Network


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

Hotspot


Dive into the research topics where Michael A. Sheard is active.

Publication


Featured researches published by Michael A. Sheard.


International Journal of Cancer | 1997

Up-regulation of Fas (CD95) in human p53wild-type cancer cells treated with ionizing radiation.

Michael A. Sheard; Borek Vojtesek; Libuše Janáková; Jan Kovarik; Jan Zaloudik

Fas is a cell‐surface protein which belongs to the tumor‐necrosis‐factor‐receptor family. Signals through Fas are able to induce apoptosis in sensitive cells, and thus modalities for regulating the level of Fas expression on tumor cells are needed. We have studied cellular responses to gamma irradiation. The level of p53 tumor‐suppressor protein was found to be elevated 3 hr after irradiation of p53wild‐type MCF‐7 breast‐carcinoma cells. Interestingly, accumulation of p53 was followed by up‐regulation of surface Fas levels between 4 and 8 hr after irradiation. The level of Fas up‐regulation was dependent on dose and, whereas elevation in the level of p53 was transient, enhancement of Fas expression was stable. Fas up‐regulation occurred coincidentally with induction of G1 cell‐cycle arrest, a post‐irradiation phenomenon known to be dependent on wild‐type‐p53 activity. We studied 9 other tumor lines, 2 with wild‐type p53, 5 with mutant p53, and 2 expressing no p53. All lines expressing wild‐type p53 were found to arrest in G1 and to up‐regulate Fas after irradiation. In contrast, all 7 p53null and p53mutant lines failed not only to arrest their cell cycles in G1 phase, but also to up‐regulate Fas levels in response to treatment. These findings demonstrate a direct correlation between wild‐type‐p53 activity and Fas up‐regulation after treatment with ionizing radiation, strongly suggesting that post‐irradiation Fas up‐regulation is dependent on wild‐type‐p53 activity. Since low doses of radiation were sufficient to modulate Fas expression, up‐regulation of the Fas death receptor may have clinical implications following radiotherapy. Int. J. Cancer 73:757–762, 1997.


Journal of Cellular Biochemistry | 2002

Release of cytokeratin-18 and -19 fragments (TPS and CYFRA 21-1) into the extracellular space during apoptosis.

Michael A. Sheard; Borek Vojtesek; Marta Šimíčková; Dalibor Valík

Serum fragments of cytokeratins‐18 and ‐19 (measured as TPS and CYFRA 21‐1, respectively) have traditionally been considered as markers of tumor proliferation, although the evidence is scarce for a causative relationship between proliferation and levels of TPS and CYFRA 21‐1. We examined whether apoptosis might produce TPS and CYFRA 21‐1 fragments. MCF‐7 breast cancer cells were treated with mitomycin C or agonistic anti‐CD95 antibody, and levels of TPS and CYFRA 21‐1 in tissue culture supernatants were compared with the frequency of cells exhibiting the following markers of cell death: intracellular cytokeratin‐18 cleavage, surface staining with annexin‐V, propidium iodide uptake, DNA fragmentation. Twenty‐four hours after inducing apoptosis, levels of TPS and CYFRA 21‐1 were elevated ≥ 4‐fold in culture supernatants. Elevations in TPS and CYFRA 21‐1 coincided with apoptosis measured by the first three cell death markers but preceded DNA fragmentation. These mitomycin C‐ and CD95‐mediated elevations were completely inhibited by co‐incubation with the caspase inhibitors Z‐VAD.fmk and Z‐IETD.fmk, respectively. We conclude that TPS and CYFRA 21‐1 can be abundantly released into the extracellular space during the intermediate stage of epithelial cell apoptosis. J. Cell. Biochem. 85: 670–677, 2002.


Clinical Cancer Research | 2013

Growth and Activation of Natural Killer Cells Ex Vivo from Children with Neuroblastoma for Adoptive Cell Therapy

Yin Liu; Hong Wei Wu; Michael A. Sheard; Richard Sposto; Srinivas S. Somanchi; Laurence J.N. Cooper; Dean A. Lee; Robert C. Seeger

Purpose: Adoptive transfer of natural killer (NK) cells combined with tumor-specific monoclonal antibodies (mAb) has therapeutic potential for malignancies. We determined if large numbers of activated NK (aNK) cells can be grown ex vivo from peripheral blood mononuclear cells (PBMC) of children with high-risk neuroblastoma using artificial antigen-presenting cells (aAPC). Experimental Design: Irradiated K562-derived Clone 9.mbIL21 aAPC were cocultured with PBMC, and propagated NK cells were characterized with flow cytometry, cytotoxicity assays, Luminex multicytokine assays, and a nonobese diabetic/severe combined immunodeficient (NOD/SCID) mouse model of disseminated neuroblastoma. Results: Coculturing patient PBMC with aAPC for 14 days induced 2,363- ± 443-fold expansion of CD56+CD3−CD14− NK cells with 83% ± 3% purity (n = 10). Results were similar to PBMC from normal donors (n = 5). Expression of DNAM-1, NKG2D, FcγRIII/CD16, and CD56 increased 6- ± 3-, 10- ± 2-, 21- ± 20-, and 18- ± 3-fold, respectively, on day 14 compared with day 0, showing activation of NK cells. In vitro, aNK cells were highly cytotoxic against neuroblastoma cell lines and killing was enhanced with GD2-specific mAb ch14.18. When mediating cytotoxicity with ch14.18, release of TNF-α, granulocyte macrophage colony-stimulating factor, IFN-γ, sCD40L, CCL2/MCP-1, CXCL9/MIG, and CXCL11/I-TAC by aNK cells increased 4-, 5-, 6-, 15-, 265-, 917-, and 363-fold (151–9,121 pg/mL), respectively, compared with aNK cells alone. Survival of NOD/SCID mice bearing disseminated neuroblastoma improved when treated with thawed and immediately intravenously infused cryopreserved aNK cells compared with untreated mice and was further improved when ch14.18 was added. Conclusion: Propagation of large numbers of aNK cells that maintain potent antineuroblastoma activities when cryopreserved supports clinical testing of adoptive cell therapy with ch14.18. Clin Cancer Res; 19(8); 2132–43. ©2013 AACR.


Cancer Research | 2013

Critical Role of STAT3 in IL-6–Mediated Drug Resistance in Human Neuroblastoma

Tasnim Ara; Rie Nakata; Michael A. Sheard; Hiroyuki Shimada; Ralf Buettner; Susan Groshen; Lingyun Ji; Hua Yu; Richard Jove; Robert C. Seeger; Yves A. DeClerck

Drug resistance is a major cause of treatment failure in cancer. Here, we have evaluated the role of STAT3 in environment-mediated drug resistance (EMDR) in human neuroblastoma. We determined that STAT3 was not constitutively active in most neuroblastoma cell lines but was rapidly activated upon treatment with interleukin (IL)-6 alone and in combination with the soluble IL-6 receptor (sIL-6R). Treatment of neuroblastoma cells with IL-6 protected them from drug-induced apoptosis in a STAT3-dependent manner because the protective effect of IL-6 was abrogated in the presence of a STAT3 inhibitor and upon STAT3 knockdown. STAT3 was necessary for the upregulation of several survival factors such as survivin (BIRC5) and Bcl-xL (BCL2L1) when cells were exposed to IL-6. Importantly, IL-6-mediated STAT3 activation was enhanced by sIL-6R produced by human monocytes, pointing to an important function of monocytes in promoting IL-6-mediated EMDR. Our data also point to the presence of reciprocal activation of STAT3 between tumor cells and bone marrow stromal cells including not only monocytes but also regulatory T cells (Treg) and nonmyeloid stromal cells. Thus, the data identify an IL-6/sIL-6R/STAT3 interactive pathway between neuroblastoma cells and their microenvironment that contributes to drug resistance.


International Journal of Cancer | 2001

Ionizing radiation as a response-enhancing agent for CD95-mediated apoptosis

Michael A. Sheard

CD95 (Fas/APO‐1) is a death receptor on the surface of a wide variety of cell types. In most cells examined, ionizing radiation acts as a response‐enhancing agent for CD95‐mediated cell death. Although DNA‐damaging radiation appears to modulate CD95‐mediated signals through multiple mechanisms, the only well‐characterized mechanism is activation of the tumor‐suppressor protein p53, which transcriptionally regulates the expression of CD95 on various cell types. The ligand for CD95 is expressed by activated lymphocytes and natural‐killer cells, which produce factors that sensitize cells resistant to CD95‐mediated cell death. Ligation of CD95 on irradiated tumor cells might be achievable using emerging modalities that reactivate the stalled anti‐tumor immune response.


PLOS ONE | 2011

Meningitic Escherichia coli K1 Penetration and Neutrophil Transmigration Across the Blood–Brain Barrier are Modulated by Alpha7 Nicotinic Receptor

Feng Chi; Lin Wang; Xueye Zheng; Chun-Hua Wu; Ambrose Jong; Michael A. Sheard; Wei Shi; Sheng-He Huang

Alpha7 nicotinic acetylcholine receptor (nAChR), an essential regulator of inflammation, is abundantly expressed in hippocampal neurons, which are vulnerable to bacterial meningitis. However, it is unknown whether α7 nAChR contributes to the regulation of these events. In this report, an aggravating role of α7 nAChR in host defense against meningitic E. coli infection was demonstrated by using α7-deficient (α7-/-) mouse brain microvascular endothelial cells (BMEC) and animal model systems. As shown in our in vitro and in vivo studies, E. coli K1 invasion and polymorphonuclear neutrophil (PMN) transmigration across the blood-brain barrier (BBB) were significantly reduced in α7-/- BMEC and α7-/- mice. Stimulation by nicotine was abolished in the α7-/- cells and animals. The same blocking effect was achieved by methyllycaconitine (α7 antagonist). The tight junction molecules occludin and ZO-1 were significantly reduced in the brain cortex of wildtype mice infected with E. coli and treated with nicotine, compared to α7-/- cells and animals. Decreased neuronal injury in the hippocampal dentate gyrus was observed in α7-/- mice with meningitis. Proinflammatory cytokines (IL-1β, IL-6, TNFα, MCP-1, MIP-1alpha, and RANTES) and adhesion molecules (CD44 and ICAM-1) were significantly reduced in the cerebrospinal fluids of the α7-/- mice with E. coli meningitis. Furthermore, α7 nAChR is the major calcium channel for nicotine- and E. coli K1-increased intracellular calcium concentrations of mouse BMEC. Taken together, our data suggest that α7 nAChR plays a detrimental role in the host defense against meningitic infection by modulation of pathogen invasion, PMN recruitment, calcium signaling and neuronal inflammation.


Journal of Immunotherapy | 2013

Membrane-bound TRAIL supplements natural killer cell cytotoxicity against neuroblastoma cells.

Michael A. Sheard; Shahab Asgharzadeh; Yin Liu; Tsen Yin Lin; Hong Wei Wu; Lingyun Ji; Susan Groshen; Dean A. Lee; Robert C. Seeger

Neuroblastoma cells have been reported to be resistant to death induced by soluble, recombinant forms of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) (CD253/TNFSF10) because of low or absent expression of caspase-8 and/or TRAIL-receptor 2 (TRAIL-R2/DR5/CD262/TNFRSF10b). However, their sensitivity to membrane-bound TRAIL on natural killer (NK) cells is not known. Comparing microarray gene expression and response to NK cell–mediated cytotoxicity, we observed a correlation between TRAIL-R2 expression and the sensitivity of 14 neuroblastoma cell lines to the cytotoxicity of NK cells activated with interleukin (IL)-2 plus IL-15. Even though most NK cytotoxicity was dependent upon perforin, the cytotoxicity was supplemented by TRAIL in 14 of 17 (82%) neuroblastoma cell lines as demonstrated using an anti-TRAIL neutralizing antibody. Similarly, a recently developed NK cell expansion system employing IL-2 plus lethally irradiated K562 feeder cells constitutively expressing membrane-bound IL-21 (K562 clone 9.mbIL21) resulted in activated NK cells derived from normal healthy donors and neuroblastoma patients that also utilized TRAIL to supplement cytotoxicity. Exogenous interferon-&ggr; upregulated expression of caspase-8 in 3 of 4 neuroblastoma cell lines and increased the contribution of TRAIL to NK cytotoxicity against 2 of the 3 lines; however, relatively little inhibition of cytotoxicity was observed when activated NK cells were treated with an anti-interferon-&ggr; neutralizing antibody. Constraining the binding of anti-TRAIL neutralizing antibody to membrane-bound TRAIL but not soluble TRAIL indicated that membrane-bound TRAIL alone was responsible for essentially all of the supplemental cytotoxicity. Together, these findings support a role for membrane-bound TRAIL in the cytotoxicity of NK cells against neuroblastoma cells.


Molecular Cancer Therapeutics | 2010

Combination of Vorinostat and Flavopiridol Is Selectively Cytotoxic to Multidrug-Resistant Neuroblastoma Cell Lines with Mutant TP53

Jen-Ming Huang; Michael A. Sheard; Lingyun Ji; Richard Sposto; Nino Keshelava

As p53 loss of function (LOF) confers high-level drug resistance in neuroblastoma, p53-independent therapies might have superior activity in recurrent neuroblastoma. We tested the activity of vorinostat, a histone deacetylase inhibitor, and flavopiridol, a pan-Cdk inhibitor, in a panel of multidrug-resistant neuroblastoma cell lines that included lines with wild-type (wt) and transcriptionally active TP53 (n = 3), mutated (mt), and LOF TP53 (n = 4) or p14ARF deletion (n = 1). The combination of vorinostat and flavopiridol was synergistic and significantly more cytotoxic (P < 0.001) in cell lines with p53-LOF and in the clones stably transfected with dominant-negative p53 plasmids. Cell cycle analysis by flow cytometry showed prominent cell-cycle arrest in G2/M (37%) for a cell line with wt TP53 (SK-N-RA) at 16 to 20 hours, while cells with mt TP53 (CHLA-90) slipped into sub-G1 at 6 to 24 hours (25%–40% specific cell death). The morphological hallmarks of mitotic cell death, including defective spindle formation and abnormal cytokinesis, were detected by confocal microscopy after the treatment with vorinostat + flavopiridol combination in CHLA-90. The combination caused reduction in the expression of G2/M proteins (cyclin B1, Mad2, MPM2) in 2 cell lines with mt TP53 but not in those with wt TP53. Plk1 expression was reduced in all treated lines. Small interfering RNA knockdown of Mad2 and cyclin B1 or Plk1 synergistically reduced the clonogenicity of CHLA-90 cells. The combination of HDAC inhibitor and flavopiridol may be a unique approach to treating neuroblastomas with p53 LOF, one that evokes induction of mitotic failure. Mol Cancer Ther; 9(12); 3289–301. ©2010 AACR.


Clinical Cancer Research | 2017

TGFβR1 Blockade with Galunisertib (LY2157299) Enhances Anti-Neuroblastoma Activity of Anti-GD2 Antibody Dinutuximab (ch14.18) with Natural Killer Cells

Hung C. Tran; Zesheng Wan; Michael A. Sheard; Jianping Sun; Jeremy R. Jackson; Jemily Malvar; Yibing Xu; Larry Wang; Richard Sposto; Eugene S. Kim; Shahab Asgharzadeh; Robert C. Seeger

