Network


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

Hotspot


Dive into the research topics where Cam T. Ha is active.

Publication


Featured researches published by Cam T. Ha.


Journal of Experimental Medicine | 2002

Murine CD9 Is the Receptor for Pregnancy-specific Glycoprotein 17

Roseann Waterhouse; Cam T. Ha; Gabriela S. Dveksler

Pregnancy-specific glycoproteins (PSGs) are a family of highly similar secreted proteins produced by the placenta. PSG homologs have been identified in primates and rodents. Members of the human and murine PSG family induce secretion of antiinflammatory cytokines in mononuclear phagocytes. For the purpose of cloning the receptor, we screened a RAW 264.7 cell cDNA expression library. The PSG17 receptor was identified as the tetraspanin, CD9. We confirmed binding of PSG17 to CD9 by ELISA, flow cytometry, alkaline phosphatase binding assays, and in situ rosetting. Anti-CD9 monoclonal antibody inhibited binding of PSG17 to CD9-transfected cells and RAW 264.7 cells. Moreover, PSG17 binding to macrophages from CD9-deficient mice was significantly reduced. We then tested whether PSG17 binds to other members of the murine tetraspanin family. PSG17 did not bind to cells transfected with CD53, CD63, CD81, CD82, or CD151, suggesting that PSG17–CD9 binding is a specific interaction. We have identified the first receptor for a murine PSG as well as the first natural ligand for a member of the tetraspanin superfamily.


Journal of Leukocyte Biology | 2005

Binding of pregnancy-specific glycoprotein 17 to CD9 on macrophages induces secretion of IL-10, IL-6, PGE2, and TGF-β1

Cam T. Ha; Roseann Waterhouse; Jennifer Wessells; Julie A. Wu; Gabriela S. Dveksler

Pregnancy‐specific glycoproteins (PSGs) are a family of secreted proteins produced by the placenta, which are believed to have a critical role in pregnancy success. Treatment of monocytes with three members of the human PSGs induces interleukin (IL)‐10, IL‐6, and transforming growth factor‐β1 (TGF‐β1) secretion. To determine whether human and murine PSGs have similar functions and use the same receptor, we treated wild‐type and CD9‐deficient macrophages with murine PSG17N and human PSG1 and ‐11. Our data show that murine PSG17N induced secretion of IL‐10, IL‐6, prostaglandin E2, and TGF‐β1 and that CD9 expression is required for the observed induction of cytokines. Therefore, the ability of PSG17 to induce anti‐inflammatory cytokines parallels that of members of the human PSG family, albeit human and murine PSGs use different receptors, as CD9‐deficient and wild‐type macrophages responded equally to human PSGs. We then proceeded to examine the signaling mechanisms responsible for the CD9‐mediated response to PSG17. Inhibition of cyclooxygenase 2 significantly reduced the PSG17N‐mediated increase in IL‐10 and IL‐6. Further characterization of the response to PSG17 indicated that cyclic adenosine monophosphate‐dependent protein kinase A (PKA) is involved in the up‐regulation of IL‐10 and IL‐6, and it is not required for the induction of TGF‐β1. Conversely, treatment of macrophages with a PKC inhibitor reduced the PSG17‐mediated induction of TGF‐β1, IL‐6, and IL‐10 significantly. The induction of anti‐inflammatory cytokines by various PSGs supports the hypothesis that these glycoproteins have an essential role in the regulation of the maternal immune response in species with hemochorial placentation.


Biology of Reproduction | 2008

Murine Pregnancy-Specific Glycoprotein 23 Induces the Proangiogenic Factors Transforming-Growth Factor Beta 1 and Vascular Endothelial Growth Factor A in Cell Types Involved in Vascular Remodeling in Pregnancy

Julie A. Wu; Briana Johnson; Yongqing Chen; Cam T. Ha; Gabriela S. Dveksler

Abstract Haemochorial placentation is a unique physiological process in which the fetal trophoblast cells remodel the maternal decidual spiral arteries to establish the fetoplacental blood supply. Pregnancy-specific glycoproteins (PSGs) are members of the carcinoembryonic antigen family. PSGs are produced by the placenta of rodents and primates and are secreted into the bloodstream. PSG23 is one of 17 members of the murine PSG family (designated PSG16 to PSG32). Previous studies determined that PSGs have immunoregulatory functions due to their ability to modulate macrophage cytokine secretion. Here we show that recombinant PSG23 induces transforming growth factor (TGF) beta1, TGFB1, and vascular endothelial growth factor A (VEGFA) in primary murine macrophages and the macrophage cell line RAW 264.7 cells. In addition, we identified new cell types that responded to PSG23 treatment. Dendritic cells, endothelial cells, and trophoblasts, which are involved in maternal vasculature remodeling during pregnancy, secreted TGFB1 and VEGFA in response to PSG23. PSG23 showed cross-reactivity with human cells, including human monocytes and the trophoblast cell line, HTR-8/SVneo cells. We analyzed the binding of PSG23 to the tetraspanin CD9, the receptor for PSG17, and found that CD9 is not essential for PSG23 binding and activity in macrophages. Overall these studies show that PSGs can modulate the secretion of important proangiogenic factors, TGFB1 and VEGFA, by different cell types involved in the development of the placenta.


PLOS ONE | 2012

REDD1 Protects Osteoblast Cells from Gamma Radiation-Induced Premature Senescence

Xiang Hong Li; Cam T. Ha; Dadin Fu; Mang Xiao

Radiotherapy is commonly used for cancer treatment. However, it often results in side effects due to radiation damage in normal tissue, such as bone marrow (BM) failure. Adult hematopoietic stem and progenitor cells (HSPC) reside in BM next to the endosteal bone surface, which is lined primarily by hematopoietic niche osteoblastic cells. Osteoblasts are relatively more radiation-resistant than HSPCs, but the mechanisms are not well understood. In the present study, we demonstrated that the stress response gene REDD1 (regulated in development and DNA damage responses 1) was highly expressed in human osteoblast cell line (hFOB) cells after γ irradiation. Knockdown of REDD1 with siRNA resulted in a decrease in hFOB cell numbers, whereas transfection of PCMV6-AC-GFP-REDD1 plasmid DNA into hFOB cells inhibited mammalian target of rapamycin (mTOR) and p21 expression and protected these cells from radiation-induced premature senescence (PS). The PS in irradiated hFOB cells were characterized by significant inhibition of clonogenicity, activation of senescence biomarker SA-β-gal, and the senescence-associated cytokine secretory phenotype (SASP) after 4 or 8 Gy irradiation. Immunoprecipitation assays demonstrated that the stress response proteins p53 and nuclear factor κ B (NFkB) interacted with REDD1 in hFOB cells. Knockdown of NFkB or p53 gene dramatically suppressed REDD1 protein expression in these cells, indicating that REDD1 was regulated by both factors. Our data demonstrated that REDD1 is a protective factor in radiation-induced osteoblast cell premature senescence.


Radiation Research | 2013

Genistein Nanoparticles Protect Mouse Hematopoietic System and Prevent Proinflammatory Factors after Gamma Irradiation

Cam T. Ha; Xiang-Hong Li; Dadin Fu; Mang Xiao; Michael R. Landauer

Previous studies demonstrated that genistein protects mice from radiation-induced bone marrow failure. To overcome genisteins extremely low water solubility, a nanoparticle suspension of genistein has been formulated for more rapid dissolution. In the current study, we evaluated the radioprotective effects of a nanoparticle formulation of genistein on survival and hematopoietic recovery in mice exposed to total-body gamma irradiation. A single intramuscular injection of a saline-based genistein nanosuspension (150 mg/kg) administered to CD2F1 mice 24 h before 9.25 Gy 60Co radiation exposure resulted in a 30-day survival rate of 95% compared to 25% in vehicle-treated animals. In mice irradiated at 7 Gy, the genistein nanosuspension increased mouse bone marrow cellularity from approximately 2.9% (vehicle treated) to 28.3% on day 7 postirradiation. Flow cytometry analysis demonstrated decreased radiation-induced hematopoietic stem and progenitor cell (HSPC, Lineage–/cKit+) death from 77.0% (vehicle) to 43.9% (genistein nanosuspension) with a significant recovery of clonogenicity 7 days after irradiation. The genistein nanosuspension also attenuated the radiation-induced elevation of proinflammatory factors interleukin 1 beta (IL-1β), IL-6 and cyclooxygenase-2 (COX-2) in mouse bone marrow and spleen, which may contribute to protecting HSPCs.


PLOS ONE | 2014

Circulating Interleukin-18 as a Biomarker of Total-Body Radiation Exposure in Mice, Minipigs, and Nonhuman Primates (NHP)

Cam T. Ha; Xiang-Hong Li; Dadin Fu; Maria Moroni; Carolyn U. Fisher; Robert Arnott; Venkataraman Srinivasan; Mang Xiao

We aim to develop a rapid, easy-to-use, inexpensive and accurate radiation dose-assessment assay that tests easily obtained samples (e.g., blood) to triage and track radiological casualties, and to evaluate the radioprotective and therapeutic effects of radiation countermeasures. In the present study, we evaluated the interleukin (IL)-1 family of cytokines, IL-1β, IL-18 and IL-33, as well as their secondary cytokines’ expression and secretion in CD2F1 mouse bone marrow (BM), spleen, thymus and serum in response to γ-radiation from sublethal to lethal doses (5, 7, 8, 9, 10, or 12 Gy) at different time points using the enzyme-linked immune sorbent assay (ELISA), immunoblotting, and cytokine antibody array. Our data identified increases of IL-1β, IL-18, and/or IL-33 in mouse thymus, spleen and BM cells after total-body irradiation (TBI). However, levels of these cytokines varied in different tissues. Interestingly, IL-18 but not IL-1β or IL-33 increased significantly (2.5–24 fold) and stably in mouse serum from day 1 after TBI up to 13 days in a radiation dose-dependent manner. We further confirmed our finding in total-body γ-irradiated nonhuman primates (NHPs) and minipigs, and demonstrated that radiation significantly enhanced IL-18 in serum from NHPs 2–4 days post-irradiation and in minipig plasma 1–3 days post-irradiation. Finally, we compared circulating IL-18 with the well known hematological radiation biomarkers lymphocyte and neutrophil counts in blood of mouse, minipigs and NHPs and demonstrated close correlations between these biomarkers in response to radiation. Our results suggest that the elevated levels of circulating IL-18 after radiation proportionally reflect radiation dose and severity of radiation injury and may be used both as a potential biomarker for triage and also to track casualties after radiological accidents as well as for therapeutic radiation exposure.


PLOS ONE | 2012

Micro-RNA30c Negatively Regulates REDD1 Expression in Human Hematopoietic and Osteoblast Cells after Gamma-Irradiation

Xiang Hong Li; Cam T. Ha; Dadin Fu; Mang Xiao

We recently demonstrated that a novel cell stress response gene REDD1 protects human fetal osteoblast cell line (hFOB) cells from γ-radiation-induced premature senescence. Here we show that levels of endogenous REDD1 are very low in human hematopoietic progenitor CD34+ cells regardless of radiation, but highly expressed in differentiated hematopoietic cells (14 day cultured CD34+ cells) in response to radiation, which might be associated with radiation tolerance of the latter cells. To further understand the mechanisms of radiation-induced damage in different cells, microRNA (miRNA)-arrays were performed using purified miRNAs from CD34+ and hFOB cells before and post-irradiation and real-time reverse transcription (RT)-PCR was used to validate the expression profiles of miRNAs in the radiation-damaged cells. The results indicate that γ-radiation downregulated 16 miRNAs in CD34+ cells and 14 in hFOB cells. Radiation-induced upregulation was observed for 15 miRNAs in CD34+ cells and 18 miRNAs in hFOB cells. The profiles of radiation-induced miRNA expression were completely different in CD34+ vs. hFOB cells. Radiation up-regulated miRNA (miR)-30b, miR-30c and miR-30d in CD34+ cells, whereas it inhibited miR-30c expression in hFOB cells. Since miR-30 has potential target sites located in the 3′untranslated region (UTR) of the REDD1 gene and radiation regulated miR-30c expression in both CD34+ and hFOB cells, we further explored the effects of miR-30c on REDD1 expression using miR-30c inhibitor and precursor (pre-miR-30c). The results show that pre-miR-30c transfection suppressed REDD1 expression in 14 day cultured CD34+ cells and hFOB cells and resulted in hFOB cell death. In contrast, inhibition of miR-30c expression significantly enhanced clonogenicity in CD34+ cells. Our data suggest that CD34+ and hFOB cells have different miRNA expression patterns after irradiation and miR-30c plays a key role in radiation-induced cell damage which might be through regulation of REDD1 expression.


Radiation Research | 2013

Delta-Tocotrienol Protects Mice from Radiation-Induced Gastrointestinal Injury

Xiang Hong Li; Sanchita P. Ghosh; Cam T. Ha; Dadin Fu; Thomas B. Elliott; David L. Bolduc; Vilmar Villa; Mark H. Whitnall; Michael R. Landauer; Mang Xiao

We recently demonstrated that natural delta-tocotrienol (DT3) significantly enhanced survival in total-body irradiated (TBI) mice, and protected mouse bone marrow cells from radiation-induced damage through Erk activation-associated mTOR survival pathways. Here, we further evaluated the effects and mechanisms of DT3 on survival of radiation-induced mouse acute gastrointestinal syndrome. DT3 (75–100 mg/kg) or vehicle was administered as a single subcutaneous injection to CD2F1 mice 24 h before 10–12 Gy 60Co total-body irradiation at a dose rate of 0.6 Gy/min and survival was monitored. In a separate group of mice, jejunum sections were stained with hematoxylin and eosin and the surviving crypts in irradiated mice were counted. Apoptosis in intestinal epithelial cells was measured by terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling (TUNEL) staining and bacterial translocation from gut to heart, spleen and liver in irradiated mice were evaluated. DT3 (75 mg/kg) significantly enhanced survival in mice that received 10, 10.5, 11 or 12 Gy TBI. Administration of DT3 protected intestinal tissue, decreased apoptotic cells in jejunum and inhibited gut bacterial translocation in irradiated mice. Furthermore, DT3 significantly inhibited radiation-induced production of pro-inflammatory factors interleukin-1β and −6 and suppressed expression of protein tyrosine kinase 6 (PTK6), a stress-induced kinase that promotes apoptosis in mouse intestinal cells. Our data demonstrate that administration of DT3 protected mice from radiation-induced gastrointestinal system damage.


PLOS ONE | 2015

Delta-Tocotrienol Suppresses Radiation-Induced MicroRNA-30 and Protects Mice and Human CD34+ Cells from Radiation Injury

Xiang Hong Li; Cam T. Ha; Dadin Fu; Michael R. Landauer; Sanchita P. Ghosh; Mang Xiao

We reported that microRNA-30c (miR-30c) plays a key role in radiation-induced human cell damage through an apoptotic pathway. Herein we further evaluated radiation-induced miR-30 expression and mechanisms of delta-tocotrienol (DT3), a radiation countermeasure candidate, for regulating miR-30 in a mouse model and human hematopoietic CD34+ cells. CD2F1 mice were exposed to 0 (control) or 7–12.5 Gy total-body gamma-radiation, and CD34+ cells were irradiated with 0, 2 or 4 Gy of radiation. Single doses of DT3 (75 mg/kg, subcutaneous injection for mice or 2 μM for CD34+ cell culture) were administrated 24 h before irradiation and animal survival was monitored for 30 days. Mouse bone marrow (BM), jejunum, kidney, liver and serum as well as CD34+ cells were collected at 1, 4, 8, 24, 48 or 72 h after irradiation to determine apoptotic markers, pro-inflammatory cytokines interleukin (IL)-1β and IL-6, miR-30, and stress response protein expression. Our results showed that radiation-induced IL-1β release and cell damage are pathological states that lead to an early expression and secretion of miR-30b and miR-30c in mouse tissues and serum and in human CD34+ cells. DT3 suppressed IL-1β and miR-30 expression, protected against radiation-induced apoptosis in mouse and human cells, and increased survival of irradiated mice. Furthermore, an anti-IL-1β antibody downregulated radiation-induced NFκBp65 phosphorylation, inhibited miR-30 expression and protected CD34+ cells from radiation exposure. Knockdown of NFκBp65 by small interfering RNA (siRNA) significantly suppressed radiation-induced miR-30 expression in CD34+ cells. Our data suggest that DT3 protects human and mouse cells from radiation damage may through suppression of IL-1β-induced NFκB/miR-30 signaling.


American Journal of Reproductive Immunology | 2008

N-glycosylation is required for binding of murine pregnancy-specific glycoproteins 17 and 19 to the receptor CD9.

Cam T. Ha; Roseann Waterhouse; James Warren; Wolfgang Zimmermann; Gabriela S. Dveksler

Murine pregnancy‐specific glycoproteins (PSGs) are encoded by 17 different genes. Different family members have different expression levels at different stages of embryonic development. It is currently unknown whether all members of this family of placentally secreted proteins have the same function and bind to the same receptor. Furthermore, the requirement of post‐translational modifications for the activity of these highly glycosylated proteins remains undetermined.

Collaboration


Dive into the Cam T. Ha's collaboration.

Top Co-Authors

Avatar

Gabriela S. Dveksler

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Mang Xiao

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Dadin Fu

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

James Warren

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Roseann Waterhouse

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Xiang Hong Li

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Julie A. Wu

Uniformed Services University of the Health Sciences

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

Xiang-Hong Li

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Sanchita P. Ghosh

Uniformed Services University of the Health Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge