Network


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

Hotspot


Dive into the research topics where Gabriel E. DiMattia is active.

Publication


Featured researches published by Gabriel E. DiMattia.


Stem Cells | 2009

Multipotent Stromal Cells Are Activated to Reduce Apoptosis in Part by Upregulation and Secretion of Stanniocalcin‐1

Gregory J. Block; Shinya Ohkouchi; Joshua W. Frenkel; Carl A. Gregory; Radhika Pochampally; Gabriel E. DiMattia; Deborah E. Sullivan; Darwin J. Prockop

Multipotent stromal cells (MSCs) have been shown to reduce apoptosis in injured cells by secretion of paracrine factors, but these factors were not fully defined. We observed that coculture of MSCs with previously UV‐irradiated fibroblasts reduced apoptosis of the irradiated cells, but fresh MSC conditioned medium was unable reproduce the effect. Comparative microarray analysis of MSCs grown in the presence or absence of UV‐irradiated fibroblasts demonstrated that the MSCs were activated by the apoptotic cells to increase synthesis and secretion of stanniocalcin‐1 (STC‐1), a peptide hormone that modulates mineral metabolism and has pleiotrophic effects that have not been fully characterized. We showed that STC‐1 was required but not sufficient for reduction of apoptosis of UV‐irradiated fibroblasts. In contrast, we demonstrated that MSC‐derived STC‐1 was both required and sufficient for reduction of apoptosis of lung cancer epithelial cells made apoptotic by incubation at low pH in hypoxia. Our data demonstrate that STC‐1 mediates the antiapoptotic effects of MSCs in two distinct models of apoptosis in vitro. STEM CELLS 2009;27:670–681


Endocrinology | 1998

Comparative analysis of mammalian stanniocalcin genes.

Robin Varghese; Chris K.C. Wong; Harminder K. Deol; Graham F. Wagner; Gabriel E. DiMattia

The recent discovery of mammalian stanniocalcin (STC) prompted an investigation of its gene structure and expression pattern to study its function and regulation. We show that both the human and mouse genes are composed of four exons spanning about 13 kb, with 85% nucleotide sequence identity in coding regions. Remarkably high sequence conservation between species also exists in the approximately 3-kb 3′-untranslated region. Comparative analysis of the 5′-untranslated region and flanking DNA from the rat and human STC genes showed long stretches of CAG trinucleotide repeats and an additional (CA)25 dinucleotide repeat unique to the rat promoter. An analysis of STC expression in the mouse showed that ovary contained the highest level of messenger RNA, with lower, but detectable, levels in most tissues. In situ hybridization revealed strong, specific hybridization over the thecal-interstitial cells of the ovarian stroma, whereas immunohistochemical analysis indicated that STC was present not only in the str...


Molecular and Cellular Endocrinology | 1998

Molecular cloning and characterization of stanniocalcin-related protein.

Gabriel E. DiMattia; Robin Varghese; Graham F. Wagner

Stanniocalcin (STC) is a glycoprotein hormone first discovered in fish and recently identified in humans and mice. In this report we have described the cloning of an STC-like cDNA, designated here as STC related protein (STCrP). Human STCrP (hSTCrP) cDNAs were isolated as expressed sequence tags (ESTs) and predicted a polypeptide of 302 amino acids, with 58%, homology to human STC (hSTC). Ten of the eleven 1/2 cysteine residues that in STC allow for dimerization of monomeric subunits were conserved in hSTCrP. By Northern analysis, three hSTCrP mRNAs were detected and were most abundant in pancreas, spleen and kidney as well as a variety of different transformed cell lines. The high degree of sequence homology suggests that STC and STCrP may have been derived from a common ancestral gene.


American Journal of Pathology | 2009

Anti-Inflammatory and Renal Protective Actions of Stanniocalcin-1 in a Model of Anti-Glomerular Basement Membrane Glomerulonephritis

Luping Huang; Gabriela Garcia; Ya Huan Lou; Qin Zhou; Luan D. Truong; Gabriel E. DiMattia; Xia Ru Lan; Hui Y. Lan; Yanlin Wang; David Sheikh-Hamad

We have previously shown that stanniocalcin-1 (STC1) inhibits the transendothelial migration of macrophages and T cells, suppresses superoxide generation in macrophages, and attenuates macrophage responses to chemoattractants. To study the effects of STC1 on inflammation, in this study we induced a macrophage- and T-cell-mediated model of anti-glomerular basement membrane disease in STC1 transgenic mice, which display elevated serum STC1 levels and preferentially express STC1 in both endothelial cells and macrophages. We examined the following parameters both at baseline and after anti-glomerular basement membrane antibody treatment: blood pressure; C(3a) levels; urine output; proteinuria; blood urea nitrogen; and kidney C(3) deposition, fibrosis, histological changes, cytokine expression, and number of T cells and macrophages. Compared with wild-type mice, after anti-glomerular basement membrane treatment STC1 transgenic mice exhibited: i) diminished infiltration of inflammatory macrophages in the glomeruli; ii) marked reduction in crescent formation and sclerotic glomeruli; iii) decreased interstitial fibrosis; iv) preservation of kidney function and lower blood pressure; v) diminished C(3) deposition in the glomeruli; and vi) reduced expression of macrophage inhibitory protein-2 and transforming growth factor-beta2 in the kidney. Compared with baseline, wild-type mice, but not STC1 transgenic mice, had higher proteinuria and a marked reduction in urine output. STC1 had minimal effects, however, on both T-cell number in the glomeruli and interstitium and on cytokine expression characteristic of either TH1 or TH2 activation. These data suggest that STC1 is a potent anti-inflammatory and renal protective protein.


Journal of Endocrinology | 2008

Stanniocalcin 2 expression is regulated by hormone signalling and negatively affects breast cancer cell viability in vitro.

Sanda Raulic; Yudith Ramos-Valdes; Gabriel E. DiMattia

Stanniocalcin 1 (STC1) and STC2 are secreted, homodimeric glycoproteins that share 30% amino acid sequence identity. Breast tumour gene profiling studies have demonstrated significantly upregulated STC2 expression in hormone-responsive positive breast tumours; therefore, the purpose of this study was to investigate STC2 hormonal regulation and function in breast cancer cells. Here we report that STC2 is expressed in a number of human breast cancer cell lines, regardless of their oestrogen (E(2)) and progesterone (P4) receptor status, and its expression is readily detectable in human and mouse mammary gland tumours. Besides E(2), retinoic acid (RA) and P4 play an important role in the regulation of STC2 expression, not only in MCF-7 but also in other breast cancer and non-breast cell lines. The expression of the related hormone, STC1, is not affected by the above hormones in breast and endometrial cancer cell lines implying a fundamental difference in regulation in cancer cell lines. The induction of STC2 expression by E(2) and RA occurs at the transcriptional level but through intermediary transcription factors. The STC2 proximal promoter region is not responsible for hormonal induction, but exhibits a high basal transcriptional activity. Constitutive STC2 expression in human breast cancer cell lines resulted in significant impairment of cell growth, migration and cell viability after serum withdrawal. In conclusion, STC2 is a downstream target of E(2), P4 and RA signalling pathways. In hormone receptor negative cell lines it can function in a paracrine/autocrine fashion to reduce cell proliferation.


Kidney International | 2012

Overexpression of stanniocalcin-1 inhibits reactive oxygen species and renal ischemia/reperfusion injury in mice

Luping Huang; Tatiana Belousova; Minyi Chen; Gabriel E. DiMattia; Dajun Liu; David Sheikh-Hamad

Reactive oxygen species, endothelial dysfunction, inflammation, and mitogen-activated protein kinases have important roles in the pathogenesis of ischemia/reperfusion kidney injury. Stanniocalcin-1 (STC1) suppresses superoxide generation in many systems through induction of mitochondrial uncoupling proteins and blocks the cytokine-induced rise in endothelial permeability. Here we tested whether transgenic overexpression of STC1 protects from bilateral ischemia/reperfusion kidney injury. This injury in wild type mice caused a halving of the creatinine clearance; severe tubular vacuolization and cast formation; increased infiltration of macrophages and T cells; higher vascular permeability; greater production of superoxide and hydrogen peroxide; and higher ratio of activated ERK/activated JNK and p38, all compared to sham-treated controls. Mice transgenic for human STC1 expression, however, had resistance to equivalent ischemia/reperfusion injury indicated as no significant change from controls in any of these parameters. Tubular epithelial cells in transgenic mice expressed higher mitochondrial uncoupling protein 2 and lower superoxide generation. Pre-treatment of transgenic mice with paraquat, a generator of reactive oxygen species, before injury restored the susceptibility to ischemia/reperfusion kidney injury, suggesting that STC1 protects by an anti-oxidant mechanism. Thus, STC1 may be a therapeutic target for ischemia/reperfusion kidney injury.


Carcinogenesis | 2012

Modulation of AKT activity is associated with reversible dormancy in ascites-derived epithelial ovarian cancer spheroids

Rohann J. M. Correa; Teresa Peart; Yudith Ramos Valdes; Gabriel E. DiMattia; Trevor G. Shepherd

Epithelial ovarian cancer (EOC) metastasis is a direct contributor to high recurrence and low survival for patients with this disease. Metastasis in EOC occurs by cell exfoliation from the primary tumor into the fluid-filled peritoneal cavity, persistence of these cells as non-adherent multicellular aggregates or spheroids and reattachment of spheroids to form secondary lesions. We have recovered native spheroids from ascites fluid and demonstrated that EOC cells within these structures exhibit reduced proliferation, yet regain the capacity to attach and reinitiate cell division. To model this process in vitro for further investigation, primary EOC cells from patient peritoneal fluid were cultured under non-adherent conditions. Here we show that these cells naturally form spheroids resembling those observed in ascites. Spheroids exhibit reduced cell proliferation and a protein expression pattern consistent with cellular quiescence: specifically, decreased phospho-AKT and p45/SKP2 with a concomitant increase in p130/RBL2 and p27(Kip1). However, when spheroids are seeded to an adherent surface, reattachment occurs rapidly and is followed by reinitiation of AKT-dependent cell proliferation. These results were strikingly consistent among numerous clinical specimens and were corroborated in the EOC cell line OVCAR3. Therefore, our data reveal that EOC cells become quiescent when forming spheroids, but reactivate proliferative mechanisms upon attachment to a permissive substratum. Overall, this work utilizes a novel in vitro model of EOC metastasis that employs primary human EOC cells and introduces the important concept of reversible dormancy in EOC pathogenesis.


Molecular and Cellular Endocrinology | 2001

Dynamic changes in stanniocalcin gene expression in the mouse uterus during early implantation.

Sasha E. Stasko; Gabriel E. DiMattia; Graham F. Wagner

Blastocyst implantation is accompanied by dramatic changes in gene expression to facilitate decidualization and remodelling of uterine architecture. Stanniocalcin (STC) is a new mammalian polypeptide hormone with roles in ion transport, reproduction and development. Here we report dynamic changes in STC mRNA and protein distributions in the early post-implantation mouse uterus. In the non-pregnant state, STC gene expression was confined to the uterine lumenal epithelium. Following implantation STC gene expression shifted to mesometrial stromal cells bordering the uterine lumen. Between E6.5-E8.5 expression shifted once more to cells of the mesometrial lateral sinusoids, and then declined thereafter. Intriguingly immunoreactive STC did not entirely co-localize with areas of high STC gene activity and instead appeared to accumulate in presumptive targets of the hormone (uterine epithelium, stromal and decidual cells, trophoblastic giant cells). STC is only the fourth gene identified as being expressed mesometrially in the uterus following implantation.


