Maria Celia Fernandez
McGill University
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Maria Celia Fernandez.
Cancer Research | 2015
Boram Ham; Ni Wang; Zarina D'Costa; Maria Celia Fernandez; Patrick Auguste; Martin Illemann; Rikke L. Eefsen; Gunilla Høyer-Hansen; Ben Vainer; Maximilien Evrard; Zu-Hua Gao; Pnina Brodt
Successful colonization by a cancer cell of a distant metastatic site requires immune escape in the new microenvironment. TNF signaling has been implicated broadly in the suppression of immune surveillance that prevents colonization at the metastatic site and therefore must be blocked. In this study, we explored how TNF signaling influences the efficiency of liver metastasis by colon and lung carcinoma in mice that are genetically deficient for the TNF receptor TNFR2. We found a marked reduction in liver metastases that correlated with a greatly reduced accumulation at metastatic sites of CD11b(+)GR-1(+) myeloid cells with enhanced arginase activity, identified as myeloid-derived suppressor cells (MDSC). Reduced infiltration of MDSC coincided with a reduction in the number of CD4(+)FoxP3(+) T regulatory cells in the tumors. Reconstitution of TNFR2-deficient mice with normal bone marrow, or adoptive transfer of TNFR2-expressing MDSC into these mice, was sufficient to restore liver metastasis to levels in wild-type mice. Conversely, treatment with TNFR2 antisense oligodeoxynucleotides reduced liver metastasis in wild-type mice. Clinically, immunohistochemical analysis of liver metastases from chemotherapy-naïve colon cancer patients confirmed the presence of CD33(+)HLA-DR(-)TNFR2(+) myeloid cells in the periphery of hepatic metastases. Overall, our findings implicate TNFR2 in supporting MDSC-mediated immune suppression and metastasis in the liver, suggesting the use of TNFR2 inhibitors as a strategy to prevent metastatic progression to liver in colon, lung, and various other types of cancer.
Molecular Human Reproduction | 2016
Donghyun Lee; Adel R. Moawad; Tania Morielli; Maria Celia Fernandez; O'Flaherty Cristian
STUDY QUESTION Do peroxiredoxins (PRDXs) control reactive oxygen species (ROS) levels during human sperm capacitation? SUMMARY ANSWER PRDXs are necessary to control the levels of ROS generated during capacitation allowing spermatozoa to achieve fertilizing ability. WHAT IS KNOWN ALREADY Sperm capacitation is an oxidative event that requires low and controlled amounts of ROS to trigger phosphorylation events. PRDXs are antioxidant enzymes that not only act as scavengers but also control ROS action in somatic cells. Spermatozoa from infertile men have lower levels of PRDXs (particularly of PRDX6), which are thiol-oxidized and therefore inactive. STUDY DESIGN, SIZE, DURATION Semen samples were obtained from a cohort of 20 healthy nonsmoker volunteers aged 22-30 years old over a period of 1 year. PARTICIPANTS/MATERIALS, SETTINGS, METHODS Sperm from healthy donors was capacitated with fetal cord serum ultrafiltrate (FCSu) in the absence or presence of thiostrepton (TSP), inhibitor of 2-Cys PRDXs or 1-Hexadecyl-3-(trifluoroethyl)-sn-glycero-2-phosphomethanol lithium (MJ33), inhibitor of calcium independent-phospholipase A2 (Ca2+-iPLA2) activity of PRDX6, added at different times of incubation. Capacitation was also induced by the dibutyryl cAMP+3-isobuty1-1-methylxanthine system. Sperm viability and motility were determined by the hypo-osmotic swelling test and computer-assisted semen analysis system, respectively. Capacitation was determined by the ability of spermatozoa to undergo the acrosome reaction triggered by lysophosphatidylcholine. Percentages of acrosome reaction were obtained using the FITC-conjugated Pisum sativum agglutinin assay. Phosphorylation of tyrosine residues and of protein kinase A (PKA) substrates were determined by sodium dodecyl sulfate polyacrylamide gel electrophoresis immunoblotting with specific antibodies. Actin polymerization was determined by phalloidin labeling. MAIN RESULTS AND THE ROLE OF CHANCE TSP and MJ33 prevented sperm capacitation and its associated actin polymerization in spermatozoa incubated with 10% FCSu (capacitation inducer) compared to non-capacitated controls (P < 0.05) without altering sperm viability. PKA substrates and tyrosine phosphorylations were prevented in FCSu-treated spermatozoa in a differential fashion depending on the type and the time of addition of the inhibitor used compared to non-capacitated controls (P < 0.05). TSP and MJ33 promoted an increase of lipid peroxidation in spermatozoa (P < 0.01) and these levels were higher in those spermatozoa incubated with the inhibitors and FCSu compared to those capacitated spermatozoa incubated without the inhibitors (P < 0.0001). Inhibition of 2-Cys PRDXs by TSP generated an oxidative stress in spermatozoa, affecting their viability compared to controls (P < 0.05). This oxidative stress was prevented by nuclephile D-penicillamine (PEN). MJ33 also promoted an increase of lipid peroxidation and impaired sperm viability compared to non-treated controls (P < 0.05) but its effect was not circumvented by PEN, suggesting that not only peroxidase but also Ca2+-iPLA2 activity of PRDX6 are necessary to guarantee viability in human spermatozoa. LARGE SCALE DATA Not applicable. LIMITATIONS REASONS FOR CAUTION We focused on the global effect of PRDXs inhibitors on human sperm capacitation and in two of its associated phosphorylation events. Thus, other phosphorylation events and mechanisms necessary for capacitation may also be affected. WIDER IMPLICATIONS OF THE FINDINGS PRDXs are the major antioxidant system in ejaculated spermatozoa and are necessary to allow spermatozoon to achieve fertilizing ability (capacitation and acrosome reaction). STUDY FUNDING/COMPETING INTEREST(S) This research was supported by Canadian Institutes of Health Research (MOP 133661) and the Fonds de Recherché en Santé Quebec (FRSQS #22151) to C.O. The authors have nothing to disclose.
British Journal of Cancer | 2017
Pieter-Jan van Dam; Eric P. van der Stok; Laure-Anne Teuwen; Gert Van den Eynden; Martin Illemann; Sophia Frentzas; A. W. Majeed; Rikke L. Eefsen; Robert R.J. Coebergh van den Braak; Anthoula Lazaris; Maria Celia Fernandez; Boris Galjart; Ole Didrik Laerum; Roni F. Rayes; Dirk J. Grünhagen; Michelle Van de paer; Yves Sucaet; Hardeep Singh Mudhar; Michael Schvimer; Hanna Nyström; Mark Kockx; Nigel C. Bird; Fernando Vidal-Vanaclocha; Peter Metrakos; Eve Simoneau; Cornelis Verhoef; Luc Dirix; Steven Van Laere; Zu-Hua Gao; Pnina Brodt
Background:Liver metastases present with distinct histopathological growth patterns (HGPs), including the desmoplastic, pushing and replacement HGPs and two rarer HGPs. The HGPs are defined owing to the distinct interface between the cancer cells and the adjacent normal liver parenchyma that is present in each pattern and can be scored from standard haematoxylin-and-eosin-stained (H&E) tissue sections. The current study provides consensus guidelines for scoring these HGPs.Methods:Guidelines for defining the HGPs were established by a large international team. To assess the validity of these guidelines, 12 independent observers scored a set of 159 liver metastases and interobserver variability was measured. In an independent cohort of 374 patients with colorectal liver metastases (CRCLM), the impact of HGPs on overall survival after hepatectomy was determined.Results:Good-to-excellent correlations (intraclass correlation coefficient >0.5) with the gold standard were obtained for the assessment of the replacement HGP and desmoplastic HGP. Overall survival was significantly superior in the desmoplastic HGP subgroup compared with the replacement or pushing HGP subgroup (P=0.006).Conclusions:The current guidelines allow for reproducible determination of liver metastasis HGPs. As HGPs impact overall survival after surgery for CRCLM, they may serve as a novel biomarker for individualised therapies.
