1
|
Abah MO, Ogenyi DO, Zhilenkova AV, Essogmo FE, Ngaha Tchawe YS, Uchendu IK, Pascal AM, Nikitina NM, Rusanov AS, Sanikovich VD, Pirogova YN, Boroda A, Moiseeva AV, Sekacheva MI. Innovative Therapies Targeting Drug-Resistant Biomarkers in Metastatic Clear Cell Renal Cell Carcinoma (ccRCC). Int J Mol Sci 2024; 26:265. [PMID: 39796121 PMCID: PMC11720203 DOI: 10.3390/ijms26010265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/09/2024] [Accepted: 12/14/2024] [Indexed: 01/13/2025] Open
Abstract
A thorough study of Clear Cell Renal Cell Carcinoma (ccRCC) shows that combining tyrosine kinase inhibitors (TKI) with immune checkpoint inhibitors (ICI) shows promising results in addressing the tumor-promoting influences of abnormal immunological and molecular biomarkers in metastatic Clear Cell Renal Cell Carcinoma (ccRCC). These abnormal biomarkers enhance drug resistance, support tumor growth, and trigger cancer-related genes. Ongoing clinical trials are testing new treatment options that appear more effective than earlier ones. However, more research is needed to confirm their long-term safety use and potential side effects. This study highlights vital molecular and immunological biomarkers associated with drug resistance in Clear Cell Renal Cell Carcinoma (ccRCC). Furthermore, this study identifies a number of promising drug candidates and biomarkers that serve as significant contributors to the enhancement of the overall survival of ccRCC patients. Consequently, this article offers pertinent insights on both recently completed and ongoing clinical trials, recommending further toxicity study for the prolonged use of this treatment strategy for patients with metastatic ccRCC, while equipping researchers with invaluable information for the progression of current treatment strategies.
Collapse
Affiliation(s)
- Moses Owoicho Abah
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
- Department of Cancer Bioinformatics and Molecular Biology, Royal Society of Clinical and Academic Researchers (ROSCAR) International, Abuja 900104, Nigeria
| | - Deborah Oganya Ogenyi
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
| | - Angelina V. Zhilenkova
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
| | - Freddy Elad Essogmo
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
| | - Yvan Sinclair Ngaha Tchawe
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
| | - Ikenna Kingsley Uchendu
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
- Medical Laboratory Science Department, Faculty of Health Science and Technology, College of Medicine, University of Nigeria, Enugu Campus, Enugu 410001, Nigeria
| | - Akaye Madu Pascal
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
| | - Natalia M. Nikitina
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
| | - Alexander S. Rusanov
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
| | - Varvara D. Sanikovich
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
| | - Yuliya N. Pirogova
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
| | - Alexander Boroda
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
| | - Aleksandra V. Moiseeva
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
| | - Marina I. Sekacheva
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.O.O.); (A.V.Z.); (F.E.E.); (Y.S.N.T.); (A.M.P.); (N.M.N.); (A.S.R.); (V.D.S.); (Y.N.P.); (A.B.); (A.V.M.); (M.I.S.)
| |
Collapse
|
2
|
Basmaeil Y, Subayyil AA, Kulayb HB, Kondkar AA, Alrodayyan M, Khatlani T. Partial Inhibition of Epithelial-to-Mesenchymal Transition (EMT) Phenotypes by Placenta-Derived DBMSCs in Human Breast Cancer Cell Lines, In Vitro. Cells 2024; 13:2131. [PMID: 39768220 PMCID: PMC11674051 DOI: 10.3390/cells13242131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/09/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Stem cell-based therapies hold significant potential for cancer treatment due to their unique properties, including migration toward tumor niche, secretion of bioactive molecules, and immunosuppression. Mesenchymal stem cells (MSCs) from adult tissues can inhibit tumor progression, angiogenesis, and apoptosis of cancer cells. We have previously reported the isolation and characterization of placenta-derived decidua basalis mesenchymal stem cells (DBMSCs), which demonstrated higher levels of pro-migratory and anti-apoptotic genes, indicating potential anti-cancer effects. In this study, we analyzed the anti-cancer effects of DBMSCs on human breast cancer cell lines MDA231 and MCF7, with MCF 10A used as control. We also investigated how these cancer cells lines affect the functional competence of DBMSCs. By co-culturing DBMSCs with cancer cells, we analyzed changes in functions of both cell types, as well as alterations in their genomic and proteomic profile. Our results showed that treatment with DBMSCs significantly reduced the functionality of MDA231 and MCF7 cells, while MCF 10A cells remained unaffected. DBMSC treatment decreased epithelial-to-mesenchymal transition (EMT)-related protein levels in MDA231 cells and modulated expression of other cancer-related genes in MDA231 and MCF7 cells. Although cancer cells reduced DBMSC proliferation, they increased their expression of anti-apoptotic genes. These findings suggest that DBMSCs can inhibit EMT-related proteins and reduce the invasive characteristics of MDA231 and MCF7 breast cancer cells, highlighting their potential as candidates for cell-based cancer therapies.
Collapse
Affiliation(s)
- Yasser Basmaeil
- Stem Cells and Regenerative Medicine Unit, Blood and Cancer Research (BCR) Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences (KSAU), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia; (Y.B.); (A.A.S.); (H.B.K.); (M.A.)
| | - Abdullah Al Subayyil
- Stem Cells and Regenerative Medicine Unit, Blood and Cancer Research (BCR) Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences (KSAU), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia; (Y.B.); (A.A.S.); (H.B.K.); (M.A.)
| | - Haya Bin Kulayb
- Stem Cells and Regenerative Medicine Unit, Blood and Cancer Research (BCR) Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences (KSAU), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia; (Y.B.); (A.A.S.); (H.B.K.); (M.A.)
| | - Altaf A. Kondkar
- Department of Ophthalmology, College of Medicine, King Saud University, Riyadh 11411, Saudi Arabia;
| | - Maha Alrodayyan
- Stem Cells and Regenerative Medicine Unit, Blood and Cancer Research (BCR) Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences (KSAU), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia; (Y.B.); (A.A.S.); (H.B.K.); (M.A.)
| | - Tanvir Khatlani
- Stem Cells and Regenerative Medicine Unit, Blood and Cancer Research (BCR) Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences (KSAU), Ministry of National Guard Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia; (Y.B.); (A.A.S.); (H.B.K.); (M.A.)
| |
Collapse
|
3
|
Baracuhy EM, Cormier O, Davola ME, Collins S, Mossman K. Virus replication is not required for oncolytic bovine herpesvirus-1 immunotherapy. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200906. [PMID: 39691853 PMCID: PMC11650296 DOI: 10.1016/j.omton.2024.200906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 10/18/2024] [Accepted: 11/14/2024] [Indexed: 12/19/2024]
Abstract
Oncolytic viruses are a promising approach for cancer treatment where viruses selectively target and kill cancer cells while also stimulating an immune response. Among viruses with this ability, bovine herpesvirus-1 (BoHV-1) has several advantages, including observations suggesting it may not require viral replication for its anti-cancer effects. We previously demonstrated that binding and penetration of enveloped virus particles are sufficient to trigger intrinsic and innate immune signaling in normal cells, while other groups have published the efficacy of non-replicating viruses as viable immunotherapies in different cancer models. In this work, we definitively show that live and UV-inactivated (UV) (non-replicating) BoHV-1-based regimens extend survival of tumor-bearing mice to similar degrees and induce infiltration of similar immune cell populations, with the exception of neutrophils. Transcriptomic analysis of tumors treated with either live or UV BoHV-1-based regimens revealed similar pathway enrichment and a subset of overlapping differentially regulated genes, suggesting live and UV BoHV-1 have similar mechanisms of activity. Last, we present a gene signature across our in vitro and in vivo models that could potentially be used to validate new BoHV-1 therapeutics. This work contributes to the growing body of literature showing that replication may not be necessary for therapeutic efficacy of viral immunotherapies.
Collapse
Affiliation(s)
- Enzo Mongiovi Baracuhy
- Center for Discovery in Cancer Research, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Olga Cormier
- Center for Discovery in Cancer Research, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Maria Eugenia Davola
- Center for Discovery in Cancer Research, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Susan Collins
- Center for Discovery in Cancer Research, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Karen Mossman
- Center for Discovery in Cancer Research, Department of Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
4
|
Sasaki Y, Sato Y, Nomura K, Wakita A, Nagaki Y, Sasamori R, Yoneya T, Takahashi T, Yamada M, Takahashi M, Terata K, Imai K. Peptidoglycan induces CXCL10 production and inhibits esophageal squamous cell carcinoma proliferation. Am J Cancer Res 2024; 14:5874-5884. [PMID: 39803645 PMCID: PMC11711533 DOI: 10.62347/nhpv3701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 11/28/2024] [Indexed: 01/16/2025] Open
Abstract
Poor oral health is an independent risk factor for upper-aerodigestive tract cancers, including esophageal squamous cell carcinoma (ESCC); thus, good oral health may reduce the risk of ESCC. We previously reported that high expression of Toll-like receptor (TLR) 6, which recognizes peptidoglycan (PGN) from Gram-positive bacteria correlates with a good prognosis after esophagectomy for ESCC. Most beneficial bacteria in the mouth are Gram-positive. We therefore hypothesized that PGN affects cancer cell proliferation and disease progression in ESCC. To test that idea, we assessed the expression of cytokine and chemokine mRNA and protein in eight ESCC cell lines. We also employed a mouse xenograft model to investigate the effect of PGN on ESCC tumor progression in vivo. We then investigated the relationship between the combined expression profiles of TLR6 and C-X-C motif chemokine ligand 10 (CXCL10) in clinical samples and 5-year overall survival (OS) and disease-specific survival (DSS) in ESCC patients after curative esophagectomy. We found that PGN significantly inhibited cell proliferation in six of eight ESCC lines and upregulated CXCL10 production via NF-κB2. In vivo, subcutaneous PGN administration tended to decrease ESCC tumor volume in mice. Combined high expression of TLR6 and CXCL10 correlated with a better prognosis in ESCC patients. This suggests that PGN reduces cell proliferation and tumor progression through a PGN-TLR-CXCL10 cascade, thereby influencing prognosis after esophagectomy for ESCC, and that improving the oral environment could potentially improve the prognosis of ESCC patients after esophagectomy.
Collapse
Affiliation(s)
- Yoshihiro Sasaki
- Division of Esophageal Surgery, Akita University HospitalAkita 010-8543, Japan
- Department of Thoracic Surgery, Akita University Graduate School of MedicineAkita 010-8543, Japan
| | - Yusuke Sato
- Division of Esophageal Surgery, Akita University HospitalAkita 010-8543, Japan
- Department of Thoracic Surgery, Akita University Graduate School of MedicineAkita 010-8543, Japan
| | - Kyoko Nomura
- Department of Environmental Health Science and Public Health, Akita University Graduate School of MedicineAkita 010-8543, Japan
| | - Akiyuki Wakita
- Division of Esophageal Surgery, Akita University HospitalAkita 010-8543, Japan
- Department of Thoracic Surgery, Akita University Graduate School of MedicineAkita 010-8543, Japan
| | - Yushi Nagaki
- Division of Esophageal Surgery, Akita University HospitalAkita 010-8543, Japan
- Department of Thoracic Surgery, Akita University Graduate School of MedicineAkita 010-8543, Japan
| | - Ryohei Sasamori
- Division of Esophageal Surgery, Akita University HospitalAkita 010-8543, Japan
- Department of Thoracic Surgery, Akita University Graduate School of MedicineAkita 010-8543, Japan
| | - Takatoshi Yoneya
- Division of Esophageal Surgery, Akita University HospitalAkita 010-8543, Japan
- Department of Thoracic Surgery, Akita University Graduate School of MedicineAkita 010-8543, Japan
| | - Tsukasa Takahashi
- Division of Esophageal Surgery, Akita University HospitalAkita 010-8543, Japan
- Department of Thoracic Surgery, Akita University Graduate School of MedicineAkita 010-8543, Japan
| | - Munehiro Yamada
- Division of Esophageal Surgery, Akita University HospitalAkita 010-8543, Japan
- Department of Thoracic Surgery, Akita University Graduate School of MedicineAkita 010-8543, Japan
| | - Mayu Takahashi
- Division of Esophageal Surgery, Akita University HospitalAkita 010-8543, Japan
- Department of Thoracic Surgery, Akita University Graduate School of MedicineAkita 010-8543, Japan
| | - Kaori Terata
- Department of Thoracic Surgery, Akita University Graduate School of MedicineAkita 010-8543, Japan
| | - Kazuhiro Imai
- Department of Thoracic Surgery, Akita University Graduate School of MedicineAkita 010-8543, Japan
| |
Collapse
|
5
|
Schonmann Y, Cohen NT, Czarnowicki T. Evaluating cancer risk in a large cohort of 25,008 patients with vitiligo: Insights from a comprehensive cohort population-based study. J Am Acad Dermatol 2024:S0190-9622(24)03284-5. [PMID: 39631697 DOI: 10.1016/j.jaad.2024.11.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/14/2024] [Accepted: 11/03/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Current research presents inconsistent results concerning the incidence of various malignancies among patients with vitiligo. OBJECTIVES To evaluate the risk of cancer in patients with vitiligo. METHODS A population-based study was conducted using Clalit Health Services database (2000-2023) using a cohort study design. Adjusted hazard ratios (HRs) were calculated using multivariate Cox regressions. RESULTS The study included 25,008 patients with vitiligo and 245,550 matched controls. The mean (SD) age of patients with vitiligo was 35.96 (22.39) years; 12,679 (50.70%) patients were men. There were 499 (95% confidence interval [CI]: 468-532) and 487 (95% CI: 476-497) incident cancer cases per 100,000 person-years among patients with and without vitiligo, respectively (adjusted HR: 1.00; 95% CI: 0.93-1.07; P = .999). Patients with vitiligo had a reduced risk of melanoma (adjusted HR: 0.70; 95% CI: 0.50-0.99; P = .0337), lung cancer (adjusted HR: 0.73; 95% CI: 0.57-0.93; P = .007), and bladder cancer (adjusted HR: 0.70; 95% CI: 0.52-0.94; P = .0138). LIMITATIONS The secondary analysis involved multiple comparisons, which could potentially influence the results. CONCLUSION Our study found that cancer incidence rates in patients with vitiligo are not elevated. Consequently, cancer screening for these patients should follow the standard guidelines recommended for the general population.
Collapse
Affiliation(s)
- Yochai Schonmann
- Department of Quality Measures and Research, Clalit Health Services, Tel Aviv, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Department of Family Medicine, Tel Aviv District, Clalit Health Services, Tel Aviv, Israel
| | - Naama T Cohen
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Tali Czarnowicki
- Dr. Phillip Frost Department of Dermatology & Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida.
| |
Collapse
|
6
|
Burnham AJ, Toma AI, Shah D, Cha T, Kaimari S, Behara M, Sekar KPC, Kamalakar A, Willett N, Botchwey E, Goudy SL. FTY720P-treated macrophages in PEG-4MAL hydrogels promote oral wound healing. Cytotherapy 2024:S1465-3249(24)00929-0. [PMID: 39665738 DOI: 10.1016/j.jcyt.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 10/21/2024] [Accepted: 11/03/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND AIMS Oral wound healing involves hemostasis, inflammation, proliferation and tissue remodeling. The oral cavity is a complex wound healing environment because of the presence of saliva, a high bacterial burden and ongoing physical trauma from eating. The inflammatory component of wound healing balances the polarization of macrophages in healing tissues between M1 inflammatory macrophages and M2 anti-inflammatory macrophages. M2 macrophages secrete anti-inflammatory and pro-regenerative cytokines and chemokines, which aid wound healing. Fingolimod, or FTY720, a Food and Drug Administration-approved sphingosine-1-phosphate modulator, has been implicated in inducing the polarization of macrophages to the M2 phenotype. In this study, we investigated whether macrophage pre-treatment with phosphorylated FTY720 (FTY720P), the bioactive form of the drug, in a PEG-4MAL hydrogel promotes improved oral wound healing in a critically sized oral mucosal defect model. METHODS AND RESULTS Using cytokine dot blots and Luminex cytokine assays (MilliporeSigma, Burlington, MA, USA), FTY720P-treated murine RAW 264.7 and human THP1-differentiated macrophages in PEG-4MAL hydrogels secreted chemokines and cytokines known to regulate inflammation (e.g., interleukin [IL] 4, IL-13) and induce M2 macrophage polarization (e.g., CCL6, CCL22), leukocyte migration (e.g., CXCL2, CCL2, CCL12, CCL22), angiogenesis (e.g., vascular endothelial growth factor) and epithelialization (e.g., IL-1, IL-17, IL-22). In vitro, FTY720P-treated cells induced chemotaxis of macrophages and fibroblasts in Transwell assays (Corning, Corning, NY, USA) and oral epithelial scratch wound closure. In a murine oronasal fistula (ONF) model of oral wound healing, the local application of FTY720P-treated macrophages in PEG-4MAL hydrogels significantly increased wound closure (75% closure) relative to non-treated cells (40% closure) and blank hydrogel controls (25% closure) (P < 0.0001). Flow cytometry of mouse palatal tissue showed that application of FTY720P-treated macrophage hydrogels to ONFs significantly increased day 7 percentage of M1 and M2 macrophages, mesenchymal stromal cells and CD19+ B cells. Significantly fewer neutrophils, monocytes, CD4+/CD8+ T cells and endothelial cells were observed in the FTY720P-treated macrophage defects, suggesting that FTY720P-treated macrophages in hydrogels promote oral wound healing via suppression of granulation and resolution of inflammation and promotion of tissue maturation. CONCLUSIONS Our data provide new insights into the use of potential FTY720P-treated macrophage therapies for oral wound healing and have clinical implications for cleft palate and oral surgery.