Purpose: Immunotherapy of high-risk neuroblastoma using the anti-GD2 antibody dinutuximab induces antibody-dependent cell-mediated cytotoxicity (ADCC). Galunisertib, an inhibitor of TGFβR1, was examined for its ability to enhance the efficacy of dinutuximab in combination with human ex vivo activated NK (aNK) cells against neuroblastoma. Experimental Design: TGFB1 and TGFBR1 mRNA expression was determined for 249 primary neuroblastoma tumors by microarray analysis. The ability of galunisertib to inhibit SMAD activity induced by neuroblastoma patient blood and bone marrow plasmas in neuroblastoma cells was tested. The impact of galunisertib on TGFβ1-induced inhibition of aNK cytotoxicity and ADCC in vitro and on anti-neuroblastoma activity in NOD-scid gamma (NSG) mice was determined. Results: Neuroblastomas express TGFB1 and TGFBR1 mRNA. Galunisertib suppressed SMAD activation in neuroblastoma cells induced by exogenous TGFβ1 or by patient blood and bone marrow plasma, and suppressed SMAD2 phosphorylation in human neuroblastoma cells growing in NSG mice. In NK cells treated in vitro with exogenous TGFβ1, galunisertib suppressed SMAD2 phosphorylation and restored the expression of DNAM-1, NKp30, and NKG2D cytotoxicity receptors and the TRAIL death ligand, the release of perforin and granzyme A, and the direct cytotoxicity and ADCC of aNK cells against neuroblastoma cells. Addition of galunisertib to adoptive cell therapy with aNK cells plus dinutuximab reduced tumor growth and increased survival of mice injected with two neuroblastoma cell lines or a patient-derived xenograft. Conclusions: Galunisertib suppresses activation of SMAD2 in neuroblastomas and aNK cells, restores NK cytotoxic mechanisms, and increases the efficacy of dinutuximab with aNK cells against neuroblastoma tumors. Clin Cancer Res; 23(3); 804–13. ©2016 AACR. See related commentary by Zenarruzabeitia et al., p. 615


Experimental Cell Research | 2015

Preservation of high glycolytic phenotype by establishing new acute lymphoblastic leukemia cell lines at physiologic oxygen concentration

Michael A. Sheard; Matthew V. Ghent; Daniel J. Cabral; Joanne C. Lee; Vazgen Khankaldyyan; Lingyun Ji; Samuel Q. Wu; Min H. Kang; Richard Sposto; Shahab Asgharzadeh; C. Patrick Reynolds

Cancer cells typically exhibit increased glycolysis and decreased mitochondrial oxidative phosphorylation, and they continue to exhibit some elevation in glycolysis even under aerobic conditions. However, it is unclear whether cancer cell lines employ a high level of glycolysis comparable to that of the original cancers from which they were derived, even if their culture conditions are changed to physiologically relevant oxygen concentrations. From three childhood acute lymphoblastic leukemia (ALL) patients we established three new pairs of cell lines in both atmospheric (20%) and physiologic (bone marrow level, 5%) oxygen concentrations. Cell lines established in 20% oxygen exhibited lower proliferation, survival, expression of glycolysis genes, glucose consumption, and lactate production. Interestingly, the effects of oxygen concentration used during cell line initiation were only partially reversible when established cell cultures were switched from one oxygen concentration to another for eight weeks. These observations indicate that ALL cell lines established at atmospheric oxygen concentration can exhibit relatively low levels of glycolysis and these levels are semi-permanent, suggesting that physiologic oxygen concentrations may be needed from the time of cell line initiation to preserve the high level of glycolysis commonly exhibited by leukemias in vivo.

Collaboration


Dive into the Michael A. Sheard's collaboration.

Top Co-Authors

Avatar

Richard Sposto

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Robert C. Seeger

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Shahab Asgharzadeh

Children's Hospital Los Angeles

View shared research outputs
Top Co-Authors

Avatar

Jemily Malvar

Children's Hospital Los Angeles

View shared research outputs
Top Co-Authors

Avatar

Jianping Sun

Children's Hospital Los Angeles

View shared research outputs
Top Co-Authors

Avatar

Susan Groshen

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Hong-Wei Wu

University of Southern California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lingyun Ji

University of Southern California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge