Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Int J Mol Sci ; 25(17)2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39273366

ABSTRACT

Angiotensin-converting enzymes (ACE) are well-known for their roles in both blood pressure regulation via the renin-angiotensin system as well as functions in fertility, immunity, hematopoiesis, and many others. The two main isoforms of ACE include ACE and ACE-2 (ACE2). Both isoforms have similar structures and mediate numerous effects on the cardiovascular system. Most remarkably, ACE2 serves as an entry receptor for SARS-CoV-2. Understanding the interaction between the virus and ACE2 is vital to combating the disease and preventing a similar pandemic in the future. Noninvasive imaging techniques such as positron emission tomography and single photon emission computed tomography could noninvasively and quantitatively assess in vivo ACE2 expression levels. ACE2-targeted imaging can be used as a valuable tool to better understand the mechanism of the infection process and the potential roles of ACE2 in homeostasis and related diseases. Together, this information can aid in the identification of potential therapeutic drugs for infectious diseases, cancer, and many ACE2-related diseases. The present review summarized the state-of-the-art radiotracers for ACE2 imaging, including their chemical design, pharmacological properties, radiochemistry, as well as preclinical and human molecular imaging findings. We also discussed the advantages and limitations of the currently developed ACE2-specific radiotracers.


Subject(s)
Angiotensin-Converting Enzyme 2 , COVID-19 , Molecular Imaging , SARS-CoV-2 , Humans , Angiotensin-Converting Enzyme 2/metabolism , Molecular Imaging/methods , COVID-19/metabolism , COVID-19/diagnostic imaging , SARS-CoV-2/metabolism , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/metabolism , Animals , Positron-Emission Tomography/methods , Tomography, Emission-Computed, Single-Photon/methods
2.
Int J Mol Sci ; 24(5)2023 Feb 24.
Article in English | MEDLINE | ID: mdl-36901893

ABSTRACT

Abusive head trauma (AHT) is a serious traumatic brain injury and the leading cause of death in children younger than 2 years. The development of experimental animal models to simulate clinical AHT cases is challenging. Several animal models have been designed to mimic the pathophysiological and behavioral changes in pediatric AHT, ranging from lissencephalic rodents to gyrencephalic piglets, lambs, and non-human primates. These models can provide helpful information for AHT, but many studies utilizing them lack consistent and rigorous characterization of brain changes and have low reproducibility of the inflicted trauma. Clinical translatability of animal models is also limited due to significant structural differences between developing infant human brains and the brains of animals, and an insufficient ability to mimic the effects of long-term degenerative diseases and to model how secondary injuries impact the development of the brain in children. Nevertheless, animal models can provide clues on biochemical effectors that mediate secondary brain injury after AHT including neuroinflammation, excitotoxicity, reactive oxygen toxicity, axonal damage, and neuronal death. They also allow for investigation of the interdependency of injured neurons and analysis of the cell types involved in neuronal degeneration and malfunction. This review first focuses on the clinical challenges in diagnosing AHT and describes various biomarkers in clinical AHT cases. Then typical preclinical biomarkers such as microglia and astrocytes, reactive oxygen species, and activated N-methyl-D-aspartate receptors in AHT are described, and the value and limitations of animal models in preclinical drug discovery for AHT are discussed.


Subject(s)
Brain Injuries, Traumatic , Brain Injuries , Child Abuse , Craniocerebral Trauma , Child , Humans , Animals , Sheep , Swine , Infant , Reproducibility of Results , Child Abuse/diagnosis , Craniocerebral Trauma/diagnosis
3.
Pediatr Radiol ; 51(6): 966-970, 2021 May.
Article in English | MEDLINE | ID: mdl-33999238

ABSTRACT

Abusive head trauma (AHT) is a leading cause of mortality and morbidity in child abuse, with a mortality rate of approximately 25%. In survivors, the prognosis remains dismal, with high prevalence of cerebral palsy, epilepsy and neuropsychiatric disorders. Early and accurate diagnosis of AHT is challenging, both clinically and radiologically, with up to one-third of cases missed on initial examination. Moreover, most of the management in AHT is supportive, reflective of the lack of clear understanding of specific pathogenic mechanisms underlying secondary insult, with approaches targeted toward decreasing intracranial hypertension and reducing cerebral metabolism, cell death and excitotoxicity. Multiple studies have elucidated the role of pro- and anti-inflammatory cytokines and chemokines with upregulation/recruitment of microglia/macrophages, oligodendrocytes and astrocytes in severe traumatic brain injury (TBI). In addition, recent studies in animal models of AHT have demonstrated significant upregulation of microglia, with a potential role of inflammatory cascade contributing to secondary insult. Despite the histological and biochemical evidence, there is a significant dearth of specific imaging approaches to identify this neuroinflammation in AHT. The primary motivation for development of such imaging approaches stems from the need to therapeutically target neuroinflammation and establish its utility in monitoring and prognostication. In the present paper, we discuss the available data suggesting the potential role of neuroinflammation in AHT and role of radiotracer imaging in aiding diagnosis and patient management.


Subject(s)
Child Abuse , Craniocerebral Trauma , Child , Child Abuse/diagnosis , Craniocerebral Trauma/diagnostic imaging , Diagnostic Imaging , Diagnostic Tests, Routine , Humans , Infant , Prognosis
4.
Anal Biochem ; 535: 25-34, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28757092

ABSTRACT

Automated cell-based high-throughput screening (HTS) is a powerful tool in drug discovery, and it is increasingly being recognized that three-dimensional (3D) models, which more closely mimic in vivo-like conditions, are desirable screening platforms. One limitation hampering the development of 3D HTS is the lack of suitable 3D culture scaffolds that can readily be incorporated into existing HTS infrastructure. We now show that ß-hairpin peptide hydrogels can serve as a 3D cell culture platform that is compatible with HTS. MAX8 ß-hairpin peptides can physically assemble into a hydrogel with defined porosity, permeability and mechanical stability with encapsulated cells. Most importantly, the hydrogels can then be injected under shear-flow and immediately reheal into a hydrogel with the same properties exhibited prior to injection. The post-injection hydrogels are cell culture compatible at physiological conditions. Using standard HTS equipment and medulloblastoma pediatric brain tumor cells as a model system, we show that automatic distribution of cell-peptide mixtures into 384-well assay plates results in evenly dispensed, viable MAX8-cell constructs suitable for commercially available cell viability assays. Since MAX8 peptides can be functionalized to mimic the microenvironment of cells from a variety of origins, MAX8 peptide gels should have broad applicability for 3D HTS drug discovery.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Culture Techniques/methods , Drug Discovery , High-Throughput Screening Assays , Hydrogels/chemical synthesis , Peptides/chemistry , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Hydrogels/chemistry , Peptides/chemical synthesis , Rheology , Structure-Activity Relationship , Tumor Cells, Cultured
5.
BMC Cell Biol ; 16: 24, 2015 Sep 18.
Article in English | MEDLINE | ID: mdl-26382850

ABSTRACT

BACKGROUND: Endocytosis of activated EGF receptor (EGFR) to specific endocytic compartments is required to terminate EGF signaling. Trafficking of EGFR relies on microtubule tracks that transport the cargo vesicle to their intermediate and final destinations and can be modulated through posttranslational modification of tubulin including acetylation. Na,K-ATPase maintains intracellular sodium homeostasis, functions as a signaling scaffold and interacts with EGFR. Na,K-ATPase also binds to and is regulated by acetylated tubulin but whether there is a functional link between EGFR, Na,K-ATPase and tubulin acetylation is not known. RESULTS: EGF-induced sodium influx regulates EGFR trafficking through increased microtubule acetylation. Increased sodium influx induced either by sodium ionophores or Na,K-ATPase blockade mimicked the EGF-induced effects on EGFR trafficking through histone deacetylase (HDAC) 6 inactivation and accumulation of acetylated tubulin. In turn, blocking sodium influx reduced tubulin acetylation and EGF-induced EGFR turnover. Knockdown of HDAC6 reversed the effect of sodium influx indicating that HDAC6 is necessary to modulate sodium-dependent tubulin acetylation. CONCLUSIONS: These studies provide a novel regulatory mechanism to attenuate EGFR signaling in which EGF modulates EGFR trafficking through intracellular sodium-mediated HDAC6 inactivation and tubulin acetylation.


Subject(s)
Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Histone Deacetylases/metabolism , Sodium/metabolism , Tubulin/metabolism , Acetylation , Biological Transport , Epidermal Growth Factor/genetics , ErbB Receptors/genetics , Histone Deacetylase 6 , Histone Deacetylases/genetics , Humans
6.
Mol Cancer ; 14: 159, 2015 Aug 19.
Article in English | MEDLINE | ID: mdl-26286140

ABSTRACT

BACKGROUND: The Sonic hedgehog (Shh) signaling pathway plays an important role in cerebellar development, and mutations leading to hyperactive Shh signaling have been associated with certain forms of medulloblastoma, a common form of pediatric brain cancer. While the fundamentals of this pathway are known, the molecular targets contributing to Shh-mediated proliferation and transformation are still poorly understood. Na,K-ATPase is a ubiquitous enzyme that maintains intracellular ion homeostasis and functions as a signaling scaffold and a cell adhesion molecule. Changes in Na,K-ATPase function and subunit expression have been reported in several cancers and loss of the ß1-subunit has been associated with a poorly differentiated phenotype in carcinoma but its role in medulloblastoma progression is not known. METHODS: Human medulloblastoma cell lines and primary cultures of cerebellar granule cell precursors (CGP) were used to determine whether Shh regulates Na,K-ATPase expression. Smo/Smo medulloblastoma were used to assess the Na,K-ATPase levels in vivo. Na,K-ATPase ß1-subunit was knocked down in DAOY cells to test its role in medulloblastoma cell proliferation and tumorigenicity. RESULTS: Na,K-ATPase ß1-subunit levels increased with differentiation in normal CGP cells. Activation of Shh signaling resulted in reduced ß1-subunit mRNA and protein levels and was mimicked by overexpression of Gli1and Bmi1, both members of the Shh signaling cascade; overexpression of Bmi1 reduced ß1-subunit promoter activity. In human medulloblastoma cells, low ß1-subunit levels were associated with increased cell proliferation and in vivo tumorigenesis. CONCLUSIONS: Na,K-ATPase ß1-subunit is a target of the Shh signaling pathway and loss of ß1-subunit expression may contribute to tumor development and progression not only in carcinoma but also in medulloblastoma, a tumor of neuronal origin.


Subject(s)
Carcinogenesis/genetics , Hedgehog Proteins/genetics , Medulloblastoma/genetics , Sodium-Potassium-Exchanging ATPase/biosynthesis , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Hedgehog Proteins/antagonists & inhibitors , Humans , Medulloblastoma/pathology , Mitogen-Activated Protein Kinase 7/biosynthesis , RNA, Messenger/biosynthesis , Signal Transduction/genetics , Sodium-Potassium-Exchanging ATPase/genetics , Transcription Factors/biosynthesis , Zinc Finger Protein GLI1
7.
Biomacromolecules ; 16(9): 2672-83, 2015 Sep 14.
Article in English | MEDLINE | ID: mdl-26225909

ABSTRACT

There is intense interest in developing novel methods for the sustained delivery of low levels of clinical therapeutics. MAX8 is a peptide-based beta-hairpin hydrogel that has unique shear thinning properties that allow for immediate rehealing after the removal of shear forces, making MAX8 an excellent candidate for injectable drug delivery at a localized injury site. The current studies examined the feasibility of using MAX8 as a delivery system for nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF), two neurotrophic growth factors currently used in experimental treatments of spinal cord injuries. Experiments determined that encapsulation of NGF and BDNF within MAX8 did not negatively impact gel formation or rehealing and that shear thinning did not result in immediate growth factor release. ELISA, microscopy, rheology, and Western blotting experiments collectively demonstrate the functional capabilities of the therapeutic-loaded hydrogels to (i) maintain a protective environment against in vitro degradation of encapsulated therapeutics for at least 28 days; and (ii) allow for sustained release of NGF and BDGF capable of initiating neurite-like extensions of PC12 cells, most likely due to NGF/BDGF signaling pathways. Importantly, while the 21 day release profiles could be tuned by adjusting the MAX8 hydrogel concentration, the initial shear thinning of the hydrogel (e.g., during injection) does not induce significant premature loss of the encapsulated therapeutic, most likely due to effective trapping of growth factors within structurally robust domains that are maintained during the application of shear forces. Together, our data suggests that MAX8 allows for greater dosage control and sustained therapeutic growth factor delivery, potentially alleviating side effects and improving the efficacy of current therapies.


Subject(s)
Drug Carriers , Hydrogels , Nerve Growth Factor , Peptides , Animals , Drug Carriers/chemistry , Drug Carriers/pharmacology , Humans , Hydrogels/chemistry , Hydrogels/pharmacology , Nerve Growth Factor/chemistry , Nerve Growth Factor/pharmacology , PC12 Cells , Peptides/chemistry , Peptides/pharmacology , Rats
8.
J Cell Sci ; 125(Pt 23): 5711-20, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23077177

ABSTRACT

Na,K-ATPase is a hetero-oligomer of an α- and a ß-subunit. The α-subunit (Na,K-α) possesses the catalytic function, whereas the ß-subunit (Na,K-ß) has cell-cell adhesion function and is localized to the apical junctional complex in polarized epithelial cells. Earlier, we identified two distinct conserved motifs on the Na,K-ß(1) transmembrane domain that mediate protein-protein interactions: a glycine zipper motif involved in the cis homo-oligomerization of Na,K-ß(1) and a heptad repeat motif that is involved in the hetero-oligomeric interaction with Na,K-α(1). We now provide evidence that knockdown of Na,K-ß(1) prevents lumen formation and induces activation of extracellular regulated kinases 1 and 2 (ERK1/2) mediated by phosphatidylinositol 3-kinase in MDCK cells grown in three-dimensional collagen cultures. These cells sustained cell proliferation in an ERK1/2-dependent manner and did not show contact inhibition at high cell densities, as revealed by parental MDCK cells. This phenotype could be rescued by wild-type Na,K-ß(1) or heptad repeat motif mutant of Na,K-ß(1), but not by the glycine zipper motif mutant that abrogates Na,K-ß(1) cis homo-oligomerization. These studies suggest that Na,K-ß(1) cis homo-oligomerization rather than hetero-oligomerization with Na,K-α(1) is involved in epithelial lumen formation. The relevance of these findings to pre-neoplastic lumen filling in epithelial cancer is discussed.


Subject(s)
Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Cell Line , Cell Proliferation , Dogs , Immunoblotting , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Protein Multimerization/genetics , Protein Multimerization/physiology , Sodium-Potassium-Exchanging ATPase/chemistry
9.
Pharmaceuticals (Basel) ; 17(6)2024 May 27.
Article in English | MEDLINE | ID: mdl-38931352

ABSTRACT

Neurofibromatosis type 1 (NF1) is a neurocutaneous disorder. Plexiform neurofibromas (PNFs) are benign tumors commonly formed in patients with NF1. PNFs have a high incidence of developing into malignant peripheral nerve sheath tumors (MPNSTs) with a 5-year survival rate of only 30%. Therefore, the accurate diagnosis and differentiation of MPNSTs from benign PNFs are critical to patient management. We studied a fluorine-18 labeled tryptophan positron emission tomography (PET) radiotracer, 1-(2-[18F]fluoroethyl)-L-tryptophan (L-[18F]FETrp), to detect NF1-associated tumors in an animal model. An ex vivo biodistribution study of L-[18F]FETrp showed a similar tracer distribution and kinetics between the wild-type and triple mutant mice with the highest uptake in the pancreas. Bone uptake was stable. Brain uptake was low during the 90-min uptake period. Static PET imaging at 60 min post-injection showed L-[18F]FETrp had a comparable tumor uptake with [18F]fluorodeoxyglucose (FDG). However, L-[18F]FETrp showed a significantly higher tumor-to-brain ratio than FDG (n = 4, p < 0.05). Sixty-minute-long dynamic PET scans using the two radiotracers showed similar kidney, liver, and lung kinetics. A dysregulated tryptophan metabolism in NF1 mice was further confirmed using immunohistostaining. L-[18F]FETrp is warranted to further investigate differentiating malignant NF1 tumors from benign PNFs. The study may reveal the tryptophan-kynurenine pathway as a therapeutic target for treating NF1.

10.
Exp Eye Res ; 115: 113-22, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23810808

ABSTRACT

Proliferative vitreo retinopathy (PVR) is associated with extracellular matrix membrane (ECM) formation on the neural retina and disruption of the multilayered retinal architecture leading to distorted vision and blindness. During disease progression in PVR, retinal pigmented epithelial cells (RPE) lose cell-cell adhesion, undergo epithelial-to-mesenchymal transition (EMT), and deposit ECM leading to tissue fibrosis. The EMT process is mediated via exposure to vitreous cytokines and growth factors such as TGF-ß2. Previous studies have shown that Na,K-ATPase is required for maintaining a normal polarized epithelial phenotype and that decreased Na,K-ATPase function and subunit levels are associated with TGF-ß1-mediated EMT in kidney cells. In contrast to the basolateral localization of Na,K-ATPase in most epithelia, including kidney, Na,K-ATPase is found on the apical membrane in RPE cells. We now show that EMT is also associated with altered Na,K-ATPase expression in RPE cells. TGF-ß2 treatment of ARPE-19 cells resulted in a time-dependent decrease in Na,K-ATPase ß1 mRNA and protein levels while Na,K-ATPase α1 levels, Na,K-ATPase activity, and intracellular sodium levels remained largely unchanged. In TGF-ß2-treated cells reduced Na,K-ATPase ß1 mRNA inversely correlated with HIF-1α levels and analysis of the Na,K-ATPase ß1 promoter revealed a putative hypoxia response element (HRE). HIF-1α bound to the Na,K-ATPase ß1 promoter and inhibiting the activity of HIF-1α blocked the TGF-ß2 mediated Na,K-ATPase ß1 decrease suggesting that HIF-1α plays a potential role in Na,K-ATPase ß1 regulation during EMT in RPE cells. Furthermore, knockdown of Na,K-ATPase ß1 in ARPE-19 cells was associated with a change in cell morphology from epithelial to mesenchymal and induction of EMT markers such as α-smooth muscle actin and fibronectin, suggesting that loss of Na,K-ATPase ß1 is a potential contributor to TGF-ß2-mediated EMT in RPE cells.


Subject(s)
Epithelial-Mesenchymal Transition/drug effects , Retinal Pigment Epithelium/enzymology , Sodium-Potassium-Exchanging ATPase/metabolism , Transforming Growth Factor beta2/pharmacology , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/physiology , Epithelium/metabolism , Fluorescent Antibody Technique, Indirect , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunoblotting , Microscopy, Confocal , Polymerase Chain Reaction , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retinaldehyde/metabolism , Smad3 Protein/metabolism , Sodium-Potassium-Exchanging ATPase/genetics , Transfection , Transforming Growth Factor beta1/pharmacology
11.
Methods Mol Biol ; 2614: 369-381, 2023.
Article in English | MEDLINE | ID: mdl-36587136

ABSTRACT

Our increased understanding of how a cell's microenvironment influences its behavior has fueled an interest in three-dimensional (3D) cell cultures for drug discovery. Particularly, scaffold-based 3D cultures are expected to recapitulate in vivo tissue stiffness and extracellular matrix composition more accurately than standard two-dimensional (2D) monolayer cultures. Here we present a 3D hydrogel cell culture setup suitable for automated screening with standard high-throughput screening (HTS) liquid handling equipment commonly found in a drug discovery laboratory. Further, we describe the steps required to validate the assay system for compound screening.


Subject(s)
Drug Discovery , Hydrogels , Drug Discovery/methods , High-Throughput Screening Assays/methods , Cell Culture Techniques/methods , Extracellular Matrix
12.
Am J Physiol Lung Cell Mol Physiol ; 302(11): L1150-8, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22345575

ABSTRACT

Diminished Na,K-ATPase expression has been reported in several carcinomas and has been linked to tumor progression. However, few studies have determined whether Na,K-ATPase function and expression are altered in lung malignancies. Because cigarette smoke (CS) is a major factor underlying lung carcinogenesis and progression, we investigated whether CS affects Na,K-ATPase activity and expression in lung cell lines. Cells exposed to CS in vitro showed a reduction of Na,K-ATPase activity. We detected the presence of reactive oxygen species (ROS) in cells exposed to CS before Na,K-ATPase inhibition, and neutralization of ROS restored Na,K-ATPase activity. We further determined whether Na,K-ATPase expression correlated with increasing grades of lung adenocarcinoma and survival of patients with smoking history. Immunohistochemical analysis of lung adenocarcinoma tissues revealed reduced Na,K-ATPase expression with increasing tumor grade. Using tissue microarray containing lung adenocarcinomas of patients with known smoking status, we found that high expression of Na,K-ATPase correlated with better survival. For the first time, these data demonstrate that CS is associated with loss of Na,K-ATPase function and expression in lung carcinogenesis, which might contribute to disease progression.


Subject(s)
Adenocarcinoma/enzymology , Lung Neoplasms/enzymology , Nicotiana , Smoke/adverse effects , Sodium-Potassium-Exchanging ATPase/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Cell Line, Tumor , Disease Progression , Disease-Free Survival , Humans , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Neoplasm Grading , Reactive Oxygen Species/metabolism , Smoking/adverse effects , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/biosynthesis
13.
BMC Cancer ; 12: 44, 2012 Jan 26.
Article in English | MEDLINE | ID: mdl-22280307