Collapse
Affiliation(s)
- Andre J Burnham
- Department of Otolaryngology-Head and Neck Surgery, Washington University School of Medicine, St Louis, Missouri, USA; Department of Otolaryngology-Head and Neck Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Afra I Toma
- Department of Otolaryngology-Head and Neck Surgery, Emory University School of Medicine, Atlanta, Georgia, USA; Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Daniel Shah
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Tim Cha
- Department of Otolaryngology-Head and Neck Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Sundus Kaimari
- Department of Otolaryngology-Head and Neck Surgery, Emory University School of Medicine, Atlanta, Georgia, USA; Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Monica Behara
- Department of Otolaryngology-Head and Neck Surgery, Emory University School of Medicine, Atlanta, Georgia, USA; Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | | | - Archana Kamalakar
- Department of Otolaryngology-Head and Neck Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Nick Willett
- Department of Bioengineering, University of Oregon, Eugene, Oregon, USA
| | - Edward Botchwey
- Department of Otolaryngology-Head and Neck Surgery, Emory University School of Medicine, Atlanta, Georgia, USA; Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Steven L Goudy
- Department of Otolaryngology-Head and Neck Surgery, Emory University School of Medicine, Atlanta, Georgia, USA; Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA; Department of Otolaryngology, Children's Healthcare of Atlanta, Atlanta, Georgia, USA.
| |
Collapse
|
7
|
Rajasekaran S, Cheng S, Gajendran N, Shekoohi S, Chesnokova L, Yu X, Witt SN. Transcriptomic analysis of melanoma cells reveals an association of α-synuclein with regulation of the inflammatory response. Sci Rep 2024; 14:27140. [PMID: 39511366 PMCID: PMC11544018 DOI: 10.1038/s41598-024-78777-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 11/04/2024] [Indexed: 11/15/2024] Open
Abstract
The Parkinson's disease protein, alpha-synuclein (α-syn/SNCA), is highly expressed in neurons and melanomas. The goal of this study was to reveal the mechanism(s) of α-syn's involvement in melanoma pathogenesis. To decipher the genes and pathways affected by α-syn, we conducted an RNA sequencing analysis of human SK-MEL-28 cells and several SK-MEL-28 SNCA-KO clones. We identified 1098 significantly up-regulated genes and 660 significantly down-regulated genes. Several of the upregulated genes are related to the immune system, i.e., the inflammatory response and the matrisome. We validated five upregulated genes (IL-1β, SAA1, IGFBP5, CXCL8, and CXCL10) by RT-qPCR and detected IGFBP5 and IL-1β in spent media of control and SNCA-KO cells. The levels of each of these secreted proteins were significantly higher in the spent media of the SNCA-KO clones than control cells. These secreted proteins quite likely activate the immune response against SNCA-KO cells. We suggest that, conversely, high levels of α-syn expression in melanoma cells helps the cells evade the immune system by inhibiting the secretion of these immune activating factors.
Collapse
Affiliation(s)
- Santhanasabapathy Rajasekaran
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Siyuan Cheng
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Nithya Gajendran
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Sahar Shekoohi
- Department of Anesthesiology, Louisiana State University Health Sciences Center at Shreveport, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Liudmila Chesnokova
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Xiuping Yu
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, USA
- Feist-Weiller Cancer Center, Louisiana State University Health Shreveport, Shreveport, USA
| | - Stephan N Witt
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, USA.
- Feist-Weiller Cancer Center, Louisiana State University Health Shreveport, Shreveport, USA.
| |
Collapse
|
8
|
Tashakori N, Mikhailova MV, Mohammedali ZA, Mahdi MS, Ali Al-Nuaimi AM, Radi UK, Alfaraj AM, Kiasari BA. Circular RNAs as a novel molecular mechanism in diagnosis, prognosis, therapeutic target, and inhibiting chemoresistance in breast cancer. Pathol Res Pract 2024; 263:155569. [PMID: 39236498 DOI: 10.1016/j.prp.2024.155569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 08/21/2024] [Accepted: 08/28/2024] [Indexed: 09/07/2024]
Abstract
Breast cancer (BC) is the most common cancer among women, characterized by significant heterogeneity. Diagnosis of the disease in the early stages and appropriate treatment plays a crucial role for these patients. Despite the available treatments, many patients due to drug resistance do not receive proper treatments. Recently, circular RNAs (circRNAs), a type of non-coding RNAs (ncRNAs), have been discovered to be involved in the progression and resistance to drugs in BC. CircRNAs can promote or inhibit malignant cells by their function. Numerous circRNAs have been discovered to be involved in the proliferation, invasion, and migration of tumor cells, as well as the progression, pathogenesis, tumor metastasis, and drug resistance of BC. Circular RNAs can also serve as a biomarker for diagnosing, predicting prognosis, and targeting therapy. In this review, we present an outline of the variations in circRNAs expression in various BCs, the functional pathways, their impact on the condition, and their uses in clinical applications.
Collapse
Affiliation(s)
- Nafiseh Tashakori
- Department of Medicine, Faculty of Internal Medicine,Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Maria V Mikhailova
- Department of Prosthetic Dentistry, I.M. Schenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | | | | | | | - Usama Kadem Radi
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar 64001, Iraq
| | | | - Bahman Abedi Kiasari
- Microbiology & Immunology Group, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| |
Collapse
|
9
|
Li Y, Zhang S. Statistical batch-aware embedded integration, dimension reduction, and alignment for spatial transcriptomics. Bioinformatics 2024; 40:btae611. [PMID: 39400541 PMCID: PMC11512591 DOI: 10.1093/bioinformatics/btae611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 09/03/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024] Open
Abstract
MOTIVATION Spatial transcriptomics (ST) technologies provide richer insights into the molecular characteristics of cells by simultaneously measuring gene expression profiles and their relative locations. However, each slice can only contain limited biological variation, and since there are almost always non-negligible batch effects across different slices, integrating numerous slices to account for batch effects and locations is not straightforward. Performing multi-slice integration, dimensionality reduction, and other downstream analyses separately often results in suboptimal embeddings for technical artifacts and biological variations. Joint modeling integrating these steps can enhance our understanding of the complex interplay between technical artifacts and biological signals, leading to more accurate and insightful results. RESULTS In this context, we propose a hierarchical hidden Markov random field model STADIA to reduce batch effects, extract common biological patterns across multiple ST slices, and simultaneously identify spatial domains. We demonstrate the effectiveness of STADIA using five datasets from different species (human and mouse), various organs (brain, skin, and liver), and diverse platforms (10x Visium, ST, and Slice-seqV2). STADIA can capture common tissue structures across multiple slices and preserve slice-specific biological signals. In addition, STADIA outperforms the other three competing methods (PRECAST, fastMNN, and Harmony) in terms of the balance between batch mixing and spatial domain identification, and it demonstrates the advantage of joint modeling when compared to STAGATE and GraphST. AVAILABILITY AND IMPLEMENTATION The source code implemented by R is available at https://github.com/zhanglabtools/STADIA and archived with version 1.01 on Zenodo https://zenodo.org/records/13637744.
Collapse
Affiliation(s)
- Yanfang Li
- NCMIS, CEMS, RCSDS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing 100190, China
| | - Shihua Zhang
- NCMIS, CEMS, RCSDS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing 100190, China
- School of Mathematical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| |
Collapse
|
10
|
Wang X, Xing Y, Zhou X, Wang C, Han S, Zhao S. Radiomics Signatures Based on Computed Tomography for Noninvasive Prediction of CXCL10 Expression and Prognosis in Ovarian Cancer. Cancer Rep (Hoboken) 2024; 7:e70030. [PMID: 39443817 PMCID: PMC11499071 DOI: 10.1002/cnr2.70030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 08/28/2024] [Accepted: 09/17/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Ovarian cancer (OC) is an aggressive gynecological tumor usually diagnosed with malignant ascites and even observed widespread metastasis or distant spread. AIMS We aimed to develop and identify radiomics models according to computed tomography (CT) for preoperative prediction of CXCL10 expression and prognosis in patients with OC. METHODS Genomic data with CT images and corresponding clinicopathological parameters were extracted from The Cancer Imaging Archive (TCIA) and The Cancer Genome Atlas (TCGA). To analyze the prognosis, we carried out the univariate Cox regression analysis (UCRA), multivariate Cox regression analysis (MCRA), and Kaplan-Meier (KM) analysis. For the data reduction, logistic regression, operator regression, least absolute shrinkage selection, radiomic feature construction, and feature selection were utilized. The predictive performance of the radiomic signatures was assessed using the analyses of the receiver operating characteristic (ROC) curve, decision curve (DCA), and precision-recall (PR) curve. To evaluate the correlation between the radiomic score (Rad-score) and CXCL10 expression, the Wilcoxon rank-sum test was applied. RESULTS Three radiomics models effectively predicted CXCL10 expression levels (AUC = 0.791, 0.748, and 0.718 for the set of training; AUC = 0.761, 0.746, and 0.701 for the set of validation). A higher Rad-score significantly correlated with upregulated CXCL10 expression. CONCLUSION CXCL10 expression can be predicted noninvasively and preoperatively via radiomic signatures based on contrast-enhanced CT images.
Collapse
Affiliation(s)
- Xiaohua Wang
- Department of Gynecology and Obstetrics, Department of GynecologyThe Second Hospital of HeBei Medical University, Affiliated Hospital of Chengde Medical UniversityShijiazhuangChina
| | - Yuanyuan Xing
- Department of Nuclear MedicineAffiliated Hospital of Chengde Medical UniversityChengdeChina
| | - Xuan Zhou
- Department of GynecologyAffiliated Hospital of Chengde Medical UniversityChengdeChina
| | - Chunhui Wang
- Department of GynecologyAffiliated Hospital of Chengde Medical UniversityChengdeChina
| | - Shuyu Han
- Department of GynecologyAffiliated Hospital of Chengde Medical UniversityChengdeChina
| | - Sufen Zhao
- Department of Gynecology and ObstetricsThe Second Hospital of HeBei Medical UniversityShijiazhuangChina
| |
Collapse
|
11
|
Hewes JL, Bhadra A, Schreck E, Goodman JT, Patel M, Zhou C, Lee JY, Bauer NR. Novel Hemodynamic, Vascular Lesion, and Cytokine/Chemokine Differences Regarding Sex in a Pulmonary Arterial Hypertension Model. Am J Respir Cell Mol Biol 2024; 71:453-463. [PMID: 38864769 PMCID: PMC11450312 DOI: 10.1165/rcmb.2023-0378oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 06/12/2024] [Indexed: 06/13/2024] Open
Abstract
Sex differences are recognized in pulmonary hypertension. However, the progression of disease with regard to vascular lesion formation and circulating cytokines/chemokines is unknown. To determine whether vascular lesion formation, changes in hemodynamics, and alterations in circulating chemokines/cytokines differ between males and females, we used a progressive model of pulmonary arterial hypertension (PAH), Sugen/hypoxia, and analyzed cohorts of male and female rats at time points suggested to indicate worsening disease. Our analysis included echocardiography for hemodynamics, morphometry, immunofluoresecence, and chemokine/cytokine analysis of plasma at each time point in both sexes. We found that male rats had significantly increased Fulton index, compared with those for females at each time point, as well as increased medial artery thickening at 8 weeks of PAH. Furthermore, females exhibited fewer obliterative vascular lesions than males at our latest time point. Our data also show increased IL-4, granulocyte-macrophage colony-stimulating factor, IL-10, and macrophage interacting protein-1α that were not observed in females, whereas females were observed to have increased RANTES (whose name derives from Regulated upon Activation, Normal T Cell Expressed and Presumably Secreted) and CXCL-10 that were not found in males. Males also have increased infiltrating macrophages in vascular lesions, compared with females. We found that development of progressive PAH in hemodynamics, morphology, and chemokine/cytokine circulation differs significantly between males and females. These data suggest a macrophage-driven pathology in males, whereas there may be T cell protection from vascular damage in females with PAH.
Collapse
Affiliation(s)
| | | | | | | | | | - Chun Zhou
- Center for Lung Biology
- Department of Physiology and Cell Biology, and
| | - Ji Young Lee
- Center for Lung Biology
- Department of Physiology and Cell Biology, and
- Pulmonary and Critical Care Medicine, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, Alabama
| | | |
Collapse
|
12
|
Ahmadi N, Ahmadi SAY, Kheirollahi A, Shahsavar F. Investigating the Relationship of G-137C, C-607A, and A-1447G Polymorphisms in the Promoter of IL-18 and CXCL10 Inflammatory Genes with Prostate Cancer in an Iranian Population. Prostate Cancer 2024; 2024:3997576. [PMID: 39359425 PMCID: PMC11446609 DOI: 10.1155/2024/3997576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 07/22/2024] [Accepted: 09/09/2024] [Indexed: 10/04/2024] Open
Abstract
Introduction Genetic and environmental factors are involved in prostate cancer. The current study was conducted to study the relationship between G-137C, C-607A, and A-1447G polymorphisms in the promoter of IL-18 and CXCL10 inflammatory genes with prostate cancer. Methods As a genetic association study with a case-control design, the genomes of people living in Khorasan, Iran, were compared in two groups of cases and controls. The genotype of the A-1447G polymorphism present in the CXCL10 gene promoter was investigated by the PCR-RFLP method. PCR-SSP was used to study the genotype of G-137C and C-607A polymorphisms present in the IL-18 gene promoter. Odds ratio (OR) and 95% confidence interval (CI) were reported. Results One mutant allele in CXCL10 A-1447G polymorphism (AG) increased the chance of cancer (OR = 4.902, 95% CI = 2.70-8.87) and two mutant alleles (GG) increased more (OR = 7.174, 95% CI = 2.48-20.68). One mutant allele in IL-18 G-137C polymorphism (CG) increased the chance of cancer (OR = 5.583, 95% CI = 3.04-10.22) and two mutant alleles (CC) increased more (OR = 9.571, 95% CI = 3.10-29.46). One mutant allele in IL-18 C607A polymorphism (CA) increased the chance of cancer (OR = 5.359, 95% CI = 2.95-9.70) and two mutant alleles (AA) increased more (OR = 7.083, 95% CI = 2.61-19.15) (P < 0.001). Conclusion According to the results, the mutant alleles in polymorphisms CXCL10 A-1447G, IL-18 G-137C, and IL-18 C-607A alleles were associated with an increased chance of prostate cancer in this population.
Collapse
Affiliation(s)
- Nahid Ahmadi
- Student Research Committee Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Seyyed Amir Yasin Ahmadi
- Preventive Medicine and Public Health Research Center Psychosocial Health Research Institute Iran University of Medical Sciences, Tehran, Iran
| | - Abdolreza Kheirollahi
- Hepatitis Research Center School of Medicine Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Farhad Shahsavar
- Hepatitis Research Center School of Medicine Lorestan University of Medical Sciences, Khorramabad, Iran
| |
Collapse
|
13
|
Kim B, Park YY, Lee JH. CXCL10 promotes melanoma angiogenesis and tumor growth. Anim Cells Syst (Seoul) 2024; 28:453-465. [PMID: 39268223 PMCID: PMC11391877 DOI: 10.1080/19768354.2024.2402024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/24/2024] [Accepted: 09/01/2024] [Indexed: 09/15/2024] Open
Abstract
Upregulation of CXC motif chemokine 10 (CXCL10) in melanoma patients has been found to be associated with melanoma progression. However, the role of endogenous CXCL10 from the host in melanoma tumor growth remains unclear. In the present study, we found that host-derived endogenous CXCL10 production was dramatically augmented during subcutaneous B16F10 melanoma tumor growth and that host ablation of CXCL10 in Cxcl10-/- mice showed a decrease in both angiogenesis and tumor growth of B16F10 melanoma in vivo. Several signaling pathways involved in production of pro-angiogenic factors and tumor growth were activated by CXCL10 in B16F10 melanoma cells. CXCL10 increased expression of pro-angiogenic factors, such as vascular endothelial growth factor (VEGF), platelet-derived growth factor subunit-B (PDGF-B), fibroblast growth factor 2 (FGF2), hepatocyte growth factor (HGF), and angiopoietin 2 (Angpt2), in B16F10 melanoma cells, resulting in enhanced tube formation and proliferation of human umbilical vein endothelial cells in vitro. In addition, CXCL10 directly enhanced B16F10 melanoma tumor growth in an in vitro three-dimensional cell culture system. Together, our findings reveal that amplified host-derived endogenous CXCL10 is critical for B16F10 melanoma angiogenesis and tumor growth. Therefore, CXCL10 might represent a therapeutic target for melanoma.
Collapse
Affiliation(s)
- Bongjun Kim
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yun-Yong Park
- Department of life Science, Chung-Ang University, Seoul, Republic of Korea
| | - Jong-Ho Lee
- Department of Health Sciences, The Graduate School of Dong-A University, Busan, Republic of Korea
- Department of Biomedical Sciences, Dong-A University, Busan, Republic of Korea
| |
Collapse
|
14
|
Bergkamp SC, Bergkamp ND, Wahadat MJ, Gruppen MP, Nassar-Sheikh Rashid A, Tas SW, Smit MJ, Versnel MA, van den Berg JM, Kamphuis S, Schonenberg-Meinema D. Learning from serum markers reflecting endothelial activation: longitudinal data in childhood-onset systemic lupus erythematosus. Lupus Sci Med 2024; 11:e001190. [PMID: 39242108 PMCID: PMC11381702 DOI: 10.1136/lupus-2024-001190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 08/04/2024] [Indexed: 09/09/2024]
Abstract
OBJECTIVES In childhood-onset SLE (cSLE), patients have an increased risk of premature atherosclerosis. The pathophysiological mechanisms for this premature atherosclerosis are not yet completely understood, but besides traditional risk factors, the endothelium plays a major role. The first aim of this study was to measure levels of SLE-associated markers involved in endothelial cell (EC) function and lipids in a cSLE cohort longitudinally in comparison with healthy controls (HC). Next aim was to correlate these levels with Systemic Lupus Erythematosus Disease Activity Index (SLEDAI) and nailfold capillaroscopic patterns. METHODS Blood serum samples, videocapillaroscopy images and patient characteristics were collected in a multicentre longitudinal cSLE cohort and from age and sex comparable HC. Disease activity was evaluated by SLEDAI. A total of 15 EC markers and six lipids were measured in two longitudinal cSLE samples (minimum interval of 6 months) and in HC. Nailfold videocapillaroscopy images were scored according to the guidelines from the EULAR Study Group on Microcirculation in Rheumatic Diseases. RESULTS In total, 47 patients with cSLE and 42 HCs were analysed. Median age at diagnosis was 15 years (IQR 12-16 years). Median time between t=1 and t=2 was 14.5 months (IQR 9-24 months). Median SLEDAI was 12 (IQR 6-18) at t=1 and 2 (IQR 1-4) at t=2. Serum levels of angiopoietin-2, CCL2, CXCL10, GAS6, pentraxin-3, thrombomodulin, VCAM-1 and vWF-A2 were elevated in cSLE compared with HC at t=1. While many elevated EC markers at t=1 normalised over time after treatment, several markers remained significantly increased compared with HC (angiopoietin-2, CCL2, CXCL10, GAS6, thrombomodulin and VCAM-1). CONCLUSION In serum from patients with cSLE different markers of endothelial activation were dysregulated. While most markers normalised during treatment, others remained elevated in a subset of patients, even during low disease activity. These results suggest a role for the dysregulated endothelium in early and later phases of cSLE, possibly also during lower disease activity. TRIAL REGISTRATION NUMBER NL60885.018.17.
Collapse
Affiliation(s)
- Sandy C Bergkamp
- Department of Paediatric Immunology, Rheumatology and Infectious Diseases, Emma Children’s Hospital, Amsterdam University Medical Centres (AUMC), University of Amsterdam, Amsterdam, The Netherlands
| | - Nick D Bergkamp
- Amsterdam Institute for Molecular and Life Sciences (AIMMS), Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Mohamed Javad Wahadat
- Department of Paediatric Rheumatology, Sophia Children’s Hospital, Erasmus University Medical Centre, Rotterdam, The Netherlands
- Department of Immunology, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Mariken P Gruppen
- Department of Paediatric Immunology, Rheumatology and Infectious Diseases, Emma Children’s Hospital, Amsterdam University Medical Centres (AUMC), University of Amsterdam, Amsterdam, The Netherlands
| | - Amara Nassar-Sheikh Rashid
- Department of Paediatric Immunology, Rheumatology and Infectious Diseases, Emma Children’s Hospital, Amsterdam University Medical Centres (AUMC), University of Amsterdam, Amsterdam, The Netherlands
- Department of Paediatrics, Zaans Medisch Centrum, Zaandam, The Netherlands
| | - Sander W Tas
- Amsterdam Rheumatology and Immunology Centre, Department of Rheumatology and Clinical Immunology, and Laboratory for Experimental Immunology, Amsterdam University Medical Centres (AUMC), University of Amsterdam, Amsterdam, The Netherlands
| | - Martine J Smit
- Amsterdam Institute for Molecular and Life Sciences (AIMMS), Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Marjan A Versnel
- Department of Immunology, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - J Merlijn van den Berg
- Department of Paediatric Immunology, Rheumatology and Infectious Diseases, Emma Children’s Hospital, Amsterdam University Medical Centres (AUMC), University of Amsterdam, Amsterdam, The Netherlands
| | - Sylvia Kamphuis
- Department of Paediatric Rheumatology, Sophia Children’s Hospital, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Dieneke Schonenberg-Meinema
- Department of Paediatric Immunology, Rheumatology and Infectious Diseases, Emma Children’s Hospital, Amsterdam University Medical Centres (AUMC), University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
15
|
Shen L, Schaefer AM, Tiruthani K, Wolf W, Lai SK. Siglec15/TGF-β bispecific antibody mediates synergistic anti-tumor response against 4T1 triple negative breast cancer in mice. Bioeng Transl Med 2024; 9:e10651. [PMID: 39553435 PMCID: PMC11561775 DOI: 10.1002/btm2.10651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 01/10/2024] [Accepted: 01/19/2024] [Indexed: 11/19/2024] Open
Abstract
An ideal tumor-specific immunomodulatory therapy should both preferentially target the tumor, while simultaneously reduce the immunosuppressive environment within the tumor. This guiding principle led us to explore engineering Siglec-15 (S15) targeted bispecific antibody (bsAb) to enhance therapy against triple negative breast cancer (TNBC). S15 appears to be exclusively expressed on macrophages and diverse tumor cells, including human and mouse 4T1 TNBC. TGF-β is a growth hormone frequently associated with increased tumor invasiveness, including in TNBC. Here, to overcome the immune-suppressive environment within TNBC tumors to enable more effective cancer therapy, we engineered a bispecific antibody (bsAb) targeting both Siglec15 and TGF-β. In mice engrafted with orthotopic 4T1 tumors, S15/TGF-β bsAb treatment was highly effective in suppressing tumor growth, not only compared to control monoclonal antibody (mAb) but also markedly more effective than mAbs against S15 alone, against TGF-β alone, as well as a cocktail of both anti-S15 and anti-TGF-β mAbs. We did not detect liver and lung metastasis in mice treated with S15/TGF-β bsAb, unlike all other treatment groups at the end of the study. The enhanced anti-tumor response observed with S15/TGF-β bsAb correlated with a less immunosuppressive environment in the tumor. These results underscore S15-targeted bsAb as a promising therapeutic strategy for TNBC, and possibly other S15 positive solid tumors.
Collapse
Affiliation(s)
- Limei Shen
- Division of Pharmacoengineering and Molecular PharmaceuticsEshelman School of Pharmacy, University of North CarolinaChapel HillNorth CarolinaUSA
| | - Alison M. Schaefer
- Department of Biomedical EngineeringUniversity of North CarolinaChapel HillNorth CarolinaUSA
| | - Karthik Tiruthani
- Division of Pharmacoengineering and Molecular PharmaceuticsEshelman School of Pharmacy, University of North CarolinaChapel HillNorth CarolinaUSA
| | - Whitney Wolf
- Division of Pharmacoengineering and Molecular PharmaceuticsEshelman School of Pharmacy, University of North CarolinaChapel HillNorth CarolinaUSA
| | - Samuel K. Lai
- Division of Pharmacoengineering and Molecular PharmaceuticsEshelman School of Pharmacy, University of North CarolinaChapel HillNorth CarolinaUSA
- Department of Biomedical EngineeringUniversity of North CarolinaChapel HillNorth CarolinaUSA
- Department of Immunology and MicrobiologyUniversity of North CarolinaChapel HillNorth CarolinaUSA
| |
Collapse
|
16
|
Mahboobnia K, Beveridge DJ, Yeoh GC, Kabir TD, Leedman PJ. MicroRNAs in Hepatocellular Carcinoma Pathogenesis: Insights into Mechanisms and Therapeutic Opportunities. Int J Mol Sci 2024; 25:9393. [PMID: 39273339 PMCID: PMC11395074 DOI: 10.3390/ijms25179393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 08/18/2024] [Accepted: 08/20/2024] [Indexed: 09/15/2024] Open
Abstract
Hepatocellular carcinoma (HCC) presents a significant global health burden, with alarming statistics revealing its rising incidence and high mortality rates. Despite advances in medical care, HCC treatment remains challenging due to late-stage diagnosis, limited effective therapeutic options, tumor heterogeneity, and drug resistance. MicroRNAs (miRNAs) have attracted substantial attention as key regulators of HCC pathogenesis. These small non-coding RNA molecules play pivotal roles in modulating gene expression, implicated in various cellular processes relevant to cancer development. Understanding the intricate network of miRNA-mediated molecular pathways in HCC is essential for unraveling the complex mechanisms underlying hepatocarcinogenesis and developing novel therapeutic approaches. This manuscript aims to provide a comprehensive review of recent experimental and clinical discoveries regarding the complex role of miRNAs in influencing the key hallmarks of HCC, as well as their promising clinical utility as potential therapeutic targets.
Collapse
Affiliation(s)
- Khadijeh Mahboobnia
- Laboratory for Cancer Medicine, Harry Perkins Institute of Medical Research, QEII Medical Centre, Perth, WA 6009, Australia
- Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia
| | - Dianne J Beveridge
- Laboratory for Cancer Medicine, Harry Perkins Institute of Medical Research, QEII Medical Centre, Perth, WA 6009, Australia
- Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia
| | - George C Yeoh
- Laboratory for Cancer Medicine, Harry Perkins Institute of Medical Research, QEII Medical Centre, Perth, WA 6009, Australia
- School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Tasnuva D Kabir
- Laboratory for Cancer Medicine, Harry Perkins Institute of Medical Research, QEII Medical Centre, Perth, WA 6009, Australia
- Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia
| | - Peter J Leedman
- Laboratory for Cancer Medicine, Harry Perkins Institute of Medical Research, QEII Medical Centre, Perth, WA 6009, Australia
- Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia
| |
Collapse
|
17
|
Otálora-Otálora BA, Payán-Gómez C, López-Rivera JJ, Pedroza-Aconcha NB, Aristizábal-Guzmán C, Isaza-Ruget MA, Álvarez-Moreno CA. Global transcriptomic network analysis of the crosstalk between microbiota and cancer-related cells in the oral-gut-lung axis. Front Cell Infect Microbiol 2024; 14:1425388. [PMID: 39228892 PMCID: PMC11368877 DOI: 10.3389/fcimb.2024.1425388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/15/2024] [Indexed: 09/05/2024] Open
Abstract
Background The diagnosis and treatment of lung, colon, and gastric cancer through the histologic characteristics and genomic biomarkers have not had a strong impact on the mortality rates of the top three global causes of death by cancer. Methods Twenty-five transcriptomic analyses (10 lung cancer, 10 gastric cancer, and 5 colon cancer datasets) followed our own bioinformatic pipeline based on the utilization of specialized libraries from the R language and DAVID´s gene enrichment analyses to identify a regulatory metafirm network of transcription factors and target genes common in every type of cancer, with experimental evidence that supports its relationship with the unlocking of cell phenotypic plasticity for the acquisition of the hallmarks of cancer during the tumoral process. The network's regulatory functional and signaling pathways might depend on the constant crosstalk with the microbiome network established in the oral-gut-lung axis. Results The global transcriptomic network analysis highlighted the impact of transcription factors (SOX4, TCF3, TEAD4, ETV4, and FOXM1) that might be related to stem cell programming and cancer progression through the regulation of the expression of genes, such as cancer-cell membrane receptors, that interact with several microorganisms, including human T-cell leukemia virus 1 (HTLV-1), the human papilloma virus (HPV), the Epstein-Barr virus (EBV), and SARS-CoV-2. These interactions can trigger the MAPK, non-canonical WNT, and IFN signaling pathways, which regulate key transcription factor overexpression during the establishment and progression of lung, colon, and gastric cancer, respectively, along with the formation of the microbiome network. Conclusion The global transcriptomic network analysis highlights the important interaction between key transcription factors in lung, colon, and gastric cancer, which regulates the expression of cancer-cell membrane receptors for the interaction with the microbiome network during the tumorigenic process.
Collapse
Affiliation(s)
| | - César Payán-Gómez
- Dirección Académica, Universidad Nacional de Colombia, Sede de La Paz, La Paz, Colombia
| | - Juan Javier López-Rivera
- Grupo de Investigación INPAC, Specialized Laboratory, Clinica Universitaria Colombia, Clínica Colsanitas S.A., Bogotá, Colombia
| | | | - Claudia Aristizábal-Guzmán
- Grupo de Investigación INPAC, Unidad de Investigación, Fundación Universitaria Sanitas, Bogotá, Colombia
| | - Mario Arturo Isaza-Ruget
- Keralty, Sanitas International Organization, Grupo de Investigación INPAC, Fundación Universitaria Sanitas, Bogotá, Colombia
| | | |
Collapse
|
18
|
Wang HR, Zhang Y, Mo YJ, Zhang Z, Chen R, Lu XB, Huang W. Reshaping tumor microenvironment by regulating local cytokines expression with a portable smart blue-light controlled device. Commun Biol 2024; 7:916. [PMID: 39080467 PMCID: PMC11289142 DOI: 10.1038/s42003-024-06566-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 07/05/2024] [Indexed: 08/02/2024] Open
Abstract
Cytokines have attracted sustained attention due to their multi-functional cellular response in immunotherapy. However, their application was limited to their short half-time, narrow therapeutic window, and undesired side effects. To address this issue, we developed a portable smart blue-light controlled (PSLC) device based on optogenetic technology. By combining this PSLC device with blue-light controlled gene modules, we successfully achieved the targeted regulation of cytokine expression within the tumor microenvironment. To alter the tumor microenvironment of solid tumors, pro-inflammatory cytokines were selected as blue-light controlled molecules. The results show that blue-light effectively regulates the expression of pro-inflammatory cytokines both in vitro and in vivo. This strategy leads to enhanced and activated tumor-infiltrating immune cells, which facilitated to overcome the immunosuppressive microenvironment, resulting in significant tumor shrinkage in tumor-bearing mice. Hence, our study offers a unique strategy for cytokine therapy and a convenient device for animal studies in optogenetic immunotherapy.
Collapse
Affiliation(s)
- Hui Rong Wang
- LiShizhen College of Traditional Chinese Medicine, Huanggang Normal University, Huanggang, Hubei, China.
- Center for Cell and Gene Circuit Design, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China.
- Department of Biology, School of Life Science, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Yi Zhang
- Department of Biology, Brandeis University, Waltham, MA, USA
| | - Yue Jian Mo
- Center for Cell and Gene Circuit Design, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
- Department of Biology, School of Life Science, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Zhan Zhang
- Department of Biology, School of Life Science, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Rui Chen
- Department of Biology, School of Life Science, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Xi Bin Lu
- Department of Biology, School of Life Science, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Wei Huang
- Center for Cell and Gene Circuit Design, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China.
- Department of Biology, School of Life Science, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| |
Collapse
|
19
|
Aloliqi AA. Insights into the Gene Expression Profile of Classical Hodgkin Lymphoma: A Study towards Discovery of Novel Therapeutic Targets. Molecules 2024; 29:3476. [PMID: 39124881 PMCID: PMC11314437 DOI: 10.3390/molecules29153476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Classical Hodgkin lymphoma (cHL) is a common B-cell cancer and a significant health concern, especially in Western and Asian countries. Despite the effectiveness of chemotherapy, many relapse cases are being reported, highlighting the need for improved treatments. This study aimed to address this issue by discovering biomarkers through the analysis of gene expression data specific to cHL. Additionally, potential anticancer inhibitors were explored to target the discovered biomarkers. This study proceeded by retrieving microarray gene expression data from cHL patients, which was then analyzed to identify significant differentially expressed genes (DEGs). Functional and network annotation of the upregulated genes revealed the active involvement of matrix metallopeptidase 12 (MMP12) and C-C motif metallopeptidase ligand 22 (CCL22) genes in the progression of cHL. Additionally, the mentioned genes were found to be actively involved in cancer-related pathways, i.e., oxidative phosphorylation, complement pathway, myc_targets_v1 pathway, TNFA signaling via NFKB, etc., and showed strong associations with other genes known to promote cancer progression. MMP12, topping the list with a logFC value of +6.6378, was selected for inhibition using docking and simulation strategies. The known anticancer compounds were docked into the active site of the MMP12 molecular structure, revealing significant binding scores of -7.7 kcal/mol and -7.6 kcal/mol for BDC_24037121 and BDC_27854277, respectively. Simulation studies of the docked complexes further supported the effective binding of the ligands, yielding MMGBSA and MMPBSA scores of -78.08 kcal/mol and -82.05 kcal/mol for MMP12-BDC_24037121 and -48.79 kcal/mol and -49.67 kcal/mol for MMP12-BDC_27854277, respectively. Our findings highlight the active role of MMP12 in the progression of cHL, with known compounds effectively inhibiting its function and potentially halting the advancement of cHL. Further exploration of downregulated genes is warranted, as associated genes may play a role in cHL. Additionally, CCL22 should be considered for further investigation due to its significant role in the progression of cHL.
Collapse
Affiliation(s)
- Abdulaziz A Aloliqi
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah 52571, Saudi Arabia
| |
Collapse
|
20
|
Suek N, Young T, Fu J. Immune cell profiling in intestinal transplantation. Hum Immunol 2024; 85:110808. [PMID: 38762429 PMCID: PMC11283363 DOI: 10.1016/j.humimm.2024.110808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 04/08/2024] [Accepted: 04/25/2024] [Indexed: 05/20/2024]
Abstract
Since the first published case study of human intestinal transplantation in 1967, there have been significant studies of intestinal transplant immunology in both animal models and humans. An improved understanding of the profiles of different immune cell subsets is critical for understanding their contributions to graft outcomes. While different studies have focused on the contribution of one or a few subsets to intestinal transplant, no study has integrated these data for a comprehensive overview of immune dynamics after intestinal transplant. Here, we provide a systematic review of the literature on different immune subsets and discuss their roles in intestinal transplant outcomes on multiple levels, focusing on chimerism and graft immune reconstitution, clonal alloreactivity, and cell phenotype. In Sections 1, 2 and 3, we lay out a shared framework for understanding intestinal transplant, focusing on the mechanisms of rejection or tolerance in the context of mucosal immunology and illustrate the unique role of the bidirectional graft-versus-host (GvH) and host-versus-graft (HvG) alloresponse. In Sections 4, 5 and 6, we further expand upon these concepts as we discuss the contribution of different cell subsets to intestinal transplant. An improved understanding of intestinal transplantation immunology will bring us closer to maximizing the potential of this important treatment.
Collapse
Affiliation(s)
- Nathan Suek
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Tyla Young
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Jianing Fu
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA.
| |
Collapse
|
21
|
Kocher D, Cao L, Guiho R, Langhammer M, Lai YL, Becker P, Hamdi H, Friedel D, Selt F, Vonhören D, Zaman J, Valinciute G, Herter S, Picard D, Rettenmeier J, Maass KK, Pajtler KW, Remke M, von Deimling A, Pusch S, Pfister SM, Oehme I, Jones DTW, Halbach S, Brummer T, Martinez-Barbera JP, Witt O, Milde T, Sigaud R. Rebound growth of BRAF mutant pediatric glioma cells after MAPKi withdrawal is associated with MAPK reactivation and secretion of microglia-recruiting cytokines. J Neurooncol 2024; 168:317-332. [PMID: 38630384 PMCID: PMC11147834 DOI: 10.1007/s11060-024-04672-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 03/28/2024] [Indexed: 06/04/2024]
Abstract
INTRODUCTION Patients with pediatric low-grade gliomas (pLGGs), the most common primary brain tumors in children, can often benefit from MAPK inhibitor (MAPKi) treatment. However, rapid tumor regrowth, also referred to as rebound growth, may occur once treatment is stopped, constituting a significant clinical challenge. METHODS Four patient-derived pediatric glioma models were investigated to model rebound growth in vitro based on viable cell counts in response to MAPKi treatment and withdrawal. A multi-omics dataset (RNA sequencing and LC-MS/MS based phospho-/proteomics) was generated to investigate possible rebound-driving mechanisms. Following in vitro validation, putative rebound-driving mechanisms were validated in vivo using the BT-40 orthotopic xenograft model. RESULTS Of the tested models, only a BRAFV600E-driven model (BT-40, with additional CDKN2A/Bdel) showed rebound growth upon MAPKi withdrawal. Using this model, we identified a rapid reactivation of the MAPK pathway upon MAPKi withdrawal in vitro, also confirmed in vivo. Furthermore, transient overactivation of key MAPK molecules at transcriptional (e.g. FOS) and phosphorylation (e.g. pMEK) levels, was observed in vitro. Additionally, we detected increased expression and secretion of cytokines (CCL2, CX3CL1, CXCL10 and CCL7) upon MAPKi treatment, maintained during early withdrawal. While increased cytokine expression did not have tumor cell intrinsic effects, presence of these cytokines in conditioned media led to increased attraction of microglia cells in vitro. CONCLUSION Taken together, these data indicate rapid MAPK reactivation upon MAPKi withdrawal as a tumor cell intrinsic rebound-driving mechanism. Furthermore, increased secretion of microglia-recruiting cytokines may play a role in treatment response and rebound growth upon withdrawal, warranting further evaluation.
Collapse
Affiliation(s)
- Daniela Kocher
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Research Center (DKFZ) Heidelberg, Clinical Cooperation Unit Pediatric Oncology, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Lei Cao
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, WC1N 1EH, London, UK
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Romain Guiho
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, WC1N 1EH, London, UK
- Nantes Université, Oniris, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000 Nantes, France
| | - Melanie Langhammer
- Institute of Molecular Medicine and Cell Research (IMMZ), Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Yun-Lu Lai
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Research Center (DKFZ) Heidelberg, Clinical Cooperation Unit Pediatric Oncology, Heidelberg, Germany
| | - Pauline Becker
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Research Center (DKFZ) Heidelberg, Clinical Cooperation Unit Pediatric Oncology, Heidelberg, Germany
- Faculty of Medicine, Heidelberg University, Heidelberg, Germany
| | - Hiba Hamdi
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, WC1N 1EH, London, UK
| | - Dennis Friedel
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Department of Neuropathology, Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Clinical Cooperation Unit Neuropathology, Heidelberg, Germany
| | - Florian Selt
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Research Center (DKFZ) Heidelberg, Clinical Cooperation Unit Pediatric Oncology, Heidelberg, Germany
- KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - David Vonhören
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Department of Neuropathology, Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Clinical Cooperation Unit Neuropathology, Heidelberg, Germany
| | - Julia Zaman
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Department of Neuropathology, Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Clinical Cooperation Unit Neuropathology, Heidelberg, Germany
| | - Gintvile Valinciute
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Research Center (DKFZ) Heidelberg, Clinical Cooperation Unit Pediatric Oncology, Heidelberg, Germany
| | - Sonja Herter
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Research Center (DKFZ) Heidelberg, Clinical Cooperation Unit Pediatric Oncology, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Daniel Picard
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
- German Cancer Consortium (DKTK), Partner site Essen/Düsseldorf, Düsseldorf, Germany
- Institute of Neuropathology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Johanna Rettenmeier
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Faculty of Medicine, Heidelberg University, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Division of Pediatric Neurooncology, Heidelberg, Germany
- Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
| | - Kendra K Maass
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Division of Pediatric Neurooncology, Heidelberg, Germany
- Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
| | - Kristian W Pajtler
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Division of Pediatric Neurooncology, Heidelberg, Germany
- Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
| | - Marc Remke
- Pediatric Hematology and Oncology, University Children's Hospital, Saarland University, Homburg, Germany
| | - Andreas von Deimling
- Department of Neuropathology, Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Clinical Cooperation Unit Neuropathology, Heidelberg, Germany
| | - Stefan Pusch
- Department of Neuropathology, Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Clinical Cooperation Unit Neuropathology, Heidelberg, Germany
| | - Stefan M Pfister
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Division of Pediatric Neurooncology, Heidelberg, Germany
| | - Ina Oehme
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Research Center (DKFZ) Heidelberg, Clinical Cooperation Unit Pediatric Oncology, Heidelberg, Germany
| | - David T W Jones
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Research Center (DKFZ) Heidelberg, Division of Pediatric Glioma Research, Heidelberg, Germany
| | - Sebastian Halbach
- Institute of Molecular Medicine and Cell Research (IMMZ), Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Consortium for Translational Cancer Research (DKTK), Freiburg, Germany, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tilman Brummer
- Institute of Molecular Medicine and Cell Research (IMMZ), Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Consortium for Translational Cancer Research (DKTK), Freiburg, Germany, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Centre for Biological Signaling Studies BIOSS, University of Freiburg, Freiburg, Germany
| | - Juan Pedro Martinez-Barbera
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, WC1N 1EH, London, UK
| | - Olaf Witt
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Research Center (DKFZ) Heidelberg, Clinical Cooperation Unit Pediatric Oncology, Heidelberg, Germany
- KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Till Milde
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany.
- German Cancer Research Center (DKFZ) Heidelberg, Clinical Cooperation Unit Pediatric Oncology, Heidelberg, Germany.
- KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany.
| | - Romain Sigaud
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany.
- German Cancer Research Center (DKFZ) Heidelberg, Clinical Cooperation Unit Pediatric Oncology, Heidelberg, Germany.
| |
Collapse
|
22
|
Cyberski TF, Singh A, Korzinkin M, Mishra V, Pun F, Shen L, Wing C, Cheng X, Baird B, Miao Y, Elkabets M, Kochanny S, Guo W, Dyer E, Pearson AT, Juloori A, Lingen M, Cole G, Zhavoronkov A, Agrawal N, Izumchenko E, Rosenberg AJ. Acquired resistance to immunotherapy and chemoradiation in MYC amplified head and neck cancer. NPJ Precis Oncol 2024; 8:114. [PMID: 38783041 PMCID: PMC11116544 DOI: 10.1038/s41698-024-00606-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
The proto-oncogene MYC encodes a nuclear transcription factor that has an important role in a variety of cellular processes, such as cell cycle progression, proliferation, metabolism, adhesion, apoptosis, and therapeutic resistance. MYC amplification is consistently observed in aggressive forms of several solid malignancies and correlates with poor prognosis and distant metastases. While the tumorigenic effects of MYC in patients with head and neck squamous cell carcinoma (HNSCC) are well known, the molecular mechanisms by which the amplification of this gene may confer treatment resistance, especially to immune checkpoint inhibitors, remains under-investigated. Here we present a unique case of a patient with recurrent/metastatic (R/M) HNSCC who, despite initial response to nivolumab-based treatment, developed rapidly progressive metastatic disease after the acquisition of MYC amplification. We conducted comparative transcriptomic analysis of this patient's tumor at baseline and upon progression to interrogate potential molecular processes through which MYC may confer resistance to immunotherapy and/or chemoradiation and used TCGA-HNSC dataset and an institutional cohort to further explore clinicopathologic features and key molecular networks associated with MYC amplification in HNSCC. This study highlights MYC amplification as a potential mechanism of immune checkpoint inhibitor resistance and suggest its use as a predictive biomarker and potential therapeutic target in R/M HNSCC.
Collapse
Affiliation(s)
- Thomas F Cyberski
- Pritzker School of Medicine, University of Chicago, Chicago, IL, USA
| | - Alka Singh
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, USA
| | | | - Vasudha Mishra
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, USA
| | - Frank Pun
- Insilico Medicine, Pak Shek Kok, Hong Kong
| | - Le Shen
- Department of Surgery, University of Chicago, Chicago, IL, USA
| | - Claudia Wing
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, USA
| | - Xiangying Cheng
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, USA
| | - Brandon Baird
- Department of Surgery, University of Chicago, Chicago, IL, USA
| | - Yuxuan Miao
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Moshe Elkabets
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Ben-Gurion University, Beer Sheva, Israel
| | - Sara Kochanny
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, USA
| | - Wenji Guo
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, USA
| | - Emma Dyer
- Harvard T.H. Chan School of Public Health, Cambridge, MA, USA
| | - Alexander T Pearson
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, USA
| | - Aditya Juloori
- Department of Radiation Oncology, University of Chicago, Chicago, IL, USA
| | - Mark Lingen
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Grayson Cole
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | | | - Nishant Agrawal
- Department of Surgery, University of Chicago, Chicago, IL, USA
| | - Evgeny Izumchenko
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, USA.
| | - Ari J Rosenberg
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
23
|
Perumalsamy H, Xiao X, Kim HY, Yoon TH. scRNA-seq analysis discovered suppression of immunomodulatory dependent inflammatory response in PMBCs exposed to silver nanoparticles. J Nanobiotechnology 2024; 22:118. [PMID: 38494495 PMCID: PMC10946150 DOI: 10.1186/s12951-024-02364-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/21/2024] [Indexed: 03/19/2024] Open
Abstract
The assessment of AgNPs toxicity in vitro and in vivo models are frequently conflicting and inaccurate. Nevertheless, single cell immunological responses in a heterogenous environment have received little attention. Therefore, in this study, we have performed in-depth analysis which clearly revealed cellular-metal ion association as well as specific immunological response. Our study didn't show significant population differences in PMBC between control and AgNPs group implying no toxicological response. To confirm it further, deep profiling identified differences in subsets and differentially expressed genes (DEGs) of monocytes, B cells and T cells. Notably, monocyte subsets showed significant upregulation of metallothionein (MT) gene expression such as MT1G, MT1X, MT1E, MT1A, and MT1F. On the other hand, downregulation of pro-inflammatory genes such as IL1β and CCL3 in both CD16 + and CD16- monocyte subsets were observed. This result indicated that AgNPs association with monocyte subsets de-promoted inflammatory responsive genes suggesting no significant toxicity observed in AgNPs treated group. Other cell types such as B cells and T cells also showed negligible differences in their subsets suggesting no toxicity response. Further, AgNPs treated group showed upregulation of cell proliferation, ribosomal synthesis, downregulation of cytokine release, and T cell differentiation inhibition. Overall, our results conclude that treatment of AgNPs to PMBC cells didn't display immunological related cytotoxicity response and thus motivate researchers to use them actively for biomedical applications.
Collapse
Affiliation(s)
- Haribalan Perumalsamy
- Center for Creative Convergence Education, Hanyang University, Seoul, 04763, Republic of Korea
- Institute of Next Generation Material Design, Hanyang University, Seoul, 04763, Republic of Korea
- Department of Chemistry, College of Natural Sciences, Hanyang University, Seoul, 04763, Republic of Korea
- Research Institute for Convergence of Basic Science, Hanyang University, Seoul, 04763, Republic of Korea
| | - Xiao Xiao
- Department of Chemistry, College of Natural Sciences, Hanyang University, Seoul, 04763, Republic of Korea
| | - Hyun-Yi Kim
- NGeneS Inc, 362, Gwangdeok 1-ro, Sangnok-gu, Ansan-si, Gyeonggi-do, 15495, Republic of Korea
| | - Tae-Hyun Yoon
- Institute of Next Generation Material Design, Hanyang University, Seoul, 04763, Republic of Korea.
- Department of Chemistry, College of Natural Sciences, Hanyang University, Seoul, 04763, Republic of Korea.
- Department of Medical and Digital Engineering, Hanyang University, Seoul, 04763, Republic of Korea.
- Research Institute for Convergence of Basic Science, Hanyang University, Seoul, 04763, Republic of Korea.
| |
Collapse
|
24
|
Avila-Bonilla RG, Martínez-Montero JP. Crosstalk between vault RNAs and innate immunity. Mol Biol Rep 2024; 51:387. [PMID: 38443657 PMCID: PMC10914904 DOI: 10.1007/s11033-024-09305-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 01/31/2024] [Indexed: 03/07/2024]
Abstract
PURPOSE Vault (vt) RNAs are noncoding (nc) RNAs transcribed by RNA polymerase III (RNA Pol III) with 5'-triphosphate (5'-PPP) termini that play significant roles and are recognized by innate immune sensors, including retinoic acid-inducible protein 1 (RIG-I). In addition, vtRNAs adopt secondary structures that can be targets of interferon-inducible protein kinase R (PKR) and the oligoadenylate synthetase (OAS)/RNase L system, both of which are important for activating antiviral defenses. However, changes in the expression of vtRNAs have been associated with pathological processes that activate proinflammatory pathways, which influence cellular events such as differentiation, aging, autophagy, apoptosis, and drug resistance in cancer cells. RESULTS In this review, we summarized the biology of vtRNAs and focused on their interactions with the innate immune system. These findings provide insights into the diverse roles of vtRNAs and their correlation with various cellular processes to improve our understanding of their biological functions.
Collapse
Affiliation(s)
- Rodolfo Gamaliel Avila-Bonilla
- Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Departamento de Genética y Biología Molecular, Av. IPN 2508, 07360, Mexico City, Mexico.
| | | |
Collapse
|
25
|
Schultze-Rhonhof L, Marzi J, Carvajal Berrio DA, Holl M, Braun T, Schäfer-Ruoff F, Andress J, Bachmann C, Templin M, Brucker SY, Schenke-Layland K, Weiss M. Human tissue-resident peritoneal macrophages reveal resistance towards oxidative cell stress induced by non-invasive physical plasma. Front Immunol 2024; 15:1357340. [PMID: 38504975 PMCID: PMC10949891 DOI: 10.3389/fimmu.2024.1357340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 02/14/2024] [Indexed: 03/21/2024] Open
Abstract
In the context of multimodal treatments for abdominal cancer, including procedures such as cytoreductive surgery and intraperitoneal chemotherapy, recurrence rates remain high, and long-term survival benefits are uncertain due to post-operative complications. Notably, treatment-limiting side effects often arise from an uncontrolled activation of the immune system, particularly peritoneally localized macrophages, leading to massive cytokine secretion and phenotype changes. Exploring alternatives, an increasing number of studies investigated the potential of plasma-activated liquids (PAL) for adjuvant peritoneal cancer treatment, aiming to mitigate side effects, preserve healthy tissue, and reduce cytotoxicity towards non-cancer cells. To assess the non-toxicity of PAL, we isolated primary human macrophages from the peritoneum and subjected them to PAL exposure. Employing an extensive methodological spectrum, including flow cytometry, Raman microspectroscopy, and DigiWest protein analysis, we observed a pronounced resistance of macrophages towards PAL. This resistance was characterized by an upregulation of proliferation and anti-oxidative pathways, countering PAL-derived oxidative stress-induced cell death. The observed cellular effects of PAL treatment on human tissue-resident peritoneal macrophages unveil a potential avenue for PAL-derived immunomodulatory effects within the human peritoneal cavity. Our findings contribute to understanding the intricate interplay between PAL and macrophages, shedding light on the promising prospects for PAL in the adjuvant treatment of peritoneal cancer.
Collapse
Affiliation(s)
| | - Julia Marzi
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine, University of Tübingen, Tübingen, Germany
- Natural and Medical Sciences Institute (NMI) at the University of Tübingen, Reutlingen, Germany
| | - Daniel Alejandro Carvajal Berrio
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine, University of Tübingen, Tübingen, Germany
| | - Myriam Holl
- Department of Women’s Health Tübingen, University of Tübingen, Tübingen, Germany
| | - Theresa Braun
- Natural and Medical Sciences Institute (NMI) at the University of Tübingen, Reutlingen, Germany
- University Development, Research and Transfer, University of Konstanz, Konstanz, Germany
| | - Felix Schäfer-Ruoff
- Natural and Medical Sciences Institute (NMI) at the University of Tübingen, Reutlingen, Germany
| | - Jürgen Andress
- Department of Women’s Health Tübingen, University of Tübingen, Tübingen, Germany
| | - Cornelia Bachmann
- Department of Women’s Health Tübingen, University of Tübingen, Tübingen, Germany
| | - Markus Templin
- Natural and Medical Sciences Institute (NMI) at the University of Tübingen, Reutlingen, Germany
| | - Sara Y. Brucker
- Department of Women’s Health Tübingen, University of Tübingen, Tübingen, Germany
| | - Katja Schenke-Layland
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine, University of Tübingen, Tübingen, Germany
- Natural and Medical Sciences Institute (NMI) at the University of Tübingen, Reutlingen, Germany
| | - Martin Weiss
- Department of Women’s Health Tübingen, University of Tübingen, Tübingen, Germany
- Natural and Medical Sciences Institute (NMI) at the University of Tübingen, Reutlingen, Germany
| |
Collapse
|
26
|
Hung HC, Mao TL, Fan MH, Huang GZ, Minhalina AP, Chen CL, Liu CL. Enhancement of Tumorigenicity, Spheroid Niche, and Drug Resistance of Pancreatic Cancer Cells in Three-Dimensional Culture System. J Cancer 2024; 15:2292-2305. [PMID: 38495500 PMCID: PMC10937281 DOI: 10.7150/jca.87494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 01/27/2024] [Indexed: 03/19/2024] Open
Abstract
The three-dimensional (3D) cell culture technique has been applied comprehensively as a variable platform for medical research, biochemical signal pathway analysis, and evaluation of anti-tumor treatment response due to an excellent recapitulation of a tumor microenvironment (TME) in the in vitro cultured cancer cells. Pancreatic cancer (PaC) is one of the toughest malignancies with a complex TME and refractory treatment response. To comprehensively study the TME of PaC, there is an eager need to develop a 3D culture model to decompose the cellular components and their cross interactions. Herein, we establish a 3D PaC culture system with cancer stem cell (CSC) and scalability properties. To validate our model, we tested the individual PaC cell and the combined effects with cancer-associated fibroblasts (CAFs) on cancer tumorigenicity, the cellular interaction through the CXCR3/CXCL10 axis, and cellular responses reflection of anti-cancer treatments. With the help of our 3D technology, a simulated malignant spheroid with important stromal populations and TME physiochemical properties may be successfully recreated. It can be used in a wide range of preclinical research and helpful in advancing basic and translational cancer biology.
Collapse
Affiliation(s)
- Hao-Chien Hung
- Department of General Surgery, Chang-Gung Memorial Hospital at Linkou, Taoyuan 33305, Taiwan
| | - Tsui-Lien Mao
- Department of Pathology, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
- Department of Pathology, National Taiwan University Hospital, Taipei 10002, Taiwan
| | - Ming-Huei Fan
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Guan-Zhi Huang
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Ainani Priza Minhalina
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Chi-Long Chen
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Department of Pathology, Taipei Medical University Hospital, Taipei 11031, Taiwan
| | - Chao-Lien Liu
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- PhD Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
27
|
Masad RJ, Idriss I, Mohamed YA, Al-Sbiei A, Bashir G, Al-Marzooq F, Altahrawi A, Fernandez-Cabezudo MJ, Al-Ramadi BK. Oral administration of Manuka honey induces IFNγ-dependent resistance to tumor growth that correlates with beneficial modulation of gut microbiota composition. Front Immunol 2024; 15:1354297. [PMID: 38444857 PMCID: PMC10912506 DOI: 10.3389/fimmu.2024.1354297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 02/01/2024] [Indexed: 03/07/2024] Open
Abstract
Background To investigate the potential of Manuka honey (MH) as an immunomodulatory agent in colorectal cancer (CRC) and dissect the underlying molecular and cellular mechanisms. Methods MH was administered orally over a 4 week-period. The effect of MH treatment on microbiota composition was studied using 16S rRNA sequencing of fecal pellets collected before and after treatment. Pretreated mice were implanted with CRC cells and followed for tumor growth. Tumors and lymphoid organs were analyzed by flow cytometry (FACS), immunohistochemistry and qRT-PCR. Efficacy of MH was also assessed in a therapeutic setting, with oral treatment initiated after tumor implantation. We utilized IFNγ-deficient mice to determine the importance of interferon signaling in MH-induced immunomodulation. Results Pretreatment with MH enhanced anti-tumor responses leading to suppression of tumor growth. Evidence for enhanced tumor immunogenicity included upregulated MHC class-II on intratumoral macrophages, enhanced MHC class-I expression on tumor cells and increased infiltration of effector T cells into the tumor microenvironment. Importantly, oral MH was also effective in retarding tumor growth when given therapeutically. Transcriptomic analysis of tumor tissue highlighted changes in the expression of various chemokines and inflammatory cytokines that drive the observed changes in tumor immunogenicity. The immunomodulatory capacity of MH was abrogated in IFNγ-deficient mice. Finally, bacterial 16S rRNA sequencing demonstrated that oral MH treatment induced unique changes in gut microbiota that may well underlie the IFN-dependent enhancement in tumor immunogenicity. Conclusion Our findings highlight the immunostimulatory properties of MH and demonstrate its potential utilization in cancer prevention and treatment.
Collapse
Affiliation(s)
- Razan J. Masad
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Ienas Idriss
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Yassir A. Mohamed
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Ashraf Al-Sbiei
- Department of Biochemistry and Molecular Biology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Ghada Bashir
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Farah Al-Marzooq
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Abeer Altahrawi
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Maria J. Fernandez-Cabezudo
- Department of Biochemistry and Molecular Biology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Basel K. Al-Ramadi
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
28
|
Lacinski RA, Dziadowicz SA, Stewart A, Chaharbakhshi E, Akhter H, Pisquiy JJ, Victory JH, Hardham JB, Chew C, Prorock A, Bao Y, Sol-Church K, Hobbs GR, Klein E, Nalesnik MA, Hu G, de Oliveira A, Santiago SP, Lindsey BA. Nanosphere pharmacodynamics improves safety of immunostimulatory cytokine therapy. iScience 2024; 27:108836. [PMID: 38303687 PMCID: PMC10831265 DOI: 10.1016/j.isci.2024.108836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/04/2023] [Accepted: 01/03/2024] [Indexed: 02/03/2024] Open
Abstract
Systemic administration of interleukin (IL)-12 induces potent anti-tumor immune responses in preclinical cancer models through the systemic activation of effector immune cells and release of proinflammatory cytokines. IL-12-loaded PLGA nanospheres (IL12ns) are hypothesized to improve therapeutic efficacy and thwart unwanted side effects observed in previous human clinical trials. Through the investigation of peripheral blood and local tissue immune responses in healthy BALB/c mice, the immune-protective pharmacodynamics of IL12ns were suggested. Nanospheres increased pro-inflammatory plasma cytokines/chemokines (IFN-γ, IL-6, TNF-α, and CXCL10) without inducing maladaptive transcriptomic signatures in circulating peripheral immune cells. Gene expression profiling revealed activation of pro-inflammatory signaling pathways in systemic tissues, the likely source of these effector cytokines. These data support that nanosphere pharmacodynamics, including shielding IL-12 from circulating immune cells, depositing peripherally in systemic immune tissues, and then slowly eluting bioactive cytokine, thereafter, are essential to safe immunostimulatory therapy.
Collapse
Affiliation(s)
- Ryan A. Lacinski
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Sebastian A. Dziadowicz
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV 26505, USA
- Bioinformatics Core, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Amanda Stewart
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Edwin Chaharbakhshi
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Halima Akhter
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV 26505, USA
- Bioinformatics Core, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - John J. Pisquiy
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Jack H. Victory
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Joshua B. Hardham
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Claude Chew
- Advanced Technology Cores, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alyson Prorock
- Genome Analysis & Technology Core, University of Virginia School of Medicine, Charlottesville, VA 22904, USA
| | - Yongde Bao
- Genome Analysis & Technology Core, University of Virginia School of Medicine, Charlottesville, VA 22904, USA
| | - Katia Sol-Church
- Genome Analysis & Technology Core, University of Virginia School of Medicine, Charlottesville, VA 22904, USA
| | - Gerald R. Hobbs
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Edwin Klein
- Division of Laboratory Animal Resources, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA
| | - Michael A. Nalesnik
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA 15260, USA
| | - Gangqing Hu
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV 26505, USA
- Bioinformatics Core, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Ana de Oliveira
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, VA 22904, USA
| | - Stell P. Santiago
- Department of Pathology, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Brock A. Lindsey
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
29
|
Hailstock T, Dai C, Aquino J, Walker KE, Chick S, Manirarora JN, Suresh R, Patil V, Renukaradhya GJ, Sullivan YB, LaBresh J, Lunney JK. Production and characterization of anti-porcine CXCL10 monoclonal antibodies. Cytokine 2024; 174:156449. [PMID: 38141459 DOI: 10.1016/j.cyto.2023.156449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/07/2023] [Accepted: 11/15/2023] [Indexed: 12/25/2023]
Abstract
Research on C-X-C motif chemokine ligand 10 (CXCL10) has been widely reported for humans and select animal species, yet immune reagents are limited for pig chemokines. Our goal is to provide veterinary immunologists and the biomedical community with new commercial immune reagents and standardized assays. Recombinant porcine CXCL10 (rPoCXCL10) protein was produced by yeast expression and used to generate a panel of α CXCL10 monoclonal antibodies (mAbs). All mAbs were assessed for cross-inhibition and reactivity to orthologous yeast expressed CXCL10 proteins. Characterization of a panel of nine α PoCXCL10 mAbs identified six distinct antigenic determinants. A sensitive quantitative sandwich ELISA was developed with anti-PoCXCL10-1.6 and -1.9 mAb; reactivity was verified with both rPoCXCL10 and native PoCXCL10, detected in supernatants of peripheral blood mononuclear cells stimulated with rPoIFNγ or PMA/Ionomycin. Immunostaining of in vitro rPoIFNγ stimulated pig spleen and blood cells verified CXCL10 + cells as CD3-CD4-CD172+, with occasional CD3-CD4 + CD172 + subsets. Comparison studies determined that α PoCXCL10-1.4 mAb was the ideal mAb clone for intracellular staining, whereas with α PoCXCL10-1.1 and -1.2 mAbs were best for immunohistochemistry analyses. These techniques and tools will be useful for evaluating swine immune development, responses to infectious diseases and vaccines, as well as for improving utility of pigs as an important biomedical model.
Collapse
Affiliation(s)
- Taylor Hailstock
- Animal Parasitic Diseases Laboratory, BARC, ARS, USDA, Beltsville, MD, USA
| | - Chaohui Dai
- Animal Parasitic Diseases Laboratory, BARC, ARS, USDA, Beltsville, MD, USA; Yangzhou University, Yangzhou, Jiangsu, China
| | - Jovan Aquino
- Animal Parasitic Diseases Laboratory, BARC, ARS, USDA, Beltsville, MD, USA
| | - Kristen E Walker
- Animal Parasitic Diseases Laboratory, BARC, ARS, USDA, Beltsville, MD, USA
| | - Shannon Chick
- Animal Parasitic Diseases Laboratory, BARC, ARS, USDA, Beltsville, MD, USA
| | - Jean N Manirarora
- Animal Parasitic Diseases Laboratory, BARC, ARS, USDA, Beltsville, MD, USA
| | - Raksha Suresh
- Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, Wooster, OH, USA
| | - Veerupaxagouda Patil
- Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, Wooster, OH, USA
| | - Gourapura J Renukaradhya
- Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, Wooster, OH, USA
| | | | | | - Joan K Lunney
- Animal Parasitic Diseases Laboratory, BARC, ARS, USDA, Beltsville, MD, USA.
| |
Collapse
|
30
|
Zou Z, Chen S, Wu Y, Ji S. The USP35-CXCR3 Axis plays an oncogenic role in JeKo-1 mantle cell lymphoma cells. Integr Biol (Camb) 2024; 16:zyae021. [PMID: 39591978 DOI: 10.1093/intbio/zyae021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 11/04/2024] [Accepted: 11/21/2024] [Indexed: 11/28/2024]
Abstract
In B cells, the chemokine receptor CXCR3 is expressed only by a subset of B cells. However, CXCR3 is highly expressed in a rare type of B-cell lymphoma known as Mantle Cell Lymphoma (MCL) and CXCR3 inhibitor impairs proliferation and induces apoptosis in the MCL cell line JeKo-1. Despite this, the mechanism responsible for maintaining high levels of CXCR3 in MCL cells remains unclear. In this study, we assessed CXCR3 expression and amplification in MCL samples and confirmed that CXCR3 is overexpressed in MCL tissues. We also observed that CXCR3 amplification is present in a small portion of MCL patients and is associated with MCL classification. We then screened ubiquitin-specific proteases (USPs) that might control the degradation of CXCR3 protein. Our investigation revealed that USP35 acts as a potent stabilizer of CXCR3 protein. Knockdown of USP35 substantially reduced the CXCR3 protein levels in JeKo-1 cells, resulting in reduced cell viability, cell cycle arrest, increased apoptosis, and mitigated migration and invasion in these cells. At the molecular level, USP35 deubiquitinates and stabilizes CXCR3. USP35 deficiency attenuated the activation of the JAK1/STAT1 pathway and reduced the expression of β-catenin and c-Myc in JeKo-1 cells. Furthermore, we observed that overexpression of CXCR3 rescued the impaired tumorigenicity of USP35-deficient JeKo-1 cells, and the mechanism may be related to the fact that USP35 promotes CXCR3 deubiquitination to stabilize its expression. These findings collectively demonstrate the oncogenic role of the USP35-CXCR3 axis in JeKo-1 MCL cells.
Collapse
Affiliation(s)
- Zongkai Zou
- Department of Pathology, Zhangzhou Affiliated Hospital of Fujian Medical University, No. 59, Shengli West Road, Xiangcheng District, Zhangzhou 363000, Fujian, China
| | - Shumin Chen
- Department of Basic Medicine, Zhangzhou Health Vocational College; Collaborative Innovation Center for Translation Medical Testing and Application Technology, No. 29, Shengli West Road, Xiangcheng District, Zhangzhou 363000, Fujian, China
| | - Yonghe Wu
- Department of Pathology, Zhangzhou Affiliated Hospital of Fujian Medical University, No. 59, Shengli West Road, Xiangcheng District, Zhangzhou 363000, Fujian, China
| | - Siling Ji
- Department of Pathology, Zhangzhou Affiliated Hospital of Fujian Medical University, No. 59, Shengli West Road, Xiangcheng District, Zhangzhou 363000, Fujian, China
| |
Collapse
|
31
|
Baginska J, Nau A, Gomez Diaz I, Giobbie-Hurder A, Weirather J, Vergara J, Abrecht C, Hallisey M, Dennis J, Severgnini M, Huezo J, Marciello I, Rahma O, Manos M, Brohl AS, Bedard PL, Renouf DJ, Sharon E, Streicher H, Ott PA, Buchbinder EI, Hodi FS. Ziv-aflibercept plus pembrolizumab in patients with advanced melanoma resistant to anti-PD-1 treatment. Cancer Immunol Immunother 2024; 73:17. [PMID: 38236249 PMCID: PMC10796592 DOI: 10.1007/s00262-023-03593-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 12/11/2023] [Indexed: 01/19/2024]
Abstract
BACKGROUND Vascular endothelial growth factor is associated with reduced immune response and impaired anti-tumor activity. Combining antiangiogenic agents with immune checkpoint inhibition can overcome this immune suppression and enhance treatment efficacy. METHODS This study investigated the combination of ziv-aflibercept anti-angiogenic therapy with pembrolizumab in patients with advanced melanoma resistant to anti-PD-1 treatment. Baseline and on-treatment plasma and PBMC samples were analyzed by multiplex protein assay and mass cytometry, respectively. RESULTS In this Phase 1B study (NCT02298959), ten patients with advanced PD-1-resistant melanoma were treated with a combination of ziv-aflibercept (at 2-4 mg/kg) plus pembrolizumab (at 2 mg/kg), administered intravenously every 2 weeks. Two patients (20%) achieved a partial response, and two patients (20%) experienced stable disease (SD) as the best response. The two responders had mucosal melanoma, while both patients with SD had ocular melanoma. The combination therapy demonstrated clinical activity and acceptable safety, despite the occurrence of adverse events. Changes in plasma analytes such as platelet-derived growth factor and PD-L1 were explored, indicating potential alterations in myeloid cell function. Higher levels of circulating CXCL10 in non-responding patients may reflect pro-tumor activity. Specific subsets of γδ T cells were associated with poor clinical outcomes, suggesting impaired γδ T-cell function in non-responding patients. CONCLUSIONS Although limited by sample size and follow-up, these findings highlight the potential of the combination of ziv-aflibercept antiangiogenic therapy with pembrolizumab in patients with advanced melanoma resistant to anti-PD-1 treatment and the need for further research to improve outcomes in anti-PD-1-resistant melanoma. TRIAL REGISTRATION NUMBER NCT02298959.
Collapse
Affiliation(s)
- Joanna Baginska
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Allison Nau
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ilana Gomez Diaz
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Jason Weirather
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Juliana Vergara
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Charlotte Abrecht
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Margaret Hallisey
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jenna Dennis
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Mariano Severgnini
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Julia Huezo
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Isabella Marciello
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Osama Rahma
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Michael Manos
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Andrew S Brohl
- Sarcoma Department and Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Philippe L Bedard
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Daniel J Renouf
- Cancer and Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Elad Sharon
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD, USA
| | - Howard Streicher
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD, USA
| | - Patrick A Ott
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Elizabeth I Buchbinder
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - F Stephen Hodi
- Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
32
|
Zheng W, Ling S, Cao Y, Shao C, Sun X. Combined use of NK cells and radiotherapy in the treatment of solid tumors. Front Immunol 2024; 14:1306534. [PMID: 38264648 PMCID: PMC10803658 DOI: 10.3389/fimmu.2023.1306534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 12/22/2023] [Indexed: 01/25/2024] Open
Abstract
Natural killer (NK) cells are innate lymphocytes possessing potent tumor surveillance and elimination activity. Increasing attention is being focused on the role of NK cells in integral antitumor strategies (especially immunotherapy). Of note, therapeutic efficacy is considerable dependent on two parameters: the infiltration and cytotoxicity of NK cells in tumor microenvironment (TME), both of which are impaired by several obstacles (e.g., chemokines, hypoxia). Strategies to overcome such barriers are needed. Radiotherapy is a conventional modality employed to cure solid tumors. Recent studies suggest that radiotherapy not only damages tumor cells directly, but also enhances tumor recognition by immune cells through altering molecular expression of tumor or immune cells via the in situ or abscopal effect. Thus, radiotherapy may rebuild a NK cells-favored TME, and thus provide a cost-effective approach to improve the infiltration of NK cells into solid tumors, as well as elevate immune-activity. Moreover, the radioresistance of tumor always hampers the response to radiotherapy. Noteworthy, the puissant cytotoxic activity of NK cells not only kills tumor cells directly, but also increases the response of tumors to radiation via activating several radiosensitization pathways. Herein, we review the mechanisms by which NK cells and radiotherapy mutually promote their killing function against solid malignancies. We also discuss potential strategies harnessing such features in combined anticancer care.
Collapse
Affiliation(s)
- Wang Zheng
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Sunkai Ling
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuandong Cao
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chunlin Shao
- Institution of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xinchen Sun
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
33
|
Ahmed SA, Mendonca P, Messeha SS, Oriaku ET, Soliman KFA. The Anticancer Effects of Marine Carotenoid Fucoxanthin through Phosphatidylinositol 3-Kinase (PI3K)-AKT Signaling on Triple-Negative Breast Cancer Cells. Molecules 2023; 29:61. [PMID: 38202644 PMCID: PMC10779870 DOI: 10.3390/molecules29010061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 12/08/2023] [Accepted: 12/12/2023] [Indexed: 01/12/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer that lacks specific targets such as estrogen, progesterone, and HER2 receptors. TNBC affects one in eight women in the United States, making up 15-20% of breast cancer cases. Patients with TNBC can develop resistance to chemotherapy over time, leading to treatment failure. Therefore, finding other options like natural products is necessary for treatment. The advantages of using natural products sourced from plants as anticancer agents are that they are less toxic, more affordable, and have fewer side effects. These products can modulate several cellular processes of the tumor microenvironment, such as proliferation, migration, angiogenesis, cell cycle arrest, and apoptosis. The phosphatidyl inositol 3-kinase (PI3K)-AKT signaling pathway is an important pathway that contributes to the survival and growth of the tumor microenvironment and is associated with these cellular processes. This current study examined the anticancer effects of fucoxanthin, a marine carotenoid isolated from brown seaweed, in the MDA-MB-231 and MDA-MB-468 TNBC cell lines. The methods used in this study include a cytotoxic assay, PI3K-AKT signaling pathway PCR arrays, and Wes analysis. Fucoxanthin (6.25 µM) + TNF-α (50 ng/mL) and TNF-α (50 ng/mL) showed no significant effect on cell viability compared to the control in both MDA-MB-231 and MDA-MB-468 cells after a 24 h treatment period. PI3K-AKT signaling pathway PCR array studies showed that in TNF-α-stimulated (50 ng/mL) MDA-MB-231 and MDA-MB-468 cells, fucoxanthin (6.25 µM) modulated the mRNA expression of 12 genes, including FOXO1, RASA1, HRAS, MAPK3, PDK2, IRS1, EIF4EBP1, EIF4B, PTK2, TIRAP, RHOA, and ELK1. Additionally, fucoxanthin significantly downregulated the protein expression of IRS1, EIF4B, and ELK1 in MDA-MB-231 cells, and no change in the protein expression of EIF4B and ELK1 was shown in MDA-MB-468 cells. Fucoxanthin upregulated the protein expression of RHOA in both cell lines. The modulation of the expression of genes and proteins of the PI3K-AKT signaling pathway may elucidate fucoxanthin's effects in cell cycle progression, apoptotic processes, migration, and proliferation, which shows that PI3K-AKT may be the possible molecular mechanism for fucoxanthin's effects. In conclusion, the results obtained in this study elucidate fucoxanthin's molecular mechanisms and indicate that fucoxanthin may be considered a promising candidate for breast cancer-targeted therapy.
Collapse
Affiliation(s)
- Shade’ A. Ahmed
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA; (S.A.A.); (E.T.O.)
| | - Patricia Mendonca
- Department of Biology, College of Science and Technology, Florida A&M University, Tallahassee, FL 32307, USA;
| | - Samia S. Messeha
- Department of Biology, College of Science and Technology, Florida A&M University, Tallahassee, FL 32307, USA;
| | - Ebenezer T. Oriaku
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA; (S.A.A.); (E.T.O.)
| | - Karam F. A. Soliman
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA; (S.A.A.); (E.T.O.)
| |
Collapse
|
34
|
Westrich JA, McNulty EE, Carpenter M, Burton M, Reed K, Nalls A, Sandoval A, Mayo C, Mathiason CK. Monitoring longitudinal immunological responses to bluetongue virus 17 in experimentally infected sheep. Virus Res 2023; 338:199246. [PMID: 37858729 PMCID: PMC10594635 DOI: 10.1016/j.virusres.2023.199246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 10/14/2023] [Accepted: 10/16/2023] [Indexed: 10/21/2023]
Abstract
Bluetongue virus (BTV) is an economically important pathogen of ruminant species with worldwide prevalence. While many BTV infections are asymptomatic, animals with symptomatic presentation deteriorate quickly with the sickest succumbing to disease within one week. Animals that survive the infection often require months to recover. The immune response to BTV infection is thought to play a central role in controlling the disease. Key to understanding BTV disease is profiling vertebrate host immunological cellular and cytokine responses. Studies to characterize immune responses in ruminants have been limited by a lack of species-specific reagents and assay technology. Here we assess the longitudinal immunological response to experimental BTV-17-California (CA) infection in sheep using the most up to date assays. We infected a cohort of sheep with BTV-17-CA and longitudinally monitored each animal for clinical disease, viremia and specific immunological parameters (B cells, T cells, monocytes) by RT-qPCR, traditional flow cytometry and/or fluorescent based antibody arrays. BTV-inoculated sheep exhibited clinical signs characteristic of bluetongue virus disease. Circulating virus was demonstrated after 8 days post inoculation (DPI) and remained detectable for the remainder of the time course (24 DPI). A distinct lymphopenia was observed between 7 and 14 DPI that rebounded to mock-inoculated control levels at 17 DPI. In addition, we observed increased expression of pro-inflammatory cytokines after 8 DPI. Taken together, we have established a model of BTV infection in sheep and have successfully monitored the longitudinal vertebrate host immunological response and viral infection progression using a combination of traditional methods and cutting-edge technology.
Collapse
Affiliation(s)
- Joseph A Westrich
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | - Erin E McNulty
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | - Molly Carpenter
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | - Mollie Burton
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | - Kirsten Reed
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | - Amy Nalls
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | - Audrey Sandoval
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | - Christie Mayo
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | - Candace K Mathiason
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA.
| |
Collapse
|
35
|
Rosati M, Terpos E, Homan P, Bergamaschi C, Karaliota S, Ntanasis-Stathopoulos I, Devasundaram S, Bear J, Burns R, Bagratuni T, Trougakos IP, Dimopoulos MA, Pavlakis GN, Felber BK. Rapid transient and longer-lasting innate cytokine changes associated with adaptive immunity after repeated SARS-CoV-2 BNT162b2 mRNA vaccinations. Front Immunol 2023; 14:1292568. [PMID: 38090597 PMCID: PMC10711274 DOI: 10.3389/fimmu.2023.1292568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/14/2023] [Indexed: 12/18/2023] Open
Abstract
Introduction Cytokines and chemokines play an important role in shaping innate and adaptive immunity in response to infection and vaccination. Systems serology identified immunological parameters predictive of beneficial response to the BNT162b2 mRNA vaccine in COVID-19 infection-naïve volunteers, COVID-19 convalescent patients and transplant patients with hematological malignancies. Here, we examined the dynamics of the serum cytokine/chemokine responses after the 3rd BNT162b2 mRNA vaccination in a cohort of COVID-19 infection-naïve volunteers. Methods We measured serum cytokine and chemokine responses after the 3rd dose of the BNT162b2 mRNA (Pfizer/BioNtech) vaccine in COVID-19 infection-naïve individuals by a chemiluminescent assay and ELISA. Anti-Spike binding antibodies were measured by ELISA. Anti-Spike neutralizing antibodies were measured by a pseudotype assay. Results Comparison to responses found after the 1st and 2nd vaccinations showed persistence of the coordinated responses of several cytokine/chemokines including the previously identified rapid and transient IL-15, IFN-γ, CXCL10/IP-10, TNF-α, IL-6 signature. In contrast to the transient (24hrs) effect of the IL-15 signature, an inflammatory/anti-inflammatory cytokine signature (CCL2/MCP-1, CCL3/MIP-1α, CCL4/MIP-1β, CXCL8/IL-8, IL-1Ra) remained at higher levels up to one month after the 2nd and 3rd booster vaccinations, indicative of a state of longer-lasting innate immune change. We also identified a systemic transient increase of CXCL13 only after the 3rd vaccination, supporting stronger germinal center activity and the higher anti-Spike antibody responses. Changes of the IL-15 signature, and the inflammatory/anti-inflammatory cytokine profile correlated with neutralizing antibody levels also after the 3rd vaccination supporting their role as immune biomarkers for effective development of vaccine-induced humoral responses. Conclusion These data revealed that repeated SARS-Cov-2 BNT162b2 mRNA vaccination induces both rapid transient as well as longer-lasting systemic serum cytokine changes associated with innate and adaptive immune responses. Clinical trial registration Clinicaltrials.gov, identifier NCT04743388.
Collapse
Affiliation(s)
- Margherita Rosati
- Human Retrovirus Pathogenesis Section, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
| | - Evangelos Terpos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Philip Homan
- Center for Cancer Research Collaborative Bioinformatics Resource, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, United States
| | - Cristina Bergamaschi
- Human Retrovirus Pathogenesis Section, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
| | - Sevasti Karaliota
- Human Retrovirus Pathogenesis Section, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
- Basic Science Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, United States
| | - Ioannis Ntanasis-Stathopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Santhi Devasundaram
- Human Retrovirus Pathogenesis Section, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
| | - Jenifer Bear
- Human Retrovirus Pathogenesis Section, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
| | - Robert Burns
- Human Retrovirus Pathogenesis Section, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
| | - Tina Bagratuni
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis P. Trougakos
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Meletios A. Dimopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - George N. Pavlakis
- Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
| | - Barbara K. Felber
- Human Retrovirus Pathogenesis Section, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
| |
Collapse
|
36
|
Hambo S, Harb H. Extracellular Vesicles and Their Role in Lung Infections. Int J Mol Sci 2023; 24:16139. [PMID: 38003329 PMCID: PMC10671184 DOI: 10.3390/ijms242216139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/07/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Lung infections are one of the most common causes of death and morbidity worldwide. Both bacterial and viral lung infections cause a vast number of infections with varying severities. Extracellular vesicles (EVs) produced by different cells due to infection in the lung have the ability to modify the immune system, leading to either better immune response or worsening of the disease. It has been shown that both bacteria and viruses have the ability to produce their EVs and stimulate the immune system for that. In this review, we investigate topics from EV biogenesis and types of EVs to lung bacterial and viral infections caused by various bacterial species. Mycobacterium tuberculosis, Staphylococcus aureus, and Streptococcus pneumoniae infections are covered intensively in this review. Moreover, various viral lung infections, including SARS-CoV-2 infections, have been depicted extensively. In this review, we focus on eukaryotic-cell-derived EVs as an important component of disease pathogenesis. Finally, this review holds high novelty in its findings and literature review. It represents the first time to cover all different information on immune-cell-derived EVs in both bacterial and viral lung infections.
Collapse
Affiliation(s)
| | - Hani Harb
- Institute for Medical Microbiology and Virology, University Hospital Dresden, Technical University Dresden, Fetscherstr. 74, 01307 Dresden, Germany;
| |
Collapse
|
37
|
Dobish KK, Wittorf KJ, Swenson SA, Bean DC, Gavile CM, Woods NT, Ghosal G, Hyde RK, Buckley SM. FBXO21 mediated degradation of p85α regulates proliferation and survival of acute myeloid leukemia. Leukemia 2023; 37:2197-2208. [PMID: 37689825 PMCID: PMC10624613 DOI: 10.1038/s41375-023-02020-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/18/2023] [Accepted: 08/31/2023] [Indexed: 09/11/2023]
Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease characterized by clonal expansion of myeloid blasts in the bone marrow (BM). Despite advances in therapy, the prognosis for AML patients remains poor, and there is a need to identify novel molecular pathways regulating tumor cell survival and proliferation. F-box ubiquitin E3 ligase, FBXO21, has low expression in AML, but expression correlates with survival in AML patients and patients with higher expression have poorer outcomes. Silencing FBXO21 in human-derived AML cell lines and primary patient samples leads to differentiation, inhibition of tumor progression, and sensitization to chemotherapy agents. Additionally, knockdown of FBXO21 leads to up-regulation of cytokine signaling pathways. Through a mass spectrometry-based proteomic analysis of FBXO21 in AML, we identified that FBXO21 ubiquitylates p85α, a regulatory subunit of the phosphoinositide 3-kinase (PI3K) pathway, for degradation resulting in decreased PI3K signaling, dimerization of free p85α and ERK activation. These findings reveal the ubiquitin E3 ligase, FBXO21, plays a critical role in regulating AML pathogenesis, specifically through alterations in PI3K via regulation of p85α protein stability.
Collapse
Affiliation(s)
- Kasidy K Dobish
- Department of Internal Medicine, Division of Hematology & Hematopoietic Malignancies, University of Utah, Salt Lake City, UT, USA
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Karli J Wittorf
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Samantha A Swenson
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Dalton C Bean
- Department of Internal Medicine, Division of Hematology & Hematopoietic Malignancies, University of Utah, Salt Lake City, UT, USA
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, USA
| | - Catherine M Gavile
- Department of Internal Medicine, Division of Hematology & Hematopoietic Malignancies, University of Utah, Salt Lake City, UT, USA
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Nicholas T Woods
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
- Eppley Institute, University of Nebraska Medical Center, Omaha, NE, USA
| | - Gargi Ghosal
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - R Katherine Hyde
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Shannon M Buckley
- Department of Internal Medicine, Division of Hematology & Hematopoietic Malignancies, University of Utah, Salt Lake City, UT, USA.
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.
- Department of Oncological Sciences, University of Utah, Salt Lake City, USA.
| |
Collapse
|
38
|
Ghaedrahmati F, Nasrolahi A, Najafi S, Mighani M, Anbiyaee O, Haybar H, Assareh AR, Kempisty B, Dzięgiel P, Azizidoost S, Farzaneh M. Circular RNAs-mediated angiogenesis in human cancers. Clin Transl Oncol 2023; 25:3101-3121. [PMID: 37039938 DOI: 10.1007/s12094-023-03178-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 03/16/2023] [Indexed: 04/12/2023]
Abstract
Circular RNAs (circRNAs) as small non-coding RNAs with cell, tissue, or organ-specific expression accomplish a broad array of functions in physiological and pathological processes such as cancer development. Angiogenesis, a complicated multistep process driving a formation of new blood vessels, speeds up tumor progression by supplying nutrients as well as energy. Abnormal expression of circRNAs reported to affect tumor development through impressing angiogenesis. Such impacts are introduced as constant with different tumorigenic features known as "hallmarks of cancer". In addition, deregulated circRNAs show possibilities to prognosis and diagnosis both in the prophecy of prognosis in malignancies and also their prejudice from healthy individuals. In the present review article, we have evaluated the angiogenic impacts and anti-angiogenic managements of circRNAs in human cancers.
Collapse
Affiliation(s)
- Farhoodeh Ghaedrahmati
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ava Nasrolahi
- Infectious Ophthalmologic Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sajad Najafi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Mighani
- School of Medicine, Golestan University of Medical Sciences, Golestan, Iran
| | - Omid Anbiyaee
- Cardiovascular Research Center, Nemazi Hospital, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Habib Haybar
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ahmad Reza Assareh
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Bartosz Kempisty
- Institute of Veterinary Medicine, Department of Veterinary Surgery, Nicolaus Copernicus University, Torun, Poland
- Department of Human Morphology and Embryology, Division of Anatomy, Wroclaw Medical University, Wrocław, Poland
- North Carolina State University College of Agriculture and Life Sciences, Raleigh, NC, 27695, US
| | - Piotr Dzięgiel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368, Wroclaw, Poland
| | - Shirin Azizidoost
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
39
|
Amador-Martínez I, Aparicio-Trejo OE, Bernabe-Yepes B, Aranda-Rivera AK, Cruz-Gregorio A, Sánchez-Lozada LG, Pedraza-Chaverri J, Tapia E. Mitochondrial Impairment: A Link for Inflammatory Responses Activation in the Cardiorenal Syndrome Type 4. Int J Mol Sci 2023; 24:15875. [PMID: 37958859 PMCID: PMC10650149 DOI: 10.3390/ijms242115875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/25/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
Cardiorenal syndrome type 4 (CRS type 4) occurs when chronic kidney disease (CKD) leads to cardiovascular damage, resulting in high morbidity and mortality rates. Mitochondria, vital organelles responsible for essential cellular functions, can become dysfunctional in CKD. This dysfunction can trigger inflammatory responses in distant organs by releasing Damage-associated molecular patterns (DAMPs). These DAMPs are recognized by immune receptors within cells, including Toll-like receptors (TLR) like TLR2, TLR4, and TLR9, the nucleotide-binding domain, leucine-rich-containing family pyrin domain-containing-3 (NLRP3) inflammasome, and the cyclic guanosine monophosphate (cGMP)-adenosine monophosphate (AMP) synthase (cGAS)-stimulator of interferon genes (cGAS-STING) pathway. Activation of these immune receptors leads to the increased expression of cytokines and chemokines. Excessive chemokine stimulation results in the recruitment of inflammatory cells into tissues, causing chronic damage. Experimental studies have demonstrated that chemokines are upregulated in the heart during CKD, contributing to CRS type 4. Conversely, chemokine inhibitors have been shown to reduce chronic inflammation and prevent cardiorenal impairment. However, the molecular connection between mitochondrial DAMPs and inflammatory pathways responsible for chemokine overactivation in CRS type 4 has not been explored. In this review, we delve into mechanistic insights and discuss how various mitochondrial DAMPs released by the kidney during CKD can activate TLRs, NLRP3, and cGAS-STING immune pathways in the heart. This activation leads to the upregulation of chemokines, ultimately culminating in the establishment of CRS type 4. Furthermore, we propose using chemokine inhibitors as potential strategies for preventing CRS type 4.
Collapse
Affiliation(s)
- Isabel Amador-Martínez
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City 04510, Mexico; (I.A.-M.); (A.K.A.-R.)
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (O.E.A.-T.); (L.G.S.-L.)
| | - Omar Emiliano Aparicio-Trejo
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (O.E.A.-T.); (L.G.S.-L.)
| | - Bismarck Bernabe-Yepes
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico;
| | - Ana Karina Aranda-Rivera
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City 04510, Mexico; (I.A.-M.); (A.K.A.-R.)
- Laboratorio F-315, Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| | - Alfredo Cruz-Gregorio
- Departamento de Fisiología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico;
| | - Laura Gabriela Sánchez-Lozada
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (O.E.A.-T.); (L.G.S.-L.)
| | - José Pedraza-Chaverri
- Laboratorio F-315, Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| | - Edilia Tapia
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (O.E.A.-T.); (L.G.S.-L.)
| |
Collapse
|
40
|
Xu YF, Wang GY, Zhang MY, Yang JG. Hub genes and their key effects on prognosis of Burkitt lymphoma. World J Clin Oncol 2023; 14:357-372. [PMID: 37970111 PMCID: PMC10631346 DOI: 10.5306/wjco.v14.i10.357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/06/2023] [Accepted: 09/18/2023] [Indexed: 10/24/2023] Open
Abstract
BACKGROUND Burkitt lymphoma (BL) is an exceptionally aggressive malignant neoplasm that arises from either the germinal center or post-germinal center B cells. Patients with BL often present with rapid tumor growth and require high-intensity multi-drug therapy combined with adequate intrathecal chemotherapy prophylaxis, however, a standard treatment program for BL has not yet been established. It is important to identify biomarkers for predicting the prognosis of BLs and discriminating patients who might benefit from the therapy. Microarray data and sequencing information from public databases could offer opportunities for the discovery of new diagnostic or therapeutic targets. AIM To identify hub genes and perform gene ontology (GO) and survival analysis in BL. METHODS Gene expression profiles and clinical traits of BL patients were collected from the Gene Expression Omnibus database. Weighted gene co-expression network analysis (WGCNA) was applied to construct gene co-expression modules, and the cytoHubba tool was used to find the hub genes. Then, the hub genes were analyzed using GO and Kyoto Encyclopedia of Genes and Genomes analysis. Additionally, a Protein-Protein Interaction network and a Genetic Interaction network were constructed. Prognostic candidate genes were identified through overall survival analysis. Finally, a nomogram was established to assess the predictive value of hub genes, and drug-gene interactions were also constructed. RESULTS In this study, we obtained 8 modules through WGCNA analysis, and there was a significant correlation between the yellow module and age. Then we identified 10 hub genes (SRC, TLR4, CD40, STAT3, SELL, CXCL10, IL2RA, IL10RA, CCR7 and FCGR2B) by cytoHubba tool. Within these hubs, two genes were found to be associated with OS (CXCL10, P = 0.029 and IL2RA, P = 0.0066) by survival analysis. Additionally, we combined these two hub genes and age to build a nomogram. Moreover, the drugs related to IL2RA and CXCL10 might have a potential therapeutic role in relapsed and refractory BL. CONCLUSION From WGCNA and survival analysis, we identified CXCL10 and IL2RA that might be prognostic markers for BL.
Collapse
Affiliation(s)
- Yan-Feng Xu
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Guan-Yun Wang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Ming-Yu Zhang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Ji-Gang Yang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| |
Collapse
|
41
|
Li J, Ramzan F, Zhong G. Investigating novel biomarkers in uterine corpus endometrial carcinoma: in silico analysis and clinical specimens validation via RT-qPCR and immunohistochemistry. Am J Cancer Res 2023; 13:4376-4400. [PMID: 37818076 PMCID: PMC10560950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/17/2023] [Indexed: 10/12/2023] Open
Abstract
The rising incidence and mortality rate of Uterine Corpus Endometrial Carcinoma (UCEC) pose significant health concerns. CC and CXC chemokines have been linked to tumorigenesis and cancer progression. Recognizing the growing significance of CC and CXC chemokines' diagnostic and prognostic significance in diverse cancer types, our objective was to comprehensively analyze the diagnostic and prognostic values of hub genes from the CC and CXC chemokines in UCEC, utilizing both in silico and clinical samples and cell lines-based approaches. In silico analyses include STRING, Cytoscape, Cytohubba, The Cancer Genome Atlas (TCGA) datasets analysis via the UALCAN, GEPIA, OncoDB, and MuTarget, SurvivalGenie, MEXPRESS, cBioPoratal, TIMER, ENCORI, and DrugBank. Meanwhile, clinical samples and cell lines based analyses include Reverse transcription-quantitative polymerase chain reaction (RT-qPCR), targeted bisulfite sequencing (bisulfite-seq) analysis, and immunohistochemistry (IHC). Through present study, we identified CCL25 (CC motif chemokine ligand 25), CXCL10 (C-X-C motif chemokine ligand 10), CXCL12 (C-X-C motif chemokine ligand 12), and CXCL16 (C-X-C motif chemokine ligand 16) as crucial hub genes among the CC and CXC chemokines. Analyzing the expression data from TCGA, we observed a significant up-regulation of CCL25, CXCL10, and CXCL16 in UCEC samples compared to controls. In contrast, we noted a significant down-regulation of CXCL12 expression in UCEC samples. On clinical UCEC samples and cell lines analysis, the significant higher expression of CCL25, CXCL10, and CXCL16 and significant lower expression of CXCL12 were also denoted in UCEC samples than the controls via RT-qPCR and IHC analyses. Moreover, in silico analysis also confirmed the abnormal promoter methylation levels of the hub genes in TCGA UCEC samples, which was later validated by the clinical samples using targeted based bisulfite-seq analysis. In addition, various additional aspects of the CCL25, CXCL10, CXCL12, and CXCL16 have also been uncovered in UCEC during the present study. Our findings offer novel insights that contribute to the clinical utility of CCL25, CXCL10, CXCL12, and CXCL16 chemokines as potential diagnostic and prognostic biomarkers in UCEC.
Collapse
Affiliation(s)
- Jie Li
- Health Management Center, The Second Affiliated Hospital of Hainan Medical UniversityHaikou 570311, Hainan, China
| | - Faiqah Ramzan
- Gomal Center of Bio-Chemistry and Biotechnology (GCBB), Gomal UniversityDera Ismail Khan 29050, Pakistan
| | - Guiping Zhong
- Health Management Center, The Second Affiliated Hospital of Hainan Medical UniversityHaikou 570311, Hainan, China
| |
Collapse
|
42
|
Juric V, Mayes E, Binnewies M, Lee T, Canaday P, Pollack JL, Rudolph J, Du X, Liu VM, Dash S, Palmer R, Jahchan NS, Ramoth ÅJ, Lacayo S, Mankikar S, Norng M, Brassell C, Pal A, Chan C, Lu E, Sriram V, Streuli M, Krummel MF, Baker KP, Liang L. TREM1 activation of myeloid cells promotes antitumor immunity. Sci Transl Med 2023; 15:eadd9990. [PMID: 37647386 DOI: 10.1126/scitranslmed.add9990] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 08/02/2023] [Indexed: 09/01/2023]
Abstract
Myeloid cells in the tumor microenvironment (TME) can exist in immunosuppressive and immunostimulatory states that impede or promote antitumor immunity, respectively. Blocking suppressive myeloid cells or increasing stimulatory cells to enhance antitumor immune responses is an area of interest for therapeutic intervention. Triggering receptor expressed on myeloid cells-1 (TREM1) is a proinflammatory receptor that amplifies immune responses. TREM1 is expressed on neutrophils, subsets of monocytes and tissue macrophages, and suppressive myeloid populations in the TME, including tumor-associated neutrophils, monocytes, and tumor-associated macrophages. Depletion or inhibition of immunosuppressive myeloid cells, or stimulation by TREM1-mediated inflammatory signaling, could be used to promote an immunostimulatory TME. We developed PY159, an afucosylated humanized anti-TREM1 monoclonal antibody with enhanced FcγR binding. PY159 is a TREM1 agonist that induces signaling, leading to up-regulation of costimulatory molecules on monocytes and macrophages, production of proinflammatory cytokines and chemokines, and enhancement of T cell activation in vitro. An antibody against mouse TREM1, PY159m, promoted antitumor efficacy in syngeneic mouse tumor models. These results suggest that PY159-mediated agonism of TREM1 on tumoral myeloid cells can promote a proinflammatory TME and offer a promising strategy for immunotherapy.
Collapse
Affiliation(s)
- Vladislava Juric
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Erin Mayes
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Mikhail Binnewies
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Tian Lee
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Pamela Canaday
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Joshua L Pollack
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Joshua Rudolph
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Xiaoyan Du
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Victoria M Liu
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Subhadra Dash
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Rachael Palmer
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Nadine S Jahchan
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Åsa Johanna Ramoth
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Sergio Lacayo
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Shilpa Mankikar
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Manith Norng
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Chris Brassell
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Aritra Pal
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Christopher Chan
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Erick Lu
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Venkataraman Sriram
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Michel Streuli
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Matthew F Krummel
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kevin P Baker
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| | - Linda Liang
- Pionyr Immunotherapeutics, 2 Tower Place, Suite 800, South San Francisco, CA 94080, USA
| |
Collapse
|
43
|
Komel T, Omerzel M, Kamensek U, Znidar K, Lampreht Tratar U, Kranjc Brezar S, Dolinar K, Pirkmajer S, Sersa G, Cemazar M. Gene Immunotherapy of Colon Carcinoma with IL-2 and IL-12 Using Gene Electrotransfer. Int J Mol Sci 2023; 24:12900. [PMID: 37629081 PMCID: PMC10454179 DOI: 10.3390/ijms241612900] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/12/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Gene immunotherapy has become an important approach in the treatment of cancer. One example is the introduction of genes encoding immunostimulatory cytokines, such as interleukin 2 and interleukin 12, which stimulate immune cells in tumours. The aim of our study was to determine the effects of gene electrotransfer of plasmids encoding interleukin 2 and interleukin 12 individually and in combination in the CT26 murine colon carcinoma cell line in mice. In the in vitro experiment, the pulse protocol that resulted in the highest expression of IL-2 and IL-12 mRNA and proteins was used for the in vivo part. In vivo, tumour growth delay and also complete response were observed in the group treated with the plasmid combination. Compared to the control group, the highest levels of various immunostimulatory cytokines and increased immune infiltration were observed in the combination group. Long-term anti-tumour immunity was observed in the combination group after tumour re-challenge. In conclusion, our combination therapy efficiently eradicated CT26 colon carcinoma in mice and also generated strong anti-tumour immune memory.
Collapse
Affiliation(s)
- Tilen Komel
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska 2, SI-1000 Ljubljana, Slovenia; (T.K.); (M.O.); (U.K.); (K.Z.); (U.L.T.); (S.K.B.); (G.S.)
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, SI-1000 Ljubljana, Slovenia
| | - Masa Omerzel
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska 2, SI-1000 Ljubljana, Slovenia; (T.K.); (M.O.); (U.K.); (K.Z.); (U.L.T.); (S.K.B.); (G.S.)
| | - Urska Kamensek
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska 2, SI-1000 Ljubljana, Slovenia; (T.K.); (M.O.); (U.K.); (K.Z.); (U.L.T.); (S.K.B.); (G.S.)
| | - Katarina Znidar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska 2, SI-1000 Ljubljana, Slovenia; (T.K.); (M.O.); (U.K.); (K.Z.); (U.L.T.); (S.K.B.); (G.S.)
| | - Ursa Lampreht Tratar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska 2, SI-1000 Ljubljana, Slovenia; (T.K.); (M.O.); (U.K.); (K.Z.); (U.L.T.); (S.K.B.); (G.S.)
| | - Simona Kranjc Brezar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska 2, SI-1000 Ljubljana, Slovenia; (T.K.); (M.O.); (U.K.); (K.Z.); (U.L.T.); (S.K.B.); (G.S.)
| | - Klemen Dolinar
- Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Zaloska 4, SI-1000 Ljubljana, Slovenia; (K.D.); (S.P.)
| | - Sergej Pirkmajer
- Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Zaloska 4, SI-1000 Ljubljana, Slovenia; (K.D.); (S.P.)
| | - Gregor Sersa
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska 2, SI-1000 Ljubljana, Slovenia; (T.K.); (M.O.); (U.K.); (K.Z.); (U.L.T.); (S.K.B.); (G.S.)
- Faculty of Health Sciences, University of Ljubljana, Zdravstvena pot 5, SI-1000 Ljubljana, Slovenia
| | - Maja Cemazar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska 2, SI-1000 Ljubljana, Slovenia; (T.K.); (M.O.); (U.K.); (K.Z.); (U.L.T.); (S.K.B.); (G.S.)
- Faculty of Health Sciences, University of Primorska, Polje 42, SI-6310 Izola, Slovenia
| |
Collapse
|
44
|
Zhang B, Huang B, Zhang X, Li S, Zhu J, Chen X, Song H, Shang D. PANoptosis-related molecular subtype and prognostic model associated with the immune microenvironment and individualized therapy in pancreatic cancer. Front Oncol 2023; 13:1217654. [PMID: 37519797 PMCID: PMC10382139 DOI: 10.3389/fonc.2023.1217654] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 06/20/2023] [Indexed: 08/01/2023] Open
Abstract
Background PANoptosis is an inflammatory type of programmed cell death regulated by PANopotosome. Mounting evidence has shown that PANoptosis could be involved in cancer pathogenesis and the tumor immune microenvironment. Nevertheless, there have been no studies on the mechanism of PANoptosis on pancreatic cancer (PC) pathogenesis. Methods We downloaded the data on transcriptomic and clinical features of PC patients from the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus databases. Additionally, the data on copy number variation (CNV), methylation and somatic mutations of genes in 33 types of cancers were obtained from TCGA. Next, we identified the PANoptosis-related molecular subtype using the consensus clustering analysis, and constructed and validated the PANoptosis-related prognostic model using LASSO and Cox regression analyses. Moreover, RT-qPCR was performed to determine the expression of genes involved in the model. Results We obtained 66 PANoptosis-related genes (PANRGs) from published studies. Of these, 24 PC-specific prognosis-related genes were identified. Pan-cancer analysis revealed complex genetic changes, including CNV, methylation, and mutation in PANRGs were identified in various cancers. By consensus clustering analysis, PC patients were classified into two PANoptosis-related patterns: PANcluster A and B. In PANcluster A, the patient prognosis was significantly worse compared to PANcluster B. The CIBERSORT algorithm showed a significant increase in the infiltration of CD8+ T cells, monocytes, and naïve B cells, in patients in PANcluster B. Additionally, the infiltration of macrophages, activated mast cells, and dendritic cells were higher in patients in PANcluster A. Patients in PANcluster A were more sensitive to erlotinib, selumetinib and trametinib, whereas patients in PANcluster B were highly sensitive to irinotecan, oxaliplatin and sorafenib. Moreover, we constructed and validated the PANoptosis-related prognostic model to predict the patient's survival. Finally, the GEPIA and Human Protein Atlas databases were analyzed, and RT-qPCR was performed. Compared to normal tissues, a significant increase in CXCL10 and ITGB6 (associated with the model) expression was observed in PC tissues. Conclusion We first identified the PANoptosis-related molecular subtypes and established a PANoptosis-related prognostic model for predicting the survival of patients with PC. These results would aid in exploring the mechanisms of PANoptosis in PC pathogenesis.
Collapse
Affiliation(s)
- Biao Zhang
- Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Bingqian Huang
- Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Xiaonan Zhang
- Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Shuang Li
- Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jingyi Zhu
- Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Xu Chen
- Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Huiyi Song
- Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Dong Shang
- Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| |
Collapse
|
45
|
Yan W, Qiu L, Yang M, Xu A, Ma M, Yuan Q, Ma X, Liang W, Li X, Lu Y. CXCL10 mediates CD8 + T cells to facilitate vessel normalization and improve the efficacy of cetuximab combined with PD-1 checkpoint inhibitors in colorectal cancer. Cancer Lett 2023:216263. [PMID: 37354983 DOI: 10.1016/j.canlet.2023.216263] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 06/05/2023] [Accepted: 06/05/2023] [Indexed: 06/26/2023]
Abstract
The immunotherapy and anti-EGFR targeted treatment occupying a pivotal position in colorectal cancer (CRC), is still limited to a group of patients who display specific molecular alterations and inevitably escape from resistance, further studies are still needed in colorectal cancer. We found that chemokine ligand 10 (CXCL10) expression correlates with intratumoral CD8+ T cell infiltration and reprograms tumor vasculatures in colorectal cancer. CXCL10 overexpression not only suppressed tumor growth but also increased CD8+ T cell infiltration and induced tumor vascular normalization in vivo. Additionally, the growth inhibition and tumor vascular normalization induced by CXCL10 can be reversed by the depletion of CD8+ T cells in vivo. Mechanically, CXCL10 interacts with VCAN to mediate tumor vascular normalization. The VCAN expression correlated inversely with the expression of CXCL10 and the infiltration of CD8+ T cells in CRC. Elevated CXCL10 expression sensitized colorectal cancer cells to cetuximab/anti-PD1 combination therapy compared with cetuximab or anti-PD1 alone. We propose that CXCL10 could be used to increase the anti-EGFR therapy and immunotherapy effect, targeting both tumor vessels and immune cells in colorectal cancer.
Collapse
Affiliation(s)
- Wei Yan
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, PR China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, 1023 South Shatai Road, Guangzhou, Guangdong, PR China.
| | - Lin Qiu
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, PR China.
| | - Meiling Yang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, PR China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, 1023 South Shatai Road, Guangzhou, Guangdong, PR China.
| | - Anran Xu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, PR China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, 1023 South Shatai Road, Guangzhou, Guangdong, PR China.
| | - Manqi Ma
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, PR China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, 1023 South Shatai Road, Guangzhou, Guangdong, PR China.
| | - Qinzi Yuan
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, PR China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, 1023 South Shatai Road, Guangzhou, Guangdong, PR China.
| | - Xiaochen Ma
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong, PR China.
| | - Wenjuan Liang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, PR China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, 1023 South Shatai Road, Guangzhou, Guangdong, PR China.
| | - Xuenong Li
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, PR China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, 1023 South Shatai Road, Guangzhou, Guangdong, PR China.
| | - Yanxia Lu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, PR China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, 1023 South Shatai Road, Guangzhou, Guangdong, PR China.
| |
Collapse
|
46
|
Lu H, Lou H, Wengert G, Paudel R, Patel N, Desai S, Crum B, Linton-Reid K, Chen M, Li D, Ip J, Mauri F, Pinato DJ, Rockall A, Copley SJ, Ghaem-Maghami S, Aboagye EO. Tumor and local lymphoid tissue interaction determines prognosis in high-grade serous ovarian cancer. Cell Rep Med 2023:101092. [PMID: 37348499 PMCID: PMC10394173 DOI: 10.1016/j.xcrm.2023.101092] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 03/29/2023] [Accepted: 05/30/2023] [Indexed: 06/24/2023]
Abstract
Tertiary lymphoid structure (TLS) is associated with prognosis in copy-number-driven tumors, including high-grade serous ovarian cancer (HGSOC), although the function of TLS and its interaction with copy-number alterations in HGSOC are not fully understood. In the current study, we confirm that TLS-high HGSOC patients show significantly better progression-free survival (PFS). We show that the presence of TLS in HGSOC tumors is associated with B cell maturation and cytotoxic tumor-specific T cell activation and proliferation. In addition, the copy-number loss of IL15 and CXCL10 may limit TLS formation in HGSOC; a list of genes that may dysregulate TLS function is also proposed. Last, a radiomics-based signature is developed to predict the presence of TLS, which independently predicts PFS in both HGSOC patients and immune checkpoint inhibitor (ICI)-treated non-small cell lung cancer (NSCLC) patients. Overall, we reveal that TLS coordinates intratumoral B cell and T cell response to HGSOC tumor, while the cancer genome evolves to counteract TLS formation and function.
Collapse
Affiliation(s)
- Haonan Lu
- Department of Surgery and Cancer, Imperial College, Hammersmith Campus, The Commonwealth Building, Du Cane Road, W12 0NN London, UK
| | - Hantao Lou
- Ludwig Cancer Research, Nuffield Department of Medicine, University of Oxford, OX3 7DQ Oxford, UK
| | - Georg Wengert
- Department of Surgery and Cancer, Imperial College, Hammersmith Campus, The Commonwealth Building, Du Cane Road, W12 0NN London, UK
| | - Reema Paudel
- Department of Surgery and Cancer, Imperial College, Hammersmith Campus, The Commonwealth Building, Du Cane Road, W12 0NN London, UK
| | - Naina Patel
- Department of Surgery and Cancer, Imperial College, Hammersmith Campus, The Commonwealth Building, Du Cane Road, W12 0NN London, UK
| | - Saral Desai
- Imperial College Healthcare NHS Trust, Du Cane Road, W12 0HS London, UK
| | - Bill Crum
- Department of Surgery and Cancer, Imperial College, Hammersmith Campus, The Commonwealth Building, Du Cane Road, W12 0NN London, UK
| | - Kristofer Linton-Reid
- Department of Surgery and Cancer, Imperial College, Hammersmith Campus, The Commonwealth Building, Du Cane Road, W12 0NN London, UK
| | - Mitchell Chen
- Department of Surgery and Cancer, Imperial College, Hammersmith Campus, The Commonwealth Building, Du Cane Road, W12 0NN London, UK; Imperial College Healthcare NHS Trust, Du Cane Road, W12 0HS London, UK
| | - Dongyang Li
- Department of Surgery and Cancer, Imperial College, Hammersmith Campus, The Commonwealth Building, Du Cane Road, W12 0NN London, UK
| | - Jacey Ip
- Department of Surgery and Cancer, Imperial College, Hammersmith Campus, The Commonwealth Building, Du Cane Road, W12 0NN London, UK; Imperial College Healthcare NHS Trust, Du Cane Road, W12 0HS London, UK
| | - Francesco Mauri
- Department of Surgery and Cancer, Imperial College, Hammersmith Campus, The Commonwealth Building, Du Cane Road, W12 0NN London, UK
| | - David J Pinato
- Department of Surgery and Cancer, Imperial College, Hammersmith Campus, The Commonwealth Building, Du Cane Road, W12 0NN London, UK; Division of Oncology, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Andrea Rockall
- Department of Surgery and Cancer, Imperial College, Hammersmith Campus, The Commonwealth Building, Du Cane Road, W12 0NN London, UK
| | - Susan J Copley
- Department of Surgery and Cancer, Imperial College, Hammersmith Campus, The Commonwealth Building, Du Cane Road, W12 0NN London, UK; Imperial College Healthcare NHS Trust, Du Cane Road, W12 0HS London, UK
| | - Sadaf Ghaem-Maghami
- Department of Surgery and Cancer, Imperial College, Hammersmith Campus, The Commonwealth Building, Du Cane Road, W12 0NN London, UK; Imperial College Healthcare NHS Trust, Du Cane Road, W12 0HS London, UK
| | - Eric O Aboagye
- Department of Surgery and Cancer, Imperial College, Hammersmith Campus, The Commonwealth Building, Du Cane Road, W12 0NN London, UK.
| |
Collapse
|
47
|
Geng T, Zheng M, Wang Y, Reseland JE, Samara A. An artificial intelligence prediction model based on extracellular matrix proteins for the prognostic prediction and immunotherapeutic evaluation of ovarian serous adenocarcinoma. Front Mol Biosci 2023; 10:1200354. [PMID: 37388244 PMCID: PMC10301747 DOI: 10.3389/fmolb.2023.1200354] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/31/2023] [Indexed: 07/01/2023] Open
Abstract
Background: Ovarian Serous Adenocarcinoma is a malignant tumor originating from epithelial cells and one of the most common causes of death from gynecological cancers. The objective of this study was to develop a prediction model based on extracellular matrix proteins, using artificial intelligence techniques. The model aimed to aid healthcare professionals to predict the overall survival of patients with ovarian cancer (OC) and determine the efficacy of immunotherapy. Methods: The Cancer Genome Atlas Ovarian Cancer (TCGA-OV) data collection was used as the study dataset, whereas the TCGA-Pancancer dataset was used for validation. The prognostic importance of 1068 known extracellular matrix proteins for OC were determined by the Random Forest algorithm and the Lasso algorithm establishing the ECM risk score. Based on the gene expression data, the differences in mRNA abundance, tumour mutation burden (TMB) and tumour microenvironment (TME) between the high- and low-risk groups were assessed. Results: Combining multiple artificial intelligence algorithms we were able to identify 15 key extracellular matrix genes, namely, AMBN, CXCL11, PI3, CSPG5, TGFBI, TLL1, HMCN2, ESM1, IL12A, MMP17, CLEC5A, FREM2, ANGPTL4, PRSS1, FGF23, and confirm the validity of this ECM risk score for overall survival prediction. Several other parameters were identified as independent prognostic factors for OC by multivariate COX analysis. The analysis showed that thyroglobulin (TG) targeted immunotherapy was more effective in the high ECM risk score group, while the low ECM risk score group was more sensitive to the RYR2 gene-related immunotherapy. Additionally, the patients with low ECM risk scores had higher immune checkpoint gene expression and immunophenoscore levels and responded better to immunotherapy. Conclusion: The ECM risk score is an accurate tool to assess the patient's sensitivity to immunotherapy and forecast OC prognosis.
Collapse
Affiliation(s)
- Tianxiang Geng
- Department of Biomaterials, FUTURE, Center for Functional Tissue Reconstruction, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Mengxue Zheng
- Laboratory of Reproductive Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Yongfeng Wang
- Department of Obstetrics and Gynecology, Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Janne Elin Reseland
- Department of Biomaterials, FUTURE, Center for Functional Tissue Reconstruction, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Athina Samara
- Department of Biomaterials, FUTURE, Center for Functional Tissue Reconstruction, Faculty of Dentistry, University of Oslo, Oslo, Norway
| |
Collapse
|
48
|
Sui X, Zhang X, Zhao J, Liu J, Li S, Zhang X, Wang J. Establishment of a prognostic model for melanoma based on necroptosis-related genes. Funct Integr Genomics 2023; 23:202. [PMID: 37314547 DOI: 10.1007/s10142-023-01129-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 06/03/2023] [Accepted: 06/07/2023] [Indexed: 06/15/2023]
Abstract
In this study, the clinical implications and potential functions of necroptosis-related genes (NRGs) in melanoma were systematically characterized. A novel NRG signature was then constructed to analyze the immune status and prognosis of patients with melanoma. The NRG signatures for melanoma prognosis were searched using the Cancer Genome Atlas (TCGA) dataset and followed by stepwise Cox regression analysis. Patients with melanoma were divided into two groups, and survival analysis, receiver operating characteristic (ROC), and univariate and multivariate analyses were subsequently performed. The correlation of risk score (RS) with tumor immunity and RT-polymerase chain reaction (PCR) was analyzed to further verify the gene signatures. Data on tumor mutational burden (TMB) and chromosomal copy number variation (CNV) were analyzed. Three NRGs were identified as prognostic risk signatures and were significantly related to overall survival (OS) in melanoma. The signatures had better diagnostic accuracy. Furthermore, analysis of mutations in the NRGs and the incidence of chromosomal CNV helped to reveal the relationship between mutations and melanoma occurrence. A nomogram was established based on RSs. The risk characteristics were significantly associated with immunity and high risk is closely correlated with melanoma development. In vitro experiments revealed that necrostatin-1 (Nec-1) promoted cell viability and repressed the expression levels of interleukin (IL)12A and proprotein convertase subtilisin/kexin type (PCSK)1. Additionally, the expression levels of IL12A, CXCL10, and PCSK1 decreased in tumor tissues of melanoma patients. NRGs exert vital roles in immunity and might be applied as a prognostic factor of melanoma.
Collapse
Affiliation(s)
- Xiaohui Sui
- Department of Traditional Chinese Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250011, China
| | - Xiaodan Zhang
- Department of Traditional Chinese Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Junde Zhao
- Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Jinxing Liu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250011, China
| | - Shuling Li
- Department of Traditional Chinese Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Xiaowen Zhang
- Haiyang Traditional Chinese Medicine Hospital, Haiyang, 265100, China
| | - Juntao Wang
- Division of Hand, Foot and Ankle Surgery, Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, 250012, China.
| |
Collapse
|
49
|
Kokinos EK, Tsymbal SA, Galochkina AV, Bezlepkina SA, Nikolaeva JV, Vershinina SO, Shtro AA, Tatarskiy VV, Shtil AA, Broude EV, Roninson IB, Dukhinova M. Inhibition of Cyclin-Dependent Kinases 8/19 Restricts Bacterial and Virus-Induced Inflammatory Responses in Monocytes. Viruses 2023; 15:1292. [PMID: 37376593 PMCID: PMC10305654 DOI: 10.3390/v15061292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/29/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
Hyperactivation of the immune system remains a dramatic, life-threatening complication of viral and bacterial infections, particularly during pneumonia. Therapeutic approaches to counteract local and systemic outbreaks of cytokine storm and to prevent tissue damage remain limited. Cyclin-dependent kinases 8 and 19 (CDK8/19) potentiate transcriptional responses to the altered microenvironment, but CDK8/19 potential in immunoregulation is not fully understood. In the present study, we investigated how a selective CDK8/19 inhibitor, Senexin B, impacts the immunogenic profiles of monocytic cells stimulated using influenza virus H1N1 or bacterial lipopolysaccharides. Senexin B was able to prevent the induction of gene expression of proinflammatory cytokines in THP1 and U937 cell lines and in human peripheral blood-derived mononuclear cells. Moreover, Senexin B substantially reduced functional manifestations of inflammation, including clustering and chemokine-dependent migration of THP1 monocytes and human pulmonary fibroblasts (HPF).
Collapse
Affiliation(s)
- Elena K Kokinos
- SCAMT Institute, ITMO University, 9 Lomonosova Street, 191002 Saint-Petersburg, Russia
| | - Sergey A Tsymbal
- SCAMT Institute, ITMO University, 9 Lomonosova Street, 191002 Saint-Petersburg, Russia
| | - Anastasia V Galochkina
- Smorodintsev Research Institute of Influenza, 15/17 Prof. Popov Street, 197376 Saint-Petersburg, Russia
| | - Svetlana A Bezlepkina
- SCAMT Institute, ITMO University, 9 Lomonosova Street, 191002 Saint-Petersburg, Russia
| | - Julia V Nikolaeva
- Smorodintsev Research Institute of Influenza, 15/17 Prof. Popov Street, 197376 Saint-Petersburg, Russia
| | - Sofia O Vershinina
- SCAMT Institute, ITMO University, 9 Lomonosova Street, 191002 Saint-Petersburg, Russia
| | - Anna A Shtro
- Smorodintsev Research Institute of Influenza, 15/17 Prof. Popov Street, 197376 Saint-Petersburg, Russia
| | - Victor V Tatarskiy
- Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilova Street, 119334 Moscow, Russia
| | - Alexander A Shtil
- Blokhin National Medical Research Center of Oncology, Kashirskoe Highway 24, 115478 Moscow, Russia
| | - Eugenia V Broude
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Sumter Street 715, Columbia, SC 29208, USA
| | - Igor B Roninson
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Sumter Street 715, Columbia, SC 29208, USA
| | - Marina Dukhinova
- SCAMT Institute, ITMO University, 9 Lomonosova Street, 191002 Saint-Petersburg, Russia
| |
Collapse
|
50
|
Tang X, Morris AJ, Deken MA, Brindley DN. Autotaxin Inhibition with IOA-289 Decreases Breast Tumor Growth in Mice Whereas Knockout of Autotaxin in Adipocytes Does Not. Cancers (Basel) 2023; 15:2937. [PMID: 37296899 PMCID: PMC10251959 DOI: 10.3390/cancers15112937] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Breast cancer cells produce negligible quantities of autotaxin. Instead, previous work indicated that adipocytes in the inflamed adipose tissue adjacent to breast tumors are a major source of autotaxin secretion that drives breast tumor growth, metastasis, and the loss of efficacy for chemotherapy and radiotherapy. To test this hypothesis, we used mice with an adipocyte-specific knock out of autotaxin. The lack of autotaxin secretion from adipocytes failed to decrease the growth of orthotopic E0771 breast tumors in syngeneic C57BL/6 mice and the growth and lung metastasis of spontaneous breast tumors in MMTV-PyMT mice. However, the inhibition of autotaxin with IOA-289 decreased the growth of E0771 tumors, indicating that another source of autotaxin is responsible for tumor growth. Tumor-associated fibroblasts and leukocytes produce the majority of autotoxin transcripts in the E0771 breast tumors, and we hypothesize that they are the main sources of ATX that drive breast tumor growth. Autotaxin inhibition with IOA-289 increased the numbers of CD8α+-T-cells in the tumors. This was accompanied by decreases in the concentrations of CXCL10, CCL2, and CXCL9 in the plasma and LIF, TGFβ1, TGFβ2, and prolactin in the tumors. Bioinformatics analysis of human breast tumor databases showed that autotaxin (ENPP2) is expressed mainly in endothelial cells and fibroblasts. Autotaxin expression correlated significantly with increases in IL-6 cytokine receptor ligand interactions, signaling by LIF, TGFβ, and prolactin. This confirms the relevance of results from autotaxin inhibition in the mouse model. We propose that inhibiting autotaxin activity that is derived from cells presenting breast tumors such as fibroblasts, leukocytes, or endothelial cells changes the tumor micro-environment in such a way as to inhibit tumor growth.
Collapse
Affiliation(s)
- Xiaoyun Tang
- Cancer Research Institute of Northern Alberta, Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2S2, Canada;
| | - Andrew J. Morris
- Central Arkansas Veterans Affairs Healthcare System and University of Arkansas for Medical Sciences, 4301 W. Markham St., Little Rock, AR 72205, USA;
| | - Marcel A. Deken
- iOnctura BV, Gustav Mahlerplein 102, 1082 MA Amsterdam, The Netherlands;
| | - David N. Brindley
- Cancer Research Institute of Northern Alberta, Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2S2, Canada;
| |
Collapse
|