Scientific Reports | 2017
Adel R. Moawad; Maria Celia Fernandez; Eleonora Scarlata; Chandra Dodia; Sheldon I. Feinstein; Aron B. Fisher; Cristian O’Flaherty
Prdx6−/− male mice are subfertile, and the deficiency or inactivation of Peroxiredoxins (PRDXs) is associated with human male infertility. We elucidate the impact of the lack of PRDX6 or inhibition of its calcium-independent phospholipase A2 (Ca2+-iPLA2) activity by MJ33 on fertilization competence of mouse spermatozoa. Sperm motility, viability, fertilization and blastocyst rates were lower in Prdx6−/− spermatozoa than in C57BL/6J wild-type (WT) controls (p ≤ 0.05). MJ33 inhibited the PRDX6 Ca2+-iPLA2 activity and reduced these parameters in WT spermatozoa compared with controls (p ≤ 0.05). Levels of lipid peroxidation and of superoxide anion (O2•─) were higher in Prdx6−/− than in WT spermatozoa (p ≤ 0.05). MJ33 increased the levels of lipid peroxidation and mitochondrial O2•─ production in treated versus non-treated WT spermatozoa. Acrosome reaction, binding to zona pellucida and fusion with the oolemma were lower in Prdx6−/− capacitated spermatozoa than WT capacitated controls and lower in WT spermatozoa treated with the PRDX6 inhibitor. In conclusion, the inhibition of the PRDX6 Ca2+-iPLA2 activity promotes an oxidative stress affecting viability, motility, and the ability of mouse spermatozoa to fertilize oocytes. Thus, PRDX6 has a critical role in the protection of the mouse spermatozoon against oxidative stress to assure fertilizing competence.
Oncotarget | 2017
Maria Celia Fernandez; Roni F. Rayes; Boram Ham; Ni Wang; Simon Milette; Martin lllemann; Nigel C. Bird; A. W. Majeed; Jun Xu; Tatiana Kisselova; Pnina Brodt
Hepatic stellate cells (HSC) play a major role in initiating the liver fibrogenic (wounding) response of the liver and can also orchestrate a pro-metastatic microenvironment in the liver in response to invading cancer cells. Here we explored the role of the hepatic stellate cells in colon carcinoma liver metastasis with emphasis on the contribution of the insulin-like growth factor (IGF) axis to their activation and function. To this end, we used mice with a Tamoxifen inducible liver IGF-I deficiency. We found that in mice with a sustained IGF-I deficiency, recruitment and activation of HSC into tumor-infiltrated areas of the liver were markedly diminished, resulting in decreased collagen deposition and reduced tumor expansion. In addition, IGF-I could rescue HSC from apoptosis induced by pro-inflammatory factors such as TNF-α known to be upregulated in the early stages of liver metastasis. Moreover, in surgical specimens, activated IGF-IR was observed on HSC-like stromal cells surrounding colorectal carcinoma liver metastases. Finally, IGF-targeting in vivo using an IGF-Trap caused a significant reduction in HSC activation in response to metastatic colon cancer cells. Therefore, our data identify IGF as a survival factor for HSC and thereby, a promoter of the pro-metastatic microenvironment in the liver. IGF-targeting could therefore provide a strategy for curtailing the pro-metastatic host response of the liver during the early stages of liver metastasis.Hepatic stellate cells (HSC) play a major role in initiating the liver fibrogenic (wounding) response of the liver and can also orchestrate a pro-metastatic microenvironment in the liver in response to invading cancer cells. Here we explored the role of the hepatic stellate cells in colon carcinoma liver metastasis with emphasis on the contribution of the insulin-like growth factor (IGF) axis to their activation and function. To this end, we used mice with a Tamoxifen inducible liver IGF-I deficiency. We found that in mice with a sustained IGF-I deficiency, recruitment and activation of HSC into tumor-infiltrated areas of the liver were markedly diminished, resulting in decreased collagen deposition and reduced tumor expansion. In addition, IGF-I could rescue HSC from apoptosis induced by pro-inflammatory factors such as TNF-α known to be upregulated in the early stages of liver metastasis. Moreover, in surgical specimens, activated IGF-IR was observed on HSC-like stromal cells surrounding colorectal carcinoma liver metastases. Finally, IGF-targeting in vivo using an IGF-Trap caused a significant reduction in HSC activation in response to metastatic colon cancer cells. Therefore, our data identify IGF as a survival factor for HSC and thereby, a promoter of the pro-metastatic microenvironment in the liver. IGF-targeting could therefore provide a strategy for curtailing the pro-metastatic host response of the liver during the early stages of liver metastasis.
Oncotarget | 2018
Roni F. Rayes; Simon Milette; Maria Celia Fernandez; Boram Ham; Ni Wang; Stephanie Perrino; Shoshana Yakar; Pnina Brodt
The growth of cancer metastases in the liver depends on a permissive interaction with the hepatic microenvironment and neutrophils can contribute to this interaction, either positively or negatively, depending on their phenotype. Here we investigated the role of IGF-I in the control of the tumor microenvironment in the liver, using mice with a conditional, liver-specific, IGF-I deficiency (iLID) induced by a single tamoxifen injection. In mice that had a sustained (3 weeks) IGF-I deficiency prior to the intrasplenic/portal inoculation of colon carcinoma MC-38 cells, we observed an increase in neutrophil accumulation in the liver relative to controls. However, unlike controls, these neutrophils did not acquire the (anti-inflammatory) tumor-promoting phenotype, as evidenced by retention of high ICAM-1 expression and nitric oxide production and low CXCR4, CCL5, and VEGF expression and arginase production, all characteristic of the (pro-inflammatory) phenotype. This coincided with an increase in apoptotic tumor cells and reduced metastasis. Neutrophils isolated from these mice also had reduced IGF-IR expression levels. These changes were not observed in iLID mice with a short-term (2 days) IGF-I depletion, despite a 70% reduction in their circulating IGF-I levels, indicating that a sustained IGF-I deficiency was necessary to alter the neutrophil phenotype. Similar results were obtained with the highly metastatic Lewis lung carcinoma subline H-59 cells and in mice injected with an IGF-Trap that blocks IGF-IR signaling by reducing ligand bioavailability. Our results implicate the IGF axis in neutrophil polarization and the induction of a pro-metastatic microenvironment in the liver.
Human Reproduction | 2018
Maria Celia Fernandez; Cristian O’Flaherty
STUDY QUESTION Are all components of the peroxiredoxins (PRDXs) system important to control the levels of reactive oxygen species (ROS) to maintain viability and DNA integrity in spermatozoa? SUMMARY ANSWER PRDX6 is the primary player of the PRDXs system for maintaining viability and DNA integrity in human spermatozoa. WHAT IS KNOWN ALREADY Mammalian spermatozoa are sensitive to high levels of ROS and PRDXs are antioxidant enzymes proven to control the levels of ROS generated during sperm capacitation to avoid oxidative damage in the spermatozoon. Low amounts of PRDXs are associated with male infertility. The absence of PRDX6 promotes sperm oxidative damage and infertility in mice. STUDY DESIGN, SIZE, DURATION Semen samples were obtained over a period of one year from a cohort of 20 healthy non-smoking volunteers aged 22-30 years old. PARTICIPANTS/MATERIALS, SETTING, METHODS Sperm from healthy donors was incubated for 2 h in the absence or presence of inhibitors for the 2-Cys PRDXs system (peroxidase, reactivation system and NADPH-enzymes suppliers) or the 1-Cys PRDX system (peroxidase and calcium independent-phospholipase A2 (Ca2+-iPLA2) activity). Sperm viability, DNA oxidation, ROS levels, mitochondrial membrane potential and 4-hydroxynonenal production were determined by flow cytometry. MAIN RESULTS AND THE ROLE OF CHANCE We observed a significant decrease in viable cells due to inhibitors of the 2-Cys PRDXs, PRDX6 Ca2+-iPLA2 activity or the PRDX reactivation system compared to controls (P ≤ 0.05). PRDX6 Ca2+-iPLA2 activity inhibition had the strongest detrimental effect on sperm viability and DNA oxidation compared to controls (P ≤ 0.05). The 2-Cys PRDXs did not compensate for the inhibition of PRDX6 peroxidase and Ca2+-iPLA2 activities. LARGE SCALE DATA Not applicable. LIMITATIONS, REASONS FOR CAUTION Players of the reactivation systems may differ among mammalian species. WIDER IMPLICATIONS OF THE FINDINGS The Ca2+-iPLA2 activity of PRDX6 is the most important and first line of defense against oxidative stress in human spermatozoa. Peroxynitrite is scavenged mainly by the PRDX6 peroxidase activity. These findings can help to design new diagnostic tools and therapies for male infertility. STUDY FUNDING/COMPETING INTEREST(S) This research was supported by The Canadian Institutes of Health Research (MOP 133661 to C.O.), and by RI MUHC-Desjardins Studentship in Child Health Research awarded to M.C.F. The authors have nothing to disclose.
Cancer Research | 2017
Jennifer Tsui; George Vaniotis; Maria Celia Fernandez; Pnina Brodt
The triple negative subtypes of breast cancer (TNBC) are associated with poor prognosis. Unlike HER2+ and hormone receptor-positive BC, TNBC do not respond to targeted therapy and chemotherapy remains the primary treatment option. There is therefore an unmet need to develop effective therapy for TNBC. The insulin-like growth factor 1 (IGF-I) axis plays a critical role in BC progression by conveying survival and growth signals. Our laboratory reported on the production of a soluble fusion protein comprised of the extracellular domain of human IGF-IR fused to the Fc portion of human IgG (the IGF-Trap). The IGF-Trap reduces the bioavailability of circulating and locally produced IGF-I, thereby limiting tumor growth. When human TNBC MDA-MB-231 cells were xenotransplanted into nude mice and treated with the IGF-Trap, we observed variability in the response as it ranged from complete tumor regression to disease stabilization and tumor progression in some mice. This suggested that MDA-MB-231 cells are heterogeneous in respect to their sensitivity to IGF-IR signaling blockade. The aim of the present study was to identify resistance mechanisms that allow the cells to progress in the face of IGF-IR signaling blockade by the IGF-Trap. We first analyzed the tyrosine kinase receptor profile of these cells and confirmed by PCR that in addition to IGF-IR, they express epidermal growth factor receptor (EGFR), c-Met, and fibroblast growth factor receptor 1 (FGFR1). They also produce IGF-I, EGF and relatively high level of FGF1 that could provide potential autocrine signaling to compensate for IGF signaling blockade. Using limiting dilution cloning, we isolated MDA-MB-231 cells with a range of IGF-IR expression levels, as confirmed by qPCR and Western blotting. We found that clones with higher basal IGF-IR activation levels, independently of expression levels had increased sensitivity to IGF-Trap treatment in the presence of serum, identifying them as IGF-addicted clonal subpopulations. Furthermore, an IGF-Trap resistant population selected from MDA-MB-231 cells by prolonged exposure to the IGF-Trap had an increased proliferation rate in the presence of the IGF-Trap as compared to unselected cells, as assessed by MTT and showed higher p-EGFR and p-ERK levels, suggesting that prolonged IGF-Trap treatment enriched an IGF-I-independent population with increased aggressiveness. Collectively these results showed that MDA-MB-231 cells are heterogeneous in respect to IGF-IR expression levels and that IGF-Trap sensitivity correlated with constitutive IGF-IR activation levels. Moreover, IGF-Trap resistance in these cells was associated with increased EGFR signaling and proliferation. Further interrogation of the gene expression profile of these cells will define a “resistance signature” with potential relevance to personalized treatment with IGF-targeting drugs. Supported by the CIHR and Mitacs. Citation Format: Jennifer Tsui, George Vaniotis, Maria Celia Fernandez, Pnina Brodt. Identification of resistance mechanisms to IGF-IR targeting in triple negative breast cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 5881. doi:10.1158/1538-7445.AM2017-5881