ABSTRACT

BACKGROUND: Curcumin (diferuloylmethane), the yellow pigment in the Asian spice turmeric, is a hydrophobic polyphenol from the rhizome of Curcuma longa. Because of its chemopreventive and chemotherapeutic potential with no discernable side effects, it has become one of the major natural agents being developed for cancer therapy. Accumulating evidence suggests that curcumin induces cell death through activation of apoptotic pathways and inhibition of cell growth and proliferation. The mitotic checkpoint, or spindle assembly checkpoint (SAC), is the major cell cycle control mechanism to delay the onset of anaphase during mitosis. One of the key regulators of the SAC is the anaphase promoting complex/cyclosome (APC/C) which ubiquitinates cyclin B and securin and targets them for proteolysis. Because APC/C not only ensures cell cycle arrest upon spindle disruption but also promotes cell death in response to prolonged mitotic arrest, it has become an attractive drug target in cancer therapy. METHODS: Cell cycle profiles were determined in control and curcumin-treated medulloblastoma and various other cancer cell lines. Pull-down assays were used to confirm curcumin binding. APC/C activity was determined using an in vitro APC activity assay. RESULTS: We identified Cdc27/APC3, a component of the APC/C, as a novel molecular target of curcumin and showed that curcumin binds to and crosslinks Cdc27 to affect APC/C function. We further provide evidence that curcumin preferably induces apoptosis in cells expressing phosphorylated Cdc27 usually found in highly proliferating cells. CONCLUSIONS: We report that curcumin directly targets the SAC to induce apoptosis preferably in cells with high levels of phosphorylated Cdc27. Our studies provide a possible molecular mechanism why curcumin induces apoptosis preferentially in cancer cells and suggest that phosphorylation of Cdc27 could be used as a biomarker to predict the therapeutic response of cancer cells to curcumin.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Cycle Proteins/metabolism , Cerebellar Neoplasms/drug therapy , Curcumin/pharmacology , Medulloblastoma/drug therapy , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cerebellar Neoplasms/metabolism , Humans , Medulloblastoma/metabolism , Phosphorylation/physiology
14.
Cancers (Basel) ; 14(14)2022 Jul 13.
Article in English | MEDLINE | ID: mdl-35884462

ABSTRACT

Central nervous system tumors are the most common pediatric solid tumors; they are also the most lethal. Unlike adults, childhood brain tumors are mostly primary in origin and differ in type, location and molecular signature. Tumor characteristics (incidence, location, and type) vary with age. Children present with a variety of symptoms, making early accurate diagnosis challenging. Neuroimaging is key in the initial diagnosis and monitoring of pediatric brain tumors. Conventional anatomic imaging approaches (computed tomography (CT) and magnetic resonance imaging (MRI)) are useful for tumor detection but have limited utility differentiating tumor types and grades. Advanced MRI techniques (diffusion-weighed imaging, diffusion tensor imaging, functional MRI, arterial spin labeling perfusion imaging, MR spectroscopy, and MR elastography) provide additional and improved structural and functional information. Combined with positron emission tomography (PET) and single-photon emission CT (SPECT), advanced techniques provide functional information on tumor metabolism and physiology through the use of radiotracer probes. Radiomics and radiogenomics offer promising insight into the prediction of tumor subtype, post-treatment response to treatment, and prognostication. In this paper, a brief review of pediatric brain cancers, by type, is provided with a comprehensive description of advanced imaging techniques including clinical applications that are currently utilized for the assessment and evaluation of pediatric brain tumors.

15.
Expert Opin Drug Discov ; 16(8): 841-850, 2021 08.
Article in English | MEDLINE | ID: mdl-33823728

ABSTRACT

INTRODUCTION: The high failure rate in drug discovery remains a costly and time-consuming challenge. Improving the odds of success in the early steps of drug development requires disease models with high biological relevance for biomarker discovery and drug development. The adoption of three-dimensional (3D) cell culture systems over traditional monolayers in cell-based assays is considered a promising step toward improving the success rate in drug discovery. AREAS COVERED: In this article, the author focuses on new technologies for 3D cell culture and their applications in cancer drug discovery. Besides the most common 3D cell-culture systems for tumor cells, the article emphasizes the need for 3D cell culture technologies that can mimic the complex tumor microenvironment and cancer stem cell niche. EXPERT OPINION: There has been a rapid increase in 3D cell culture technologies in recent years in an effort to more closely mimic in vivo physiology. Each 3D cell culture system has its own strengths and weaknesses with regard to in vivo tumor growth and the tumor microenvironment. This requires careful consideration of which 3D cell culture system is chosen for drug discovery and should be based on factors like drug target and tumor origin.


