Network


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

Hotspot


Dive into the research topics where Bolormaa Baljinnyam is active.

Publication


Featured researches published by Bolormaa Baljinnyam.


Cellular Signalling | 2014

Secreted Frizzled-related protein potentiation versus inhibition of Wnt3a/β-catenin signaling.

Charles P. Xavier; Maria Melikova; Yoshiro Chuman; Aykut Üren; Bolormaa Baljinnyam; Jeffrey S. Rubin

Wnt signaling regulates a variety of cellular processes during embryonic development and in the adult. Many of these activities are mediated by the Frizzled family of seven-pass transmembrane receptors, which bind Wnts via a conserved cysteine-rich domain (CRD). Secreted Frizzled-related proteins (sFRPs) contain an amino-terminal, Frizzled-like CRD and a carboxyl-terminal, heparin-binding netrin-like domain. Previous studies identified sFRPs as soluble Wnt antagonists that bind directly to Wnts and prevent their interaction with Frizzleds. However, subsequent observations suggested that sFRPs and Frizzleds form homodimers and heterodimers via their respective CRDs, and that sFRPs can stimulate signal transduction. Here, we present evidence that sFRP1 either inhibits or enhances signaling in the Wnt3a/β-catenin pathway, depending on its concentration and the cellular context. Nanomolar concentrations of sFRP1 increased Wnt3a signaling, while higher concentrations blocked it in HEK293 cells expressing a SuperTopFlash reporter. sFRP1 primarily augmented Wnt3a/β-catenin signaling in C57MG cells, but it behaved as an antagonist in L929 fibroblasts. sFRP1 enhanced reporter activity in L cells that were engineered to stably express Frizzled 5, though not Frizzled 2. This implied that the Frizzled expression pattern could determine the response to sFRP1. Similar results were obtained with sFRP2 in HEK293, C57MG and L cell reporter assays. CRDsFRP1 mimicked the potentiating effect of sFRP1 in multiple settings, contradicting initial expectations that this domain would inhibit Wnt signaling. Moreover, CRDsFRP1 showed little avidity for Wnt3a compared to sFRP1, implying that the mechanism for potentiation by CRDsFRP1 probably does not require an interaction with Wnt protein. Together, these findings demonstrate that sFRPs can either promote or suppress Wnt/β-catenin signaling, depending on cellular context, concentration and most likely the expression pattern of Fzd receptors.


Journal of Biological Chemistry | 2015

Systematic mapping of WNT-FZD protein interactions reveals functional selectivity by distinct WNT-FZD pairs.

Jacomijn P. Dijksterhuis; Bolormaa Baljinnyam; Karen Stanger; Hakki Ogun Sercan; Yun Ji; Osler Andres; Jeffrey S. Rubin; Rami N. Hannoush; Gunnar Schulte

Background: WNT-FZD specificity and signaling outcome have not been systematically mapped so far. Results: WNTs show selective binding to FZDs, and respective WNT-FZD pairs exert functional selectivity in different downstream signaling pathways. Conclusion: The WNT-FZD signaling system provides ligand-receptor selectivity. Significance: Understanding WNT-FZD selectivity is crucial for development of WNT pathway inhibitors and further understanding of molecular interactions within the WNT receptor signaling complex. The seven-transmembrane-spanning receptors of the FZD1–10 class are bound and activated by the WNT family of lipoglycoproteins, thereby inducing a complex network of signaling pathways. However, the specificity of the interaction between mammalian WNT and FZD proteins and the subsequent signaling cascade downstream of the different WNT-FZD pairs have not been systematically addressed to date. In this study, we determined the binding affinities of various WNTs for different members of the FZD family by using bio-layer interferometry and characterized their functional selectivity in a cell system. Using purified WNTs, we show that different FZD cysteine-rich domains prefer to bind to distinct WNTs with fast on-rates and slow off-rates. In a 32D cell-based system engineered to overexpress FZD2, FZD4, or FZD5, we found that WNT-3A (but not WNT-4, -5A, or -9B) activated the WNT-β-catenin pathway through FZD2/4/5 as measured by phosphorylation of LRP6 and β-catenin stabilization. Surprisingly, different WNT-FZD pairs showed differential effects on phosphorylation of DVL2 and DVL3, revealing a previously unappreciated DVL isoform selectivity by different WNT-FZD pairs in 32D cells. In summary, we present extensive mapping of WNT-FZD cysteine-rich domain interactions complemented by analysis of WNT-FZD pair functionality in a unique cell system expressing individual FZD isoforms. Differential WNT-FZD binding and selective functional readouts suggest that endogenous WNT ligands evolved with an intrinsic natural bias toward different downstream signaling pathways, a phenomenon that could be of great importance in the design of FZD-targeting drugs.


Journal of Cellular Physiology | 2012

Rspo2/Int7 regulates invasiveness and tumorigenic properties of mammary epithelial cells

Malgorzata Klauzinska; Bolormaa Baljinnyam; Ahmed Raafat; Jaime Rodriguez-Canales; Luigi Strizzi; Yoshimi Endo Greer; Jeffrey S. Rubin; Robert Callahan

Rspo2 was identified as a novel common integration site (CIS) for the mouse mammary tumor virus (MMTV) in viral induced mouse mammary tumors. Here we show that Rspo2 modulates Wnt signaling in mouse mammary epithelial cells. Co‐expression of both genes resulted in an intermediate growth phenotype on plastic and had minor effects on the growth‐promoting properties of Wnt1 in soft agar. However, individual Rspo2 and Wnt1 HC11 transfectants as well as the double transfectant were tumorigenic in athymic nude mice, with tumors from each line having distinctive histological characteristics. Rspo2 and Rspo2/Wnt1 tumors contained many spindle cells, consistent with an epithelial–mesenchymal transformation (EMT) phenotype. When Rspo2 and Rspo2/Wnt1 tumor cells were transferred into naïve mice, they exhibited greater metastatic activity than cells derived from Wnt1 tumors. For comparison, C57MG/Wnt1/Rspo2 co‐transfectants exhibited invasive properties in three‐dimensional (3D) Matrigel cultures that were not seen with cells transfected only with Wnt1 or Rspo2. Use of Dickkopf‐1, a specific antagonist of the Wnt/β‐catenin pathway, or short hairpin RNA targeting β‐catenin expression demonstrated that the invasive activity was not mediated by β‐catenin. Our results indicate that Rspo2 and Wnt1 have mutually distinct effects on mammary epithelial cell growth and these effects are context‐dependent. While Rspo2 and Wnt1 act synergistically in the β‐catenin pathway, other mechanisms are responsible for the invasive properties of stable double transfectants observed in 3D Matrigel cultures. J. Cell. Physiol. 227: 1960–1971, 2012.


Stem Cells and Development | 2010

Proliferation and Pluripotency of Human Embryonic Stem Cells Maintained on Type I Collagen

Meredith B. Jones; Chia H. Chu; James Pendleton; Michael J. Betenbaugh; Joseph Shiloach; Bolormaa Baljinnyam; Jeffrey S. Rubin; Michael J. Shamblott

Human embryonic stem cells (hESC) require a balance of growth factors and signaling molecules to proliferate and retain pluripotency. Conditioned medium (CM) from a human embryonic germ-cell-derived cell culture, SDEC, was observed to support the growth of hESC on type I collagen (COL I) and on Matrigel (MAT) biomatricies. After 1 month, the population doubling of hESC grown in SDEC CM on COL I was equivalent to that of hESC grown in mouse embryonic fibroblast (MEF) CM on MAT. hESC grown in SDEC CM on COL I expressed OCT4, NANOG, SSEA-4, alkaline phosphatase (AP), and TRA-1-60; retained a normal karyotype; and were capable of forming teratomas. DNA microarray analysis was used to compare the transcriptional profiles of SDEC and the less supportive WI38 and Detroit 551 human cell lines. The mRNA level of secreted frizzled-related protein (sFRP-1), a known antagonist of the WNT/β-catenin signaling pathway, was significantly reduced in SDEC as compared with the other 2 cell lines, whereas the mRNA levels of prostaglandin-endoperoxide synthase 2 (PTGS2 or COX-2) and prostaglandin I₂ synthase (PGIS), two prostaglandin biosynthesis genes, were significantly increased in SDEC. The level of sFRP-1 protein was significantly reduced, and levels of 2 prostaglandins that are downstream products of PTGS2 and PGIS, prostaglandin E₂ and 6-keto-prostaglandin F(1α), were significantly elevated in SDEC CM compared with WI38, Detroit 551, and MEF CM. Further, addition of purified sFRP-1 to SDEC CM reduced the proliferation of hESC grown on COL I as well as MAT in a dose-dependent manner.


PLOS ONE | 2012

Recombinant R-spondin2 and Wnt3a Up- and Down- Regulate Novel Target Genes in C57MG Mouse Mammary Epithelial Cells

Bolormaa Baljinnyam; Malgorzata Klauzinska; Saad Saffo; Robert Callahan; Jeffrey S. Rubin

R-spondins (Rspos) comprise a family of four secreted proteins that have important roles in cell proliferation, cell fate determination and organogenesis. Rspos typically exert their effects by potentiating the Wnt/β-catenin signaling pathway. To systematically investigate the impact of Rspo/Wnt on gene expression, we performed a microarray analysis using C57MG mouse mammary epithelial cells treated with recombinant Rspo2 and/or Wnt3a. We observed the up- and down-regulation of several previously unidentified target genes, including ones that encode proteins involved in immune responses, effectors of other growth factor signaling pathways and transcription factors. Dozens of these changes were validated by quantitative real time RT-PCR. Time course experiments showed that Rspo2 typically had little or no effect on Wnt-dependent gene expression at 3 or 6 h, but enhanced expression at 24 h, consistent with biochemical data indicating that Rspo2 acts primarily to sustain rather than acutely increase Wnt pathway activation. Up-regulation of gene expression was inhibited by pre-treatment with Dickkopf1, a Wnt/β-catenin pathway antagonist, and by siRNA knockdown of β-catenin expression. While Dickkopf1 blocked Rspo2/Wnt3a-dependent down-regulation, a number of down-regulated genes were not affected by β-catenin knockdown, suggesting that in these instances down-regulation was mediated by a β-catenin-independent mechanism.


Bioorganic & Medicinal Chemistry | 2018

Discovery and optimization of piperazine-1-thiourea-based human phosphoglycerate dehydrogenase inhibitors

Jason M. Rohde; Kyle R. Brimacombe; Li Liu; Michael E. Pacold; Adam Yasgar; Dorian M. Cheff; Tobie D. Lee; Ganesha Rai; Bolormaa Baljinnyam; Zhuyin Li; Anton Simeonov; Matthew D. Hall; Min Shen; David M. Sabatini; Matthew B. Boxer

Proliferating cells, including cancer cells, obtain serine both exogenously and via the metabolism of glucose. By catalyzing the first, rate-limiting step in the synthesis of serine from glucose, phosphoglycerate dehydrogenase (PHGDH) controls flux through the biosynthetic pathway for this important amino acid and represents a putative target in oncology. To discover inhibitors of PHGDH, a coupled biochemical assay was developed and optimized to enable high-throughput screening for inhibitors of human PHGDH. Feedback inhibition was minimized by coupling PHGDH activity to two downstream enzymes (PSAT1 and PSPH), providing a marked improvement in enzymatic turnover. Further coupling of NADH to a diaphorase/resazurin system enabled a red-shifted detection readout, minimizing interference due to compound autofluorescence. With this protocol, over 400,000 small molecules were screened for PHGDH inhibition, and following hit validation and triage work, a piperazine-1-thiourea was identified. Following rounds of medicinal chemistry and SAR exploration, two probes (NCT-502 and NCT-503) were identified. These molecules demonstrated improved target activity and encouraging ADME properties, enabling in vitro assessment of the biological importance of PHGDH, and its role in the fate of serine in PHGDH-dependent cancer cells. This manuscript reports the assay development and medicinal chemistry leading to the development of NCT-502 and -503 reported in Pacold et al. (2016).


Journal of Medicinal Chemistry | 2018

Discovery of Orally Bioavailable, Quinoline-Based Aldehyde Dehydrogenase 1A1 (ALDH1A1) Inhibitors with Potent Cellular Activity

Shyh-Ming Yang; Natalia Martínez; Adam Yasgar; Carina Danchik; C. Johansson; Yuhong Wang; Bolormaa Baljinnyam; Amy Wang; Xin Xu; Pranav Shah; Dorian M. Cheff; Xinran S. Wang; Jacob S. Roth; Madhu Lal-Nag; J E Dunford; U. Oppermann; Vasilis Vasiliou; Anton Simeonov; Ajit Jadhav; David J. Maloney

Aldehyde dehydrogenases (ALDHs) are responsible for the metabolism of aldehydes (exogenous and endogenous) and possess vital physiological and toxicological functions in areas such as CNS, inflammation, metabolic disorders, and cancers. Overexpression of certain ALDHs (e.g., ALDH1A1) is an important biomarker in cancers and cancer stem cells (CSCs) indicating the potential need for the identification and development of small molecule ALDH inhibitors. Herein, a newly designed series of quinoline-based analogs of ALDH1A1 inhibitors is described. Extensive medicinal chemistry optimization and biological characterization led to the identification of analogs with significantly improved enzymatic and cellular ALDH inhibition. Selected analogs, e.g., 86 (NCT-505) and 91 (NCT-506), demonstrated target engagement in a cellular thermal shift assay (CETSA), inhibited the formation of 3D spheroid cultures of OV-90 cancer cells, and potentiated the cytotoxicity of paclitaxel in SKOV-3-TR, a paclitaxel resistant ovarian cancer cell line. Lead compounds also exhibit high specificity over other ALDH isozymes and unrelated dehydrogenases. The in vitro ADME profiles and pharmacokinetic evaluation of selected analogs are also highlighted.


bioRxiv | 2018

Molecular basis for activation of lecithin:cholesterol acyltransferase by a compound that increases HDL cholesterol

Kelly A. Manthei; Shyh-Ming Yang; Bolormaa Baljinnyam; Louise Chang; Alisa Glukhova; Wenmin Yuan; Lita Freeman; David J. Maloney; Anna Schwendeman; Alan T. Remaley; Ajit Jadhav; John J. G. Tesmer

Lecithin:cholesterol acyltransferase (LCAT) and LCAT-activating small molecules are being investigated as treatments for coronary heart disease (CHD) and familial LCAT deficiency (FLD). Herein we report the crystal structure of LCAT bound to a potent activator and an acyl intermediate-like inhibitor, thereby revealing an active conformation of LCAT and that the activator is bound exclusively to its membrane-binding domain (MBD). Functional studies indicate that the compound does not modulate the affinity of LCAT for HDL, but instead stabilizes residues in the MBD and likely facilitates channeling of substrates into the active site. By demonstrating that these activators increase the activity of an FLD variant, we show that compounds targeting the MBD have therapeutic potential. In addition, our data better define the acyl binding site of LCAT and pave the way for rational design of LCAT agonists and improved biotherapeutics for augmenting or restoring reverse cholesterol transport in CHD and FLD patients.


Expert Opinion on Drug Discovery | 2018

Testing for drug-human serum albumin binding using fluorescent probes and other methods

Michael Ronzetti; Bolormaa Baljinnyam; Adam Yasgar; Anton Simeonov

ABSTRACT Introduction: Drug plasma protein binding remains highly relevant to research and drug development, making the assessment and profiling of compound affinity to plasma proteins essential to drug discovery efforts. Although there are a number of fully-characterized methods, they lack the throughput to handle large numbers of compounds. As the evaluation of adsorption, distribution, metabolism, and excretion is addressed earlier in the drug development timeline, the need for higher-throughput methods has grown. Areas Covered: This review will highlight recent developments on methods for profiling drug plasma binding, with an emphasis on fluorescent probes and emerging high-throughput methodologies. Expert Opinion: There have been a number of high-throughput assays developed in recent years to meet the scaled up demands for compound profiling. Ultimately, the selection of assay technology relies on a number of factors, such as capabilities of the laboratory and the breadth and amount of data required. Fluorescent probe displacement assays are highly flexible and amenable to high-throughput screening, easily scaling up to handle large compound libraries. Recent developments in fluorescence technologies, such as homogenous time-resolved fluorescence and probes utilizing the aggregation-induced emission effect, have improved the sensitivity of these assays. Other technologies, such as microscale thermophoresis and quantitative structure-activity relationship modeling, are gaining popularity as alternative techniques for drug plasma protein binding characterization.


ACS Chemical Biology | 2017

Cell Lysate-Based AlphaLISA Deubiquitinase Assay Platform for Identification of Small Molecule Inhibitors

Christine A. Ott; Bolormaa Baljinnyam; Alexey V. Zakharov; Ajit Jadhav; Anton Simeonov; Zhihao Zhuang

The deubiquitinases, or DUBs, are associated with various human diseases, including neurological disorders, cancer, and viral infection, making them excellent candidates for pharmacological intervention. Drug discovery campaigns against DUBs require enzymatic deubiquitination assays amenable for high-throughput screening (HTS). Although several DUB substrates and assays have been developed in recent years, they are largely limited to recombinantly purified DUBs. Many DUBs are large multidomain proteins that are difficult to obtain recombinantly in sufficient quantities for HTS. Therefore, an assay that obviates the need of recombinant protein generation and also recapitulates a physiologically relevant environment is highly desirable. Such an assay will open doors for drug discovery against many therapeutically relevant, but currently inaccessible, DUBs. Here, we report a cell lysate DUB assay based on AlphaLISA technology for high throughput screening. This assay platform uses a biotin-tagged ubiquitin probe and a HA-tagged DUB expressed in human cells. The assay was validated and adapted to a 1536-well format, which enabled a screening against UCHL1 as proof of principle using a library of 15 000 compounds. We expect that the new platform can be readily adapted to other DUBs to allow the identification of more potent and selective small molecule inhibitors and chemical probes.

Collaboration


Dive into the Bolormaa Baljinnyam's collaboration.

Top Co-Authors

Avatar

Anton Simeonov

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Jeffrey S. Rubin

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Adam Yasgar

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Ajit Jadhav

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Malgorzata Klauzinska

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Robert Callahan

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

David J. Maloney

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Dorian M. Cheff

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Shyh-Ming Yang

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Jeffrey S. Rubin

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge