Network


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

Hotspot


Dive into the research topics where Ognoon Mungunsukh is active.

Publication


Featured researches published by Ognoon Mungunsukh.


International Journal of Radiation Biology | 2008

Genistein induces radioprotection by hematopoietic stem cell quiescence

Thomas A. Davis; Ognoon Mungunsukh; Stephen R. Zins; Regina M. Day; Michael R. Landauer

Purpose: In this study we addressed whether genistein-induced radioprotection in mice is associated with alterations of the cell cycle of hematopoietic stem and progenitor cells. Materials and methods: C57BL/6J female mice received a single subcutaneous injection of genistein (200 mg/kg) 24 h prior to a lethal dose (7.75 Gy, 60Co) of total body irradiation. Proliferation-associated Ki-67 protein/7-aminoactinomycin-D (Ki67/7AAD) cell cycle staining was used to differentiate between G0, G1, and S/G2/M in bone marrow cell populations negative for expression of mature hematopoietic lineage marker cells but positive for expression of stem cell antigen-1 and tyrosine kinase receptor for stem cell factor (Lin−Sca-1+cKit+, LSK+). Quantitative real-time polymerase chain reaction (qRT-PCR) microarrays were utilized to examine cell cycle specific genes. Results: At 24 h following radiation exposure, a greater percentage of LSK+ in genistein-treated mice accumulated in the G0 phase of the cell cycle, whereas a large percentage of LSK+ bone marrow cells from untreated and vehicle (PEG-400)-treated mice progressed into the G1 and S/G2/M phases. Moreover, the absolute number of marrow total LSK+, long-term LSK+, and short-term LSK+ increased 2.8, 12.1, and 4.2-fold, respectively, at 7 days post-irradiation in genistein-treated vs. untreated irradiated mice. Lin− cells from genistein-treated mice expressed fewer DNA damage responsive and cell cycle checkpoint genes than LSK+ from untreated or vehicle-treated mice. Conclusion: Pretreatment with genistein provides in vivo protection from acute myelotoxicity through extended quiescence followed by reduced senescence of marrow repopulating LSK+.


Journal of Cell Science | 2010

Angiotensin-II-induced apoptosis requires regulation of nucleolin and Bcl-xL by SHP-2 in primary lung endothelial cells

Young H. Lee; Ognoon Mungunsukh; Rebecca L. Tutino; Ana P. Marquez; Regina M. Day

Angiotensin II (Ang II) is a key proapoptotic factor in fibrotic tissue diseases. However, the mechanism of Ang-II-induced cell death in endothelial cells has not been previously elucidated. Using the neutral comet assay and specific receptor antagonists and agonists, we found that Ang-II-mediated apoptosis in primary pulmonary endothelial cells required the AT2 receptor. Ang II caused cytochrome c release from the mitochondria concurrent with caspase-3 activation and DNA fragmentation, and apoptosis was suppressed by an inhibitor of Bax-protein channel formation, implicating mitochondrial-mediated apoptosis. There was no evidence that the extrinsic apoptotic pathway was involved, because caspase-9, but not caspase-8, was activated by Ang-II treatment. Apoptosis required phosphoprotein phosphatase activation, and inhibition of the SHP-2 phosphatase (encoded by Ptpn11) blocked cell death. Reduced levels of anti-apoptotic Bcl-2-family members can initiate intrinsic apoptosis, and we found that Ang-II treatment lowered cytosolic Bcl-xL protein levels. Because the protein nucleolin has been demonstrated to bind Bcl-xL mRNA and prevent its degradation, we investigated the role of nucleolin in Ang-II-induced loss of Bcl-xL. RNA-immunoprecipitation experiments revealed that Ang II reduced the binding of nucleolin to Bcl-xL mRNA in an AU-rich region implicated in instability of Bcl-xL mRNA. Inhibition of SHP-2 prevented Ang-II-induced degradation of Bcl-xL mRNA. Taken together, our findings suggest that nucleolin is a primary target of Ang-II signaling, and that Ang-II-activated SHP-2 inhibits nucleolin binding to Bcl-xL mRNA, thus affecting the equilibrium between pro- and anti-apoptotic members of the Bcl-2 family.


International Journal of Radiation Biology | 2013

X-irradiation induces ER stress, apoptosis, and senescence in pulmonary artery endothelial cells

Ronald Allan M. Panganiban; Ognoon Mungunsukh; Regina M. Day

Abstract Purpose: The use of clinical radiation for cancer treatment is limited by damage to underlying normal tissue including to the vascular endothelium. We investigated the mechanisms of X-ray-induced cell damage to endothelial cells. Methods: We evaluated necrosis, apoptosis, cellular senescence, and the contribution of endoplasmic reticulum (ER) stress in pulmonary artery endothelial cells (PAEC) irradiated with X-rays (2–50 Gray [Gy]). Results: Clonogenic assays showed that 10 Gy induced ∼99.9% loss of cell viability. No necrosis was detected using lactate dehydrogenase assays, but a low population underwent extrinsic and intrinsic apoptosis, as indicated by the activation of caspases 3, 8, and 9 as well as by neutral comet assay. A majority of PAEC underwent accelerated senescence, as indicated by morphological changes, increased 21 kD cyclin-dependent kinase inhibitor (p21/waf1), decreased sirtuin 1 (SIRT1), and elevated senescence-associated β-galactosidase (SA-β-gal). ER stress was detected by assays for glucose-regulated protein 78 (GRP78), CCAAT/enhancer-binding protein homologous protein (CHOP), and growth arrest and DNA damage-inducible protein 34 (GADD34) mRNA, and transient phosphorylation of eukaryotic translation initiation factor 2 alpha (eIF2α). The ER stress inhibitor salubrinal blocked ∼50% of apoptosis with no effect on senescence. Conclusions: X-rays primarily induced cellular senescence with limited levels of apoptosis in endothelial cells. ER stress contributed to apoptosis but not to senescence.


American Journal of Physiology-lung Cellular and Molecular Physiology | 2010

Bleomycin induces the extrinsic apoptotic pathway in pulmonary endothelial cells

Ognoon Mungunsukh; Autumn J. Griffin; Young H. Lee; Regina M. Day

Bleomycin, a chemotherapeutic agent, can cause pulmonary fibrosis in humans and is commonly used to induce experimental pulmonary fibrosis in rodents. In cell culture, bleomycin causes single- and double-stranded DNA breaks and produces reactive oxidative species, both of which require iron (Fe(2+)) and O(2). The mechanism of bleomycin-induced apoptosis is controversial due to its complexity. We investigated bleomycin apoptotic signaling events in primary pulmonary endothelial cells. Time course experiments revealed that bleomycin induced apoptosis within 4 h. Caspase-8, the initiator caspase for the extrinsic pathway, was activated within 2 h and preceded activation of the effector caspases-3 and -6 (4 h). Caspase-9, the initiator of the intrinsic pathway and release of cytochrome c from the mitochondria were not detected at these time points. Bleomycin induced the expression of Bcl-2 and Bcl-x(L), Bcl-2 family member proteins that protect cells from the mitochondria-dependent intrinsic apoptosis. Real-time quantitative RT-PCR results demonstrated that, at 4-8 h, bleomycin induced expression of TNF and TNF receptor family genes known to induce the extrinsic apoptotic pathway. Silencing of the death receptor adaptor protein Fas-associated death domain by short interfering RNA significantly reduced bleomycin-induced apoptosis. Apoptosis was also abrogated by caspase-8 inhibition, but only slightly reduced by caspase-3 inhibition. Together, these data suggest that bleomycin initiates apoptosis via the extrinsic pathway.


Molecular Biology of the Cell | 2013

Transforming growth factor-β1 selectively inhibits hepatocyte growth factor expression via a micro-RNA-199–dependent posttranscriptional mechanism

Ognoon Mungunsukh; Regina M. Day

Hepatocyte growth factor (HGF) is a multipotent endogenous repair factor. The profibrotic cytokine transforming growth factor (TGF)-β1 inhibits HGF expression by a micro-RNA-199 (miR-199)-dependent posttranscriptional mechanism. In contrast, NK2, a truncated isoform of HGF that inhibits normal repair, is protected from TGF-β1–induced downregulation by miR-199.


Experimental Hematology | 2011

Captopril modulates hypoxia-inducible factors and erythropoietin responses in a murine model of total body irradiation.

Michal Barshishat-Kupper; Ognoon Mungunsukh; Ashlee J. Tipton; Elizabeth A. McCart; Ronald A.M. Panganiban; Thomas A. Davis; Michael R. Landauer; Regina M. Day

OBJECTIVE Our laboratory reported that the angiotensin converting enzyme inhibitor captopril improves erythroid recovery from total body irradiation (TBI) in mice when administered after irradiation. However, captopril administered before TBI attenuates erythroid recovery. Here we investigate captopril and radiation regulation of erythropoietin (EPO) and thrombopoietin (TPO), key effectors of erythroid progenitor proliferation and differentiation. MATERIALS AND METHODS C57BL/6 mice, nonirradiated or exposed to 7.5 Gy TBI ((60)Co, 0.6 Gy/min) were untreated or administered captopril. Plasma EPO and TPO levels were measured by enzyme-linked immunosorbent assay. Gene expression of EPO was determined by quantitative reverse transcription polymerase chain reaction. The hypoxia-inducible factors (HIF)-1α and -2α were measured by immunoblotting. RESULTS In nonirradiated mice, continuous captopril administration in the water transiently reduced reticulocytes and red blood cells after 7 and 10 days, respectively. EPO plasma levels and gene expression were reduced below detectable limits after 2 days of captopril treatment, but recovered within 7 days. HIF-1α and HIF-2α were activated preceding reticulocyte and red blood cell recovery. TBI, which ablates early and late-stage erythroid progenitors, activated both HIFs and increased EPO and TPO. Captopril treatment postirradiation suppressed radiation-induced HIF activation and EPO expression. In contrast, captopril administration for 7 days before TBI resulted in earlier EPO induction and activation. Captopril treatment lowered TPO levels in nonirradiated mice, but had minimal effects on radiation-induced TPO. CONCLUSIONS In nonirradiated mice, captopril biphasically regulates EPO via HIF activation. TBI ablates erythroid progenitors, resulting in hypoxia, HIF activation, and increased EPO expression that are modulated by captopril treatment. These data suggest that short-term suppression of radiation-induced EPO immediately after TBI is favorable for erythroid recovery.


Molecular Biology of the Cell | 2010

Hepatocyte Growth Factor Inhibits Apoptosis by the Profibrotic Factor Angiotensin II via Extracellular Signal-regulated Kinase 1/2 in Endothelial Cells and Tissue Explants

Young H. Lee; Ana P. Marquez; Ognoon Mungunsukh; Regina M. Day

Hepatocyte growth factor (HGF) is a potent repair factor, promoting normal repair and preventing aberrant repair processes. Angiotensin (Ang) II is up-regulated in abnormal repair, such as fibrotic organ diseases. HGF also blocked Ang II-induced caspase apoptosis in primary endothelial cells and tissue explants in an extracellular signal-regulated kinase-dependent manner.


Molecular Pharmacology | 2014

Mechanism of Erythropoietin Regulation by Angiotensin II

Yong-Chul Kim; Ognoon Mungunsukh; Elizabeth A. McCart; Peter J. Roehrich; Daniel K. Yee; Regina M. Day

Erythropoietin (EPO) is the primary regulator of red blood cell development. Although hypoxic regulation of EPO has been extensively studied, the mechanism(s) for basal regulation of EPO are not well understood. In vivo studies in healthy human volunteers and animal models indicated that angiotensin II (Ang II) and angiotensin converting enzyme inhibitors regulated blood EPO levels. In the current study, we found that Ang II induced EPO expression in situ in murine kidney slices and in 786-O kidney cells in culture as determined by reverse transcription polymerase chain reaction. We further investigated the signaling mechanism of Ang II regulation of EPO in 786-O cells. Pharmacological inhibitors of Ang II type 1 receptor (AT1R) and extracellular signal-regulated kinase 1/2 (ERK1/2) suppressed Ang II transcriptional activation of EPO. Inhibitors of AT2R or Src homology 2 domain–containing tyrosine phosphatase had no effect. Coimmunoprecipiation experiments demonstrated that p21Ras was constitutively bound to the AT1R; this association was increased by Ang II but was reduced by the AT1R inhibitor telmisartan. Transmembrane domain (TM) 2 of AT1R is important for G protein–dependent ERK1/2 activation, and mutant D74E in TM2 blocked Ang II activation of ERK1/2. Ang II signaling induced the nuclear translocation of the Egr-1 transcription factor, and overexpression of dominant-negative Egr-1 blocked EPO promoter activation by Ang II. These data identify a novel pathway for basal regulation of EPO via AT1R-mediated Egr-1 activation by p21Ras-mitogen-activated protein kinase/ERK kinase-ERK1/2. Our current data suggest that Ang II, in addition to regulating blood volume and pressure, may be a master regulator of erythropoiesis.


American Journal of Physiology-lung Cellular and Molecular Physiology | 2010

A tandem repeat of a fragment of Listeria monocytogenes internalin B protein induces cell survival and proliferation

Ognoon Mungunsukh; Young H. Lee; Ana P. Marquez; Fabiola Cecchi; Donald P. Bottaro; Regina M. Day

Hepatocyte growth factor (HGF) is critical for tissue homeostasis and repair in many organs including the lung, heart, kidney, liver, nervous system, and skin. HGF is a heterodimeric protein containing 20 disulfide bonds distributed among an amino-terminal hairpin, four kringle domains, and a serine protease-like domain. Due to its complex structure, recombinant production of HGF in prokaryotes requires denaturation and refolding, processes that are impractical for large-scale manufacture. Thus, pharmaceutical quantities of HGF are not available despite its potential applications. A fragment of the Listeria monocytogenes internalin B protein from amino acids 36-321 (InlB₃₆₋₃₂₁) was demonstrated to bind to and partially activate the HGF receptor Met. InlB₃₆₋₃₂₁ has a stable β-sheet structure and is easily produced in its native conformation by Escherichia coli. We cloned InlB₃₆₋₃₂₁ (1×InlB₃₆₋₃₂₁) and engineered a head-to-tail repeat of InlB₃₆₋₃₂₁ with a linker peptide (2×InlB₃₆₋₃₂₁); 1×InlB₃₆₋₃₂₁ and 2×InlB₃₆₋₃₂₁ were purified from E. coli. Both 1× and 2×InlB₃₆₋₃₂₁ activated the Met tyrosine kinase. We subsequently compared signal transduction of the two proteins in primary lung endothelial cells. 2×InlB₃₆₋₃₂₁ activated ERK1/2, STAT3, and phosphatidylinositol 3-kinase/Akt pathways, whereas 1×InlB₃₆₋₃₂₁ activated only STAT3 and ERK1/2. The 2×InlB₃₆₋₃₂₁ promoted improved motility compared with 1×InlB₃₆₋₃₂₁ and additionally stimulated proliferation equivalent to full-length HGF. Both the 1× and 2×InlB₃₆₋₃₂₁ prevented apoptosis by the profibrotic peptide angiotensin II in cell culture and ex vivo lung slice cultures. The ease of large-scale production and capacity of 2×InlB₃₆₋₃₂₁ to mimic HGF make it a potential candidate as a pharmaceutical agent for tissue repair.


Cellular Signalling | 2016

The hepatocyte growth factor isoform NK2 activates motogenesis and survival but not proliferation due to lack of Akt activation

Ognoon Mungunsukh; Young H. Lee; Donald P. Bottaro; Regina M. Day

Hepatocyte growth factor (HGF) is a pleiotrophic factor involved in cellular proliferation, migration and morphogenesis. HGF is required for normal tissue and organ development during embryogenesis, but in the adult HGF has been demonstrated to drive normal tissue repair and inhibit fibrotic remodeling. HGF has two naturally occurring human isoforms as a result of alternative splicing, NK1 and NK2. While NK1 has been defined as an agonist for HGF receptor, Met, NK2 is defined as a partial Met antagonist. Furthermore, under conditions of fibrotic remodeling, NK2 is still expressed while full length HGF is suppressed. Furthermore, the mechanism by which NK2 partially signals through Met is not completely understood. Here, we investigated the mitogenic, motogenic, and anti-apoptotic activities of NK2 compared with full length HGF in primary human bronchial epithelial cells (BEpC) and bovine pulmonary artery endothelial cells (PAEC). In human BEpC, NK2 partial activated Met, inducing Met phosphorylation at Y1234/1235 in the tyrosine-kinase domain but not at Y1349 site in the multifunctional docking domain. Partial phosphorylation of Met by NK2 resulted in activation of MAPK and STAT3, but not AKT. This correlated with motogenesis and survival in a MAPK-dependent manner, but not cell proliferation. Overexpression of a constitutively active AKT complemented NK2 signaling, allowing NK2 to induce cell proliferation. These data indicate that NK2 and HGF drive motogenic and anti-apoptotic signaling but only HGF drives cell proliferation by activating AKT-pathway signaling. These results have implications for the biological consequences of differential regulation of the two isoforms under pro-fibrotic conditions.

Collaboration


Dive into the Ognoon Mungunsukh's collaboration.

Top Co-Authors

Avatar

Regina M. Day

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Young H. Lee

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Ana P. Marquez

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Elizabeth A. McCart

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Donald P. Bottaro

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Michael R. Landauer

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Thomas A. Davis

Naval Medical Research Center

View shared research outputs
Top Co-Authors

Avatar

Yong-Chul Kim

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Ashlee J. Tipton

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Aviva J. Symes

Uniformed Services University of the Health Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge