101
|
McCormick TS, Hejal RB, Leal LO, Ghannoum MA. GM-CSF: Orchestrating the Pulmonary Response to Infection. Front Pharmacol 2022; 12:735443. [PMID: 35111042 PMCID: PMC8803133 DOI: 10.3389/fphar.2021.735443] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 12/13/2021] [Indexed: 01/18/2023] Open
Abstract
This review summarizes the structure and function of the alveolar unit, comprised of alveolar macrophage and epithelial cell types that work in tandem to respond to infection. Granulocyte-macrophage colony-stimulating factor (GM-CSF) helps to maintain the alveolar epithelium and pulmonary immune system under physiological conditions and plays a critical role in restoring homeostasis under pathologic conditions, including infection. Given the emergence of novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and global spread of coronavirus disease 2019 (COVID-19), with subsequent acute respiratory distress syndrome, understanding basic lung physiology in infectious diseases is especially warranted. This review summarizes clinical and preclinical data for GM-CSF in respiratory infections, and the rationale for sargramostim (yeast-derived recombinant human [rhu] GM-CSF) as adjunctive treatment for COVID-19 and other pulmonary infectious diseases.
Collapse
Affiliation(s)
- Thomas S. McCormick
- Center for Medical Mycology, Department of Dermatology, Case Western Reserve University, Cleveland, OH, United States
| | - Rana B. Hejal
- Medical Intensive Care Unit, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
- Pulmonary and Critical Care Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Luis O. Leal
- Partner Therapeutics, Lexington, MA, United States
| | - Mahmoud A. Ghannoum
- Center for Medical Mycology, Department of Dermatology, Case Western Reserve University, Cleveland, OH, United States
- University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| |
Collapse
|
102
|
Viswanath DI, Liu HC, Huston DP, Chua CYX, Grattoni A. Emerging biomaterial-based strategies for personalized therapeutic in situ cancer vaccines. Biomaterials 2022; 280:121297. [PMID: 34902729 PMCID: PMC8725170 DOI: 10.1016/j.biomaterials.2021.121297] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 11/19/2021] [Accepted: 11/29/2021] [Indexed: 01/03/2023]
Abstract
Landmark successes in oncoimmunology have led to development of therapeutics boosting the host immune system to eradicate local and distant tumors with impactful tumor reduction in a subset of patients. However, current immunotherapy modalities often demonstrate limited success when involving immunologically cold tumors and solid tumors. Here, we describe the role of various biomaterials to formulate cancer vaccines as a form of cancer immunotherapy, seeking to utilize the host immune system to activate and expand tumor-specific T cells. Biomaterial-based cancer vaccines enhance the cancer-immunity cycle by harnessing cellular recruitment and activation against tumor-specific antigens. In this review, we discuss biomaterial-based vaccine strategies to induce lymphocytic responses necessary to mediate anti-tumor immunity. We focus on strategies that selectively attract dendritic cells via immunostimulatory gradients, activate them against presented tumor-specific antigens, and induce effective cross-presentation to T cells in secondary lymphoid organs, thereby generating immunity. We posit that personalized cancer vaccines are promising targets to generate long-term systemic immunity against patient- and tumor-specific antigens to ensure long-term cancer remission.
Collapse
Affiliation(s)
- Dixita Ishani Viswanath
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA; Texas A&M University College of Medicine, Bryan & Houston, TX, USA
| | - Hsuan-Chen Liu
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA
| | - David P Huston
- Texas A&M University College of Medicine, Bryan & Houston, TX, USA
| | | | - Alessandro Grattoni
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA; Department of Surgery, Houston Methodist Hospital, Houston, TX, USA; Department of Radiation Oncology, Houston Methodist Hospital, Houston, TX, USA.
| |
Collapse
|
103
|
Czajka-Francuz P, Cisoń-Jurek S, Czajka A, Kozaczka M, Wojnar J, Chudek J, Francuz T. Systemic Interleukins' Profile in Early and Advanced Colorectal Cancer. Int J Mol Sci 2021; 23:124. [PMID: 35008550 PMCID: PMC8745135 DOI: 10.3390/ijms23010124] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 12/18/2021] [Accepted: 12/20/2021] [Indexed: 02/05/2023] Open
Abstract
Tumor microenvironment (TME) is characterized by mutual interactions of the tumor, stromal and immune cells. Early and advanced colorectal tumors differ in structure and present altered serum cytokine levels. Mutual crosstalk among TME infiltrating cells may shift the balance into immune suppressive or pro-inflammatory, antitumor response this way influencing patients' prognosis. Cancer-related inflammation affects all the body and this way, the systemic level of cytokines could reflect TME processes. Despite numerous studies, it is still not known how systemic cytokines levels change during colorectal cancer (CRC) tumor development. Better understanding tumor microenvironment processes could help in planning therapeutic interventions and more accurate patient prognosis. To contribute to the comprehension of these processes within TME, we reviewed cytokines levels from clinical trials in early and advanced colorectal cancer. Presented data were analyzed in the context of experimental studies and studies analyzing tumor infiltration with immune cells. The review summarizes clinical data of cytokines secreted by tumor microenvironment cells: lymphocytes T helper 1 (Th1), lymphocytes T helper 2 (Th2), lymphocytes T helper 17 (Th17), regulatory T cells (Treg cells), regulatory T cells (Breg cells), M1/M2 macrophages, N1/N2 neutrophils, myeloid-derived suppressor cells (MDSC), dendritic cells (DC), innate lymphoid cells (ILC) natural killer (NK) cells and tumor cells.
Collapse
Affiliation(s)
- Paulina Czajka-Francuz
- Department of Internal Medicine and Oncological Chemotherapy, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-027 Katowice, Poland; (S.C.-J.); (J.W.); (J.C.); (T.F.)
| | - Sylwia Cisoń-Jurek
- Department of Internal Medicine and Oncological Chemotherapy, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-027 Katowice, Poland; (S.C.-J.); (J.W.); (J.C.); (T.F.)
| | - Aleksander Czajka
- Department of General Surgery, Vascular Surgery, Angiology and Phlebology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-635 Katowice, Poland;
| | - Maciej Kozaczka
- Department of Radiotherapy and Chemotherapy, National Institute of Oncology, Public Research Institute in Gliwice, 44-101 Gliwice, Poland;
| | - Jerzy Wojnar
- Department of Internal Medicine and Oncological Chemotherapy, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-027 Katowice, Poland; (S.C.-J.); (J.W.); (J.C.); (T.F.)
| | - Jerzy Chudek
- Department of Internal Medicine and Oncological Chemotherapy, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-027 Katowice, Poland; (S.C.-J.); (J.W.); (J.C.); (T.F.)
| | - Tomasz Francuz
- Department of Internal Medicine and Oncological Chemotherapy, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-027 Katowice, Poland; (S.C.-J.); (J.W.); (J.C.); (T.F.)
- Department of Biochemistry, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-752 Katowice, Poland
| |
Collapse
|
104
|
Hellenbrand DJ, Quinn CM, Piper ZJ, Morehouse CN, Fixel JA, Hanna AS. Inflammation after spinal cord injury: a review of the critical timeline of signaling cues and cellular infiltration. J Neuroinflammation 2021; 18:284. [PMID: 34876174 PMCID: PMC8653609 DOI: 10.1186/s12974-021-02337-2] [Citation(s) in RCA: 293] [Impact Index Per Article: 73.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 11/30/2021] [Indexed: 03/02/2023] Open
Abstract
Traumatic spinal cord injury (SCI) is a devastating neurological condition that results in a loss of motor and sensory function. Although extensive research to develop treatments for SCI has been performed, to date, none of these treatments have produced a meaningful amount of functional recovery after injury. The primary injury is caused by the initial trauma to the spinal cord and results in ischemia, oxidative damage, edema, and glutamate excitotoxicity. This process initiates a secondary injury cascade, which starts just a few hours post-injury and may continue for more than 6 months, leading to additional cell death and spinal cord damage. Inflammation after SCI is complex and driven by a diverse set of cells and signaling molecules. In this review, we utilize an extensive literature survey to develop the timeline of local immune cell and cytokine behavior after SCI in rodent models. We discuss the precise functional roles of several key cytokines and their effects on a variety of cell types involved in the secondary injury cascade. Furthermore, variations in the inflammatory response between rats and mice are highlighted. Since current SCI treatment options do not successfully initiate functional recovery or axonal regeneration, identifying the specific mechanisms attributed to secondary injury is critical. With a more thorough understanding of the complex SCI pathophysiology, effective therapeutic targets with realistic timelines for intervention may be established to successfully attenuate secondary damage.
Collapse
Affiliation(s)
- Daniel J Hellenbrand
- Department of Neurological Surgery, School of Medicine and Public Health (UWSMPH), University of Wisconsin, 600 Highland Ave, Madison, WI, 53792, USA
| | - Charles M Quinn
- Department of Neurological Surgery, School of Medicine and Public Health (UWSMPH), University of Wisconsin, 600 Highland Ave, Madison, WI, 53792, USA
| | - Zachariah J Piper
- Department of Neurological Surgery, School of Medicine and Public Health (UWSMPH), University of Wisconsin, 600 Highland Ave, Madison, WI, 53792, USA
| | - Carolyn N Morehouse
- Department of Neurological Surgery, School of Medicine and Public Health (UWSMPH), University of Wisconsin, 600 Highland Ave, Madison, WI, 53792, USA
| | - Jordyn A Fixel
- Department of Neurological Surgery, School of Medicine and Public Health (UWSMPH), University of Wisconsin, 600 Highland Ave, Madison, WI, 53792, USA
| | - Amgad S Hanna
- Department of Neurological Surgery, School of Medicine and Public Health (UWSMPH), University of Wisconsin, 600 Highland Ave, Madison, WI, 53792, USA.
| |
Collapse
|
105
|
Wang S, Pei S, Wasi M, Parajuli A, Yee A, You L, Wang L. Moderate tibial loading and treadmill running, but not overloading, protect adult murine bone from destruction by metastasized breast cancer. Bone 2021; 153:116100. [PMID: 34246808 PMCID: PMC8478818 DOI: 10.1016/j.bone.2021.116100] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 06/18/2021] [Accepted: 06/29/2021] [Indexed: 12/15/2022]
Abstract
Osteolytic bone lesions, which develop in many metastatic breast cancer patients, impair bone integrity and lead to adverse skeletal related events that are difficult to treat and sometimes fatal. Moderate mechanical loading has been shown to suppress osteolysis in young mice with breast cancer. In this study, we aimed to investigate the dose-dependent effects of mechanical loading on protecting the integrity of adult skeletons with breast cancer. Localized tibial loading and aerobic treadmill running with three levels of varying intensity were tested in a syngeneic mammary tumor bone metastasis model. Adult C57BL/6J female mice (14-week-old, N = 88 mice) received intra-tibial injections of Py8119 triple-negative murine breast cancer cells or PBS and underwent 4 to 5 weeks of exercise or acted as sedentary/non-loaded controls. The bone structure was monitored longitudinally with weekly in vivo micro-computed tomography imaging, while the cellular responses in bone and marrow were examined using immunohistochemistry. Moderate treadmill running (16 m/min, 50 min/day, 5 days/week, and 5 weeks) and tibial loading (4.5 N, 630 με, 4 Hz, 300 cycles/day, 5 days/week, and 4 weeks) suppressed tumor-induced bone destruction, as evaluated by full-thickness perforation of tibial cortex and the volume of osteolytic lesions in the cortex. In contrast, tibial loading at higher magnitude (8 N, 1100 με) induced woven bone and accelerated bone destruction, compared with the non-loaded controls. The three exercise regimens differentially affected osteocyte apoptosis, osteocyte hypoxia, osteoclast activity, bone marrow vasculature, and tumor proliferation. In conclusion, the relationship between exercise intensity and the risk of breast cancer-induced osteolysis was found to follow a J-shaped curve in a preclinical model, suggesting the need to optimize exercise parameters in order to harness the skeletal benefits of exercise in metastatic breast cancers.
Collapse
Affiliation(s)
- Shubo Wang
- Center for Biomechanical Engineering Research, Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Shaopeng Pei
- Center for Biomechanical Engineering Research, Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Murtaza Wasi
- Center for Biomechanical Engineering Research, Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Ashutosh Parajuli
- Center for Biomechanical Engineering Research, Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Albert Yee
- Division of Orthopaedics, Department of Surgery, Sunnybrook Health Sciences Centre and the University of Toronto, Toronto, Ontario, Canada
| | - Lidan You
- Department of Mechanical and Industrial Engineering, Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Liyun Wang
- Center for Biomechanical Engineering Research, Department of Mechanical Engineering, University of Delaware, Newark, DE, USA.
| |
Collapse
|
106
|
Arghiani N, Nissan T, Matin MM. Role of microRNAs in COVID-19 with implications for therapeutics. Biomed Pharmacother 2021; 144:112247. [PMID: 34601190 PMCID: PMC8463393 DOI: 10.1016/j.biopha.2021.112247] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 02/09/2023] Open
Abstract
COVID-19 is a pneumonia-like disease with highly transmittable and pathogenic properties caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which infects both animals and humans. Although many efforts are currently underway to test possible therapies, there is no specific FDA approved drug against SARS-CoV-2 yet. miRNA-directed gene regulation controls the majority of biological processes. In addition, the development and progression of several human diseases are associated with dysregulation of miRNAs. In this regard, it has been shown that changes in miRNAs are linked to severity of COVID-19 especially in patients with respiratory diseases, diabetes, heart failure or kidney problems. Therefore, targeting these small noncoding-RNAs could potentially alleviate complications from COVID-19. Here, we will review the roles and importance of host and RNA virus encoded miRNAs in COVID-19 pathogenicity and immune response. Then, we focus on potential miRNA therapeutics in the patients who are at increased risk for severe disease.
Collapse
Affiliation(s)
- Nahid Arghiani
- Department of Molecular Biosciences, the Wenner-Gren Institute, Stockholm University, Stockholm, Sweden; School of Life Science, Department of Biochemistry and Biomedicine, University of Sussex, Brighton, United Kingdom
| | - Tracy Nissan
- Department of Molecular Biosciences, the Wenner-Gren Institute, Stockholm University, Stockholm, Sweden; School of Life Science, Department of Biochemistry and Biomedicine, University of Sussex, Brighton, United Kingdom.
| | - Maryam M Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran; Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran; Stem Cell and Regenerative Medicine Research Group, Iranian Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran.
| |
Collapse
|
107
|
Gas6/TAM Signalling Negatively Regulates Inflammatory Induction of GM-CSF in Mouse Brain Microglia. Cells 2021; 10:cells10123281. [PMID: 34943789 PMCID: PMC8699038 DOI: 10.3390/cells10123281] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/17/2021] [Accepted: 11/21/2021] [Indexed: 12/16/2022] Open
Abstract
Microglia and astrocytes are the main CNS glial cells responsible for the neuroinflammatory response, where they release a plethora of cytokines into the CNS inflammatory milieu. The TAM (Tyro3, Axl, Mer) receptors and their main ligand Gas6 are regulators of this response, however, the underlying mechanisms remain to be determined. We investigated the ability of Gas6 to modulate the CNS glial inflammatory response to lipopolysaccharide (LPS), a strong pro-inflammatory agent, through a qPCR array that explored Toll-like receptor signalling pathway-associated genes in primary cultured mouse microglia. We identified the Csf2 gene, encoding granulocyte-macrophage colony-stimulating factor (GM-CSF), as a major Gas6 target gene whose induction by LPS was markedly blunted by Gas6. Both the Csf2 gene induction and the suppressive effect of Gas6 on this were emulated through measurement of GM-CSF protein release by cells. We found distinct profiles of GM-CSF induction in different glial cell types, with microglia being most responsive during inflammation. Also, Gas6 markedly inhibited the LPS-stimulated nuclear translocation of NF-κB p65 protein in microglia. These results illustrate microglia as a major resident CNS cellular source of GM-CSF as part of the neuroinflammatory response, and that Gas6/TAM signalling inhibits this response through suppression of NF-κB signalling.
Collapse
|
108
|
Lee YY, Irfan M, Quah Y, Saba E, Kim SD, Park SC, Jeong MG, Kwak YS, Rhee MH. The increasing hematopoietic effect of the combined treatment of Korean Red Ginseng and Colla corii asini on cyclophosphamide-induced immunosuppression in mice. J Ginseng Res 2021; 45:591-598. [PMID: 34803429 PMCID: PMC8587481 DOI: 10.1016/j.jgr.2021.02.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 02/17/2021] [Indexed: 12/22/2022] Open
Abstract
Background Hematopoiesis is the production of blood cells from hematopoietic stem cells (HSCs) that reside in the bone marrow. Cyclophosphamide (CTX) is a chemotherapy drug that suppresses the immune system. Korean Red Ginseng (KRG) and Colla corii asini (CCA) have been traditionally used for boosting the immune system. Methods HSCs in the bone marrow, and immune cell subtype in splenocytes, PBMCs, and thymocytes were investigated. Serum levels of hematopoietic-related markers were analyzed using ELISA. Protein expression in spleen tissue was analyzed using western blot analysis. Hematoxylin & eosin staining in the femurs of mice were also conducted. Results The combination of KRG and CCA with a ratio of 3:2 increased HSCs, CD3 and CD8+ T cells in the circulation, and CD3 T cells in the spleen. A ratio of 2:3 (KRG:CCA) increased the thymic regulatory T cells and recovered the CD3 T cells in the spleen and circulation while recovering proteins in the JAK-STAT pathway in the spleen. Overall, blood cell population and differentiating factors vital for cell differentiation were also significantly recovered by all combinations especially in ratios of 3:2 and 2:3. Conclusion A ratio of 3:2 (KRG:CCA) is the most ideal combination as it recovered the HSC population in the bone marrow of mice.
Collapse
Affiliation(s)
- Yuan Yee Lee
- College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Muhammad Irfan
- College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Oral Biology, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Yixian Quah
- College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Evelyn Saba
- Department of Veterinary Biomedical Sciences, Faculty of Veterinary and Animal Sciences, Pir Mehr Ali Shah Arid Agriculture University, Rawalpindi, Pakistan
| | - Sung-Dae Kim
- College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Seung-Chun Park
- College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Myung-Gyun Jeong
- R&D Headquarters, Korea Ginseng Cooperation, Daejeon, Republic of Korea
| | - Yi-Seong Kwak
- R&D Headquarters, Korea Ginseng Cooperation, Daejeon, Republic of Korea
- Corresponding author. Department of Veterinary Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, Republic of Korea.
| | - Man Hee Rhee
- College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea
- Corresponding author. R&D Headquarters, Korea Ginseng Cooperation, Daejeon, 34520, Republic of Korea.
| |
Collapse
|
109
|
Reece MD, Taylor RR, Song C, Gavegnano C. Targeting Macrophage Dysregulation for Viral Infections: Novel Targets for Immunomodulators. Front Immunol 2021; 12:768695. [PMID: 34790202 PMCID: PMC8591232 DOI: 10.3389/fimmu.2021.768695] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/13/2021] [Indexed: 12/20/2022] Open
Abstract
A major barrier to human immunodeficiency virus (HIV-1) cure is the latent viral reservoir, which persists despite antiretroviral therapy (ART), including across the non-dividing myeloid reservoir which is found systemically in sanctuary sites across tissues and the central nervous system (CNS). Unlike activated CD4+ T cells that undergo rapid cell death during initial infection (due to rapid viral replication kinetics), viral replication kinetics are delayed in non-dividing myeloid cells, resulting in long-lived survival of infected macrophages and macrophage-like cells. Simultaneously, persistent inflammation in macrophages confers immune dysregulation that is a key driver of co-morbidities including cardiovascular disease (CVD) and neurological deficits in people living with HIV-1 (PLWH). Macrophage activation and dysregulation is also a key driver of disease progression across other viral infections including SARS-CoV-2, influenza, and chikungunya viruses, underscoring the interplay between macrophages and disease progression, pathogenesis, and comorbidity in the viral infection setting. This review discusses the role of macrophages in persistence and pathogenesis of HIV-1 and related comorbidities, SARS-CoV-2 and other viruses. A special focus is given to novel immunomodulatory targets for key events driving myeloid cell dysregulation and reservoir maintenance across a diverse array of viral infections.
Collapse
Affiliation(s)
- Monica D Reece
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, United States
| | - Ruby R Taylor
- Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Colin Song
- Department of Chemistry, Emory University, Atlanta, GA, United States
| | - Christina Gavegnano
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, United States
| |
Collapse
|
110
|
Jiang Y, Li F, Li Y, Duan J, Di C, Zhu Y, Zhao J, Jia X, Qu J. CD69 mediates the protective role of adipose tissue-derived mesenchymal stem cells against Pseudomonas aeruginosa pulmonary infection. Clin Transl Med 2021; 11:e563. [PMID: 34841721 PMCID: PMC8567058 DOI: 10.1002/ctm2.563] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/15/2021] [Accepted: 08/19/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Our previous study shows that Adipose tissue-derived mesenchymal stem cells (ASCs) are a promising strategy for cell-based therapy against pulmonary infection with Pseudomonas aeruginosa (P. aeruginosa), but the underlying mechanisms remain unclear. METHODS cDNA microarray assay was performed to explore the transcriptome of ASCs primed by P. aeruginosa. Small interfering RNA (siRNA) was constructed to select the receptor candidates for P. aeruginosa recognition and granulocyte-macrophage colony-stimulating factor (GM-CSF) production in ASCs. The soluble protein chimeras containing the extracellular domain of human CD69 fused to the Fc region of human immunoglobulin IgG1 were used as a probe to validate the recognition of P. aeruginosa. The association between CD69 and extracellular regulated protein kinases 1/2 (ERK1/2) was explored via co-immunoprecipitation, siRNA, and inhibitor. The murine models of P. aeruginosa pneumonia treated with WT-ASCs, GM-CSF-/- -ASCs Cd69-/- -ASCs or Erk1-/- -ASCs were used to determine the role of GM-CSF, CD69, and ERK1 in ASCs against P. aeruginosa infection. RESULTS We showed that C-type lectin receptor CD69 mediated the protective effects of ASCs partly through GM-CSF. CD69 could specifically recognize P. aeruginosa and regulate GM-CSF secretion of ASCs. CD69 regulated the production of GM-CSF via ERK1 in ASCs after P. aeruginosa infection. Moreover, the Administration of ASCs with deficiency of CD69 or ERK1 completely blocked its protective effects in a murine model of P. aeruginosa pneumonia. CONCLUSIONS CD69 recognizes P. aeruginosa and further facilitates ERK1 activation, which plays a crucial role in ASCs-based therapy against P. aeruginosa pneumonia. CD69 may be a novel target molecule to improve ASCs-based therapy against P. aeruginosa infection.
Collapse
Affiliation(s)
- Yanshan Jiang
- Department of Respiratory and Critical Care MedicineRuijin Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200025China
- Institute of Respiratory DiseasesSchool of MedicineShanghai Jiao Tong UniversityShanghai200025China
- Shanghai Key Laboratory of Emergency PreventionDiagnosis and Treatment of Respiratory Infectious DiseasesShanghai200025China
- Clinical Medicine Scientific and Technical Innovation CenterShanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Fan Li
- Clinical Medicine Scientific and Technical Innovation CenterShanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Yanan Li
- Department of Respiratory and Critical Care MedicineRuijin Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200025China
- Institute of Respiratory DiseasesSchool of MedicineShanghai Jiao Tong UniversityShanghai200025China
- Shanghai Key Laboratory of Emergency PreventionDiagnosis and Treatment of Respiratory Infectious DiseasesShanghai200025China
- Clinical Medicine Scientific and Technical Innovation CenterShanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Jielin Duan
- Clinical Medicine Scientific and Technical Innovation CenterShanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Caixia Di
- Department of Respiratory and Critical Care MedicineRuijin Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200025China
- Institute of Respiratory DiseasesSchool of MedicineShanghai Jiao Tong UniversityShanghai200025China
- Shanghai Key Laboratory of Emergency PreventionDiagnosis and Treatment of Respiratory Infectious DiseasesShanghai200025China
| | - Yinggang Zhu
- Department of Pulmonary and Critical Care MedicineHuadong HospitalFudan UniversityShanghaiChina
| | - Jingya Zhao
- Department of Respiratory and Critical Care MedicineRuijin Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200025China
- Institute of Respiratory DiseasesSchool of MedicineShanghai Jiao Tong UniversityShanghai200025China
- Shanghai Key Laboratory of Emergency PreventionDiagnosis and Treatment of Respiratory Infectious DiseasesShanghai200025China
| | - Xinming Jia
- Clinical Medicine Scientific and Technical Innovation CenterShanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Jieming Qu
- Department of Respiratory and Critical Care MedicineRuijin Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200025China
- Institute of Respiratory DiseasesSchool of MedicineShanghai Jiao Tong UniversityShanghai200025China
- Shanghai Key Laboratory of Emergency PreventionDiagnosis and Treatment of Respiratory Infectious DiseasesShanghai200025China
| |
Collapse
|
111
|
SARS-CoV-2 Exposed Mesenchymal Stromal Cell from Congenital Pulmonary Airway Malformations: Transcriptomic Analysis and the Expression of Immunomodulatory Genes. Int J Mol Sci 2021; 22:ijms222111814. [PMID: 34769246 PMCID: PMC8584055 DOI: 10.3390/ijms222111814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/28/2021] [Accepted: 10/29/2021] [Indexed: 12/22/2022] Open
Abstract
The inflammatory response plays a central role in the complications of congenital pulmonary airway malformations (CPAM) and severe coronavirus disease 2019 (COVID-19). The aim of this study was to evaluate the transcriptional changes induced by SARS-CoV-2 exposure in pediatric MSCs derived from pediatric lung (MSCs-lung) and CPAM tissues (MSCs-CPAM) in order to elucidate potential pathways involved in SARS-CoV-2 infection in a condition of exacerbated inflammatory response. MSCs-lung and MSCs-CPAM do not express angiotensin-converting enzyme 2 (ACE2) and transmembrane serine protease 2 (TRMPSS2). SARS-CoV-2 appears to be unable to replicate in MSCs-CPAM and MSCs-lung. MSCs-lung and MSCs-CPAM maintained the expression of stemness markers MSCs-lung show an inflammatory response (IL6, IL1B, CXCL8, and CXCL10), and the activation of Notch3 non-canonical pathway; this route appears silent in MSCs-CPAM, and cytokine genes expression is reduced. Decreased value of p21 in MSCs-lung suggested no cell cycle block, and cells did not undergo apoptosis. MSCs-lung appears to increase genes associated with immunomodulatory function but could contribute to inflammation, while MSCs-CPAM keeps stable or reduce the immunomodulatory receptors expression, but they also reduce their cytokines expression. These data indicated that, independently from their perilesional or cystic origin, the MSCs populations already present in a patient affected with CPAM are not permissive for SARS-CoV-2 entry, and they will not spread the disease in case of infection. Moreover, these MSCs will not undergo apoptosis when they come in contact with SARS-CoV-2; on the contrary, they maintain their staminality profile.
