Product Citations: 6

A superantigen-based MHC class II targeted cancer immunotherapy for the treatment of acute myeloid leukemia

Preprint on Research Square on 18 April 2025 by Dolloff, N., Golick, L., et al.

Abstract The treatment of acute myeloid leukemia (AML) remains a challenge due to disease heterogeneity, which undermines efforts to develop targeted therapeutics, leaving conventional chemotherapy as the standard of care (SOC). Sialic acid binding Ig-like lectin 3 (CD33) is a myeloid cell surface glycoprotein that is highly expressed on AML blasts. However, while ~90% of AML cases express CD33, 50% of these patients harbor a single nucleotide polymorphism (SNP) that eliminates the antibody binding epitope for existing CD33-targeted antibody therapeutics. In this study, we developed an immunotherapy (M2T-CD33) that targets CD33 directly to MHC class II molecules on antigen presenting cells for enhanced presentation to the immune system. We found that M2T-CD33 induces a robust polyclonal anti-CD33 humoral response composed of the full immunoglobulin repertoire. M2T-CD33 induced an anti-AML response in a syngeneic mouse model that was dependent on CD4+ and CD8+ T cells. The immune response was elicited against both the full length and spliced version of CD33 and showed no evidence of toxicity at concentrations 40-fold higher than the efficacious dose. Finally, M2T-CD33 was enhanced by combinations with anti-PD-1 therapy. These experiments demonstrate the preclinical potential of M2T-CD33 in AML and emphasize the importance of MHCII for cancer immunotherapy.

  • Cancer Research
  • Immunology and Microbiology

Recent studies have reported that circular RNAs (circRNAs) play a crucial regulatory role in a variety of human diseases. However, the roles of circRNAs in pathological osteogenesis in ankylosing spondylitis (AS) remain unclear. We conducted circRNA and miRNA expression profiling of osteogenically differentiated bone marrow-derived mesenchymal stem cells (BMSCs) of patients with AS compared with those of healthy donors (HDs) by RNA sequencing (RNA-seq). Results showed that a total of 31806 circRNAs were detected in the BMSC samples, of which 418 circRNAs were significantly differentially expressed (DE) with a fold change ≥2 and p value <0.05. Among these, 204 circRNAs were upregulated, and 214 were downregulated. GO and KEGG analyses demonstrated that the DE circRNAs were mainly involved in the regulation of biological processes of the cell matrix adhesion and the TGF-beta signaling pathway, which are closely related to AS. circRNA-miRNA interaction networks related to the TGF-beta signaling pathway were established. The results of qRT-PCR showed that has_circ_0070562 was significantly up-regulated in AS-MSCs. In vitro experiments showed that silencing of has_circ_0070562 weakened osteogenesis of AS-BMSCs. In conclusion, we identified numerous circRNAs that were dysregulated in AS-BMSCs compared with HD-BMSCs. Bioinformatic analyses suggested that these dysregulated circRNAs might play important functional roles in AS-BMSCs osteogenesis. Circ_0070562 functioned as a pro-ostegenic factor and might serve as a potential biomarker and a therapeutic target for AS.
Copyright © 2022 Wang, Chen, Zeng, Gu, Wang, Yu, Wu and Shen.

  • FC/FACS
  • Homo sapiens (Human)
  • Genetics
  • Stem Cells and Developmental Biology

Progesterone-induced blocking factor improves blood pressure, inflammation, and pup weight in response to reduced uterine perfusion pressure (RUPP).

In American Journal of Physiology - Regulatory, Integrative and Comparative Physiology on 1 May 2021 by Cottrell, J. N., Witcher, A. C., et al.

Preeclampsia (PE) is characterized by new-onset hypertension in association with elevated natural killer (NK) cells and inflammatory cytokines, which are likely culprits for decreased fetal weight during PE pregnancies. As progesterone increases during normal pregnancy, it stimulates progesterone-induced blocking factor (PIBF). PIBF has been shown to decrease inflammation and cytolytic NK cells, both of which are increased during PE. We hypothesized that PIBF reduces inflammation as a mechanism to improve hypertension in the preclinical reduced uterine perfusion pressure (RUPP) rat model of PE. PIBF (2.0 µg/mL) was administered intraperitoneally on gestational day 15 to either RUPP or normal pregnant (NP) rats. On day 18, carotid catheters were inserted. Mean arterial blood pressure (MAP) and samples were collected on day 19. MAP in NP rats (n = 11) was 100 ± 2 mmHg and 105 ± 3 mmHg in NP + PIBF rats (n = 8) and 122 ± 1 mmHg in RUPP rats (n = 10), which improved to 110 ± 2 mmHg in RUPP + PIBF rats (n = 11), P < 0.05. Pup weight was 2.4 ± 0.1 g in NP, 2.5 ± 0.1 g in NP + PIBF, 1.9 ± 0.1 g in RUPP, and improved to 2.1 ± 0.1 g in RUPP + PIBF rats. Circulating and placental cytolytic NK cells, IL-17, and IL-6 were significantly reduced while IL-4 and T helper (TH) 2 cells were significantly increased in RUPP rats after PIBF administration. Importantly, vasoactive pathways preproendothelin-1, nitric oxide, and soluble fms-Like tyrosine Kinase-1 (sFlt-1) were normalized in RUPP + PIBF rats compared with RUPP rats, P < 0.05. Our findings suggest that PIBF normalized IL-4/TH2 cells, which was associated with improved inflammation, fetal growth restriction, and blood pressure in the RUPP rat model of PE.

  • Cardiovascular biology
  • Endocrinology and Physiology
  • Immunology and Microbiology

Immunophenotypic Analysis of CAR-T Cells.

In Methods in Molecular Biology (Clifton, N.J.) on 11 November 2019 by de Azevedo, J. T. C., Mizukami, A., et al.

CAR-T cell immunotherapy is a promising therapeutic modality for cancer patients. The success of CAR-T cell therapy has been associated with the phenotype, activation and functional profiling of infused CAR-T cells. Therefore, immunophenotypic characterization of CAR-T cells during bioprocess is crucial for cell quality control and ultimately for improved antitumor efficacy. In this chapter, we propose a flow cytometry panel to characterize the immunophenotype of the CAR-T subsets.

  • Biochemistry and Molecular biology
  • Immunology and Microbiology

The pathology of bone tissue during peri-implantitis.

In Journal of Dental Research on 1 February 2015 by Schminke, B., Vom Orde, F., et al.

Dental implants are one of the most frequently used treatment options for tooth replacement. Approximately 30% of patients with dental implants develop peri-implantitis, which is an oral inflammatory disease that leads to loss of the supporting tissues, predominately the bone. For the development of future therapeutic strategies, it is essential to understand the molecular pathophysiology of human dental peri-implant infections. Here, we describe the gene and protein expression patterns of peri-implantitis bone tissue compared with healthy peri-implant bone tissue. Furthermore, cells from the osteoblastic lineage derived from peri-implantitis samples were immortalized and characterized. We applied microarray, quantitative reverse transcription polymerase chain reaction, fluorescence-activated cell sorting, and Western blot analyses. The levels of typical bone matrix molecules, including SPP1, BGLAP, and COL9A1, in patients with peri-implantitis were reduced, while the inflammation marker interleukin 8 (IL8) was highly expressed. RUNX2, one of the transcription factors of mature osteoblasts, was also decreased in peri-implantitis. Finally, the human telomerase reverse transcriptase immortalized cell line from peri-implantitis exhibited a more fibro-osteoblastic character than did the healthy control.
© International & American Associations for Dental Research 2014.

  • Pathology
View this product on CiteAb