92 results on '"Peter C. Hart"'
Search Results
2. A biofunctional review of C-reactive protein (CRP) as a mediator of inflammatory and immune responses: differentiating pentameric and modified CRP isoform effects
- Author
-
Margaret E. Olson, Mary G. Hornick, Ashley Stefanski, Haya R. Albanna, Alesia Gjoni, Griffin D. Hall, Peter C. Hart, Ibraheem M. Rajab, and Lawrence A. Potempa
- Subjects
CRP isoforms ,mCRP ,inflammation ,C-reactive protein ,innate immunity ,complement ,Immunologic diseases. Allergy ,RC581-607 - Abstract
C-reactive protein (CRP) is an acute phase, predominantly hepatically synthesized protein, secreted in response to cytokine signaling at sites of tissue injury or infection with the physiological function of acute pro-inflammatory response. Historically, CRP has been classified as a mediator of the innate immune system, acting as a pattern recognition receptor for phosphocholine-containing ligands. For decades, CRP was envisioned as a single, non-glycosylated, multi-subunit protein arranged non-covalently in cyclic symmetry around a central void. Over the past few years, however, CRP has been shown to exist in at least three distinct isoforms: 1.) a pentamer of five identical globular subunits (pCRP), 2.) a modified monomer (mCRP) resulting from a conformational change when subunits are dissociated from the pentamer, and 3.) a transitional isoform where the pentamer remains intact but is partially changed to express mCRP structural characteristics (referred to as pCRP* or mCRPm). The conversion of pCRP into mCRP can occur spontaneously and is observed under commonly used experimental conditions. In careful consideration of experimental design used in published reports of in vitro pro- and anti-inflammatory CRP bioactivities, we herein provide an interpretation of how distinctive CRP isoforms may have affected reported results. We argue that pro-inflammatory amplification mechanisms are consistent with the biofunction of mCRP, while weak anti-inflammatory mechanisms are consistent with pCRP. The interplay of each CRP isoform with specific immune cells (platelets, neutrophils, monocytes, endothelial cells, natural killer cells) and mechanisms of the innate immune system (complement), as well as differences in mCRP and pCRP ligand recognition and effector functions are discussed. This review will serve as a revised understanding of the structure-function relationship between CRP isoforms as related to inflammation and innate immunity mechanisms.
- Published
- 2023
- Full Text
- View/download PDF
3. C-Reactive Protein and Cancer: Interpreting the Differential Bioactivities of Its Pentameric and Monomeric, Modified Isoforms
- Author
-
Lawrence A. Potempa, Ibraheem M. Rajab, Margaret E. Olson, and Peter C. Hart
- Subjects
CRP isoforms ,mCRP ,cancer ,inflammation ,C-reactive protein ,anti-cancer ,Immunologic diseases. Allergy ,RC581-607 - Abstract
C-reactive protein (CRP) was first recognized in the 1940s as a protein that appeared in blood during acute episodes of infectious disease. Its presence and pharmacodynamics were found in essentially all diseases that involved tissue damage and inflammation. Identified as a major component of the innate, unlearned immunity, it became a useful diagnostic marker for the extent of inflammation during disease exacerbation or remission. Efforts to define its true biological role has eluded clear definition for over a half-century. Herein, a unifying concept is presented that explains both pro-inflammatory and anti-inflammatory activities of CRP. This concept involves the recognition and understanding that CRP can be induced to undergo a pronounced, non-proteolytic reorganization of its higher-level protein structures into conformationally distinct isomers with distinctive functional activities. This process occurs when the non-covalently associated globular subunits of the pentameric isoform (“pCRP”) are induced to dissociate into a monomeric isoform (“mCRP”). mCRP consistently and potently provides pro-inflammatory activation and amplification activities. pCRP provides weak anti-inflammatory activities consistent with low-level chronic inflammation. mCRP can spontaneously form in purified pCRP reagents in ways that are not immediately recognized during purification and certification analyses. By now understanding the factors that influence pCRP dissociate into mCRP, many published reports investigating CRP as a biological response modifier of host defense can be reevaluated to include a discussion of how each CRP isoform may have affected the generated results. Specific attention is given to in vitro and in vivo studies of CRP as an anti-cancer agent.
- Published
- 2021
- Full Text
- View/download PDF
4. C-Reactive Protein and Cancer—Diagnostic and Therapeutic Insights
- Author
-
Peter C. Hart, Ibraheem M. Rajab, May Alebraheem, and Lawrence A. Potempa
- Subjects
C-reactive protein ,monomeric C-reactive protein ,inflammation ,tumor microenvironment ,acute phase response ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Cancer disease describes any pathology involving uncontrolled cell growth. As cells duplicate, they can remain localized in defined tissues, forming tumor masses and altering their microenvironmental niche, or they can disseminate throughout the body in a metastatic process affecting multiple tissues and organs. As tumors grow and metastasize, they affect normal tissue integrity and homeostasis which signals the body to trigger the acute phase inflammatory response. C-reactive protein (CRP) is a predominant protein of the acute phase response; its blood levels have long been used as a minimally invasive index of any ongoing inflammatory response, including that occurring in cancer. Its diagnostic significance in assessing disease progression or remission, however, remains undefined. By considering the recent understanding that CRP exists in multiple isoforms with distinct biological activities, a unified model is advanced that describes the relevance of CRP as a mediator of host defense responses in cancer. CRP in its monomeric, modified isoform (mCRP) modulates inflammatory responses by inserting into activated cell membranes and stimulating platelet and leukocyte responses associated with acute phase responses to tumor growth. It also binds components of the extracellular matrix in involved tissues. Conversely, CRP in its pentameric isoform (pCRP), which is the form quantified in diagnostic measurements of CRP, is notably less bioactive with weak anti-inflammatory bioactivity. Its accumulation in blood is associated with a continuous, low-level inflammatory response and is indicative of unresolved and advancing disease, as occurs in cancer. Herein, a novel interpretation of the diagnostic utility of CRP is presented accounting for the unique properties of the CRP isoforms in the context of the developing pro-metastatic tumor microenvironment.
- Published
- 2020
- Full Text
- View/download PDF
5. How C-Reactive Protein Structural Isoforms With Distinctive Bioactivities Affect Disease Progression
- Author
-
Ibraheem M. Rajab, Peter C. Hart, and Lawrence A. Potempa
- Subjects
CRP - C-reactive protein ,MCRP ,inflammation ,conformational isoforms ,blood kinetics ,modified/monomeric (mCRP) ,Immunologic diseases. Allergy ,RC581-607 - Abstract
C-reactive protein (CRP) is a widely known, hepatically synthesized protein whose blood levels change rapidly and pronouncedly in response to any tissue damaging event associated with an inflammatory response. The synthesis and secretion of CRP is stimulated by interleukin-6, an early pleiotropic cytokine released by macrophages, endothelial, and other cells that are activated when localized normal tissue structures are compromised by trauma or disease. Serum CRP levels can change rapidly and robustly from 10-100-fold within 6–72 h of any tissue damaging event. Elevated blood levels correlate with the onset and extent of both activated inflammation and the acute phase biochemical response to the tissue insult. Because its functional bioactivity as the prototypic acute phase reactant has eluded clear definition for decades, diagnosticians of various conditions and diseases use CRP blood levels as a simple index for ongoing inflammation. In many pathologies, which involves many different tissues, stages of disease, treatments, and responses to treatments, its interpretive diagnostic value requires a deeper understanding of the localized tissue processes and events that contribute signals which regulate protective or pathological host defense bioactivities. This report presents concepts that describe how local tissue activation events can lead to a non-proteolytic, conformational rearrangement of CRP into a unique isoform with distinctive solubility, antigenicity, binding reactivities and bioactivities from that protein widely known and measured in serum. By describing factors that control the expression, tissue localization, half-life and pro-inflammatory amplification activity of this CRP isoform, a unifying explanation for the diagnostic significance of CRP measurement in disease is advanced.
- Published
- 2020
- Full Text
- View/download PDF
6. Mesothelial Cell HIF1α Expression Is Metabolically Downregulated by Metformin to Prevent Oncogenic Tumor-Stromal Crosstalk
- Author
-
Peter C. Hart, Hilary A. Kenny, Niklas Grassl, Karen M. Watters, Lacey M. Litchfield, Fabian Coscia, Ivana Blaženović, Lisa Ploetzky, Oliver Fiehn, Matthias Mann, Ernst Lengyel, and Iris L. Romero
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: The tumor microenvironment (TME) plays a pivotal role in cancer progression, and, in ovarian cancer (OvCa), the primary TME is the omentum. Here, we show that the diabetes drug metformin alters mesothelial cells in the omental microenvironment. Metformin interrupts bidirectional signaling between tumor and mesothelial cells by blocking OvCa cell TGF-β signaling and mesothelial cell production of CCL2 and IL-8. Inhibition of tumor-stromal crosstalk by metformin is caused by the reduced expression of the tricarboxylic acid (TCA) enzyme succinyl CoA ligase (SUCLG2). Through repressing this TCA enzyme and its metabolite, succinate, metformin activated prolyl hydroxylases (PHDs), resulting in the degradation of hypoxia-inducible factor 1α (HIF1α) in mesothelial cells. Disruption of HIF1α-driven IL-8 signaling in mesothelial cells by metformin results in reduced OvCa invasion in an organotypic 3D model. These findings indicate that tumor-promoting signaling between mesothelial and OvCa cells in the TME can be targeted using metformin. : Hart et al. identify that the type 2 diabetes drug metformin inhibits ovarian cancer invasion by targeting crosstalk between cancer cells and adjacent normal stromal mesothelial cells, making the microenvironment less hospitable to cancer growth. Keywords: mesothelial cells, tumor microenvironment, ovarian cancer, metformin, HIF1, SUCLG2, IL-8, TGF, omental metastasis, succinate
- Published
- 2019
- Full Text
- View/download PDF
7. Fig S3 from Loss of BRCA1 in the Cells of Origin of Ovarian Cancer Induces Glycolysis: A Window of Opportunity for Ovarian Cancer Chemoprevention
- Author
-
Iris L. Romero, Ernst Lengyel, Olufunmilayo I. Olopade, S. Diane Yamada, Yusuke Nakamura, Ryuji Hamamoto, Ricardo R. Lastra, Stephanie M. McGregor, Mark A. Eckert, Peter C. Hart, and Tatsuyuki Chiyoda
- Abstract
Fig S3 shows BRCA1 knockdown in IOSE and FT does not increase HK1 expression or AMPK activity.
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Figure 6 from SPHK1 Is a Novel Target of Metformin in Ovarian Cancer
- Author
-
Iris L. Romero, Ernst Lengyel, Jason W. Locasale, Stephanie M. McGregor, Ricardo Lastra, Rachel Loth, Chun-Yi Chiang, Marion Curtis, Melanie Weigert, Xiaojing Liu, Tatsuyuki Chiyoda, and Peter C. Hart
- Abstract
Silencing SPHK1 promotes resistance to metformin-induced cytotoxicity.
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Figure 2 from SPHK1 Is a Novel Target of Metformin in Ovarian Cancer
- Author
-
Iris L. Romero, Ernst Lengyel, Jason W. Locasale, Stephanie M. McGregor, Ricardo Lastra, Rachel Loth, Chun-Yi Chiang, Marion Curtis, Melanie Weigert, Xiaojing Liu, Tatsuyuki Chiyoda, and Peter C. Hart
- Abstract
Analysis of SPHK1 expression in multiple OvCa cell lines.
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Figure 7 from SPHK1 Is a Novel Target of Metformin in Ovarian Cancer
- Author
-
Iris L. Romero, Ernst Lengyel, Jason W. Locasale, Stephanie M. McGregor, Ricardo Lastra, Rachel Loth, Chun-Yi Chiang, Marion Curtis, Melanie Weigert, Xiaojing Liu, Tatsuyuki Chiyoda, and Peter C. Hart
- Abstract
SPHK1 expression does not alter other markers of metformin sensitivity.
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Figure 8 from SPHK1 Is a Novel Target of Metformin in Ovarian Cancer
- Author
-
Iris L. Romero, Ernst Lengyel, Jason W. Locasale, Stephanie M. McGregor, Ricardo Lastra, Rachel Loth, Chun-Yi Chiang, Marion Curtis, Melanie Weigert, Xiaojing Liu, Tatsuyuki Chiyoda, and Peter C. Hart
- Abstract
Knockdown of SPHK1 in OvCa cells.
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Figure 1 from SPHK1 Is a Novel Target of Metformin in Ovarian Cancer
- Author
-
Iris L. Romero, Ernst Lengyel, Jason W. Locasale, Stephanie M. McGregor, Ricardo Lastra, Rachel Loth, Chun-Yi Chiang, Marion Curtis, Melanie Weigert, Xiaojing Liu, Tatsuyuki Chiyoda, and Peter C. Hart
- Abstract
Analysis of S1P-related enzymes in two OvCa cell lines.
- Published
- 2023
- Full Text
- View/download PDF
13. Supplementary Figure 3 from SPHK1 Is a Novel Target of Metformin in Ovarian Cancer
- Author
-
Iris L. Romero, Ernst Lengyel, Jason W. Locasale, Stephanie M. McGregor, Ricardo Lastra, Rachel Loth, Chun-Yi Chiang, Marion Curtis, Melanie Weigert, Xiaojing Liu, Tatsuyuki Chiyoda, and Peter C. Hart
- Abstract
SPHK1 promotes proliferation, migration and clonogenicity in a representative HGSOC cell line.
- Published
- 2023
- Full Text
- View/download PDF
14. Supplementary Figure Legends from Loss of BRCA1 in the Cells of Origin of Ovarian Cancer Induces Glycolysis: A Window of Opportunity for Ovarian Cancer Chemoprevention
- Author
-
Iris L. Romero, Ernst Lengyel, Olufunmilayo I. Olopade, S. Diane Yamada, Yusuke Nakamura, Ryuji Hamamoto, Ricardo R. Lastra, Stephanie M. McGregor, Mark A. Eckert, Peter C. Hart, and Tatsuyuki Chiyoda
- Abstract
Legends for supplementary figures.
- Published
- 2023
- Full Text
- View/download PDF
15. Supplementary Figure 4 from SPHK1 Is a Novel Target of Metformin in Ovarian Cancer
- Author
-
Iris L. Romero, Ernst Lengyel, Jason W. Locasale, Stephanie M. McGregor, Ricardo Lastra, Rachel Loth, Chun-Yi Chiang, Marion Curtis, Melanie Weigert, Xiaojing Liu, Tatsuyuki Chiyoda, and Peter C. Hart
- Abstract
Metformin inhibits induction of SPHK1 transcription by HIFs.
- Published
- 2023
- Full Text
- View/download PDF
16. Data from SPHK1 Is a Novel Target of Metformin in Ovarian Cancer
- Author
-
Iris L. Romero, Ernst Lengyel, Jason W. Locasale, Stephanie M. McGregor, Ricardo Lastra, Rachel Loth, Chun-Yi Chiang, Marion Curtis, Melanie Weigert, Xiaojing Liu, Tatsuyuki Chiyoda, and Peter C. Hart
- Abstract
The role of phospholipid signaling in ovarian cancer is poorly understood. Sphingosine-1-phosphate (S1P) is a bioactive metabolite of sphingosine that has been associated with tumor progression through enhanced cell proliferation and motility. Similarly, sphingosine kinases (SPHK), which catalyze the formation of S1P and thus regulate the sphingolipid rheostat, have been reported to promote tumor growth in a variety of cancers. The findings reported here show that exogenous S1P or overexpression of SPHK1 increased proliferation, migration, invasion, and stem-like phenotypes in ovarian cancer cell lines. Likewise, overexpression of SPHK1 markedly enhanced tumor growth in a xenograft model of ovarian cancer, which was associated with elevation of key markers of proliferation and stemness. The diabetes drug, metformin, has been shown to have anticancer effects. Here, we found that ovarian cancer patients taking metformin had significantly reduced serum S1P levels, a finding that was recapitulated when ovarian cancer cells were treated with metformin and analyzed by lipidomics. These findings suggested that in cancer the sphingolipid rheostat may be a novel metabolic target of metformin. In support of this, metformin blocked hypoxia-induced SPHK1, which was associated with inhibited nuclear translocation and transcriptional activity of hypoxia-inducible factors (HIF1α and HIF2α). Further, ovarian cancer cells with high SPHK1 were found to be highly sensitive to the cytotoxic effects of metformin, whereas ovarian cancer cells with low SPHK1 were resistant. Together, the findings reported here show that hypoxia-induced SPHK1 expression and downstream S1P signaling promote ovarian cancer progression and that tumors with high expression of SPHK1 or S1P levels might have increased sensitivity to the cytotoxic effects of metformin.Implications:Metformin targets sphingolipid metabolism through inhibiting SPHK1, thereby impeding ovarian cancer cell migration, proliferation, and self-renewal.
- Published
- 2023
- Full Text
- View/download PDF
17. Supplementary Figure 5 from SPHK1 Is a Novel Target of Metformin in Ovarian Cancer
- Author
-
Iris L. Romero, Ernst Lengyel, Jason W. Locasale, Stephanie M. McGregor, Ricardo Lastra, Rachel Loth, Chun-Yi Chiang, Marion Curtis, Melanie Weigert, Xiaojing Liu, Tatsuyuki Chiyoda, and Peter C. Hart
- Abstract
Metformin prevents nuclear localization of HIF1α in Kuramochi cells.
- Published
- 2023
- Full Text
- View/download PDF
18. Nuclear-localized, iron-bound superoxide dismutase-2 antagonizes epithelial lineage programs to promote stemness of breast cancer cells via a histone demethylase activity
- Author
-
Diego R. Coelho, Flavio R. Palma, Veronica Paviani, Chenxia He, Jeanne M. Danes, Yunping Huang, Juliana C. P. Calado, Peter C. Hart, Cristina M. Furdui, Leslie B. Poole, Matthew J. Schipma, and Marcelo G. Bonini
- Subjects
Cell Nucleus ,Histone Demethylases ,Multidisciplinary ,Superoxide Dismutase ,Iron ,Neoplastic Stem Cells ,Breast Neoplasms ,Hydrogen Peroxide ,Protein Processing, Post-Translational - Abstract
The dichotomous behavior of superoxide dismutase-2 (SOD2) in cancer biology has long been acknowledged and more recently linked to different posttranslational forms of the enzyme. However, a distinctive activity underlying its tumor-promoting function is yet to be described. Here, we report that acetylation, one of such posttranslational modifications (PTMs), increases SOD2 affinity for iron, effectively changing the biochemical function of this enzyme from that of an antioxidant to a demethylase. Acetylated, iron-bound SOD2 localizes to the nucleus, promoting stem cell gene expression via removal of suppressive epigenetic marks such as H3K9me3 and H3K927me3. Particularly, H3K9me3 was specifically removed from regulatory regions upstream of Nanog and Oct-4, two pluripotency factors involved in cancer stem cell reprogramming. Phenotypically, cells expressing nucleus-targeted SOD2 (NLS-SOD2) have increased clonogenicity and metastatic potential. FeSOD2 operating as H3 demethylase requires H 2 O 2 as substrate, which unlike cofactors of canonical demethylases (i.e., oxygen and 2-oxoglutarate), is more abundant in tumor cells than in normal tissue. Therefore, our results indicate that FeSOD2 is a demethylase with unique activities and functions in the promotion of cancer evolution toward metastatic phenotypes.
- Published
- 2023
19. Systemic maternal inflammation promotes ASD via IL-6 and IFN-γ
- Author
-
Daniel Majerczyk, Elizabeth G. Ayad, Kari L. Brewton, Pichrasmei Saing, and Peter C. Hart
- Subjects
Inflammation ,Autism Spectrum Disorder ,Interleukin-6 ,Placenta ,Biophysics ,Cell Biology ,Biochemistry ,Interferon-gamma ,Pregnancy ,Prenatal Exposure Delayed Effects ,Animals ,Humans ,Cytokines ,Female ,Molecular Biology - Abstract
Autism spectrum disorder (ASD) is a neurological disorder that manifests during early development, impacting individuals through their ways of communicating, social behaviors, and their ability to perform day-to-day activities. There have been different proposed mechanisms on how ASD precipitates within a patient, one of which being the impact cytokines have on fetal development once a mother’s immune system has been activated (referred to as maternal immune activation, MIA). The occurrence of ASD has long been associated with elevated levels of several cytokines, including interleukin-6 (IL-6) and interferon gamma (IFN-γ). These proinflammatory cytokines can achieve high systemic levels in response to immune activating pathogens from various extrinsic sources. Transfer of cytokines such as IL-6 across the placental barrier allows accumulation in the fetus, potentially inducing neuroinflammation and consequently altering neurodevelopmental processes. Individuals who have been later diagnosed with ASD have been observed to have elevated levels of IL-6 and other proinflammatory cytokines during gestation. Moreover, the outcome of MIA has been associated with neurological effects such as impaired social interaction and an increase in repetitive behavior in animal models, supporting a mechanistic link between gestational inflammation and development of ASD-like characteristics. The present review attempts to provide a concise overview of the available preclinical and clinical data that suggest cross-talk between IL-6 and IFN-γ through both extrinsic and intrinsic factors as a central mechanism of MIA that may promote the development of ASD.
- Published
- 2022
20. The Natural Product β-Escin Targets Cancer and Stromal Cells of the Tumor Microenvironment to Inhibit Ovarian Cancer Metastasis
- Author
-
Manish S. Patankar, Madhu Lal, Kasjusz Kordylewicz, Bikash R. Pattnaik, Hilary A. Kenny, Yousef Alharbi, Niklas Grassl, Karen M. Watters, Min Shen, Yen-Ju Chen, Betul Kara, Peter C. Hart, Ernst Lengyel, and Marc Ferrer
- Subjects
Cancer Research ,Stromal cell ,endocrine system diseases ,Lactate dehydrogenase A ,high-throughput screening ,Article ,Metastasis ,Extracellular matrix ,Digitoxin ,medicine ,tumor microenvironment ,metastasis ,Cell adhesion ,education ,Ouabain ,RC254-282 ,Tumor microenvironment ,education.field_of_study ,Chemistry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Cancer ,medicine.disease ,female genital diseases and pregnancy complications ,ovarian cancer ,Oncology ,Cancer research ,Ovarian cancer ,β-escin - Abstract
Simple Summary β-escin, a component of horse chestnut seed extract, was first identified as an inhibitor of ovarian cancer (OvCa) adhesion/invasion in our high-throughput screening program using a three-dimensional organotypic model assembled from primary human cells and extracellular matrix. The goal of the study presented here is to determine if β-escin and structurally-similar compounds have a therapeutic potential against OvCa metastasis. β-escin and cardiac glycosides inhibit ovarian cancer adhesion/invasion to the omental microenvironment in vivo, and β-escin inhibits ovarian cancer metastasis in the prevention and intervention setting. Additionally, β-escin was found to decrease the stemness of ovarian cancer cells, inhibit extracellular matrix production in the tumor microenvironment, and inhibit HIF1α stability in ovarian cancer cells and the tumor microenvironment. This study reveals that the natural compound β-escin has therapeutic potential because of its ability to prevent OvCa dissemination by targeting both cancer and stromal cells in the OvCa tumor microenvironment. Abstract The high mortality of OvCa is caused by the wide dissemination of cancer within the abdominal cavity. OvCa cells metastasize to the peritoneum, which is covered by mesothelial cells, and invade into the underlying stroma, composed of extracellular matrices (ECM) and stromal cells. In a study using a three-dimensional quantitative high-throughput screening platform (3D-qHTS), we found that β-escin, a component of horse chestnut seed extract, inhibited OvCa adhesion/invasion. Here, we determine whether β-escin and structurally similar compounds have a therapeutic potential against OvCa metastasis. Different sources of β-escin and horse chestnut seed extract inhibited OvCa cell adhesion/invasion, both in vitro and in vivo. From a collection of 160 structurally similar compounds to β-escin, we found that cardiac glycosides inhibited OvCa cell adhesion/invasion and proliferation in vitro, and inhibited adhesion/invasion and metastasis in vivo. Mechanistically, β-escin and the cardiac glycosides inhibited ECM production in mesothelial cells and fibroblasts. The oral administration of β-escin inhibited metastasis in both OvCa prevention and intervention mouse models. Specifically, β-escin inhibited ECM production in the omental tumors. Additionally, the production of HIF1α-targeted proteins, lactate dehydrogenase A, and hexokinase 2 in omental tumors was blocked by β-escin. This study reveals that the natural compound β-escin has a therapeutic potential because of its ability to prevent OvCa dissemination by targeting both cancer and stromal cells in the OvCa tumor microenvironment.
- Published
- 2021
- Full Text
- View/download PDF
21. Modeling the Early Steps of Ovarian Cancer Dissemination in an Organotypic Culture of the Human Peritoneal Cavity
- Author
-
Peter C, Hart, Preety, Bajwa, and Hilary A, Kenny
- Subjects
Ovarian Neoplasms ,Mice ,Cell Line, Tumor ,Tumor Microenvironment ,Animals ,Humans ,Female ,Carcinoma, Ovarian Epithelial ,Peritoneal Cavity ,Epithelium - Abstract
The majority of ovarian cancer patients present clinically with wide-spread metastases throughout the peritoneal cavity, metastasizing to the mesothelium-lined peritoneum and visceral adipose depots within the abdomen. This unique metastatic tumor microenvironment is comprised of multiple cell types, including mesothelial cells, fibroblasts, adipocytes, macrophages, neutrophils, and T lymphocytes. Modeling advancements, including complex 3D systems and organoids, coupled with 2D cocultures, in vivo mouse models, and ex vivo human tissue cultures have greatly enhanced our understanding of the tumor-stroma interactions that are required for successful metastasis of ovarian cancer cells. However, advanced multifaceted model systems that incorporate frequency and spatial distribution of all cell types present in the tumor microenvironment of ovarian cancer are needed to enhance our knowledge of ovarian cancer biology in order to identify methods for preventing and treating metastatic disease. This review highlights the utility of recently developed modeling approaches, summarizes some of the resulting progress using these techniques, and suggests how these strategies may be implemented to elucidate signaling processes among cell types of the tumor microenvironment that promote ovarian cancer metastasis.
- Published
- 2021
22. Insights into the Use of C-Reactive Protein as a Diagnostic Index of Disease Severity in COVID-19 Infections
- Author
-
Rafael Fernandez-Botran, Ibraheem M. Rajab, Lawrence A. Potempa, Peter C Hart, and Jose Bordon
- Subjects
Pneumonia, Viral ,030231 tropical medicine ,Inflammation ,Stimulation ,Perspective Piece ,Betacoronavirus ,03 medical and health sciences ,COVID-19 Testing ,0302 clinical medicine ,Virology ,medicine ,Humans ,Protein Isoforms ,Pandemics ,biology ,Clinical Laboratory Techniques ,SARS-CoV-2 ,Septic shock ,business.industry ,C-reactive protein ,Acute-phase protein ,COVID-19 ,Prognosis ,medicine.disease ,Pneumonia ,C-Reactive Protein ,Infectious Diseases ,Coagulation ,Immunology ,biology.protein ,Parasitology ,medicine.symptom ,Coronavirus Infections ,business ,Cytokine storm - Abstract
Approximately 20% of patients infected with SARS-CoV-2 (COVID-19) develop potentially life-threatening pathologies involving hyperinflammation, cytokine storm, septic shock complications, coagulation dysfunction, and multiple organ failure. Blood levels of the prototypic acute phase reactant, C-reactive protein (CRP), which is hepatically synthesized and released in response to interleukin-6 stimulation, is markedly elevated in patients with COVID-19. Markedly high CRP levels correlate with poor prognosis for survival. Insights into CRP structure–function relationships have uncovered both pro- and anti-inflammatory isoforms that may be used to monitor the extent of tissue damage associated with COVID-19 pathologies and prognoses. Herein, rationale is given for interpretation of CRP blood levels as a simple, rapid, and cost-effective way to assess disease severity and help guide therapeutic options in COVID-19 patients.
- Published
- 2020
- Full Text
- View/download PDF
23. SOD2 acetylation on lysine 68 promotes stem cell reprogramming in breast cancer
- Author
-
Chenxia He, Benjamin N. Gantner, Andre L. P de Abreu, Joseph O'Brien, Jacek Zielonka, Marcelo G. Bonini, Lalage M. Wakefield, Binwu Tang, David Gius, Angela Mathison, Erich Stauder, Raul Urrutia, Jeanne M. Danes, Jonna Frasor, Peter C. Hart, Douglas Ganini, Yueming Zhu, and Yunping Huang
- Subjects
Homeobox protein NANOG ,Multidisciplinary ,SOX2 ,Acetylation ,SOD2 ,Tumor initiation ,Biology ,Stem cell ,skin and connective tissue diseases ,Reprogramming ,Transcription factor ,Cell biology - Abstract
Mitochondrial superoxide dismutase (SOD2) suppresses tumor initiation but promotes invasion and dissemination of tumor cells at later stages of the disease. The mechanism of this functional switch remains poorly defined. Our results indicate that as SOD2 expression increases acetylation of lysine 68 ensues. Acetylated SOD2 promotes hypoxic signaling via increased mitochondrial reactive oxygen species (mtROS). mtROS, in turn, stabilize hypoxia-induced factor 2α (HIF2α), a transcription factor upstream of "stemness" genes such as Oct4, Sox2, and Nanog. In this sense, our findings indicate that SOD2K68Ac and mtROS are linked to stemness reprogramming in breast cancer cells via HIF2α signaling. Based on these findings we propose that, as tumors evolve, the accumulation of SOD2K68Ac turns on a mitochondrial pathway to stemness that depends on HIF2α and may be relevant for the progression of breast cancer toward poor outcomes.
- Published
- 2019
- Full Text
- View/download PDF
24. Inhibition of fascin in cancer and stromal cells blocks ovarian cancer metastasis
- Author
-
Xin-Yun Huang, Peter C. Hart, Anthony G. Montag, Hilary A. Kenny, Betul Kara, Sean McGuire, Sarah Fazal, Kristen Wroblewski, and Ernst Lengyel
- Subjects
0301 basic medicine ,Stromal cell ,endocrine system diseases ,Mice, Nude ,macromolecular substances ,Carcinoma, Ovarian Epithelial ,Epithelium ,Article ,Metastasis ,Small Molecule Libraries ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cancer-Associated Fibroblasts ,Cell Movement ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Neoplasm Metastasis ,RNA, Small Interfering ,Fascin ,Tumor microenvironment ,biology ,business.industry ,Microfilament Proteins ,Obstetrics and Gynecology ,Cancer ,medicine.disease ,Xenograft Model Antitumor Assays ,female genital diseases and pregnancy complications ,030104 developmental biology ,Oncology ,Gene Knockdown Techniques ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,biology.protein ,Female ,Stromal Cells ,Carrier Proteins ,Ovarian cancer ,business ,Filopodia - Abstract
Objective Ovarian cancer (OvCa) metastasis requires the coordinated motility of both cancer and stromal cells. Cellular movement is a dynamic process that involves the synchronized assembly of f-actin bundles into cytoskeletal protrusions by fascin. Fascin directly binds f-actin and is an integral component of filopodia, lamellapodia and stress fibers. Here, we examine the expression pattern and function of fascin in the cancer and stromal cells of OvCa tumors. Methods Fascin expression was evaluated in human cells and tissues using immunohistochemistry and immunofluorescence. The functional role of fascin in cancer and stromal cells was assessed with in vitro functional assays, an ex vivo colonization assay and in vivo metastasis assays using siRNA/shRNA and an inhibitor. The effect of fascin inhibition on Cdc42 and Rac1 activity was evaluated using GTPase activity assays and immunofluorescence. Results Fascin expression was found to be higher in the stromal cell, when compared to the cancer cell, compartment of ovarian tumors. The low expression of fascin in the cancer cells of the primary tumor indicated a favorable prognosis for non-serous OvCa patients. In vitro, both knockdown and pharmacologic inhibition of fascin decreased the migration of cancer and stromal cells. The inhibition of fascin impaired Cdc42 and Rac1 activity in cancer cells, and cytoskeletal reorganization in the cancer and stromal cells. Inhibition of fascin ex vivo blocked OvCa cell colonization of human omental tissue and in vivo prevented and reduced OvCa metastases in mice. Likewise, knockdown of fascin specifically in the OvCa cells using a fascin-specific lentiviral-shRNA also blocked metastasis in vivo. Conclusion This study reveals the therapeutic potential of pharmacologically inhibiting fascin in both cancer and stromal cells of the OvCa tumor microenvironment.
- Published
- 2019
- Full Text
- View/download PDF
25. Effective breast cancer combination therapy targeting BACH1 and mitochondrial metabolism
- Author
-
Mohamad Elbaz, Marsha Rich Rosner, Daniel C. Rabe, Juan Liu, Daniel K. Nomura, Jielin Yan, Jiyoung Lee, Elizabeth A. Grossman, Jason W. Locasale, Payal Tiwari, Casey Frankenberger, Jorge Andrade, Ali Ekrem Yesilkanal, Sydney M. Sanderson, Joseph Wynne, Marcelo G. Bonini, Christie Kang, Peter C. Hart, and Felicia D. Rustandy
- Subjects
0301 basic medicine ,Citric Acid Cycle ,Mice, Nude ,Triple Negative Breast Neoplasms ,Biology ,Article ,Electron Transport ,Small hairpin RNA ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Transcription (biology) ,Gene expression ,medicine ,Animals ,Humans ,Gene ,Transcription factor ,Multidisciplinary ,medicine.disease ,Xenograft Model Antitumor Assays ,Phenotype ,Metformin ,Mitochondria ,Basic-Leucine Zipper Transcription Factors ,Glucose ,030104 developmental biology ,030220 oncology & carcinogenesis ,Proteolysis ,Cancer research ,Hemin ,Heterografts ,Female ,Reprogramming - Abstract
Mitochondrial metabolism is an attractive target for cancer therapy1,2. Reprogramming metabolic pathways could improve the ability of metabolic inhibitors to suppress cancers with limited treatment options, such as triple-negative breast cancer (TNBC)1,3. Here we show that BTB and CNC homology1 (BACH1)4, a haem-binding transcription factor that is increased in expression in tumours from patients with TNBC, targets mitochondrial metabolism. BACH1 decreases glucose utilization in the tricarboxylic acid cycle and negatively regulates transcription of electron transport chain (ETC) genes. BACH1 depletion by shRNA or degradation by hemin sensitizes cells to ETC inhibitors such as metformin5,6, suppressing growth of both cell line and patient-derived tumour xenografts. Expression of a haem-resistant BACH1 mutant in cells that express a short hairpin RNA for BACH1 rescues the BACH1 phenotype and restores metformin resistance in hemin-treated cells and tumours7. Finally, BACH1 gene expression inversely correlates with ETC gene expression in tumours from patients with breast cancer and in other tumour types, which highlights the clinical relevance of our findings. This study demonstrates that mitochondrial metabolism can be exploited by targeting BACH1 to sensitize breast cancer and potentially other tumour tissues to mitochondrial inhibitors. The transcription factor BACH1, which targets mitochondrial metabolism, is expressed at high levels in several types of cancer; reducing its expression in tumours makes them more susceptible to treatment with mitochondrial inhibitors.
- Published
- 2019
- Full Text
- View/download PDF
26. Modeling the Early Steps of Ovarian Cancer Dissemination in an Organotypic Culture of the Human Peritoneal Cavity
- Author
-
Peter C Hart, Preety Bajwa, and Hilary A. Kenny
- Subjects
Tumor microenvironment ,Cell type ,business.industry ,medicine.disease ,Metastasis ,Peritoneal cavity ,medicine.anatomical_structure ,Peritoneum ,In vivo ,medicine ,Cancer research ,Ovarian cancer ,business ,Ex vivo - Abstract
The majority of ovarian cancer patients present clinically with wide-spread metastases throughout the peritoneal cavity, metastasizing to the mesothelium-lined peritoneum and visceral adipose depots within the abdomen. This unique metastatic tumor microenvironment is comprised of multiple cell types, including mesothelial cells, fibroblasts, adipocytes, macrophages, neutrophils, and T lymphocytes. Modeling advancements, including complex 3D systems and organoids, coupled with 2D cocultures, in vivo mouse models, and ex vivo human tissue cultures have greatly enhanced our understanding of the tumor-stroma interactions that are required for successful metastasis of ovarian cancer cells. However, advanced multifaceted model systems that incorporate frequency and spatial distribution of all cell types present in the tumor microenvironment of ovarian cancer are needed to enhance our knowledge of ovarian cancer biology in order to identify methods for preventing and treating metastatic disease. This review highlights the utility of recently developed modeling approaches, summarizes some of the resulting progress using these techniques, and suggests how these strategies may be implemented to elucidate signaling processes among cell types of the tumor microenvironment that promote ovarian cancer metastasis.
- Published
- 2021
- Full Text
- View/download PDF
27. Possible Role of Metformin as an Immune Modulator in the Tumor Microenvironment of Ovarian Cancer
- Author
-
Daniel Majerczyk, Faye K Tsogas, and Peter C Hart
- Subjects
Oncology ,medicine.medical_specialty ,Disease ,Type 2 diabetes ,Review ,macrophage ,Catalysis ,Inorganic Chemistry ,lcsh:Chemistry ,myeloid-derived suppressor cell ,Immune system ,Internal medicine ,omentum ,medicine ,Animals ,Humans ,Hypoglycemic Agents ,Immunologic Factors ,tumor microenvironment ,Physical and Theoretical Chemistry ,Molecular Biology ,lcsh:QH301-705.5 ,Spectroscopy ,Ovarian Neoplasms ,Tumor microenvironment ,business.industry ,Organic Chemistry ,neutrophil ,T cell ,General Medicine ,medicine.disease ,Computer Science Applications ,Metformin ,ovarian cancer ,lcsh:Biology (General) ,lcsh:QD1-999 ,Tumor progression ,Myeloid-derived Suppressor Cell ,Female ,sense organs ,Ovarian cancer ,business ,metformin ,medicine.drug - Abstract
Growing evidence suggests that the immune component of the tumor microenvironment (TME) may be highly involved in the progression of high-grade serous ovarian cancer (HGSOC), as an immunosuppressive TME is associated with worse patient outcomes. Due to the poor prognosis of HGSOC, new therapeutic strategies targeting the TME may provide a potential path forward for preventing disease progression to improve patient survival. One such postulated approach is the repurposing of the type 2 diabetes medication, metformin, which has shown promise in reducing HGSOC tumor progression in retrospective epidemiological analyses and through numerous preclinical studies. Despite its potential utility in treating HGSOC, and that the immune TME is considered as a key factor in the disease’s progression, little data has definitively shown the ability of metformin to target this component of the TME. In this brief review, we provide a summary of the current understanding of the effects of metformin on leukocyte function in ovarian cancer and, coupled with data from other related disease states, posit the potential mechanisms by which the drug may enhance the anti-tumorigenic effects of immune cells to improve HGSOC patient survival.
- Published
- 2021
28. C-Reactive Protein and Cancer—Diagnostic and Therapeutic Insights
- Author
-
May Alebraheem, Peter C Hart, Ibraheem M. Rajab, and Lawrence A. Potempa
- Subjects
lcsh:Immunologic diseases. Allergy ,0301 basic medicine ,Immunology ,Context (language use) ,Inflammation ,Review ,monomeric C-reactive protein ,C-reactive protein ,Extracellular matrix ,03 medical and health sciences ,0302 clinical medicine ,Neoplasms ,acute phase response ,medicine ,Humans ,tumor microenvironment ,Immunology and Allergy ,Acute-Phase Reaction ,Tumor microenvironment ,biology ,business.industry ,Cell growth ,Acute-phase protein ,Cancer ,medicine.disease ,030104 developmental biology ,inflammation ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,medicine.symptom ,lcsh:RC581-607 ,business ,Biomarkers - Abstract
Cancer disease describes any pathology involving uncontrolled cell growth. As cells duplicate, they can remain localized in defined tissues, forming tumor masses and altering their microenvironmental niche, or they can disseminate throughout the body in a metastatic process affecting multiple tissues and organs. As tumors grow and metastasize, they affect normal tissue integrity and homeostasis which signals the body to trigger the acute phase inflammatory response. C-reactive protein (CRP) is a predominant protein of the acute phase response; its blood levels have long been used as a minimally invasive index of any ongoing inflammatory response, including that occurring in cancer. Its diagnostic significance in assessing disease progression or remission, however, remains undefined. By considering the recent understanding that CRP exists in multiple isoforms with distinct biological activities, a unified model is advanced that describes the relevance of CRP as a mediator of host defense responses in cancer. CRP in its monomeric, modified isoform (mCRP) modulates inflammatory responses by inserting into activated cell membranes and stimulating platelet and leukocyte responses associated with acute phase responses to tumor growth. It also binds components of the extracellular matrix in involved tissues. Conversely, CRP in its pentameric isoform (pCRP), which is the form quantified in diagnostic measurements of CRP, is notably less bioactive with weak anti-inflammatory bioactivity. Its accumulation in blood is associated with a continuous, low-level inflammatory response and is indicative of unresolved and advancing disease, as occurs in cancer. Herein, a novel interpretation of the diagnostic utility of CRP is presented accounting for the unique properties of the CRP isoforms in the context of the developing pro-metastatic tumor microenvironment.
- Published
- 2020
- Full Text
- View/download PDF
29. How C-Reactive Protein Structural Isoforms With Distinctive Bioactivities Affect Disease Progression
- Author
-
Lawrence A. Potempa, Peter C Hart, and Ibraheem M. Rajab
- Subjects
0301 basic medicine ,Gene isoform ,lcsh:Immunologic diseases. Allergy ,Antigenicity ,Protein Conformation ,medicine.medical_treatment ,Immunology ,Inflammation ,Disease ,03 medical and health sciences ,0302 clinical medicine ,modified/monomeric (mCRP) ,conformational isoforms ,medicine ,Immunology and Allergy ,Humans ,Protein Isoforms ,Secretion ,CRP - C-reactive protein ,blood kinetics ,biology ,Chemistry ,C-reactive protein ,Acute-phase protein ,MCRP ,030104 developmental biology ,Cytokine ,C-Reactive Protein ,inflammation ,biology.protein ,Disease Progression ,medicine.symptom ,lcsh:RC581-607 ,030215 immunology - Abstract
C-reactive protein (CRP) is a widely known, hepatically synthesized protein whose blood levels change rapidly and pronouncedly in response to any tissue damaging event associated with an inflammatory response. The synthesis and secretion of CRP is stimulated by interleukin-6, an early pleiotropic cytokine released by macrophages, endothelial, and other cells that are activated when localized normal tissue structures are compromised by trauma or disease. Serum CRP levels can change rapidly and robustly from 10-100-fold within 6-72 h of any tissue damaging event. Elevated blood levels correlate with the onset and extent of both activated inflammation and the acute phase biochemical response to the tissue insult. Because its functional bioactivity as the prototypic acute phase reactant has eluded clear definition for decades, diagnosticians of various conditions and diseases use CRP blood levels as a simple index for ongoing inflammation. In many pathologies, which involves many different tissues, stages of disease, treatments, and responses to treatments, its interpretive diagnostic value requires a deeper understanding of the localized tissue processes and events that contribute signals which regulate protective or pathological host defense bioactivities. This report presents concepts that describe how local tissue activation events can lead to a non-proteolytic, conformational rearrangement of CRP into a unique isoform with distinctive solubility, antigenicity, binding reactivities and bioactivities from that protein widely known and measured in serum. By describing factors that control the expression, tissue localization, half-life and pro-inflammatory amplification activity of this CRP isoform, a unifying explanation for the diagnostic significance of CRP measurement in disease is advanced.
- Published
- 2020
- Full Text
- View/download PDF
30. Selected mitochondrial DNA landscapes activate the SIRT3 axis of the UPRmt to promote metastasis
- Author
-
Sneha Grandhi, Jerry E. Chipuk, Thomas LaFramboise, M D'Aurello, Janine H. Santos, Doris Germain, Luena Papa, Marcelo G. Bonini, Abdul Kader Sagar, Peter C. Hart, Giovanni Manfredi, M Sersinghe, Timothy C. Kenny, M Ragazzi, Kevin W. Eliceiri, and Amanjot Kaur Riar
- Subjects
0301 basic medicine ,Cancer Research ,Mitochondrial DNA ,SIRT3 ,SOD2 ,Repressor ,Breast Neoplasms ,Biology ,DNA, Mitochondrial ,03 medical and health sciences ,Cell Line, Tumor ,Sirtuin 3 ,Mitochondrial unfolded protein response ,Genetics ,Humans ,Neoplasm Metastasis ,Enhancer ,Molecular Biology ,Superoxide Dismutase ,Forkhead Box Protein O3 ,Mitochondria ,030104 developmental biology ,Proteostasis ,Cancer cell ,Unfolded Protein Response ,Cancer research ,Original Article ,Female - Abstract
By causing mitochondrial DNA (mtDNA) mutations and oxidation of mitochondrial proteins, reactive oxygen species (ROS) leads to perturbations in mitochondrial proteostasis. Several studies have linked mtDNA mutations to metastasis of cancer cells but the nature of the mtDNA species involved remains unclear. Our data suggests that no common mtDNA mutation identifies metastatic cells; rather the metastatic potential of several ROS-generating mutations is largely determined by their mtDNA genomic landscapes, which can act either as an enhancer or repressor of metastasis. However, mtDNA landscapes of all metastatic cells are characterized by activation of the SIRT/FOXO/SOD2 axis of the mitochondrial unfolded protein response (UPRmt). The UPRmt promotes a complex transcription program ultimately increasing mitochondrial integrity and fitness in response to oxidative proteotoxic stress. Using SOD2 as a surrogate marker of the UPRmt, we found that in primary breast cancers, SOD2 is significantly increased in metastatic lesions. We propose that the ability of selected mtDNA species to activate the UPRmt is a process that is exploited by cancer cells to maintain mitochondrial fitness and facilitate metastasis.
- Published
- 2017
- Full Text
- View/download PDF
31. Loss of BRCA1 in the Cells of Origin of Ovarian Cancer Induces Glycolysis: A Window of Opportunity for Ovarian Cancer Chemoprevention
- Author
-
Ricardo R. Lastra, Ryuji Hamamoto, Iris L. Romero, S. Diane Yamada, Tatsuyuki Chiyoda, Yusuke Nakamura, Peter C. Hart, Stephanie M. McGregor, Mark A. Eckert, Ernst Lengyel, and Olufunmilayo I. Olopade
- Subjects
0301 basic medicine ,Cancer Research ,endocrine system diseases ,DNA repair ,Carcinoma, Ovarian Epithelial ,medicine.disease_cause ,Chemoprevention ,Article ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Cell Line, Tumor ,medicine ,Humans ,Gene silencing ,Neoplasms, Glandular and Epithelial ,skin and connective tissue diseases ,STAT3 ,Fallopian Tubes ,Ovarian Neoplasms ,Mutation ,Aspirin ,Cyclooxygenase 2 Inhibitors ,biology ,BRCA1 Protein ,Cancer ,Epithelial Cells ,medicine.disease ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Female ,Ovarian cancer ,Carcinogenesis ,Glycolysis - Abstract
Mutations in the breast cancer susceptibility gene 1 (BRCA1) are associated with an increased risk of developing epithelial ovarian cancer. However, beyond the role of BRCA1 in DNA repair, little is known about other mechanisms by which BRCA1 impairment promotes carcinogenesis. Given that altered metabolism is now recognized as important in the initiation and progression of cancer, we asked whether the loss of BRCA1 changes metabolism in the cells of origin of ovarian cancer. The findings show that silencing BRCA1 in ovarian surface epithelial and fallopian tube cells increased glycolysis. Furthermore, when these cells were transfected with plasmids carrying deleterious BRCA1 mutations (5382insC or the P1749R), there was an increase in hexokinase-2 (HK2), a key glycolytic enzyme. This effect was mediated by MYC and the STAT3. To target the metabolic phenotype induced by loss of BRCA1, a drug-repurposing approach was used and aspirin was identified as an agent that counteracted the increase in HK2 and the increase in glycolysis induced by BRCA1 impairment. Evidence from this study indicates that the tumor suppressor functions of BRCA1 extend beyond DNA repair to include metabolic endpoints and identifies aspirin as an ovarian cancer chemopreventive agent capable of reversing the metabolic derangements caused by loss of BRCA1. Cancer Prev Res; 10(4); 255–66. ©2017 AACR.
- Published
- 2017
- Full Text
- View/download PDF
32. SPHK1 is a novel target of metformin in ovarian cancer
- Author
-
Iris L. Romero, Jason W. Locasale, Marion Curtis, Peter C. Hart, Stephanie M. McGregor, Ricardo R. Lastra, Tatsuyuki Chiyoda, Xiaojing Liu, Melanie Weigert, Chun-Yi Chiang, Ernst Lengyel, and Rachel M. Loth
- Subjects
0301 basic medicine ,Cancer Research ,endocrine system diseases ,Mice, Nude ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Cell Movement ,Sphingosine ,medicine ,Basic Helix-Loop-Helix Transcription Factors ,Animals ,Humans ,Hypoglycemic Agents ,Molecular Targeted Therapy ,Molecular Biology ,Ovarian Neoplasms ,Kinase ,business.industry ,Cell growth ,Cancer ,medicine.disease ,Hypoxia-Inducible Factor 1, alpha Subunit ,Sphingolipid ,Xenograft Model Antitumor Assays ,female genital diseases and pregnancy complications ,Metformin ,Phosphotransferases (Alcohol Group Acceptor) ,030104 developmental biology ,Oncology ,chemistry ,Tumor progression ,030220 oncology & carcinogenesis ,Cancer research ,lipids (amino acids, peptides, and proteins) ,Female ,Lysophospholipids ,Ovarian cancer ,business ,medicine.drug - Abstract
The role of phospholipid signaling in ovarian cancer is poorly understood. Sphingosine-1-phosphate (S1P) is a bioactive metabolite of sphingosine that has been associated with tumor progression through enhanced cell proliferation and motility. Similarly, sphingosine kinases (SPHK), which catalyze the formation of S1P and thus regulate the sphingolipid rheostat, have been reported to promote tumor growth in a variety of cancers. The findings reported here show that exogenous S1P or overexpression of SPHK1 increased proliferation, migration, invasion, and stem-like phenotypes in ovarian cancer cell lines. Likewise, overexpression of SPHK1 markedly enhanced tumor growth in a xenograft model of ovarian cancer, which was associated with elevation of key markers of proliferation and stemness. The diabetes drug, metformin, has been shown to have anticancer effects. Here, we found that ovarian cancer patients taking metformin had significantly reduced serum S1P levels, a finding that was recapitulated when ovarian cancer cells were treated with metformin and analyzed by lipidomics. These findings suggested that in cancer the sphingolipid rheostat may be a novel metabolic target of metformin. In support of this, metformin blocked hypoxia-induced SPHK1, which was associated with inhibited nuclear translocation and transcriptional activity of hypoxia-inducible factors (HIF1α and HIF2α). Further, ovarian cancer cells with high SPHK1 were found to be highly sensitive to the cytotoxic effects of metformin, whereas ovarian cancer cells with low SPHK1 were resistant. Together, the findings reported here show that hypoxia-induced SPHK1 expression and downstream S1P signaling promote ovarian cancer progression and that tumors with high expression of SPHK1 or S1P levels might have increased sensitivity to the cytotoxic effects of metformin.Implications:Metformin targets sphingolipid metabolism through inhibiting SPHK1, thereby impeding ovarian cancer cell migration, proliferation, and self-renewal.
