimicked by AMPA (glutamate/AMPA-receptor agonist) and blocked by CNQX (glutamate/AMPA-receptor antagonist). Importantly, glutamate also increased gelatinase MMP-9 secretion by T-lymphoma. Finally, ex vivo pre-treatment of T-leukemia with glutamate enhanced their subsequent in vivo engraftment into chick embryo liver and chorioallantoic membrane. Together, these findings reveal that glutamate elevates cancer associated proteins and activity in T-cell cancers and by doing so may facilitate their growth and spread, especially to and within the nervous system. If so, glutamate receptors in T-cell malignancies should be blocked. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
29. Albumin–Methotrexate Prodrug Analogues That Undergo Intracellular Reactivation Following Entrance into Cancerous Glioma Cells.
- Author
-
Cooper, Itzik, Schnaider-Beeri, Michal, Fridkin, Mati, and Shechter, Yoram
- Subjects
PRODRUGS ,GLIOMAS ,TETRAHYDROFOLATE dehydrogenase ,CARRIER proteins ,SERUM albumin ,PEPTIDE bonds - Abstract
A family of monomodified bovine serum albumin (BSA) linked to methotrexate (MTX) through a variety of spacers was prepared. All analogues were found to be prodrugs having low MTX-inhibitory potencies toward dihydrofolate reductase in a cell-free system. The optimal conjugates regenerated their antiproliferative efficacies following entrance into cancerous glioma cell lines and were significantly superior to MTX in an insensitive glioma cell line. A BSA–MTX conjugate linked through a simple ethylene chain spacer, containing a single peptide bond located 8.7 Å distal to the protein back bone, and apart from the covalently linked MTX by about 12 Å, was most effective. The inclusion of an additional disulfide bond in the spacer neither enhanced nor reduced the killing potency of this analogue. Disrupting the native structure of the carrier protein in the conjugates significantly reduced their antiproliferative activity. In conclusion, we have engineered BSA–MTX prodrug analogues which undergo intracellular reactivation and facilitate antiproliferative activities following their entrance into glioma cells. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
30. Albumin-EDTA-Vanadium Is a Powerful Anti-Proliferative Agent, Following Entrance into Glioma Cells via Caveolae-Mediated Endocytosis.
- Author
-
Cooper, Itzik, Ravid, Orly, Rand, Daniel, Atrakchi, Dana, Shemesh, Chen, Bresler, Yael, Ben-Nissan, Gili, Sharon, Michal, Fridkin, Mati, and Shechter, Yoram
- Subjects
GLIOMAS ,TWENTY twenties ,SERUM albumin ,VANADIUM ,ENDOCYTOSIS ,CELL lines ,CANCER cells - Abstract
Human serum albumin (HSA) is efficiently taken up by cancer cells as a source of carbon and energy. In this study, we prepared a monomodified derivative of HSA covalently linked to an EDTA derivative and investigated its efficacy to shuttle weakly anti-proliferative EDTA associating ligands such as vanadium, into a cancer cell line. HSA-S-MAL-(CH
2 )2 -NH-CO-EDTA was found to associate both with the vanadium anion (+5) and the vanadium cation (+4) with more than thrice the associating affinity of those ligands toward EDTA. Both conjugates internalized into glioma tumor cell line via caveolae-mediated endocytosis pathway and showed potent anti-proliferative capacities. IC50 values were in the range of 0.2 to 0.3 µM, potentiating the anti-proliferative efficacies of vanadium (+4) and vanadium (+5) twenty to thirty fold, respectively. HSA-EDTA-VO++ in particular is a cancer permeable prodrug conjugate. The associated vanadium (+4) is not released, nor is it active anti-proliferatively prior to its engagement with the cancerous cells. The bound vanadium (+4) dissociates from the conjugate under acidic conditions with half maximal value at pH 5.8. In conclusion, the anti-proliferative activity feature of vanadium can be amplified and directed toward a cancer cell line. This is accomplished using a specially designed HSA-EDTA-shuttling vehicle, enabling vanadium to be anti-proliferatively active at the low micromolar range of concentration. [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
31. Endothelial Iron Homeostasis Regulates Blood-Brain Barrier Integrity via the HIF2α—Ve-Cadherin Pathway.
- Author
-
Rand, Daniel, Ravid, Orly, Atrakchi, Dana, Israelov, Hila, Bresler, Yael, Shemesh, Chen, Omesi, Liora, Liraz-Zaltsman, Sigal, Gosselet, Fabien, Maskrey, Taber S., Schnaider Beeri, Michal, Wipf, Peter, Cooper, Itzik, and Neuhaus, Winfried
- Subjects
BLOOD-brain barrier ,MITOCHONDRIAL membranes ,CENTRAL nervous system ,HOMEOSTASIS ,IRON chelates ,MEMBRANE permeability (Biology) ,IRON - Abstract
The objective of this study was to investigate the molecular response to damage at the blood-brain barrier (BBB) and to elucidate critical pathways that might lead to effective treatment in central nervous system (CNS) pathologies in which the BBB is compromised. We have used a human, stem-cell derived in-vitro BBB injury model to gain a better understanding of the mechanisms controlling BBB integrity. Chemical injury induced by exposure to an organophosphate resulted in rapid lipid peroxidation, initiating a ferroptosis-like process. Additionally, mitochondrial ROS formation (MRF) and increase in mitochondrial membrane permeability were induced, leading to apoptotic cell death. Yet, these processes did not directly result in damage to barrier functionality, since blocking them did not reverse the increased permeability. We found that the iron chelator, Desferal© significantly decreased MRF and apoptosis subsequent to barrier insult, while also rescuing barrier integrity by inhibiting the labile iron pool increase, inducing HIF2α expression and preventing the degradation of Ve-cadherin specifically on the endothelial cell surface. Moreover, the novel nitroxide JP4-039 significantly rescued both injury-induced endothelium cell toxicity and barrier functionality. Elucidating a regulatory pathway that maintains BBB integrity illuminates a potential therapeutic approach to protect the BBB degradation that is evident in many neurological diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
32. Non-Invasive Low Pulsed Electrical Fields for Inducing BBB Disruption in Mice—Feasibility Demonstration.
- Author
-
Sharabi, Shirley, Last, David, Daniels, Dianne, Fabian, Ido Didi, Atrakchi, Dana, Bresler, Yael, Liraz-Zaltsman, Sigal, Cooper, Itzik, Mardor, Yael, and Declèves, Xavier
- Subjects
BLOOD-brain barrier ,CENTRAL nervous system ,ELECTRIC fields ,MICE - Abstract
The blood–brain barrier (BBB) is a major hurdle for the treatment of central nervous system disorders, limiting passage of both small and large therapeutic agents from the blood stream into the brain. Thus, means for inducing BBB disruption (BBBd) are urgently needed. Here, we studied the application of low pulsed electrical fields (PEFs) for inducing BBBd in mice. Mice were treated by low PEFs using electrodes pressed against both sides of the skull (100–400 square 50 µs pulses at 4 Hz with different voltages). BBBd as a function of treatment parameters was evaluated using MRI-based treatment response assessment maps (TRAMs) and Evans blue extravasation. A 3D numerical model of the mouse brain and electrodes was constructed using finite element software, simulating the electric fields distribution in the brain and ensuring no significant temperature elevation. BBBd was demonstrated immediately after treatment and significant linear regressions were found between treatment parameters and the extent of BBBd. The maximal induced electric field in the mice brains, calculated by the numerical model, ranged between 62.4 and 187.2 V/cm for the minimal and maximal applied voltages. These results demonstrate the feasibility of inducing significant BBBd using non-invasive low PEFs, well below the threshold for electroporation. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
33. Publisher Correction: TCF7L2 polymorphisms are associated with amygdalar volume in elderly individuals with Type 2 Diabetes.
- Author
-
Ganmore, Ithamar, Livny, Abigail, Ravona-Springer, Ramit, Cooper, Itzik, Alkelai, Anna, Shelly, Shahar, Tsarfaty, Galia, Heymann, Anthony, Schnaider Beeri, Michal, and Greenbaum, Lior
- Subjects
TYPE 2 diabetes ,TRANSCRIPTION factors - Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
34. TCF7L2 polymorphisms are associated with amygdalar volume in elderly individuals with Type 2 Diabetes.
- Author
-
Ganmore, Ithamar, Livny, Abigail, Ravona-Springer, Ramit, Cooper, Itzik, Alkelai, Anna, Shelly, Shahar, Tsarfaty, Galia, Heymann, Anthony, Schnaider Beeri, Michal, and Greenbaum, Lior
- Subjects
TYPE 2 diabetes ,SINGLE nucleotide polymorphisms ,TRANSCRIPTION factors ,DISEASE susceptibility ,GENETIC polymorphisms - Abstract
The association between several Single Nucleotide Polymorphisms (SNPs) within the transcription factor 7-like 2 (TCF7L2) gene and Type 2 Diabetes (T2D) as well as additional T2D-related traits is well established. Since alteration in total and regional brain volumes are consistent findings among T2D individuals, we studied the association of four T2D susceptibility SNPS within TCF7L2 (rs7901695, rs7903146, rs11196205, and rs12255372) with volumes of white matter hyperintensities (WMH), gray matter, and regional volumes of amygdala and hippocampus obtained from structural MRI among 191 T2D elderly Jewish individuals. Under recessive genetic model (controlling for age, sex and intracranial volume), we found that for all four SNPs, carriers of two copies of the T2D risk allele (homozygous genotype) had significantly smaller amygdalar volume: rs7901695- CC genotype vs. CT + TT genotypes, p = 0.002; rs7903146-TT vs. TC + CC, p = 0.003; rs11196205- CC vs. CG + GG, p = 0.0003; and rs12255372- TT vs. TG + GG, p = 0.003. Adjusting also for T2D-related covariates, body mass index (BMI), and ancestry did not change the results substantively (rs7901695, p = 0.003; rs7903146, p = 0.005; rs11196205, p = 0.001; and rs12255372, p = 0.005). Conditional analysis demonstrated that only rs11196205 was independently associated with amygdalar volume at a significant level. Separate analysis of left and right amygdala revealed stronger results for left amygdalar volume. Taken together, we report association of TCF7L2 SNPs with amygdalar volume among T2D elderly Jewish patients. Further studies in other populations are required to support these findings and reach more definitive conclusions. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
35. COMBINATION THERAPY OF TYPE 2 DIABETES MEDICATIONS AS A TREATMENT TARGET FOR ALZHEIMER DISEASE.
- Author
-
Atrakchi-Baranes, Dana, Lubitz, Irit, Chen, Shemesh, Katsel, Pavel, Liraz-Zaltsman, Sigal, Licht, Tali, Sharvit-Ginon, Inbal, Goldman, Shirin Elhaik, Cooper, Itzik, Haroutunian, Vahram, and Beeri, Michal Schnaider
- Published
- 2017
- Full Text
- View/download PDF
36. The Alzheimer's disease risk loci bin1 is associated with poorer episodic memory performance in cognitively normal type 2 diabetes elderly.
- Author
-
Greenbaum, Lior, Ravona-Springer, Ramit, Lubitz, Irit, Schmeidler, James, Cooper, Itzik, Silverman, Jeremy M., Heymann, Anthony, and Beeri, Michal Schnaider
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.