Cancer Research | 2015
Maria Celia Fernandez; Roni F. Rayes; Jun Xu; Tatiana Kisseleva; Shoshana Yakar; Pnina Brodt
Proceedings: AACR 106th Annual Meeting 2015; April 18-22, 2015; Philadelphia, PA Background: The hepatic stellate cells (HSC) play a major role in orchestrating the livers fibrogenic (wounding) response and have been identified as an important player in the prometastatic microenvironment in the liver. Here we analyzed the role of the IGF axis in the recruitment and activation of HSC during the early stages of colorectal carcinoma (CRC) liver metastasis. Methods: Murine CRC MC-38 cells were inoculated via the intrasplenic/portal route into female mice with a conditional liver IGF-1 deletion (iLID), induced by a single tamoxifen (Tx) injection 2 days or 3 weeks earlier. Vehicle (sunflower oil) injected iLID mice or Tx - injected wild type mice were used as controls. Experimental liver fibrosis was achieved by repeated carbon tetrachloride (CCl4) administration. The stromal response of the liver was analyzed using immunohistochemistry with emphasis on HSC recruitment and activation. In addition, in vitro assays were utilized to explore the role of IGF-1 in HSC activation. Results: In iLID mice treated with Tx, a 75% reduction in circulating IGF-1 levels could be observed within 24 hr and it persisted for the duration of the experiments. When injected with MC-38 cells, 3 weeks post Tx injection, these mice developed significantly fewer liver metastases than non-treated controls, while no reduction in the number of metastases was seen in WT mice injected with Tx or in oil-injected iLID controls. Interestingly, we observed that Tx treatment 48 hr prior to tumor injection, failed to reduce liver metastasis in iLID mice, although their circulating IGF-1 levels were markedly reduced, suggesting that the loss of direct paracrine IGF-1 effects on the tumor cells was not sufficient to inhibit tumor growth in the liver and that other effects on the hepatic microenvironment were at play. Analysis of HSC recruitment and activation subsequently revealed a significant reduction in HSC activation around micrometastases as compared to controls. This was evident as early as 3 days post tumor inoculation, persisted for the duration of the experimental period (16-18 days) and corresponded with reduced IGF-1 receptor and Akt activation in these cells. In vitro studies confirmed that IGF-1 could directly activate isolated HSC and rescue them from serum-depletion induced apoptosis. Finally, in iLID mice with sustained IGF-1 depletion, a significant reduction in HSC-mediated collagen deposition was observed following continuous treatment with CCl4, confirming the role of IGF-1 in HSC activation in a second tumor-free model. Conclusion: Our results show that a sustained reduction in circulating IGF-1 levels altered HSC recruitment and activation to tumor sites and reduced tumor cell growth in the liver. We identify IGF-1 as a regulator of HSC function and the response of the microenvironment to invading cancer cells, thereby affecting metastatic expansion. Supported by CIHR Grant MOP 81201 (to PB) and MICRTP Fellowship (to MCF). Note: This abstract was not presented at the meeting. Citation Format: Maria C. Fernandez, Roni F. Rayes, Jun Xu, Tatiana Kisseleva, Shoshana Yakar, Pnina Brodt. The IGF axis regulates hepatic stellate cell recruitment and activation during colorectal carcinoma liver metastasis. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 5081. doi:10.1158/1538-7445.AM2015-5081
Trends in cancer research | 2016
Boram Ham; Maria Celia Fernandez; D'Costa Z; Pnina Brodt