- Published
- 2019
33. Mesothelial Cell HIF1α Expression Is Metabolically Downregulated by Metformin to Prevent Oncogenic Tumor-Stromal Crosstalk
- Author
-
Niklas Grassl, Iris L. Romero, Matthias Mann, Karen M. Watters, Ivana Blaženović, Oliver Fiehn, Lisa Ploetzky, Lacey M. Litchfield, Ernst Lengyel, Hilary A. Kenny, Peter C. Hart, and Fabian Coscia
- Subjects
0301 basic medicine ,endocrine system diseases ,Carcinogenesis ,Cell ,Medical Physiology ,Inbred C57BL ,Mice ,0302 clinical medicine ,Succinate-CoA Ligases ,Tumor Cells, Cultured ,Tumor Microenvironment ,2.1 Biological and endogenous factors ,SUCLG2 ,Aetiology ,lcsh:QH301-705.5 ,Cancer ,Ovarian Neoplasms ,mesothelial cells ,Cultured ,Chemistry ,Diabetes ,digestive, oral, and skin physiology ,Metformin ,TGF ,Tumor Cells ,Ovarian Cancer ,medicine.anatomical_structure ,Female ,Hypoxia-Inducible Factor 1 ,medicine.drug ,Stromal cell ,HIF1 ,alpha Subunit ,General Biochemistry, Genetics and Molecular Biology ,Article ,Prolyl Hydroxylases ,03 medical and health sciences ,Rare Diseases ,medicine ,Animals ,Humans ,Hypoglycemic Agents ,Interleukin 8 ,omental metastasis ,Tumor microenvironment ,IL-8 ,medicine.disease ,Hypoxia-Inducible Factor 1, alpha Subunit ,succinate ,Mice, Inbred C57BL ,030104 developmental biology ,lcsh:Biology (General) ,Cancer research ,Biochemistry and Cell Biology ,Stromal Cells ,Ovarian cancer ,030217 neurology & neurosurgery ,Mesothelial Cell ,Transforming growth factor - Abstract
Summary: The tumor microenvironment (TME) plays a pivotal role in cancer progression, and, in ovarian cancer (OvCa), the primary TME is the omentum. Here, we show that the diabetes drug metformin alters mesothelial cells in the omental microenvironment. Metformin interrupts bidirectional signaling between tumor and mesothelial cells by blocking OvCa cell TGF-β signaling and mesothelial cell production of CCL2 and IL-8. Inhibition of tumor-stromal crosstalk by metformin is caused by the reduced expression of the tricarboxylic acid (TCA) enzyme succinyl CoA ligase (SUCLG2). Through repressing this TCA enzyme and its metabolite, succinate, metformin activated prolyl hydroxylases (PHDs), resulting in the degradation of hypoxia-inducible factor 1α (HIF1α) in mesothelial cells. Disruption of HIF1α-driven IL-8 signaling in mesothelial cells by metformin results in reduced OvCa invasion in an organotypic 3D model. These findings indicate that tumor-promoting signaling between mesothelial and OvCa cells in the TME can be targeted using metformin. : Hart et al. identify that the type 2 diabetes drug metformin inhibits ovarian cancer invasion by targeting crosstalk between cancer cells and adjacent normal stromal mesothelial cells, making the microenvironment less hospitable to cancer growth. Keywords: mesothelial cells, tumor microenvironment, ovarian cancer, metformin, HIF1, SUCLG2, IL-8, TGF, omental metastasis, succinate
- Published
- 2019
- Full Text
- View/download PDF
34. SOD2 and the Mitochondrial UPR: Partners Regulating Cellular Phenotypic Transitions
- Author
-
Chenxia He, Peter C. Hart, Marcelo G. Bonini, and Doris Germain
- Subjects
0301 basic medicine ,Cell signaling ,Somatic cell ,SOD2 ,Mitochondrion ,Biochemistry ,Article ,Superoxide dismutase ,03 medical and health sciences ,Mitochondrial unfolded protein response ,Animals ,Humans ,Molecular Biology ,chemistry.chemical_classification ,Genetics ,Reactive oxygen species ,biology ,Superoxide Dismutase ,Hydrogen Peroxide ,Mitochondria ,Cell biology ,Phenotype ,030104 developmental biology ,chemistry ,Unfolded Protein Response ,Unfolded protein response ,biology.protein ,Reactive Oxygen Species - Abstract
ATP and reactive oxygen species (ROS) are signaling molecules that control cellular function and phenotype. Mitochondria produce both ATP and ROS. Since the electrons needed to generate either ATP or ROS originate from NADH/FADH2, the mechanism through which electrons flow towards oxygen determines yields and whether ATP or ROS prevails. Alterations in the electron flow impact cells dramatically, such as by supporting specialization (which requires high ATP) or imposing dedifferentiation. High ROS, facilitated by enzymes such as superoxide dismutase 2 (SOD2) that enhance mitochondrial hydrogen peroxide (mtH2O2), are normally linked to dedifferentiation of somatic cells. Here we propose that combined high mtH2O2 and mitochondrial unfolded protein response (UPR(mt)) activation are essential for somatic dedifferentiation programs and the acquisition of stem-like properties in reparative processes and disease.
- Published
- 2016
- Full Text
- View/download PDF
35. Caveolin-1 regulates cancer cell metabolism via scavenging Nrf2 and suppressing MnSOD-driven glycolysis
- Author
-
Peter C. Hart, Angela L. Tyner, Richard D. Minshall, Mao Mao, Marcelo G. Bonini, Kristine Ansenberger-Fricano, Ayesha N. Shajahan-Haq, and Bianca Altrão Ratti
- Subjects
0301 basic medicine ,Gerontology ,MnSOD ,Cell type ,caveolin-1 ,NF-E2-Related Factor 2 ,animal diseases ,Blotting, Western ,Caveolin 1 ,tumor progression ,Breast Neoplasms ,Mammary Neoplasms, Animal ,Biology ,Nrf2 ,Cell Line ,03 medical and health sciences ,Mice ,breast cancer ,Downregulation and upregulation ,Animals ,Humans ,Transcription factor ,Kelch-Like ECH-Associated Protein 1 ,Microscopy, Confocal ,Superoxide Dismutase ,Intracellular Signaling Peptides and Proteins ,AMPK ,Prognosis ,Survival Analysis ,030104 developmental biology ,Oncology ,Anaerobic glycolysis ,Tumor progression ,Cancer cell ,Cancer research ,MCF-7 Cells ,RNA Interference ,Glycolysis ,Research Paper ,Protein Binding - Abstract
Aerobic glycolysis is an indispensable component of aggressive cancer cell metabolism. It also distinguishes cancer cells from most healthy cell types in the body. Particularly for this reason, targeting the metabolism to improve treatment outcomes has long been perceived as a potentially valuable strategy. In practice, however, our limited knowledge of why and how metabolic reprogramming occurs has prevented progress towards therapeutic interventions that exploit the metabolic peculiarities of tumors. We recently described that in breast cancer, MnSOD upregulation is both necessary and sufficient to activate glycolysis. Here, we focused on determining the molecular mechanisms of MnSOD upregulation. We found that Caveolin-1 (Cav-1) is a central component of this mechanism due to its suppressive effects of NF-E2-related factor 2 (Nrf2), a transcription factor upstream of MnSOD. In transformed MCF10A(Er/Src) cells, Cav-1 loss preceded the activation of Nrf2 and its induction of MnSOD expression. Consistently, with previous observations, MnSOD expression secondary to Nrf2 activation led to an increase in the glycolytic rate dependent on mtH2O2 production and the activation of AMPK. Moreover, rescue of Cav-1 expression in a breast cancer cell line (MCF7) suppressed Nrf2 and reduced MnSOD expression. Experimental data were reinforced by epidemiologic nested case-control studies showing that Cav-1 and MnSOD are inversely expressed in cases of invasive ductal carcinoma, with low Cav-1 and high MnSOD expression being associated with lower 5-year survival rates and molecular subtypes with poorest prognosis.
- Published
- 2015
36. NOS1-derived nitric oxide promotes NF-κB transcriptional activity through inhibition of suppressor of cytokine signaling-1
- Author
-
Richard D. Minshall, Peter C. Hart, Mirza S. Baig, Saurabh Chatterjee, Asrar B. Malik, Sofia Zaichick, Michelle L. Block, Jing Deng, Stephen M. Vogel, Marcia Edilaine Lopes Consolaro, John W. Christman, Andre L. P de Abreu, Mao Mao, Uzma Saqib, Benjamin N. Gantner, Marcelo G. Bonini, and Farnaz R. Bakhshi
- Subjects
Lipopolysaccharides ,Nitric Oxide Synthase Type III ,NOS1 ,medicine.medical_treatment ,Molecular Sequence Data ,Immunology ,Nitric Oxide Synthase Type II ,Mice, Transgenic ,Suppressor of Cytokine Signaling Proteins ,Inflammation ,Nitric Oxide Synthase Type I ,Lung injury ,Biology ,Nitric Oxide ,Article ,Proinflammatory cytokine ,Nitric oxide ,chemistry.chemical_compound ,Suppressor of Cytokine Signaling 1 Protein ,Sepsis ,medicine ,Animals ,Humans ,Immunology and Allergy ,Mice, Knockout ,Suppressor of cytokine signaling 1 ,Macrophages ,NF-kappa B ,NF-κB ,Molecular biology ,Mice, Inbred C57BL ,Toll-Like Receptor 4 ,Cytokine ,chemistry ,Cytokines ,medicine.symptom - Abstract
NOS1−/− mice show reduced inflammatory responses and tissue damage in experimental sepsis models. Baig et al show that NOS1-derived NO production in macrophages leads to proteolysis of SOCS1 to alleviate its repression of NFkB transcriptional activity in response to TLR4-mediated responses., The NF-κB pathway is central to the regulation of inflammation. Here, we demonstrate that the low-output nitric oxide (NO) synthase 1 (NOS1 or nNOS) plays a critical role in the inflammatory response by promoting the activity of NF-κB. Specifically, NOS1-derived NO production in macrophages leads to proteolysis of suppressor of cytokine signaling 1 (SOCS1), alleviating its repression of NF-κB transcriptional activity. As a result, NOS1−/− mice demonstrate reduced cytokine production, lung injury, and mortality when subjected to two different models of sepsis. Isolated NOS1−/− macrophages demonstrate similar defects in proinflammatory transcription on challenge with Gram-negative bacterial LPS. Consistently, we found that activated NOS1−/− macrophages contain increased SOCS1 protein and decreased levels of p65 protein compared with wild-type cells. NOS1-dependent S-nitrosation of SOCS1 impairs its binding to p65 and targets SOCS1 for proteolysis. Treatment of NOS1−/− cells with exogenous NO rescues both SOCS1 degradation and stabilization of p65 protein. Point mutation analysis demonstrated that both Cys147 and Cys179 on SOCS1 are required for its NO-dependent degradation. These findings demonstrate a fundamental role for NOS1-derived NO in regulating TLR4-mediated inflammatory gene transcription, as well as the intensity and duration of the resulting host immune response.
- Published
- 2015
- Full Text
- View/download PDF
37. Mangenese superoxide dismutase (MnSOD) promotes stem‐like cell phenotypes in breast cancer
- Author
-
Marcelo G. Bonini, Micaela Vargas, Chenxia He, Peter C. Hart, Kraig Thieraud, Kristine Fricano, and André Luelsdorf Pimenta de Abreu
- Subjects
Stem like cell ,Superoxide dismutase ,Breast cancer ,biology ,Genetics ,medicine ,Cancer research ,biology.protein ,medicine.disease ,Molecular Biology ,Biochemistry ,Phenotype ,Biotechnology - Published
- 2017
- Full Text
- View/download PDF
38. Allele-specific interaction between glutathione peroxidase 1 and manganese superoxide dismutase affects the levels of Bcl-2, Sirt3 and E-cadherin
- Author
-
Dede N. Ekoue, Sofia Zaichick, Peter C. Hart, Soumen Bera, Emmanuel Ansong, Alan M. Diamond, Frederick E. Domann, and Marcelo G. Bonini
- Subjects
0301 basic medicine ,GPX1 ,Biology ,Transfection ,Biochemistry ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Breast cancer ,Glutathione Peroxidase GPX1 ,Antigens, CD ,Cell Line, Tumor ,Sirtuin 3 ,medicine ,Humans ,Phosphorylation ,Cell Engineering ,Alleles ,chemistry.chemical_classification ,Glutathione Peroxidase ,Polymorphism, Genetic ,Cadherin ,Superoxide ,Superoxide Dismutase ,Glutathione peroxidase ,Cancer ,General Medicine ,Hydrogen Peroxide ,medicine.disease ,Cadherins ,Molecular biology ,Mitochondria ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,chemistry ,Proto-Oncogene Proteins c-bcl-2 ,MCF-7 Cells ,Ectopic expression ,Female ,Proto-Oncogene Proteins c-akt ,Plasmids ,Protein Binding ,Signal Transduction - Abstract
Manganese superoxide dismutase (MnSOD) is a mitochondrial-resident enzyme that reduces superoxide to hydrogen peroxide (H2O2), which can be further reduced to water by glutathione peroxidase (GPX1). Data from human studies have indicated that common polymorphisms in both of these proteins are associated with the risk of several cancers, including breast cancer. Moreover, polymorphisms in MnSOD and GPX1 were shown to interact to increase the risk of breast cancer. To gain an understanding of the molecular mechanisms behind these observations, we engineered human MCF-7 breast cancer cells to exclusively express GPX1 and/or MnSOD alleles and investigated the consequences on the expression of several proteins associated with cancer aetiology. Little or no effect was observed on the ectopic expression of these genes on the phosphorylation of Akt, although allele-specific effects and interactions were observed for the impact on the levels of Bcl-2, E-cadherin and Sirt3. The patterns observed were not consistent with the steady-state levels of H2O2 determined in the transfected cells. These results indicate plausible contributing factors to the effects of allelic variations on cancer risk observed in human epidemiological studies.
- Published
- 2017
- Full Text
- View/download PDF
39. Redox control of enzymatic functions: The electronics of life's circuitry
- Author
-
Mao Mao, André Luelsdorf Pimenta de Abreu, Marcia Edilaine Lopes Consolaro, Peter C. Hart, Alyssa M. Master, and Marcelo G. Bonini
- Subjects
chemistry.chemical_classification ,Cell signaling ,Reactive oxygen species ,Methionine ,Cellular respiration ,Chemistry ,Clinical Biochemistry ,Context (language use) ,Cell Biology ,Biochemistry ,chemistry.chemical_compound ,Downregulation and upregulation ,Second messenger system ,Genetics ,Molecular Biology ,Reactive nitrogen species - Abstract
The field of redox biology has changed tremendously over the past 20 years. Formerly regarded as bi-products of the aerobic metabolism exclusively involved in tissue damage, reactive oxygen species (ROS) are now recognized as active participants of cell signaling events in health and in disease. In this sense, ROS and the more recently defined reactive nitrogen species (RNS) are, just like hormones and second messengers, acting as fundamental orchestrators of cell signaling pathways. The chemical modification of enzymes by ROS and RNS (that result in functional enzymatic alterations) accounts for a considerable fraction of the transient and persistent perturbations imposed by variations in oxidant levels. Upregulation of ROS and RNS in response to stress is a common cellular response that foments adaptation to a variety of physiologic alterations (hypoxia, hyperoxia, starvation, and cytokine production). Frequently, these are beneficial and increase the organisms' resistance against subsequent acute stress (preconditioning). Differently, the sustained ROS/RNS-dependent rerouting of signaling produces irreversible alterations in cellular functioning, often leading to pathogenic events. Thus, the duration and reversibility of protein oxidations define whether complex organisms remain "electronically" healthy. Among the 20 essential amino acids, four are particularly susceptible to oxidation: cysteine, methionine, tyrosine, and tryptophan. Here, we will critically review the mechanisms, implications, and repair systems involved in the redox modifications of these residues in proteins while analyzing well-characterized prototypic examples. Occasionally, we will discuss potential consequences of amino acid oxidation and speculate on the biologic necessity for such events in the context of adaptative redox signaling. © 2014 IUBMB Life, 66(3):167-181, 2014.
- Published
- 2014
- Full Text
- View/download PDF
40. Acetylation activates an alternative function of SOD2 as a stemness factor in breast cancer
- Author
-
Douglas Ganini da Silva, Peter C. Hart, David Gius, Marcelo G. Bonini, Chenxia He, and Yueming Zhu
- Subjects
Chemistry ,SOD2 ,Cancer ,Tumor initiation ,medicine.disease ,Biochemistry ,law.invention ,Breast cancer ,SOX2 ,Acetylation ,law ,Cancer stem cell ,Physiology (medical) ,cardiovascular system ,Cancer research ,medicine ,Suppressor ,skin and connective tissue diseases - Abstract
Mitochondrial superoxide dismutase (SOD2) displays a dichotomous role in cancer, being a suppressor of tumor initiation while stimulating cancer progression later in the established disease. The mechanistic basis of this switch remains unknown. Our results indicate that an increase in SOD2 expression beyond a defined threshold leads to its accumulation in an acetylated state and the activation of redox stress responses including, as reported here, the activation of hypoxic signaling. Specifically, we found that increased expression of SOD2/Ac-SOD2 stabilizes hypoxia-induced factor 2α (HIF2α) in a H2O2-dependent manner. Consequently, the SOD2/HIF2α promotes core stemness gene (i.e. Oct4 and SOX2) expression increasing the cancer stem cell (CSC) subpopulation, tumorigenicity and invasiveness of breast cancer cells. Based on these findings we propose that as tumors evolve to advanced stages a mitochondrial pathway dependent on SOD2/ROS and HIF2α is activated and contributes to CSC formation. It is also proposed that a SOD2high/HIF2α+ may identify cells in breast cancer with tumor initiating capacity and high metastatic potential.
- Published
- 2018
- Full Text
- View/download PDF
41. List of Contributors
- Author
-
Ottavio Arancio, Nancy Bartolotti, Adam W. Bero, Jacqueline A. Bonds, Marcelo G. Bonini, Guojun Bu, Xu Chen, Toby Cumming, Jole Fiorito, Li Gan, David Gate, Walter Gulisano, Peter C. Hart, Jacob M. Haus, Katherine A. Jackman, Takahisa Kanekiyo, Orly Lazarov, Sylvia Lombardo, Robert A. Marr, Keri Martinowich, Eliezer Masliah, Alyson A. Miller, Richard D. Minshall, Russell Nicholls, Agostino Palmeri, Rosita Purgatorio, Daniela Puzzo, Amanda R. Rabinowitz, Meredith C. Reichert, David P. Salmon, Robert J. Schloesser, Douglas H. Smith, Giuseppina Tesco, Terrence Town, and Li-Huei Tsai
- Published
- 2016
- Full Text
- View/download PDF
42. Mouse Models for Studying Depression-Like States and Antidepressant Drugs
- Author
-
Carisa L. Bergner, Allan V. Kalueff, Justin L. LaPorte, Rupert J. Egan, Amanda N. Smolinsky, Brett D. Dufour, and Peter C. Hart
- Subjects
0301 basic medicine ,business.industry ,Brain dysfunction ,Anhedonia ,Disease ,Pharmacology ,Bioinformatics ,medicine.disease ,Comorbidity ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Medicine ,Antidepressant ,Chronic stress ,medicine.symptom ,business ,030217 neurology & neurosurgery ,Depression (differential diagnoses) - Abstract
Depression is a common psychiatric disorder, with diverse symptoms and high comorbidity with other brain dysfunctions. Due to this complexity, little is known about the neural and genetic mechanisms involved in depression pathogenesis. In a large proportion of patients, current antidepressant treatments are often ineffective and/or have undesirable side effects, fueling the search for more effective drugs. Animal models mimicking various symptoms of depression are indispensable in studying the biological mechanisms of this disease. Here, we summarize several popular methods for assessing depression-like symptoms in mice, and their utility in screening antidepressant drugs.
- Published
- 2016
- Full Text
- View/download PDF
43. Type 2 Diabetes Mellitus as a Risk Factor for Alzheimer’s Disease
- Author
-
Richard D. Minshall, Jacqueline A. Bonds, Peter C. Hart, Orly Lazarov, Marcelo G. Bonini, and Jacob M. Haus
- Subjects
biology ,business.industry ,Insulin ,medicine.medical_treatment ,Type 2 Diabetes Mellitus ,Disease ,medicine.disease ,Bioinformatics ,Insulin receptor ,Insulin resistance ,Diabetes mellitus ,Immunology ,medicine ,biology.protein ,Risk factor ,Cognitive decline ,business - Abstract
Type 2 diabetes mellitus (T2DM) is an acknowledged risk factor for the development of Alzheimer’s disease (AD). However, molecular mechanisms linking these disorders remain poorly defined. This chapter will review recent advancements in the understanding of the etiologic causes of AD occurring in a subgroup of diabetic patients based on studies using a variety of animal models of insulin resistance, and T2DM. We will also focus on vascular deterioration and defective insulin signaling in the brain as primary causes of molecular, vascular, and neuronal changes that translate into cognitive decline, memory loss, and other features characteristic of AD.
- Published
- 2016
- Full Text
- View/download PDF
44. Experimental Models of Anxiety for Drug Discovery and Brain Research
- Author
-
Peter C. Hart, Carisa L. Bergner, Amanda N. Smolinsky, Brett D. Dufour, Rupert J. Egan, Justin L. LaPorte, and Allan V. Kalueff
- Published
- 2016
- Full Text
- View/download PDF
45. Behavioral and physiological effects of acute ketamine exposure in adult zebrafish
- Author
-
Molly Hook, John DiLeo, Andrew Roth, Louis Monnig, Jeremy Green, Evan J. Kyzar, Mimi Pham, Peter C. Hart, Alexander V. Allain, Kate Rhymes, Russell Riehl, Roshan Razavi, Siddharth Gaikwad, and Allan V. Kalueff
- Subjects
Male ,animal structures ,Hydrocortisone ,Sedation ,Video Recording ,Danio ,Gene Expression ,Anxiety ,Motor Activity ,Pharmacology ,Toxicology ,Polymerase Chain Reaction ,Receptors, N-Methyl-D-Aspartate ,c-Fos ,Cellular and Molecular Neuroscience ,Glutamatergic ,Developmental Neuroscience ,medicine ,Animals ,Ketamine ,Habituation ,Zebrafish ,Behavior, Animal ,Dose-Response Relationship, Drug ,biology ,Antagonist ,Brain ,biology.organism_classification ,High-Throughput Screening Assays ,biology.protein ,Female ,medicine.symptom ,Psychology ,Excitatory Amino Acid Antagonists ,Proto-Oncogene Proteins c-fos ,Neuroscience ,medicine.drug - Abstract
Ketamine is a non-competitive glutamatergic antagonist used to induce sedation and analgesia. In sub-anesthetic doses, it induces hyperlocomotion, impairs memory and evokes stereotypic circling in rodents. Zebrafish (Danio rerio) emerged as a promising new animal model to screen the effects of psychotropic compounds. Here, we investigated the effects of sub-anesthetic doses of ketamine on anxiety, locomotion, habituation and social behavior of adult zebrafish. Acute 20-min exposure to 20 and 40 mg/L (but not 2 mg/L) of ketamine reduced anxiety, impaired intra-session habituation, evoked circular swimming and disrupted zebrafish shoaling. Additionally, ketamine reduced whole-body cortisol levels and elevated brain c-fos expression in zebrafish. Our findings demonstrate the sensitivity of zebrafish to behavioral and physiological effects of sub-anesthetic doses of ketamine, further supporting the utility of this species as a model for neuropharmacological research, including testing ketamine and related drugs.
- Published
- 2011
- Full Text
- View/download PDF
46. Zebrafish models to study drug abuse-related phenotypes
- Author
-
Siddharth Gaikwad, Marco Elegante, Allan V. Kalueff, Eli Utterback, David Tien, Keith Wong, Nadine Wu, Evan J. Kyzar, Valerie Piet, Jonathan Cachat, Adam Stewart, and Peter C. Hart
- Subjects
Drug ,animal structures ,Substance-Related Disorders ,media_common.quotation_subject ,Danio ,Pharmacology ,medicine ,Animals ,Humans ,Adverse effect ,Zebrafish ,media_common ,Study drug ,biology ,General Neuroscience ,Addiction ,fungi ,biology.organism_classification ,medicine.disease ,Phenotype ,Substance Withdrawal Syndrome ,Substance abuse ,Disease Models, Animal ,Psychology ,Neuroscience - Abstract
Mounting evidence implicates the zebrafish (Danio rerio) as a promising model species for reward and addiction research. Modeling drug abuse-related behavior in both adult and larval zebrafish produced a wealth of clinically translatable data, also demonstrating their sensitivity to various drugs of abuse and the ability to develop tolerance. Several studies have also applied withdrawal paradigms to model the adverse effects of drug abuse in zebrafish. In this review, we summarize recent findings of a wide spectrum of zebrafish drug abuse-related behavioral and physiological phenotypes, discuss the existing challenges, and outline potential future directions of research in this field.
- Published
- 2011
- Full Text
- View/download PDF
47. Abstract A28: Beta-escin inhibits ovarian cancer metastasis by targeting the tumor microenvironment
- Author
-
Marc Ferrer, Min Shen, Karen M. Watters, Ernst Lengyel, Betul Kara, Peter C. Hart, Chun-Yi Chiang, Hilary A. Kenny, and Madhu Lal
- Subjects
Cancer Research ,Tumor microenvironment ,education.field_of_study ,Stromal cell ,Population ,Cancer ,Biology ,medicine.disease ,Metastasis ,Oncology ,Cancer cell ,medicine ,Cancer research ,Cytokine secretion ,education ,Ovarian cancer - Abstract
The high mortality rate that results from ovarian cancer (OvCa) is caused by the wide dissemination of cancer cells within the abdominal cavity. OvCa cells metastasize to the peritoneum, which is covered by a single layer of mesothelial cells, and invade into the underlying stroma, composed of extracellular matrices including fibronectin (FN) and stromal cells such as fibroblasts. Using a three-dimensional organotypic quantitative high-throughput screening platform (3D-qHTS) modeling the peritoneum, we discovered that beta-escin inhibited OvCa adhesion and invasion to the peritoneum. Beta-escin is a natural triterpenoid saponin from Chinese horse chestnut seeds and has a variety of known pharmacologic effects. We hypothesize that beta-escin targets both the cancer cells and the stromal cells to inhibit the metastatic niche. The therapeutic effects of beta-escin and horse chestnut seed extract were tested in vivo using xenograft and syngeneic models of OvCa prevention and intervention. The mechanisms of beta-escin action in cancer cells and microenvironmental cells was explored in vitro using OvCa cell lines and primary human mesothelial cells and fibroblasts, and in vivo using different mouse models of metastasis. Moreover, a collection of 160 analogs of beta-escin were gathered and screened using our 3D-qHTS platform for inhibitory activity in OvCa cell adhesion/invasion. Our results reveal that beta-escin and horse chestnut extract taken orally inhibit metastasis in both OvCa prevention and intervention models. In addition, beta-escin mechanistically depresses the pluripotent stem cell population, inflammatory cytokine secretion, HIF-1alpha expression, and fibronectin production, while it increases autophagy in the tumor microenvironment. Furthermore, three distant analogs of beta-escin inhibited OvCa metastasis, and all three are cardiac glycosides. Taken together, we reveal a potential therapeutic value for beta-escin and/or analogs of beta-escin in preventing and treating OvCa dissemination. Citation Format: Hilary A. Kenny, Madhu Lal, Min Shen, Betul Kara, Chun-Yi Chiang, Karen Watters, Peter Hart, Marc Ferrer, Ernst Lengyel. Beta-escin inhibits ovarian cancer metastasis by targeting the tumor microenvironment. [abstract]. In: Proceedings of the AACR Conference: Addressing Critical Questions in Ovarian Cancer Research and Treatment; Oct 1-4, 2017; Pittsburgh, PA. Philadelphia (PA): AACR; Clin Cancer Res 2018;24(15_Suppl):Abstract nr A28.
- Published
- 2018
- Full Text
- View/download PDF
48. Abstract 5497: Effective combination therapy for breast cancer targeting BACH1 and mitochondrial metabolism
- Author
-
Jielin Yan, Jiyoung Lee, Daniel C. Rabe, Peter C. Hart, Felicia D. Rustandy, Jason W. Locasale, Mohamad Elbaz, Marcelo G. Bonini, Elizabeth A. Grossman, Marsha Rich Rosner, Christie Kang, Daniel K. Nomura, Casey Frankenberger, and Ali Ekrem Yesilkanal
- Subjects
Cancer Research ,Combination therapy ,business.industry ,Cancer ,medicine.disease ,Small hairpin RNA ,Breast cancer ,Oncology ,Gene expression ,Transcriptional regulation ,medicine ,Cancer research ,business ,Transcription factor ,Triple-negative breast cancer - Abstract
Oxidative phosphorylation is an attractive target for cancer therapy. Reprogramming metabolic pathways by promoting oxidative phosphorylation could improve the ability of metabolic inhibitors to suppress cancers with limited treatment options like triple negative breast cancer (TNBC). Here we show that BACH1, a heme-binding transcription factor whose expression is enriched in patients with TNBC, inhibits oxidative phosphorylation through direct transcriptional regulation of electron transport chain (ETC) gene expression. Treatment of cells with hemin, which induces BACH1 degradation, mimics BACH1 depletion with shRNA. Pretreatment of TNBC tumors with BACH1 shRNA or hemin overcame resistance to metformin, an anti-diabetic drug, and abolished the growth of both cell line and patient-derived tumor xenografts. BACH1 gene expression inversely correlated with ETC gene expression in breast cancer patients as well as other tumor types, highlighting the clinical relevance. This study demonstrates that oxidative phosphorylation represents an Achilles heel that can be exploited through targeting BACH1 to sensitize breast cancer and potentially other tumor tissues to mitochondrial inhibitors. Citation Format: Jiyoung Lee, Ali Yesilkanal, Casey Frankenberger, Mohamad Elbaz, Daniel Rabe, Jielin Yan, Felicia Rustandy, Peter Hart, Christie Kang, Elizabeth Grossman, Jason Locasale, Daniel Nomura, Marcelo Bonini, Marsha Rosner. Effective combination therapy for breast cancer targeting BACH1 and mitochondrial metabolism [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 5497.
- Published
- 2018
- Full Text
- View/download PDF
49. Abstract 5474: Activity of the S1P pathway promotes ovarian cancer and serves as a novel metabolic target of metformin
- Author
-
Tatsuyuki Chiyoda, Ricardo R. Lastra, Xiaojing Liu, Ernst Lengyel, Chun-Yi Chiang, Iris L. Romero, Marion Curtis, Peter C. Hart, Stephanie M. McGregor, and Jason W. Locasale
- Subjects
Cancer Research ,endocrine system diseases ,Sphingosine ,Kinase ,business.industry ,Cancer ,medicine.disease ,Sphingolipid ,Metformin ,chemistry.chemical_compound ,Oncology ,chemistry ,Tumor progression ,Cancer research ,medicine ,Receptor ,Ovarian cancer ,business ,medicine.drug - Abstract
Sphingosine-1-phosphate (S1P), a metabolite of the phospholipid sphingosine, is a bioactive lipid that directly interacts with specific G-protein-coupled receptors to transduce signals that regulate growth, proliferation and motility. Sphingosine kinases (SPHK1 and 2) catalyze the formation of S1P from sphingosine, thereby regulating S1P homeostasis and the sphingolipid rheostat. Recently, using TCGA data, we noted SPHK1 amplification in several gynecologic cancers and that high expression of SPHK1 was associated with increased mortality in high-grade serous ovarian cancer (HGSOC). The findings reported here show that exposure of HGSOC tumor cells to exogenous S1P, or overexpression of SPHK1, induced migration, proliferation and clonogenecity in multiple ovarian cancer cell lines in vitro. Likewise, in a xenograft mouse model of ovarian cancer, overexpression of SPHK1 markedly enhanced tumor growth. In prior pre-clinical studies, we have demonstrated a strong protective effect of the diabetes medication metformin in ovarian cancer. Interestingly, we found that patients with HGSOC that use metformin for diabetes have reduced serum S1P levels compared to controls, suggesting that the sphingolipid rheostat may be a novel metabolic target of metformin in ovarian cancer. Supporting this, we identified that SPHK1 expression in ovarian cancer cell lines is reduced by treatment with metformin, and further that the reduction of SPHK1 by metformin was mediated through inhibition of transcriptional activity of hypoxia-inducible factors (HIF1α and HIF2α). Finally, we show that overexpression of SPHK1 in HGSOC cell lines enhanced the cytotoxic effects of metformin. Taken together, our data indicates that hypoxia-induced SPHK1 expression and downstream S1P signaling promote tumor progression in HGSOC, and that tumors utilizing this pathway may be particularly vulnerable to the anti-cancer effects of metformin. Citation Format: Peter C. Hart, Tatsuyuki Chiyoda, Marion Curtis, Xiaojing Liu, Chun-Yi Chiang, Stephanie McGregor, Ricardo Lastra, Jason Locasale, Ernst Lengyel, Iris L. Romero. Activity of the S1P pathway promotes ovarian cancer and serves as a novel metabolic target of metformin [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 5474.
- Published
- 2018
- Full Text
- View/download PDF
50. Qui non proficit, deficit: Experimental models for ‘integrative’ research of affective disorders
- Author
-
Justin L. LaPorte, Peter C. Hart, Carisa L. Bergner, Jonathan Cachat, Rupert J. Egan, Allan V. Kalueff, and Peter R. Canavello
- Subjects
Mice, Knockout ,Genotype ,Models, Genetic ,Mood Disorders ,Research ,Brain ,Translational research ,Social Environment ,Epigenesis, Genetic ,Rats ,Developmental psychology ,Disease Models, Animal ,Mice ,Psychiatry and Mental health ,Clinical Psychology ,Phenotype ,Animals ,Humans ,Genetic Predisposition to Disease ,Biological psychiatry ,Psychology ,Cognitive psychology - Abstract
Experimental models are an important tool for the study of biological mechanisms of psychiatric disorders. Although encouraging progress has been made in biological psychiatry of affective disorders, there remain numerous methodological, conceptual, and translational challenges in this field. Mounting clinical data support the view that psychiatric disorders as spectra, rather than as discrete or isolated illnesses. This requires new theories as well as new animal paradigms for “integrative” modeling of psychiatric disorders and their spectra. Here we discuss recent “integrative” experimental models and concepts that promise to advance translational research of affective disorders.
- Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.