Subject(s)
Antineoplastic Agents , Neoplasms , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Culture Techniques, Three Dimensional , Drug Discovery , Humans , Neoplasms/drug therapy , Spheroids, Cellular , Tumor Microenvironment
16.
Front Oncol ; 11: 620831, 2021.
Article in English | MEDLINE | ID: mdl-33869004

ABSTRACT

After leukemia, tumors of the brain and spine are the second most common form of cancer in children. Despite advances in treatment, brain tumors remain a leading cause of death in pediatric cancer patients and survivors often suffer from life-long consequences of side effects of therapy. The 5-year survival rates, however, vary widely by tumor type, ranging from over 90% in more benign tumors to as low as 20% in the most aggressive forms such as glioblastoma. Even within historically defined tumor types such as medulloblastoma, molecular analysis identified biologically heterogeneous subgroups each with different genetic alterations, age of onset and prognosis. Besides molecularly driven patient stratification to tailor disease risk to therapy intensity, such a diversity demonstrates the need for more precise and disease-relevant pediatric brain cancer models for research and drug development. Here we give an overview of currently available in vitro and in vivo pediatric brain tumor models and discuss the opportunities that new technologies such as 3D cultures and organoids that can bridge limitations posed by the simplicity of monolayer cultures and the complexity of in vivo models, bring to accommodate better precision in drug development for pediatric brain tumors.

17.
J Vis Exp ; (175)2021 09 21.
Article in English | MEDLINE | ID: mdl-34633385

ABSTRACT

The kynurenine pathway (KP) is a primary route for tryptophan metabolism. Evidence strongly suggests that metabolites of the KP play a vital role in tumor proliferation, epilepsy, neurodegenerative diseases, and psychiatric illnesses due to their immune-modulatory, neuro-modulatory, and neurotoxic effects. The most extensively used positron emission tomography (PET) agent for mapping tryptophan metabolism, α-[11C]methyl-L-tryptophan ([11C]AMT), has a short half-life of 20 min with laborious radiosynthesis procedures. An onsite cyclotron is required to radiosynthesize [11C]AMT. Only a limited number of centers produce [11C]AMT for preclinical studies and clinical investigations. Hence, the development of an alternative imaging agent that has a longer half-life, favorable in vivo kinetics, and is easy to automate is urgently needed. The utility and value of 1-(2-[18F]fluoroethyl)-L-tryptophan, a fluorine-18-labeled tryptophan analog, has been reported in preclinical applications in cell line-derived xenografts, patient-derived xenografts, and transgenic tumor models. This paper presents a protocol for the radiosynthesis of 1-(2-[18F]fluoroethyl)-L-tryptophan using a one-pot, two-step strategy. Using this protocol, the radiotracer can be produced in a 20 ± 5% (decay corrected at the end of synthesis, n > 20) radiochemical yield, with both radiochemical purity and enantiomeric excess of over 95%. The protocol features a small precursor amount with no more than 0.5 mL of reaction solvent in each step, low loading of potentially toxic 4,7,13,16,21,24-hexaoxa-1,10-diazabicyclo[8.8.8]hexacosane (K222), and an environmentally benign and injectable mobile phase for purification. The protocol can be easily configured to produce 1-(2-[18F]fluoroethyl)-L-tryptophan for clinical investigation in a commercially available module.


Subject(s)
Radiopharmaceuticals , Tryptophan , Humans , Kynurenine , Positron-Emission Tomography , Radiochemistry
18.
Sci Rep ; 10(1): 3800, 2020 03 02.
Article in English | MEDLINE | ID: mdl-32123231

ABSTRACT

In vivo positron emission tomography (PET) imaging is a key modality to evaluate disease status of brain tumors. In recent years, tremendous efforts have been made in developing PET imaging methods for pediatric brain tumors. Carbon-11 labelled tryptophan derivatives are feasible as PET imaging probes in brain tumor patients with activation of the kynurenine pathway, but the short half-life of carbon-11 limits its application. Using a transgenic mouse model for the sonic hedgehog (Shh) subgroup of medulloblastoma, here we evaluated the potential of the newly developed 1-(2-[18F]fluoroethyl)-L-tryptophan (1-L-[18F]FETrp) as a PET imaging probe for this common malignant pediatric brain tumor. 1-L-[18F]FETrp was synthesized on a PETCHEM automatic synthesizer with good chemical and radiochemical purities and enantiomeric excess values. Imaging was performed in tumor-bearing Smo/Smo medulloblastoma mice with constitutive actvation of the Smoothened (Smo) receptor using a PerkinElmer G4 PET-X-Ray scanner. Medulloblastoma showed significant and specific accumulation of 1-L-[18F]FETrp. 1-L-[18F]FETrp also showed significantly higher tumor uptake than its D-enantiomer, 1-D-[18F]FETrp. The uptake of 1-L-[18F]FETrp in the normal brain tissue was low, suggesting that 1-L-[18F]FETrp may prove a valuable PET imaging probe for the Shh subgroup of medulloblastoma and possibly other pediatric and adult brain tumors.


Subject(s)
Brain Neoplasms/diagnostic imaging , Fluorine Radioisotopes/chemistry , Medulloblastoma/diagnostic imaging , Radiopharmaceuticals/chemistry , Tryptophan/analogs & derivatives , Animals , Biological Transport , Fluorine Radioisotopes/metabolism , Humans , Medulloblastoma/metabolism , Mice , Mice, Transgenic , Positron-Emission Tomography , Radiopharmaceuticals/metabolism , Tryptophan/metabolism
19.
Front Pediatr ; 7: 314, 2019.
Article in English | MEDLINE | ID: mdl-31417884

ABSTRACT

The development of therapies aimed at leukemia has progressed substantially in the past years but childhood acute myeloid leukemia (AML) remains one of the most challenging cancers to treat. Genomic profiling of AML has greatly enhanced our understanding of the genetic and epigenetic landscape of this high-risk leukemia. With it comes the opportunity to develop targeted therapies that are expected to be more effective and less toxic than current treatment regimens. Nevertheless, often overlooked in leukemia drug discovery are the dynamic interactions between leukemic cells and the bone marrow environment. The interplay between leukemic cells, stromal cells and the extracellular matrix plays critical roles in the development, progression and relapse of AML as well as in drug response and the development of resistance. Here we will review pediatric leukemia with a special focus on acute myeloid disease in children, and discuss the tumor microenvironment in the context of drug resistance and leukemia stem cell survival. We will emphasize how three-dimensional (3D) cell-based drug discovery may offer hope for both the identification and advancement of more effective treatment options for patients suffering from this devastating disease.

20.
Mol Neurobiol ; 56(11): 7557-7571, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31062247

ABSTRACT

The Na,K-ATPase, consisting of a catalytic α-subunit and a regulatory ß-subunit, is a ubiquitously expressed ion pump that carries out the transport of Na+ and K+ across the plasma membranes of most animal cells. In addition to its pump function, Na,K-ATPase serves as a signaling scaffold and a cell adhesion molecule. Of the three ß-subunit isoforms, ß1 is found in almost all tissues, while ß2 expression is mostly restricted to brain and muscle. In cerebellar granule cells, the ß2-subunit, also known as adhesion molecule on glia (AMOG), has been linked to neuron-astrocyte adhesion and granule cell migration, suggesting its role in cerebellar development. Nevertheless, little is known about molecular pathways that link the ß2-subunit to its cellular functions. Using cerebellar granule precursor cells, we found that the ß2-subunit, but not the ß1-subunit, negatively regulates the expression of a key activator of the Hippo/YAP signaling pathway, Merlin/neurofibromin-2 (NF2). The knockdown of the ß2-subunit resulted in increased Merlin/NF2 expression and affected downstream targets of Hippo signaling, i.e., increased YAP phosphorylation and decreased expression of N-Ras. Further, the ß2-subunit knockdown altered the kinetics of epidermal growth factor receptor (EGFR) signaling in a Merlin-dependent mode and impaired EGF-induced reorganization of the actin cytoskeleton. Therefore, our studies for the first time provide a functional link between the Na,K-ATPase ß2-subunit and Merlin/NF2 and suggest a role for the ß2-subunit in regulating cytoskeletal dynamics and Hippo/YAP signaling during neuronal differentiation.


Subject(s)
Cerebellum/cytology , Cytoplasmic Granules/metabolism , Epidermal Growth Factor/metabolism , Neurofibromin 2/metabolism , Protein Subunits/metabolism , Signal Transduction , Sodium-Potassium-Exchanging ATPase/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Adhesion/drug effects , Cell Cycle Proteins/metabolism , Cell Differentiation/drug effects , Cell Line , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Ion Transport/drug effects , Isoenzymes/metabolism , Mice, Inbred C57BL , Models, Biological , Monomeric GTP-Binding Proteins/metabolism , Phosphorylation/drug effects , Protein Binding/drug effects , YAP-Signaling Proteins
SELECTION OF CITATIONS
SEARCH DETAIL