Molecular and Cellular Biology | 2009

A Functional Connection between pRB and Transforming Growth Factor β in Growth Inhibition and Mammary Gland Development

Sarah M. Francis; Jacqueline Bergsied; Christian E. Isaac; Courtney H. Coschi; Alison L. Martens; Carlo V. Hojilla; Subrata Chakrabarti; Gabriel E. DiMattia; Rama Khoka; Jean Y. J. Wang; Frederick A. Dick

ABSTRACT Transforming growth factor β (TGF-β) is a crucial mediator of breast development, and loss of TGF-β-induced growth arrest is a hallmark of breast cancer. TGF-β has been shown to inhibit cyclin-dependent kinase (CDK) activity, which leads to the accumulation of hypophosphorylated pRB. However, unlike other components of TGF-β cytostatic signaling, pRB is thought to be dispensable for mammary development. Using gene-targeted mice carrying subtle missense changes in pRB (Rb1ΔL and Rb1NF), we have discovered that pRB plays a critical role in mammary gland development. In particular, Rb1 mutant female mice have hyperplastic mammary epithelium and defects in nursing due to insensitivity to TGF-β growth inhibition. In contrast with previous studies that highlighted the inhibition of cyclin/CDK activity by TGF-β signaling, our experiments revealed that active transcriptional repression of E2F target genes by pRB downstream of CDKs is also a key component of TGF-β cytostatic signaling. Taken together, our work demonstrates a unique functional connection between pRB and TGF-β in growth control and mammary gland development.


Transgenic Research | 2010

Human stanniocalcin-1 or -2 expressed in mice reduces bone size and severely inhibits cranial intramembranous bone growth.

Jennifer L. Johnston; Yudith Ramos-Valdes; Lee-Anne Stanton; Sadia Ladhani; Frank Beier; Gabriel E. DiMattia

Stanniocalcin-1 (STC1) and -2 (STC2) are highly related, secreted, homodimeric glycoproteins that are significantly upregulated by different forms of stress including high phosphate levels. Transgenic mice that constitutively express either human STC1 or STC2 exhibit intra-uterine growth restriction and permanent post-natal growth retardation. STC1 is expressed in chondrocytic and osteoblastic cells during murine development and can enhance differentiation of calvarial cells in culture. Therefore, there is mounting evidence that stanniocalcins (STCs) modulate bone development in vivo. To further define the effects of stanniocalcins on skeletal development, we performed a series of measurements on components of the axial, appendicular, and cranial skeleton in transgenic and wildtype mice. We show that skeletal growth is retarded and that the intramembranous bones of the cranium exhibit a particularly severe delay in suture closure. The posterior frontal suture remains patent throughout the lifetime of human STC1 and STC2 transgenic mice. We did not observe significant effects on chondrogenesis: however, calvarial cells exhibited reduced viability, proliferation and delayed differentiation, indicating that developing osteoblasts are particularly sensitive to the levels of STCs. Given the evidence linking STC1 to cellular phosphate homeostasis, we assessed the expression of a variety of phosphate regulators in transgenic and wildtype calvarial cells and found significantly lower levels of Mepe, Dmp1, Sfrp4 in transgenic cells without a change in Pit1 or Pit2. Collectively these data support a direct regulatory role for STCs in osteoblasts and suggest that overexposure to these factors inhibits normal skeletal development without significant changes in patterning.

Collaboration


Dive into the Gabriel E. DiMattia's collaboration.

Top Co-Authors

Avatar

Trevor G. Shepherd

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Graham F. Wagner

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Yudith Ramos Valdes

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Rohann J. M. Correa

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Teresa Peart

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Yudith Ramos-Valdes

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Luping Huang

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Akira Sugimoto

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Elena N. Fazio

University of Western Ontario

View shared research outputs
Researchain Logo
Decentralizing Knowledge