Collapse
|
112
|
Humeau J, Le Naour J, Galluzzi L, Kroemer G, Pol JG. Trial watch: intratumoral immunotherapy. Oncoimmunology 2021; 10:1984677. [PMID: 34676147 PMCID: PMC8526014 DOI: 10.1080/2162402x.2021.1984677] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 09/20/2021] [Indexed: 02/06/2023] Open
Abstract
While chemotherapy and radiotherapy remain the first-line approaches for the management of most unresectable tumors, immunotherapy has emerged in the past two decades as a game-changing treatment, notably with the clinical success of immune checkpoint inhibitors. Immunotherapies aim at (re)activating anticancer immune responses which occur in two main steps: (1) the activation and expansion of tumor-specific T cells following cross-presentation of tumor antigens by specialized myeloid cells (priming phase); and (2) the immunological clearance of malignant cells by these antitumor T lymphocytes (effector phase). Therapeutic vaccines, adjuvants, monoclonal antibodies, cytokines, immunogenic cell death-inducing agents including oncolytic viruses, anthracycline-based chemotherapy and radiotherapy, as well as adoptive cell transfer, all act at different levels of this cascade to (re)instate cancer immunosurveillance. Intratumoral delivery of these immunotherapeutics is being tested in clinical trials to promote superior antitumor immune activity in the context of limited systemic toxicity.
Collapse
Affiliation(s)
- Juliette Humeau
- Equipe labellisée par la Ligue contre le cancer, INSERM U1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, QC H3C 3J7, Canada
- Department of Medicine, Université de Montréal, Montreal, Quebec H3C 3J7, Canada
| | - Julie Le Naour
- Equipe labellisée par la Ligue contre le cancer, INSERM U1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Kremlin Bicêtre, France
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA
| | - Guido Kroemer
- Equipe labellisée par la Ligue contre le cancer, INSERM U1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Kremlin Bicêtre, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
- Institut Universitaire de France, Paris, France
- Karolinska Institute, Department of Women’s and Children’s Health, Karolinska University Hospital, Stockholm, Sweden
- Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China
| | - Jonathan G. Pol
- Equipe labellisée par la Ligue contre le cancer, INSERM U1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Kremlin Bicêtre, France
| |
Collapse
|
113
|
Skerrett-Byrne DA, Nixon B, Bromfield EG, Breen J, Trigg NA, Stanger SJ, Bernstein IR, Anderson AL, Lord T, Aitken RJ, Roman SD, Robertson SA, Schjenken JE. Transcriptomic analysis of the seminal vesicle response to the reproductive toxicant acrylamide. BMC Genomics 2021; 22:728. [PMID: 34625024 PMCID: PMC8499523 DOI: 10.1186/s12864-021-07951-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 08/14/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The seminal vesicles synthesise bioactive factors that support gamete function, modulate the female reproductive tract to promote implantation, and influence developmental programming of offspring phenotype. Despite the significance of the seminal vesicles in reproduction, their biology remains poorly defined. Here, to advance understanding of seminal vesicle biology, we analyse the mouse seminal vesicle transcriptome under normal physiological conditions and in response to acute exposure to the reproductive toxicant acrylamide. Mice were administered acrylamide (25 mg/kg bw/day) or vehicle control daily for five consecutive days prior to collecting seminal vesicle tissue 72 h following the final injection. RESULTS A total of 15,304 genes were identified in the seminal vesicles with those encoding secreted proteins amongst the most abundant. In addition to reproductive hormone pathways, functional annotation of the seminal vesicle transcriptome identified cell proliferation, protein synthesis, and cellular death and survival pathways as prominent biological processes. Administration of acrylamide elicited 70 differentially regulated (fold-change ≥1.5 or ≤ 0.67) genes, several of which were orthogonally validated using quantitative PCR. Pathways that initiate gene and protein synthesis to promote cellular survival were prominent amongst the dysregulated pathways. Inflammation was also a key transcriptomic response to acrylamide, with the cytokine, Colony stimulating factor 2 (Csf2) identified as a top-ranked upstream driver and inflammatory mediator associated with recovery of homeostasis. Early growth response (Egr1), C-C motif chemokine ligand 8 (Ccl8), and Collagen, type V, alpha 1 (Col5a1) were also identified amongst the dysregulated genes. Additionally, acrylamide treatment led to subtle changes in the expression of genes that encode proteins secreted by the seminal vesicle, including the complement regulator, Complement factor b (Cfb). CONCLUSIONS These data add to emerging evidence demonstrating that the seminal vesicles, like other male reproductive tract tissues, are sensitive to environmental insults, and respond in a manner with potential to exert impact on fetal development and later offspring health.
Collapse
Affiliation(s)
- David A Skerrett-Byrne
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, NSW, 2305, Australia
| | - Brett Nixon
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, NSW, 2305, Australia
| | - Elizabeth G Bromfield
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, NSW, 2305, Australia.,Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University, 3584 CM, Utrecht, The Netherlands
| | - James Breen
- The Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia.,South Australian Genomics Centre (SAGC), South Australian Health & Medical Research Institute (SAHMRI), Adelaide, SA, 5000, Australia.,Computational & Systems Biology Program, Precision Medicine Theme, South Australian Health & Medical Research Institute (SAHMRI), Adelaide, SA, 5000, Australia.,Adelaide Medical School, Faculty of Health & Medical Sciences, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Natalie A Trigg
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, NSW, 2305, Australia
| | - Simone J Stanger
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, NSW, 2305, Australia
| | - Ilana R Bernstein
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, NSW, 2305, Australia
| | - Amanda L Anderson
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, NSW, 2305, Australia
| | - Tessa Lord
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, NSW, 2305, Australia
| | - R John Aitken
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, NSW, 2305, Australia
| | - Shaun D Roman
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, NSW, 2305, Australia
| | - Sarah A Robertson
- The Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia.,Adelaide Medical School, Faculty of Health & Medical Sciences, University of Adelaide, Adelaide, SA, 5005, Australia
| | - John E Schjenken
- Priority Research Centre for Reproductive Science, School of Environmental and Life Sciences, Discipline of Biological Sciences, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia. .,Hunter Medical Research Institute, Pregnancy and Reproduction Program, New Lambton Heights, NSW, 2305, Australia.
| |
Collapse
|
114
|
Zhang C, Röder J, Scherer A, Bodden M, Pfeifer Serrahima J, Bhatti A, Waldmann A, Müller N, Oberoi P, Wels WS. Bispecific antibody-mediated redirection of NKG2D-CAR natural killer cells facilitates dual targeting and enhances antitumor activity. J Immunother Cancer 2021; 9:jitc-2021-002980. [PMID: 34599028 PMCID: PMC8488744 DOI: 10.1136/jitc-2021-002980] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/16/2021] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Natural killer group 2D (NKG2D) is an activating receptor of natural killer (NK) cells and other lymphocytes that mediates lysis of malignant cells through recognition of stress-induced ligands such as MICA and MICB. Such ligands are broadly expressed by cancer cells of various origins and serve as targets for adoptive immunotherapy with effector cells endogenously expressing NKG2D or carrying an NKG2D-based chimeric antigen receptor (CAR). However, shedding or downregulation of NKG2D ligands (NKG2DL) can prevent NKG2D activation, resulting in escape of cancer cells from NKG2D-dependent immune surveillance. METHODS To enable tumor-specific targeting of NKG2D-expressing effector cells independent of membrane-anchored NKG2DLs, we generated a homodimeric recombinant antibody which harbors an N-terminal single-chain fragment variable (scFv) antibody domain for binding to NKG2D, linked via a human IgG4 Fc region to a second C-terminal scFv antibody domain for recognition of the tumor-associated antigen ErbB2 (HER2). The ability of this molecule, termed NKAB-ErbB2, to redirect NKG2D-expressing effector cells to ErbB2-positive tumor cells of different origins was investigated using peripheral blood mononuclear cells, ex vivo expanded NK cells, and NK and T cells engineered with an NKG2D-based chimeric receptor. RESULTS On its own, bispecific NKAB-ErbB2 increased lysis of ErbB2-positive breast carcinoma cells by peripheral blood-derived NK cells endogenously expressing NKG2D more effectively than an ErbB2-specific IgG1 mini-antibody able to induce antibody-dependent cell-mediated cytotoxicity via activation of CD16. Furthermore, NKAB-ErbB2 synergized with NK-92 cells or primary T cells engineered to express an NKG2D-CD3ζ chimeric antigen receptor (NKAR), leading to targeted cell killing and greatly enhanced antitumor activity, which remained unaffected by soluble MICA known as an inhibitor of NKG2D-mediated natural cytotoxicity. In an immunocompetent mouse glioblastoma model mimicking low or absent NKG2DL expression, the combination of NKAR-NK-92 cells and NKAB-ErbB2 effectively suppressed outgrowth of ErbB2-positive tumors, resulting in treatment-induced endogenous antitumor immunity and cures in the majority of animals. CONCLUSIONS Our results demonstrate that combining an NKAB antibody with effector cells expressing an activating NKAR receptor represents a powerful and versatile approach to simultaneously enhance tumor antigen-specific as well as NKG2D-CAR and natural NKG2D-mediated cytotoxicity, which may be particularly useful to target tumors with heterogeneous target antigen expression.
Collapse
Affiliation(s)
- Congcong Zhang
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany.,German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, Frankfurt, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Frankfurt Cancer Institute, Goethe University, Frankfurt, Germany
| | - Jasmin Röder
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany.,Frankfurt Cancer Institute, Goethe University, Frankfurt, Germany
| | - Anne Scherer
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - Malena Bodden
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | | | - Anita Bhatti
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - Anja Waldmann
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - Nina Müller
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - Pranav Oberoi
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany.,German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, Frankfurt, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Winfried S Wels
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany .,German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, Frankfurt, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Frankfurt Cancer Institute, Goethe University, Frankfurt, Germany
| |
Collapse
|
115
|
Wagner AD, Wittkop U, Thalmann J, Willmen T, Gödecke V, Hodam J, Ronicke S, Zenke M. Glucocorticoid Effects on Tissue Residing Immune Cells in Giant Cell Arteritis: Importance of GM-CSF. Front Med (Lausanne) 2021; 8:709404. [PMID: 34557501 PMCID: PMC8452956 DOI: 10.3389/fmed.2021.709404] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 08/17/2021] [Indexed: 11/13/2022] Open
Abstract
Giant cell arteritis (GCA) is a systemic granulomatous vasculitis clinically characterized by a prompt response to glucocorticoid therapy. Dendritic cells (DCs) play a central role in the pathogenesis of the disease and are increased in temporal arteries from GCA patients. The aim of this study was to determine the effects of glucocorticoid therapy on granulomatous infiltrates and on peripheral DCs of GCA patients. Immunohistochemical staining of temporal artery specimens from 41 GCA patients revealed a rapid reduction of the number of DCs after initiation of glucocorticoid treatment. TUNEL staining was performed to quantify apoptotic S100+ DC, CD3+ T cells, and CD68+ macrophages in the granulomatous infiltrates. An increase of apoptotic cells up to 9 ± 2% after 4–5 days of glucocorticoid therapy and up to 27 ± 5% (p < 0.001, compared to earlier timepoints) after 6–10 days was detected. A decrease of CCL19 and CCL21 expression was observed after starting glucocorticoid therapy. Granulocyte-macrophage colony-stimulating factor (GM-CSF) expression also significantly decreased under glucocorticoid therapy. No GM-CSF expression was detected in the control specimens. Glucocorticoid therapy leads to a rapid, time-dependent reduction of DCs in temporal arteries from GCA patients and reduction of mediators for cell migration. Our data suggest GM-CSF as a novel therapeutic target of GCA.
Collapse
Affiliation(s)
- Annette D Wagner
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Ulrike Wittkop
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Jessica Thalmann
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Tina Willmen
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Vega Gödecke
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Justyna Hodam
- Deutsches Rotes Kreuz (DRK) Clinic for Child and Adolescent Psychiatry, Bad Neuenahr, Germany
| | - Simon Ronicke
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Martin Zenke
- Department of Cell Biology, Institute for Biomedical Engineering, Rheinisch-Westfälische Technische Hochschule Aachen (RWTH) Aachen University Medical School, Aachen, Germany.,Helmholtz Institute for Biomedical Engineering, Rheinisch-Westfälische Technische Hochschule Aachen (RWTH) Aachen University, Aachen, Germany
| |
Collapse
|
116
|
Sinaga DS, Ho SL, Lu CA, Yu SM, Huang LF. Knockdown expression of a MYB-related transcription factor gene, OsMYBS2, enhances production of recombinant proteins in rice suspension cells. PLANT METHODS 2021; 17:99. [PMID: 34560901 PMCID: PMC8464127 DOI: 10.1186/s13007-021-00799-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 09/12/2021] [Indexed: 05/04/2023]
Abstract
BACKGROUND Transgenic plant suspension cells show economic potential for the production of valuable bioproducts. The sugar starvation-inducible rice αAmy3 promoter, together with its signal peptide, is widely applied to produce recombinant proteins in rice suspension cells. The OsMYBS2 transcription factor was shown recently to reduce activation of the αAmy3 promoter by competing for the binding site of the TA box of the αAmy3 promoter with the potent OsMYBS1 activator. In this study, rice suspension cells were genetically engineered to silence OsMYBS2 to enhance the production of recombinant proteins. RESULTS The mouse granulocyte-macrophage colony-stimulating factor (mGM-CSF) gene was controlled by the αAmy3 promoter and expressed in OsMYBS2-silenced transgenic rice suspension cells. Transcript levels of the endogenous αAmy3 and the transgene mGM-CSF were increased in the OsMYBS2-silenced suspension cells. The highest yield of recombinant mGM-CSF protein attained in the OsMYBS2-silenced transgenic suspension cells was 69.8 µg/mL, which is 2.5-fold that of non-silenced control cells. The yield of recombinant mGM-CSF was further increased to 118.8 µg/mL in cultured cells derived from homozygous F5 seeds, which was 5.1 times higher than that of the control suspension cell line. CONCLUSIONS Our results demonstrate that knockdown of the transcription factor gene OsMYBS2 increased the activity of the αAmy3 promoter and improved the yield of recombinant proteins secreted in rice cell suspension cultures.
Collapse
Affiliation(s)
- Desyanti Saulina Sinaga
- Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan City, 320, Taiwan, ROC
- Department of Life Sciences, National Central University, Taoyuan City, 320, Taiwan, ROC
| | - Shin-Lon Ho
- Department of Agronomy, National Chiayi University, Chiayi City, 600, Taiwan, ROC
| | - Chung-An Lu
- Department of Life Sciences, National Central University, Taoyuan City, 320, Taiwan, ROC
| | - Su-May Yu
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei City, 115, Taiwan, ROC
| | - Li-Fen Huang
- Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan City, 320, Taiwan, ROC.
| |
Collapse
|
117
|
Mol S, Hafkamp FMJ, Varela L, Simkhada N, Taanman-Kueter EW, Tas SW, Wauben MHM, Groot Kormelink T, de Jong EC. Efficient Neutrophil Activation Requires Two Simultaneous Activating Stimuli. Int J Mol Sci 2021; 22:ijms221810106. [PMID: 34576270 PMCID: PMC8467451 DOI: 10.3390/ijms221810106] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 09/12/2021] [Accepted: 09/15/2021] [Indexed: 12/13/2022] Open
Abstract
Neutrophils are abundantly present in the synovium and synovial fluid of patients suffering from arthritis. Neutrophils can be activated by a multitude of stimuli and the current dogma states that this is a two-step process, consisting of a priming step followed by an activation step. Considering that neutrophil activation occurs in an inflammatory environment, where multiple stimuli are present, we argue that a two-step process is highly unlikely. Here, we indeed demonstrate that neutrophils require simultaneous ligation of two different receptors for efficient activation. We isolated human peripheral blood neutrophils and cultured them with various combinations of stimuli (GM-CSF, fMLF, TNF, and LPS). Next, we evaluated essential neutrophil functions, including degranulation and ROS production using flow cytometry, mediator release using ELISA, NETosis by a live cell imaging method, phagocytosis by imaging flow cytometry, and extracellular vesicle (EV) release quantified by high-resolution flow cytometry. Exposure of neutrophils to any combination of stimuli, but not to single stimuli, resulted in significant degranulation, and mediator and EV release. Furthermore, ROS production increased substantially by dual stimulation, yet appeared to be more dependent on the type of stimulation than on dual stimulation. Phagocytosis was induced to its maximum capacity by a single stimulus, while NETosis was not induced by any of the used physiological stimuli. Our data indicate that neutrophil activation is tightly regulated and requires activation by two simultaneous stimuli, which is largely independent of the combination of stimuli.
Collapse
Affiliation(s)
- Sanne Mol
- Department Experimental Immunology, Amsterdam UMC, Location AMC, 1105 AZ Amsterdam, The Netherlands; (S.M.); (F.M.J.H.); (N.S.); (E.W.T.-K.); (S.W.T.); (T.G.K.)
- Department Biomolecular Health Sciences, Faculty Veterinary Medicine, Utrecht University, 3508 TD Utrecht, The Netherlands; (L.V.); (M.H.M.W.)
| | - Florianne M. J. Hafkamp
- Department Experimental Immunology, Amsterdam UMC, Location AMC, 1105 AZ Amsterdam, The Netherlands; (S.M.); (F.M.J.H.); (N.S.); (E.W.T.-K.); (S.W.T.); (T.G.K.)
| | - Laura Varela
- Department Biomolecular Health Sciences, Faculty Veterinary Medicine, Utrecht University, 3508 TD Utrecht, The Netherlands; (L.V.); (M.H.M.W.)
| | - Neena Simkhada
- Department Experimental Immunology, Amsterdam UMC, Location AMC, 1105 AZ Amsterdam, The Netherlands; (S.M.); (F.M.J.H.); (N.S.); (E.W.T.-K.); (S.W.T.); (T.G.K.)
| | - Esther W. Taanman-Kueter
- Department Experimental Immunology, Amsterdam UMC, Location AMC, 1105 AZ Amsterdam, The Netherlands; (S.M.); (F.M.J.H.); (N.S.); (E.W.T.-K.); (S.W.T.); (T.G.K.)
| | - Sander W. Tas
- Department Experimental Immunology, Amsterdam UMC, Location AMC, 1105 AZ Amsterdam, The Netherlands; (S.M.); (F.M.J.H.); (N.S.); (E.W.T.-K.); (S.W.T.); (T.G.K.)
- Amsterdam Rheumatology and Immunology Center, Department of Rheumatology and Clinical Immunology, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Marca H. M. Wauben
- Department Biomolecular Health Sciences, Faculty Veterinary Medicine, Utrecht University, 3508 TD Utrecht, The Netherlands; (L.V.); (M.H.M.W.)
| | - Tom Groot Kormelink
- Department Experimental Immunology, Amsterdam UMC, Location AMC, 1105 AZ Amsterdam, The Netherlands; (S.M.); (F.M.J.H.); (N.S.); (E.W.T.-K.); (S.W.T.); (T.G.K.)
| | - Esther C. de Jong
- Department Experimental Immunology, Amsterdam UMC, Location AMC, 1105 AZ Amsterdam, The Netherlands; (S.M.); (F.M.J.H.); (N.S.); (E.W.T.-K.); (S.W.T.); (T.G.K.)
- Correspondence: ; Tel.: +31-2-0566-4963
| |
Collapse
|
118
|
Aharon A, Horn G, Bar-Lev TH, Zagagi Yohay E, Waks T, Levin M, Deshet Unger N, Avivi I, Globerson Levin A. Extracellular Vesicles Derived from Chimeric Antigen Receptor-T Cells: A Potential Therapy for Cancer. Hum Gene Ther 2021; 32:1224-1241. [PMID: 34494460 DOI: 10.1089/hum.2021.192] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Chimeric antigen receptor (CAR)-T cells are genetically engineered T cells, directed against a tumor-associated antigen. Extracellular vesicles (EVs) derived from CAR-T cells (CAR-T EVs) may preserve CAR-T activity and overcome one of the major obstacles responsible for CAR-T cell failure in patients with solid tumors. This study aimed to compare CAR-T EVs with their parental cells and explore their cell penetration and cytotoxic activity. Anti-HER-2 CARs were stimulated with specific target cells. EVs were isolated from the cell media and characterized for their content and functions. We found that CAR-T EVs contained a mixture of small and large EVs. Stimulated anti-HER-2+ CAR-T EVs expressed lower cytokine levels compared with their parental CAR-T cells (such as interferon gamma). Higher levels of granzyme B were found in CAR-T EVs (≥20 × ) compared with EVs from unstimulated cells (p < 0.001). Anti-HER-2+ CAR-T EVs bound and penetrated specifically into HER-2 expressing target cells. Similar cytotoxic effects measured by caspase-3/7 activity were found in CAR-T cells and their derived EVs. However, while the CAR-T cells induced massive apoptosis during the first 24 h, CAR-T EVs required 60 - 90 h. In summary, CAR-T EVs provide a novel potent immunotherapy approach that may be effective against solid tumors.
Collapse
Affiliation(s)
- Anat Aharon
- Hematology Research Laboratory for Extracellular Vesicles, Hematology Division, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Galit Horn
- Immunology Laboratory, Research & Development Department, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Tali Hana Bar-Lev
- Hematology Research Laboratory for Extracellular Vesicles, Hematology Division, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Einav Zagagi Yohay
- Hematology Research Laboratory for Extracellular Vesicles, Hematology Division, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Tova Waks
- Immunology Laboratory, Research & Development Department, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel.,Immunology Department, The Weizmann Institute, Rehovot, Israel
| | - Maya Levin
- Hematology Research Laboratory for Extracellular Vesicles, Hematology Division, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Naamit Deshet Unger
- Immunology Laboratory, Research & Development Department, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Irit Avivi
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Hematology Division, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Anat Globerson Levin
- Immunology Laboratory, Research & Development Department, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel.,Dotan Center for Advanced Therapies, Tel-Aviv Sourasky Medical Center and Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
119
|
Reilly EC, Sportiello M, Emo KL, Amitrano AM, Jha R, Kumar ABR, Laniewski NG, Yang H, Kim M, Topham DJ. CD49a Identifies Polyfunctional Memory CD8 T Cell Subsets that Persist in the Lungs After Influenza Infection. Front Immunol 2021; 12:728669. [PMID: 34566986 PMCID: PMC8462271 DOI: 10.3389/fimmu.2021.728669] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/23/2021] [Indexed: 12/13/2022] Open
Abstract
CD8 T cell memory offers critical antiviral protection, even in the absence of neutralizing antibodies. The paradigm is that CD8 T cell memory within the lung tissue consists of a mix of circulating TEM cells and non-circulating TRM cells. However, based on our analysis, the heterogeneity within the tissue is much higher, identifying TCM, TEM, TRM, and a multitude of populations which do not perfectly fit these classifications. Further interrogation of the populations shows that TRM cells that express CD49a, both with and without CD103, have increased and diverse effector potential compared with CD49a negative populations. These populations function as a one-man band, displaying antiviral activity, chemokine production, release of GM-CSF, and the ability to kill specific targets in vitro with delayed kinetics compared with effector CD8 T cells. Together, this study establishes that CD49a defines multiple polyfunctional CD8 memory subsets after clearance of influenza infection, which act to eliminate virus in the absence of direct killing, recruit and mature innate immune cells, and destroy infected cells if the virus persists.
Collapse
Affiliation(s)
- Emma C. Reilly
- Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| | - Mike Sportiello
- Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| | - Kris Lambert Emo
- Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| | - Andrea M. Amitrano
- Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| | - Rakshanda Jha
- Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| | - Ashwin B. R. Kumar
- Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| | - Nathan G. Laniewski
- Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, United States
| | - Hongmei Yang
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY, United States
| | - Minsoo Kim
- Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| | - David J. Topham
- Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
120
|
Khalid Z, He L, Yu Q, Breedlove C, Joiner K, Toro H. Enhanced Protection by Recombinant Newcastle Disease Virus Expressing Infectious Bronchitis Virus Spike Ectodomain and Chicken Granulocyte-Macrophage Colony-Stimulating Factor. Avian Dis 2021; 65:364-372. [PMID: 34427409 DOI: 10.1637/aviandiseases-d-21-00032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/18/2021] [Indexed: 11/05/2022]
Abstract
We previously reported that recombinant Newcastle disease virus LaSota (rLS) expressing infectious bronchitis virus (IBV) Arkansas (Ark)-type trimeric spike (S) ectodomain (Se; rLS/ArkSe) provides suboptimal protection against IBV challenge. We have now developed rLS expressing chicken granulocyte-macrophage colony-stimulating factor (GMCSF) and IBV Ark Se in an attempt to enhance vaccine effectiveness. In the current study, we first compared protection conferred by vaccination with rLS/ArkSe and rLS/ArkSe.GMCSF. Vaccinated chickens were challenged with virulent Ark, and protection was determined by clinical signs, viral load, and tracheal histomorphometry. Results showed that coexpression of GMCSF and the Se from rLS significantly reduced tracheal viral load and tracheal lesions compared with chickens vaccinated with rLS/ArkSe. In a second experiment, we evaluated enhancement of cross-protection of a Massachusetts (Mass) attenuated vaccine by priming or boosting with rLS/ArkSe.GMCSF. Vaccinated chickens were challenged with Ark, and protection was evaluated. Results show that priming or boosting with the recombinant virus significantly increased cross-protection conferred by Mass against Ark virulent challenge. Greater reductions of viral loads in both trachea and lachrymal fluids were observed in chickens primed with rLS/ArkSe.GMCSF and boosted with Mass. Consistently, Ark Se antibody levels measured with recombinant Ark Se protein-coated ELISA plates 14 days after boost were significantly higher in these chickens. Unexpectedly, the inverse vaccination scheme, that is, priming with Mass and boosting with the recombinant vaccine, proved somewhat less effective. We concluded that a prime and boost strategy by using rLS/ArkSe.GMCSF and the worldwide ubiquitous Mass attenuated vaccine provides enhanced cross-protection. Thus, rLS/GMCSF coexpressing the Se of regionally relevant IBV serotypes could be used in combination with live Mass to protect against regionally circulating IBV variant strains.
Collapse
Affiliation(s)
- Z Khalid
- Department of Pathobiology, Auburn University College of Veterinary Medicine, Auburn, AL 36830
| | - L He
- United States Department of Agriculture, Agricultural Research Service, United States National Poultry Research Center, Athens, GA 30605.,The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, Luoyang 471023, Henan, China
| | - Q Yu
- United States Department of Agriculture, Agricultural Research Service, United States National Poultry Research Center, Athens, GA 30605
| | - C Breedlove
- Department of Pathobiology, Auburn University College of Veterinary Medicine, Auburn, AL 36830
| | - K Joiner
- Department of Pathobiology, Auburn University College of Veterinary Medicine, Auburn, AL 36830
| | - H Toro
- Department of Pathobiology, Auburn University College of Veterinary Medicine, Auburn, AL 36830,
| |
Collapse
|
121
|
Rocconi RP, Stanbery L, Madeira da Silva L, Barrington RA, Aaron P, Manning L, Horvath S, Wallraven G, Bognar E, Walter A, Nemunaitis J. Long-Term Follow-Up of Gemogenovatucel-T (Vigil) Survival and Molecular Signals of Immune Response in Recurrent Ovarian Cancer. Vaccines (Basel) 2021; 9:vaccines9080894. [PMID: 34452019 PMCID: PMC8402348 DOI: 10.3390/vaccines9080894] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/03/2021] [Accepted: 08/06/2021] [Indexed: 12/20/2022] Open
Abstract
Aim: To determine the relationship between gene expression profile (GEP) and overall survival (OS) by NanoString following treatment with Vigil. Patients and Methods: Recurrent ovarian cancer patients (n = 21) enrolled in prior clinical trials. Results: GEP stratified by TISHIGH vs. TISLOW demonstrated OS benefit (NR vs. 5.8 months HR 0.23; p = 0.0379), and in particular, MHC-II elevated baseline expression was correlated with OS advantage (p = 0.038). Moreover, 1-year OS was 75% in TISHIGH patients vs. 25% in TISLOW (p = 0.03795). OS was also correlated with positive γ-IFN ELISPOT response, 36.8 vs. 23.0 months (HR 0.19, p = 0.0098). Conclusion: Vigil demonstrates OS benefit in correlation with TISHIGH score, elevated MHC-II expression and positive γ-IFN ELISPOT in recurrent ovarian cancer patients.
Collapse
Affiliation(s)
- Rodney P. Rocconi
- Mitchell Cancer Institute, Division of Gynecologic Oncology, University of South Alabama, Mobile, AL 36604, USA;
| | - Laura Stanbery
- Gradalis, Inc., 2545 Golden Bear Drive, Suite 110, Carrollton, TX 75006, USA; (L.S.); (P.A.); (L.M.); (S.H.); (G.W.); (E.B.)
| | - Luciana Madeira da Silva
- Department of Microbiology and Immunology, University of South Alabama, Mobile, AL 36688, USA; (L.M.d.S.); (R.A.B.)
| | - Robert A. Barrington
- Department of Microbiology and Immunology, University of South Alabama, Mobile, AL 36688, USA; (L.M.d.S.); (R.A.B.)
| | - Phylicia Aaron
- Gradalis, Inc., 2545 Golden Bear Drive, Suite 110, Carrollton, TX 75006, USA; (L.S.); (P.A.); (L.M.); (S.H.); (G.W.); (E.B.)
| | - Luisa Manning
- Gradalis, Inc., 2545 Golden Bear Drive, Suite 110, Carrollton, TX 75006, USA; (L.S.); (P.A.); (L.M.); (S.H.); (G.W.); (E.B.)
| | - Staci Horvath
- Gradalis, Inc., 2545 Golden Bear Drive, Suite 110, Carrollton, TX 75006, USA; (L.S.); (P.A.); (L.M.); (S.H.); (G.W.); (E.B.)
| | - Gladice Wallraven
- Gradalis, Inc., 2545 Golden Bear Drive, Suite 110, Carrollton, TX 75006, USA; (L.S.); (P.A.); (L.M.); (S.H.); (G.W.); (E.B.)
| | - Ernest Bognar
- Gradalis, Inc., 2545 Golden Bear Drive, Suite 110, Carrollton, TX 75006, USA; (L.S.); (P.A.); (L.M.); (S.H.); (G.W.); (E.B.)
| | | | - John Nemunaitis
- Gradalis, Inc., 2545 Golden Bear Drive, Suite 110, Carrollton, TX 75006, USA; (L.S.); (P.A.); (L.M.); (S.H.); (G.W.); (E.B.)
- Correspondence:
| |
Collapse
|
122
|
Agina OA, Cheah KT, Sayuti NSA, Shaari MR, Isa NMM, Ajat M, Zamri-Saad M, Mazlan M, Hamzah H. High Granulocyte-Macrophage Colony Stimulating Factor to Interleukin 10 Ratio and Marked Antioxidant Enzyme Activities Predominate in Symptomatic Cattle Naturally Infected with Candidatus Mycoplasma haemobos, Theileria orientalis, Theileria sinensis and Trypanosoma evansi. Animals (Basel) 2021; 11:ani11082235. [PMID: 34438696 PMCID: PMC8388426 DOI: 10.3390/ani11082235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/26/2021] [Accepted: 05/27/2021] [Indexed: 11/16/2022] Open
Abstract
The aim of this study was to measure the serum proinflammatory (IL-12, GM-CSF & IFN-γ) to anti-inflammatory (IL-10, IL-4) cytokine ratio, oxidant (MDA) level and antioxidant enzyme (SOD; GPx) activities after blood parasite infections. The blood and serum samples were obtained from 130 cattle and screened for identity of the infecting blood parasites by conventional PCR. The following blood parasite species were detected: Candidatus Mycoplasma haemobos (70/130); Theileria orientalis (65/130); Theileria sinensis (32/130); Anaplasma marginale (49/130); Anaplasma platys (7/130); and Trypanosoma evansi (4/130). The GM-CSF/IL-10 ratio showed significantly higher values in all the symptomatic blood parasite infected cattle groups except for symptomatic A. platys infected cattle groups. Anti-inflammatory cytokine immune responses were notable findings in symptomatic and asymptomatic cattle infected with C. M. haemobos and T. orientalis characterized by low serum IL-12:IL-10, IFN-γ:IL-10, IL-12:IL-4 and IFN-γ:IL-4 (p < 0.05). Therefore, high serum GM-CSF:IL:10 in the symptomatic blood parasite infected cattle, low serum IL-12:IL-10, IFN-γ:IL-10, IL-12:IL-4 and IFN-γ:IL-4 ratios in asymptomatic cattle, high MDA level, and increased antioxidant enzyme activities could be useful predictive tools for outcome of natural blood parasite infections in cattle.
Collapse
Affiliation(s)
- Onyinyechukwu Ada Agina
- Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Malaysia; (K.T.C.); (N.S.A.S.); (N.M.M.I.); (M.M.)
- Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, University of Nigeria, Nsukka 410001, Nigeria
- Correspondence: (O.A.A.); (H.H.); Tel.: +234-70-3901-0464 (O.A.A.); +60-19-2846-897 (H.H.)
| | - Kim Tho Cheah
- Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Malaysia; (K.T.C.); (N.S.A.S.); (N.M.M.I.); (M.M.)
| | - Nurul Syahirah Ahmad Sayuti
- Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Malaysia; (K.T.C.); (N.S.A.S.); (N.M.M.I.); (M.M.)
| | - Mohd Rosly Shaari
- Animal Science Research Centre, Malaysian Agricultural Research and Development Institute, Serdang 43400, Malaysia;
| | - Nur Mahiza Md Isa
- Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Malaysia; (K.T.C.); (N.S.A.S.); (N.M.M.I.); (M.M.)
| | - Mokrish Ajat
- Department of Pre-Clinical Sciences, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Malaysia;
| | - Mohd Zamri-Saad
- Centre for Ruminant Diseases, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Malaysia;
| | - Mazlina Mazlan
- Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Malaysia; (K.T.C.); (N.S.A.S.); (N.M.M.I.); (M.M.)
| | - Hazilawati Hamzah
- Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Malaysia; (K.T.C.); (N.S.A.S.); (N.M.M.I.); (M.M.)
- Correspondence: (O.A.A.); (H.H.); Tel.: +234-70-3901-0464 (O.A.A.); +60-19-2846-897 (H.H.)
| |
Collapse
|
123
|
Vanni VS, Villanacci R, Salmeri N, Papaleo E, Delprato D, Ottolina J, Rovere-Querini P, Ferrari S, Viganò P, Candiani M. Concomitant autoimmunity may be a predictor of more severe stages of endometriosis. Sci Rep 2021; 11:15372. [PMID: 34321558 PMCID: PMC8319416 DOI: 10.1038/s41598-021-94877-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 06/25/2021] [Indexed: 12/14/2022] Open
Abstract
Pathogenesis of endometriosis is still unclear and a role of both innate and adaptive immune system has been postulated. Some recent findings have revealed an increased risk to have concomitant autoimmune disease in women with endometriosis, but no study so far has investigated whether this association could affect endometriosis severity and stage. We retrospectively reviewed medical patients' notes of women with a confirmed diagnosis of endometriosis who referred to our endometriosis outpatient clinic between January 2015 and December 2019. Cases (endometriosis and an autoimmune disease) were matched in a 1:3 ratio by age and study period with controls (endometriosis without history of autoimmunity). At univariate logistic analysis, concomitant autoimmunity (OR 2.63, 95% CI 1.64-4.21, p < 0.001) and the number of laparoscopic procedures performed (OR 2.81, 95% CI 1.45-5.43, p = 0.002) emerged as factors significantly associated with the likelihood of stage IV endometriosis. In the multivariate logistic regression model, concomitant autoimmunity remained a significant predictor of stage IV endometriosis (OR 2.54, 95% CI 1.57-4.10, p = 0.004), whereas the association between the number of laparoscopic procedures performed and stage IV endometriosis was found to be of borderline-significance (OR 2.70, 95% 1.37-5.30, p = 0.050). Our findings suggest that endometriosis is more severe in patients who are also affected by autoimmune disturbances after controlling for relevant confounders.
Collapse
Affiliation(s)
- Valeria Stella Vanni
- Gynecology/Obstetrics Unit, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy.
- Vita-Salute San Raffaele University, 20132, Milan, Italy.
| | - Roberta Villanacci
- Gynecology/Obstetrics Unit, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Noemi Salmeri
- Gynecology/Obstetrics Unit, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Enrico Papaleo
- Gynecology/Obstetrics Unit, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Diana Delprato
- Gynecology/Obstetrics Unit, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Jessica Ottolina
- Gynecology/Obstetrics Unit, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Patrizia Rovere-Querini
- Vita-Salute San Raffaele University, 20132, Milan, Italy
- Division of Immunology, Transplantation & Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Stefano Ferrari
- Gynecology/Obstetrics Unit, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Paola Viganò
- Reproductive Sciences Lab, Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Massimo Candiani
- Gynecology/Obstetrics Unit, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| |
Collapse
|
124
|
Darden DB, Ghita GL, Wang Z, Stortz JA, Lopez MC, Cox MC, Hawkins RB, Rincon JC, Kelly LS, Fenner BP, Ozrazgat-Baslanti T, Leeuwenburgh C, Bihorac A, Loftus TJ, Moore FA, Brakenridge SC, Baker HV, Bacher R, Mohr AM, Moldawer LL, Efron PA. Chronic Critical Illness Elicits a Unique Circulating Leukocyte Transcriptome in Sepsis Survivors. J Clin Med 2021; 10:3211. [PMID: 34361995 PMCID: PMC8348105 DOI: 10.3390/jcm10153211] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/16/2021] [Accepted: 07/19/2021] [Indexed: 12/15/2022] Open
Abstract
Surgical sepsis has evolved into two major subpopulations: patients who rapidly recover, and those who develop chronic critical illness (CCI). Our primary aim was to determine whether CCI sepsis survivors manifest unique blood leukocyte transcriptomes in late sepsis that differ from transcriptomes among sepsis survivors with rapid recovery. In a prospective cohort study of surgical ICU patients, genome-wide expression analysis was conducted on total leukocytes in human whole blood collected on days 1 and 14 from sepsis survivors who rapidly recovered or developed CCI, defined as ICU length of stay ≥ 14 days with persistent organ dysfunction. Both sepsis patients who developed CCI and those who rapidly recovered exhibited marked changes in genome-wide expression at day 1 which remained abnormal through day 14. Although summary changes in gene expression were similar between CCI patients and subjects who rapidly recovered, CCI patients exhibited differential expression of 185 unique genes compared with rapid recovery patients at day 14 (p < 0.001). The transcriptomic patterns in sepsis survivors reveal an ongoing immune dyscrasia at the level of the blood leukocyte transcriptome, consistent with persistent inflammation and immune suppression. Furthermore, the findings highlight important genes that could compose a prognostic transcriptomic metric or serve as therapeutic targets among sepsis patients that develop CCI.
Collapse
Affiliation(s)
- Dijoia B. Darden
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, USA; (D.B.D.); (J.A.S.); (M.C.C.); (R.B.H.); (J.C.R.); (L.S.K.); (B.P.F.); (T.J.L.); (F.A.M.); (S.C.B.); (A.M.M.); (L.L.M.)
| | - Gabriela L. Ghita
- Department of Biostatistics, University of Florida, Gainesville, FL 32610, USA; (G.L.G.); (Z.W.); (R.B.)
| | - Zhongkai Wang
- Department of Biostatistics, University of Florida, Gainesville, FL 32610, USA; (G.L.G.); (Z.W.); (R.B.)
| | - Julie A. Stortz
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, USA; (D.B.D.); (J.A.S.); (M.C.C.); (R.B.H.); (J.C.R.); (L.S.K.); (B.P.F.); (T.J.L.); (F.A.M.); (S.C.B.); (A.M.M.); (L.L.M.)
| | - Maria-Cecilia Lopez
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, USA; (M.-C.L.); (H.V.B.)
| | - Michael C. Cox
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, USA; (D.B.D.); (J.A.S.); (M.C.C.); (R.B.H.); (J.C.R.); (L.S.K.); (B.P.F.); (T.J.L.); (F.A.M.); (S.C.B.); (A.M.M.); (L.L.M.)
| | - Russell B. Hawkins
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, USA; (D.B.D.); (J.A.S.); (M.C.C.); (R.B.H.); (J.C.R.); (L.S.K.); (B.P.F.); (T.J.L.); (F.A.M.); (S.C.B.); (A.M.M.); (L.L.M.)
| | - Jaimar C. Rincon
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, USA; (D.B.D.); (J.A.S.); (M.C.C.); (R.B.H.); (J.C.R.); (L.S.K.); (B.P.F.); (T.J.L.); (F.A.M.); (S.C.B.); (A.M.M.); (L.L.M.)
| | - Lauren S. Kelly
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, USA; (D.B.D.); (J.A.S.); (M.C.C.); (R.B.H.); (J.C.R.); (L.S.K.); (B.P.F.); (T.J.L.); (F.A.M.); (S.C.B.); (A.M.M.); (L.L.M.)
| | - Brittany P. Fenner
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, USA; (D.B.D.); (J.A.S.); (M.C.C.); (R.B.H.); (J.C.R.); (L.S.K.); (B.P.F.); (T.J.L.); (F.A.M.); (S.C.B.); (A.M.M.); (L.L.M.)
| | - Tezcan Ozrazgat-Baslanti
- Department of Aging and Geriatric Research, University of Florida, Gainesville, FL 32610, USA; (T.O.-B.); (C.L.)
| | - Christiaan Leeuwenburgh
- Department of Aging and Geriatric Research, University of Florida, Gainesville, FL 32610, USA; (T.O.-B.); (C.L.)
| | - Azra Bihorac
- Department of Medicine, University of Florida College of Medicine, Gainesville, FL 32610, USA;
| | - Tyler J. Loftus
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, USA; (D.B.D.); (J.A.S.); (M.C.C.); (R.B.H.); (J.C.R.); (L.S.K.); (B.P.F.); (T.J.L.); (F.A.M.); (S.C.B.); (A.M.M.); (L.L.M.)
| | - Frederick A. Moore
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, USA; (D.B.D.); (J.A.S.); (M.C.C.); (R.B.H.); (J.C.R.); (L.S.K.); (B.P.F.); (T.J.L.); (F.A.M.); (S.C.B.); (A.M.M.); (L.L.M.)
| | - Scott C. Brakenridge
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, USA; (D.B.D.); (J.A.S.); (M.C.C.); (R.B.H.); (J.C.R.); (L.S.K.); (B.P.F.); (T.J.L.); (F.A.M.); (S.C.B.); (A.M.M.); (L.L.M.)
| | - Henry V. Baker
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, USA; (M.-C.L.); (H.V.B.)
| | - Rhonda Bacher
- Department of Biostatistics, University of Florida, Gainesville, FL 32610, USA; (G.L.G.); (Z.W.); (R.B.)
| | - Alicia M. Mohr
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, USA; (D.B.D.); (J.A.S.); (M.C.C.); (R.B.H.); (J.C.R.); (L.S.K.); (B.P.F.); (T.J.L.); (F.A.M.); (S.C.B.); (A.M.M.); (L.L.M.)
| | - Lyle L. Moldawer
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, USA; (D.B.D.); (J.A.S.); (M.C.C.); (R.B.H.); (J.C.R.); (L.S.K.); (B.P.F.); (T.J.L.); (F.A.M.); (S.C.B.); (A.M.M.); (L.L.M.)
| | - Philip A. Efron
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, USA; (D.B.D.); (J.A.S.); (M.C.C.); (R.B.H.); (J.C.R.); (L.S.K.); (B.P.F.); (T.J.L.); (F.A.M.); (S.C.B.); (A.M.M.); (L.L.M.)
| |
Collapse
|
125
|
Oncolytic HSV: Underpinnings of Tumor Susceptibility. Viruses 2021; 13:v13071408. [PMID: 34372614 PMCID: PMC8310378 DOI: 10.3390/v13071408] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/03/2021] [Accepted: 07/14/2021] [Indexed: 12/12/2022] Open
Abstract
Oncolytic herpes simplex virus (oHSV) is a therapeutic modality that has seen substantial success for the treatment of cancer, though much remains to be improved. Commonly attenuated through the deletion or alteration of the γ134.5 neurovirulence gene, the basis for the success of oHSV relies in part on the malignant silencing of cellular pathways critical for limiting these viruses in healthy host tissue. However, only recently have the molecular mechanisms underlying the success of these treatments begun to emerge. Further clarification of these mechanisms can strengthen rational design approaches to develop the next generation of oHSV. Herein, we review our current understanding of the molecular basis for tumor susceptibility to γ134.5-attenuated oHSV, with particular focus on the malignant suppression of nucleic acid sensing, along with strategies meant to improve the clinical efficacy of these therapeutic viruses.
Collapse
|
126
|
Cosenza M, Sacchi S, Pozzi S. Cytokine Release Syndrome Associated with T-Cell-Based Therapies for Hematological Malignancies: Pathophysiology, Clinical Presentation, and Treatment. Int J Mol Sci 2021; 22:ijms22147652. [PMID: 34299273 PMCID: PMC8305850 DOI: 10.3390/ijms22147652] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/15/2021] [Accepted: 07/15/2021] [Indexed: 01/08/2023] Open
Abstract
Cytokines are a broad group of small regulatory proteins with many biological functions involved in regulating the hematopoietic and immune systems. However, in pathological conditions, hyperactivation of the cytokine network constitutes the fundamental event in cytokine release syndrome (CRS). During the last few decades, the development of therapeutic monoclonal antibodies and T-cell therapies has rapidly evolved, and CRS can be a serious adverse event related to these treatments. CRS is a set of toxic adverse events that can be observed during infection or following the administration of antibodies for therapeutic purposes and, more recently, during T-cell-engaging therapies. CRS is triggered by on-target effects induced by binding of chimeric antigen receptor (CAR) T cells or bispecific antibody to its antigen and by subsequent activation of bystander immune and non-immune cells. CRS is associated with high circulating concentrations of several pro-inflammatory cytokines, including interleukins, interferons, tumor necrosis factors, colony-stimulating factors, and transforming growth factors. Recently, considerable developments have been achieved with regard to preventing and controlling CRS, but it remains an unmet clinical need. This review comprehensively summarizes the pathophysiology, clinical presentation, and treatment of CRS caused by T-cell-engaging therapies utilized in the treatment of hematological malignancies.
Collapse
|
127
|
Mohapatra S, Kundu AK, Mishra SR, Senapati S, Jyotiranjan T, Panda G. HSF1 and GM-CSF expression, its association with cardiac health, and assessment of organ function during heat stress in crossbred Jersey cattle. Res Vet Sci 2021; 139:200-210. [PMID: 34358923 DOI: 10.1016/j.rvsc.2021.07.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/16/2021] [Accepted: 07/14/2021] [Indexed: 11/25/2022]
Abstract
The study aimed to evaluate the differential expression of HSF1 and GM-CSF mRNA in PBMCs and correlate it with myocardial injury in crossbred Jersey heifers during heat stress. The study also assessed the effect of heat stress on cardiac electrical activity, vascular health, liver function and kidney function. The experiment was conducted in two phases: for heat stressed animals; HS in June (THI ranged from 80.0 to 89.8) and for control group i.e. not exposed to heat stress in January (THI ranged between 70.1 and 71.4). Results of the study revealed that the relative abundance of HSF1 and GM-CSF mRNA increased significantly (P < 0.05) in HS. Serum cardiac biomarkers such as CK-MB, AST and CRP were significantly elevated (P < 0.05) in HS. cTnI was detected 'positive' in nineteen out of twenty four cases in HS. Correlation of HSF1 and GM-CSF expression with concentration of LDH, CKMB, CRP and AST in HS was negative but non-significant (P > 0.05). Significant (P < 0.05) ECG findings in HS were increased heart rate, decreased RR interval, decreased PR interval, decreased QRS amplitude and decreased amplitude of P wave. Marked reduction (P < 0.05) in serum cholesterol and triglyceride levels was observed in HS. ALP, AST, bilirubin and urea levels in serum were significantly elevated (P < 0.05) in HS. In conclusion, cardiac enzymes in serum were significantly elevated in HS indicating myocardial injury. HSF1 and GM-CSF mRNA expression alone was inadequate in conferring cytoprotection to cardiac cells in HS. Cardiac electrical activity, vascular status, liver and kidney function were significantly altered in HS.
Collapse
Affiliation(s)
- Swagat Mohapatra
- Department of Veterinary Physiology, C.V.Sc. & A.H., O.U.A.T., Bhubaneswar, Odisha, India
| | - Akshaya Kumar Kundu
- Department of Veterinary Physiology, C.V.Sc. & A.H., O.U.A.T., Bhubaneswar, Odisha, India
| | - Smruti Ranjan Mishra
- Department of Veterinary Physiology, C.V.Sc. & A.H., O.U.A.T., Bhubaneswar, Odisha, India
| | | | - Tushar Jyotiranjan
- Department of Veterinary Physiology, C.V.Sc. & A.H., O.U.A.T., Bhubaneswar, Odisha, India
| | - Gopalaxmi Panda
- Department of Biochemistry, SCB Medical College, Cuttack, Odisha, India.
| |
Collapse
|
128
|
Zhang Y, Zhao Y, Li Q, Wang Y. Macrophages, as a Promising Strategy to Targeted Treatment for Colorectal Cancer Metastasis in Tumor Immune Microenvironment. Front Immunol 2021; 12:685978. [PMID: 34326840 PMCID: PMC8313969 DOI: 10.3389/fimmu.2021.685978] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 06/24/2021] [Indexed: 12/16/2022] Open
Abstract
The tumor immune microenvironment plays a vital role in the metastasis of colorectal cancer. As one of the most important immune cells, macrophages act as phagocytes, patrol the surroundings of tissues, and remove invading pathogens and cell debris to maintain tissue homeostasis. Significantly, macrophages have a characteristic of high plasticity and can be classified into different subtypes according to the different functions, which can undergo reciprocal phenotypic switching induced by different types of molecules and signaling pathways. Macrophages regulate the development and metastatic potential of colorectal cancer by changing the tumor immune microenvironment. In tumor tissues, the tumor-associated macrophages usually play a tumor-promoting role in the tumor immune microenvironment, and they are also associated with poor prognosis. This paper reviews the mechanisms and stimulating factors of macrophages in the process of colorectal cancer metastasis and intends to indicate that targeting macrophages may be a promising strategy in colorectal cancer treatment.
Collapse
Affiliation(s)
- Yingru Zhang
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiyang Zhao
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qi Li
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Wang
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
129
|
Ellingsen EB, Mangsbo SM, Hovig E, Gaudernack G. Telomerase as a Target for Therapeutic Cancer Vaccines and Considerations for Optimizing Their Clinical Potential. Front Immunol 2021; 12:682492. [PMID: 34290704 PMCID: PMC8288190 DOI: 10.3389/fimmu.2021.682492] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 06/21/2021] [Indexed: 12/14/2022] Open
Abstract
Telomerase-based therapeutic cancer vaccines (TCVs) have been under clinical investigation for the past two decades. Despite past failures, TCVs have gained renewed enthusiasm for their potential to improve the efficacy of checkpoint inhibition. Telomerase stands as an attractive target for TCVs due to its almost universal presence in cancer and its essential function promoting tumor growth. Herein, we review tumor telomerase biology that may affect the efficacy of therapeutic vaccination and provide insights on optimal vaccine design and treatment combinations. Tumor types possessing mechanisms of increased telomerase expression combined with an immune permissive tumor microenvironment are expected to increase the therapeutic potential of telomerase-targeting cancer vaccines. Regardless, rational treatment combinations, such as checkpoint inhibitors, are likely necessary to bring out the true clinical potential of TCVs.
Collapse
Affiliation(s)
- Espen Basmo Ellingsen
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo, Norway.,Faculty of Medicine, University of Oslo, Oslo, Norway.,Research and Development, Ultimovacs ASA, Oslo, Norway
| | - Sara M Mangsbo
- Research and Development, Ultimovacs AB, Uppsala, Sweden.,Department of Pharmaceutical Biosciences, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Eivind Hovig
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo, Norway.,Centre for Bioinformatics, Department of Informatics, University of Oslo, Oslo, Norway
| | | |
Collapse
|
130
|
Jenner AL, Aogo RA, Alfonso S, Crowe V, Deng X, Smith AP, Morel PA, Davis CL, Smith AM, Craig M. COVID-19 virtual patient cohort suggests immune mechanisms driving disease outcomes. PLoS Pathog 2021; 17:e1009753. [PMID: 34260666 PMCID: PMC8312984 DOI: 10.1371/journal.ppat.1009753] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 07/26/2021] [Accepted: 06/24/2021] [Indexed: 12/11/2022] Open
Abstract
To understand the diversity of immune responses to SARS-CoV-2 and distinguish features that predispose individuals to severe COVID-19, we developed a mechanistic, within-host mathematical model and virtual patient cohort. Our results suggest that virtual patients with low production rates of infected cell derived IFN subsequently experienced highly inflammatory disease phenotypes, compared to those with early and robust IFN responses. In these in silico patients, the maximum concentration of IL-6 was also a major predictor of CD8+ T cell depletion. Our analyses predicted that individuals with severe COVID-19 also have accelerated monocyte-to-macrophage differentiation mediated by increased IL-6 and reduced type I IFN signalling. Together, these findings suggest biomarkers driving the development of severe COVID-19 and support early interventions aimed at reducing inflammation.
Collapse
Affiliation(s)
- Adrianne L. Jenner
- Sainte-Justine University Hospital Research Centre, Montréal, Québec, Canada
- Department of Mathematics and Statistics, Université de Montréal, Montréal, Québec, Canada
| | - Rosemary A. Aogo
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Sofia Alfonso
- Department of Physiology, McGill University, Montréal, Québec, Canada
| | - Vivienne Crowe
- Department of Mathematics and Statistics, Concordia University, Montréal, Québec, Canada
| | - Xiaoyan Deng
- Sainte-Justine University Hospital Research Centre, Montréal, Québec, Canada
- Department of Mathematics and Statistics, Université de Montréal, Montréal, Québec, Canada
| | - Amanda P. Smith
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Penelope A. Morel
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Courtney L. Davis
- Natural Science Division, Pepperdine University, Malibu, California, United States of America
| | - Amber M. Smith
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Morgan Craig
- Sainte-Justine University Hospital Research Centre, Montréal, Québec, Canada
- Department of Mathematics and Statistics, Université de Montréal, Montréal, Québec, Canada
- Department of Physiology, McGill University, Montréal, Québec, Canada
| |
Collapse
|
131
|
Valigura HC, Leatherwood JL, Martinez RE, Norton S, White-Springer SH. Dietary supplementation of a Saccharomyces cerevisiae fermentation product attenuates exercise-induced stress markers in young horses. J Anim Sci 2021; 99:6310836. [PMID: 34181712 PMCID: PMC8521742 DOI: 10.1093/jas/skab199] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 06/25/2021] [Indexed: 11/13/2022] Open
Abstract
Mitigation of exercise-induced stress is of key interest in determining ways to optimize performance horse health. To test the hypothesis that dietary supplementation of a Saccharomyces cerevisiae fermentation product would decrease markers of exercise-induced stress and inflammation in young horses, Quarter Horse yearlings (mean ± SD; 9 ± 1 mo) were randomly assigned to receive either no supplementation (CON; n = 8) or 21 g/d S. cerevisiae fermentation product (10.5 g/feeding twice daily; SCFP; n = 10) top-dressed on a basal diet of custom-formulated grain as well as ad libitum Coastal bermudagrass hay. After 8 wk of dietary treatments, horses underwent a 2-h submaximal exercise test (SET) on a free-stall mechanical exerciser. Serum was collected before dietary treatment supplementation (week 0), at week 8 pre-SET, and 0, 1, and 6 h post-SET and analyzed for concentrations of cortisol and serum amyloid A (SAA) by commercial enzyme-linked immunosorbent assay (ELISA) and for cytokine concentrations by commercial bead-based ELISA. Data were analyzed using linear models with repeated measures in SAS v9.4. From week 0 to 8 (pre-SET), serum cortisol decreased (P = 0.01) and SAA did not change, but neither were affected by diet. Serum concentrations of all cytokines decreased from week 0 to 8 (P ≤ 0.008), but granulocyte colony-stimulating factor (G-CSF), granulocyte-macrophage colony-stimulating factor, and interleukin-8 (IL-8) decreased to a greater extent in CON than in SCFP horses (P ≤0.003). In response to the week 8 SET, serum cortisol increased in all horses (P < 0.0001) but returned to pre-SET levels by 1 h post-SET in horses receiving SCFP. At 6 h post-SET, cortisol concentrations in CON horses returned to pre-SET concentrations, whereas cortisol declined further in SCFP horses to below pre-SET levels (P = 0.0002) and lower than CON (P = 0.003) at that time point. SAA increased at 6 h post-SET in CON (P < 0.0001) but was unchanged through 6 h in SCFP horses. All cytokines except G-CSF increased in response to the SET (P < 0.0001) but showed differing response patterns. Concentrations of IL-1β, IL-6, and tumor necrosis factor-alpha were lesser (P ≤ 0.05), and concentrations of G-CSF and IL-18 tended to be lesser (P ≤ 0.09) in SCFP compared with CON horses throughout recovery from the SET. In summary, 8 wk of dietary supplementation with 21 g/d of SCFP may mitigate cellular stress following a single, prolonged submaximal exercise bout in young horses.
Collapse
Affiliation(s)
- Hannah C Valigura
- Department of Animal Science, Texas A&M University and Texas A&M AgriLife Research, College Station, TX, 77843, US
| | - Jessica L Leatherwood
- Department of Animal Science, Texas A&M University and Texas A&M AgriLife Research, College Station, TX, 77843, US
| | - Rafael E Martinez
- Department of Animal Science, Texas A&M University and Texas A&M AgriLife Research, College Station, TX, 77843, US
| | | | - Sarah H White-Springer
- Department of Animal Science, Texas A&M University and Texas A&M AgriLife Research, College Station, TX, 77843, US
| |
Collapse
|
132
|
Harris KM, Clements MA, Kwilasz AJ, Watkins LR. T cell transgressions: Tales of T cell form and function in diverse disease states. Int Rev Immunol 2021; 41:475-516. [PMID: 34152881 PMCID: PMC8752099 DOI: 10.1080/08830185.2021.1921764] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/17/2021] [Accepted: 04/20/2021] [Indexed: 01/03/2023]
Abstract
Insights into T cell form, function, and dysfunction are rapidly evolving. T cells have remarkably varied effector functions including protecting the host from infection, activating cells of the innate immune system, releasing cytokines and chemokines, and heavily contributing to immunological memory. Under healthy conditions, T cells orchestrate a finely tuned attack on invading pathogens while minimizing damage to the host. The dark side of T cells is that they also exhibit autoreactivity and inflict harm to host cells, creating autoimmunity. The mechanisms of T cell autoreactivity are complex and dynamic. Emerging research is elucidating the mechanisms leading T cells to become autoreactive and how such responses cause or contribute to diverse disease states, both peripherally and within the central nervous system. This review provides foundational information on T cell development, differentiation, and functions. Key T cell subtypes, cytokines that create their effector roles, and sex differences are highlighted. Pathological T cell contributions to diverse peripheral and central disease states, arising from errors in reactivity, are highlighted, with a focus on multiple sclerosis, rheumatoid arthritis, osteoarthritis, neuropathic pain, and type 1 diabetes.
Collapse
Affiliation(s)
- Kevin M. Harris
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado, Boulder, CO U.S.A
| | - Madison A. Clements
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado, Boulder, CO U.S.A
| | - Andrew J. Kwilasz
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado, Boulder, CO U.S.A
| | - Linda R. Watkins
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado, Boulder, CO U.S.A
| |
Collapse
|
133
|
Kiełbasa A, Gadzała-Kopciuch R, Buszewski B. Cytokines-Biogenesis and Their Role in Human Breast Milk and Determination. Int J Mol Sci 2021; 22:6238. [PMID: 34207900 PMCID: PMC8229712 DOI: 10.3390/ijms22126238] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 06/01/2021] [Accepted: 06/04/2021] [Indexed: 12/13/2022] Open
Abstract
Cytokines play a huge role in many biological processes. Their production, release and interactions are subject to a very complex mechanism. Cytokines are produced by all types of cells, they function very differently and they are characterized by synergism in action, antagonism, and aggregation activity, opposing action of one cytokine, overlapping activity, induction of another cytokine, inhibition of cytokine synthesis at the mRNA level as well as autoregulation-stimulation or inhibition of own production. The predominance of pro-inflammatory cytokines leads to a systemic inflammatory response, and anti-inflammatory-to an anti-inflammatory response. They regulate the organism's immune response and protect it against sudden disturbances in homeostasis. The synthesis and activity of cytokines are influenced by the central nervous system through the endocrine system (pituitary gland, adrenal glands).
Collapse
Affiliation(s)
- Anna Kiełbasa
- Department of Environmental Chemistry and Bioanalysis, Faculty of Chemistry, Nicolaus Copernicus University in Toruń, ul. Gagarina 7, 87-100 Toruń, Poland; (A.K.); (B.B.)
| | - Renata Gadzała-Kopciuch
- Department of Environmental Chemistry and Bioanalysis, Faculty of Chemistry, Nicolaus Copernicus University in Toruń, ul. Gagarina 7, 87-100 Toruń, Poland; (A.K.); (B.B.)
- Interdisciplinary Centre of Modern Technologies, Group for Separation and Bioanalytical Methods (Bio-Sep) Nicolaus Copernicus University in Toruń, ul. Wileńska 4, 87-100 Toruń, Poland
| | - Bogusław Buszewski
- Department of Environmental Chemistry and Bioanalysis, Faculty of Chemistry, Nicolaus Copernicus University in Toruń, ul. Gagarina 7, 87-100 Toruń, Poland; (A.K.); (B.B.)
- Interdisciplinary Centre of Modern Technologies, Group for Separation and Bioanalytical Methods (Bio-Sep) Nicolaus Copernicus University in Toruń, ul. Wileńska 4, 87-100 Toruń, Poland
| |
Collapse
|
134
|
Cham J, Zhang L, Kwek S, Paciorek A, He T, Fong G, Oh DY, Fong L. Combination immunotherapy induces distinct T-cell repertoire responses when administered to patients with different malignancies. J Immunother Cancer 2021; 8:jitc-2019-000368. [PMID: 32376721 PMCID: PMC7223469 DOI: 10.1136/jitc-2019-000368] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2020] [Indexed: 12/18/2022] Open
Abstract
Background CTLA-4 blockade with ipilimumab is Food and Drug Administration-approved for melanoma as a monotherapy and has been shown to modulate the circulating T-cell repertoire. We have previously reported clinical trials combining CTLA-4 blockade with granulocyte-macrophage colony-stimulating factor (GM-CSF) in metastatic melanoma patients and in metastatic castration resistant prostate cancer (mCRPC) patients. Here, we investigate the effect that cancer type has on circulating T cells in metastatic melanoma and mCRPC patients, treated with ipilimumab and GM-CSF. Methods We used next-generation sequencing of T-cell receptors (TCR) to compare the circulating T cells of melanoma and mCRPC patients receiving the same treatment with ipilimumab and GM-CSF by Wilcoxon rank sum test. Flow cytometry was utilized to investigate specific T-cell populations. TCR sequencing results were correlated with each T-cell subpopulation by Spearman’s rank correlation coefficient. Of note, 14 metastatic melanoma patients had samples available for TCR sequencing and 21 had samples available for flow cytometry analysis; 37 mCRPC patients had samples available for sequencing of whom 22 have TCR data available at both timepoints; 20 of these patients had samples available for flow cytometry analysis and 16 had data available at both timepoints. Results While melanoma and mCRPC patients had similar pretreatment circulating T-cell counts, treatment induces greater expansion of circulating T cells in melanoma patients. Metastatic melanoma patients have a higher proportion of clones that increased more than fourfold after the treatment compared with mCRPC patients (18.9% vs 11.0%, p=0.017). Additionally, melanoma patients compared with mCRPC patients had a higher ratio of convergent frequency (1.22 vs 0.60, p=0.012). Decreases in clonality induced by treatment are associated with baseline CD8+ T-cell counts in both patient groups, but are more pronounced in the melanoma patients (r=−0.81, p<0.001 vs r=−0.59, p=0.02). Trial registration numbers NCT00064129; NCT01363206.
Collapse
Affiliation(s)
- Jason Cham
- Medicine, University of California San Francisco, San Francisco, California, USA
| | - Li Zhang
- Medicine, University of California San Francisco, San Francisco, California, USA
| | - Serena Kwek
- Medicine, University of California San Francisco, San Francisco, California, USA
| | - Alan Paciorek
- Medicine, University of California San Francisco, San Francisco, California, USA
| | - Tao He
- San Francisco State University, San Francisco, California, USA
| | - Grant Fong
- Medicine, University of California San Francisco, San Francisco, California, USA
| | - David Y Oh
- Medicine, University of California San Francisco, San Francisco, California, USA
| | - Lawrence Fong
- Medicine, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
135
|
Zhang N, Foiret J, Kheirolomoom A, Liu P, Feng Y, Tumbale S, Raie M, Wu B, Wang J, Fite BZ, Dai Z, Ferrara KW. Optimization of microbubble-based DNA vaccination with low-frequency ultrasound for enhanced cancer immunotherapy. ADVANCED THERAPEUTICS 2021; 4. [PMID: 34632048 DOI: 10.1002/adtp.202100033] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Immunotherapy is an important cancer treatment strategy; nevertheless, the lack of robust immune cell infiltration in the tumor microenvironment remains a factor in limiting patient response rates. In vivo gene delivery protocols can amplify immune responses and sensitize tumors to immunotherapies, yet non-viral transfection methods often sacrifice transduction efficiency for improved safety tolerance. To improve transduction efficiency, we optimized a strategy employing low ultrasound transmission frequency-induced bubble oscillation to introduce plasmids into tumor cells. Differential centrifugation isolated size-specific microbubbles. The diameter of the small microbubble population was 1.27 ± 0.89 μm and that of larger population was 4.23 ± 2.27 μm. Upon in vitro insonation with the larger microbubble population, 29.7% of cancer cells were transfected with DNA plasmids, higher than that with smaller microbubbles (18.9%, P <0.05) or positive control treatments with a commercial transfection reagent (12%, P < 0.01). After 48 h, gene expression increased more than two-fold in tumors treated with large, as compared with small, microbubbles. Furthermore, the immune response, including tumor infiltration of CD8+ T cells and F4/80+ macrophages, was enhanced. We believe that this safe and efficacious method can improve preclinical procedures and outcomes for DNA vaccines in cancer immunotherapy in the future.
Collapse
Affiliation(s)
- Nisi Zhang
- Department of Radiology, Stanford University, Palo Alto, CA, USA
| | - Josquin Foiret
- Department of Radiology, Stanford University, Palo Alto, CA, USA
| | | | - Pei Liu
- Department of Radiology, Stanford University, Palo Alto, CA, USA
| | - Yi Feng
- Department of Radiology, Stanford University, Palo Alto, CA, USA
| | - Spencer Tumbale
- Department of Radiology, Stanford University, Palo Alto, CA, USA
| | - Marina Raie
- Department of Radiology, Stanford University, Palo Alto, CA, USA
| | - Bo Wu
- Department of Radiology, Stanford University, Palo Alto, CA, USA
| | - James Wang
- Department of Radiology, Stanford University, Palo Alto, CA, USA
| | - Brett Z Fite
- Department of Radiology, Stanford University, Palo Alto, CA, USA
| | - Zhifei Dai
- Department of Engineering, Peking University, Beijing, China
| | | |
Collapse
|
136
|
Tay BQ, Wright Q, Ladwa R, Perry C, Leggatt G, Simpson F, Wells JW, Panizza BJ, Frazer IH, Cruz JLG. Evolution of Cancer Vaccines-Challenges, Achievements, and Future Directions. Vaccines (Basel) 2021; 9:vaccines9050535. [PMID: 34065557 PMCID: PMC8160852 DOI: 10.3390/vaccines9050535] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/14/2021] [Accepted: 05/15/2021] [Indexed: 02/06/2023] Open
Abstract
The development of cancer vaccines has been intensively pursued over the past 50 years with modest success. However, recent advancements in the fields of genetics, molecular biology, biochemistry, and immunology have renewed interest in these immunotherapies and allowed the development of promising cancer vaccine candidates. Numerous clinical trials testing the response evoked by tumour antigens, differing in origin and nature, have shed light on the desirable target characteristics capable of inducing strong tumour-specific non-toxic responses with increased potential to bring clinical benefit to patients. Novel delivery methods, ranging from a patient’s autologous dendritic cells to liposome nanoparticles, have exponentially increased the abundance and exposure of the antigenic payloads. Furthermore, growing knowledge of the mechanisms by which tumours evade the immune response has led to new approaches to reverse these roadblocks and to re-invigorate previously suppressed anti-tumour surveillance. The use of new drugs in combination with antigen-based therapies is highly targeted and may represent the future of cancer vaccines. In this review, we address the main antigens and delivery methods used to develop cancer vaccines, their clinical outcomes, and the new directions that the vaccine immunotherapy field is taking.
Collapse
Affiliation(s)
- Ban Qi Tay
- Faculty of Medicine, Diamantina Institute, University of Queensland, Brisbane, QLD 4102, Australia; (B.Q.T.); (Q.W.); (G.L.); (F.S.); (J.W.W.); (I.H.F.)
| | - Quentin Wright
- Faculty of Medicine, Diamantina Institute, University of Queensland, Brisbane, QLD 4102, Australia; (B.Q.T.); (Q.W.); (G.L.); (F.S.); (J.W.W.); (I.H.F.)
| | - Rahul Ladwa
- Department of Medical Oncology, Princess Alexandra Hospital, Brisbane, QLD 4102, Australia;
- Faculty of Medicine, University of Queensland, Woolloongabba, QLD 4102, Australia; (C.P.); (B.J.P.)
| | - Christopher Perry
- Faculty of Medicine, University of Queensland, Woolloongabba, QLD 4102, Australia; (C.P.); (B.J.P.)
- Department of Otolaryngology, Princess Alexandra Hospital, Brisbane, QLD 4102, Australia
| | - Graham Leggatt
- Faculty of Medicine, Diamantina Institute, University of Queensland, Brisbane, QLD 4102, Australia; (B.Q.T.); (Q.W.); (G.L.); (F.S.); (J.W.W.); (I.H.F.)
| | - Fiona Simpson
- Faculty of Medicine, Diamantina Institute, University of Queensland, Brisbane, QLD 4102, Australia; (B.Q.T.); (Q.W.); (G.L.); (F.S.); (J.W.W.); (I.H.F.)
| | - James W. Wells
- Faculty of Medicine, Diamantina Institute, University of Queensland, Brisbane, QLD 4102, Australia; (B.Q.T.); (Q.W.); (G.L.); (F.S.); (J.W.W.); (I.H.F.)
| | - Benedict J. Panizza
- Faculty of Medicine, University of Queensland, Woolloongabba, QLD 4102, Australia; (C.P.); (B.J.P.)
- Department of Otolaryngology, Princess Alexandra Hospital, Brisbane, QLD 4102, Australia
| | - Ian H. Frazer
- Faculty of Medicine, Diamantina Institute, University of Queensland, Brisbane, QLD 4102, Australia; (B.Q.T.); (Q.W.); (G.L.); (F.S.); (J.W.W.); (I.H.F.)
| | - Jazmina L. G. Cruz
- Faculty of Medicine, Diamantina Institute, University of Queensland, Brisbane, QLD 4102, Australia; (B.Q.T.); (Q.W.); (G.L.); (F.S.); (J.W.W.); (I.H.F.)
- Correspondence: ; Tel.: +61-0478912737
| |
Collapse
|
137
|
Patel A, Carr MJ, Sun J, Zager JS. In-transit metastatic cutaneous melanoma: current management and future directions. Clin Exp Metastasis 2021; 39:201-211. [PMID: 33999365 DOI: 10.1007/s10585-021-10100-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 04/22/2021] [Indexed: 12/22/2022]
Abstract
Management of in-transit melanoma encompasses a variety of possible treatment pathways and modalities. Depending on the location of disease, number of lesions, burden of disease and patient preference and characteristics, some treatments may be more beneficial than others. After full body radiographic staging is performed to rule out metastatic disease, curative therapy may be performed through surgical excision, intraarterial regional perfusion and infusion therapies, intralesional injections, systemic therapies or various combinations of any of these. While wide excision is limited in indication to superficial lesions that are few in number, the other listed therapies may be effective in treating unresectable disease. Where intraarterial perfusion based therapies have been shown to successfully treat extremity disease, injectable therapies can be used in lesions of the head and neck. Although systemic therapies for in-transit melanoma have limited specific data to support their primary use for in-transit disease, there are patients who may not be eligible for any of the other options, and current clinical trials are exploring the use of concurrent and sequential use of regional and systemic therapies with early results suggesting a synergistic benefit for oncologic response and outcomes.
Collapse
Affiliation(s)
- Ayushi Patel
- Department of Oncologic Sciences, University of South Florida Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, USA
| | - Michael J Carr
- Department of Cutaneous Oncology, Moffitt Cancer Center, 10920 North McKinley Drive, Tampa, FL, 33612, USA
| | - James Sun
- Department of Cutaneous Oncology, Moffitt Cancer Center, 10920 North McKinley Drive, Tampa, FL, 33612, USA.,Department of Surgery, University Hospitals, Cleveland Medical Center, Cleveland, OH, USA
| | - Jonathan S Zager
- Department of Oncologic Sciences, University of South Florida Morsani College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, USA. .,Department of Cutaneous Oncology, Moffitt Cancer Center, 10920 North McKinley Drive, Tampa, FL, 33612, USA.
| |
Collapse
|
138
|
The transition of M-CSF-derived human macrophages to a growth-promoting phenotype. Blood Adv 2021; 4:5460-5472. [PMID: 33166408 DOI: 10.1182/bloodadvances.2020002683] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/27/2020] [Indexed: 12/12/2022] Open
Abstract
Stimulated macrophages are potent producers of inflammatory mediators. This activity is highly regulated, in part, by resolving molecules to prevent tissue damage. In this study, we demonstrate that inflammation induced by Toll-like receptor stimulation is followed by the upregulation of receptors for adenosine (Ado) and prostaglandin E2 (PGE2), which help terminate macrophage activation and initiate tissue remodeling and angiogenesis. Macrophages can be hematopoietically derived from monocytes in response to 2 growth factors: macrophage colony-stimulating factor (M-CSF) and granulocyte-macrophage colony-stimulating factor (GM-CSF). We examine how exposure to either of these differentiation factors shapes the macrophage response to resolving molecules. We analyzed the transcriptomes of human monocyte-derived macrophages stimulated in the presence of Ado or PGE2 and demonstrated that, in macrophages differentiated in M-CSF, Ado and PGE2 induce a shared transcriptional program involving the downregulation of inflammatory mediators and the upregulation of growth factors. In contrast, macrophages generated in GM-CSF fail to convert to a growth-promoting phenotype, which we attribute to the suppression of receptors for Ado and PGE2 and lower production of these endogenous regulators. These observations indicate that M-CSF macrophages are better prepared to transition to a program of tissue repair, whereas GM-CSF macrophages undergo more profound activation. We implicate the differential sensitivity to pro-resolving mediators as a contributor to these divergent phenotypes. This research highlights a number of molecular targets that can be exploited to regulate the strength and duration of macrophage activation.
Collapse
|
139
|
Nold C, Esteves K, Jensen T, Vella AT. Granulocyte-macrophage colony-stimulating factor initiates amniotic membrane rupture and preterm birth in a mouse model. Am J Reprod Immunol 2021; 86:e13424. [PMID: 33772943 DOI: 10.1111/aji.13424] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/05/2021] [Accepted: 03/23/2021] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVE Preterm premature rupture of membranes is associated with 30% of all preterm births. The weakening of amniotic membranes is associated with an increase in matrix metallopeptidases (MMPs) along with a decrease in their inhibitors, tissue inhibitor metallopeptidases (TIMPs). Additionally, granulocyte-macrophage colony-stimulating factor (GM-CSF) has been shown to weaken fetal membranes in-vitro. We hypothesize pregnant mice treated with GM-CSF lead to increased MMPs:TIMPs resulting in membrane rupture and preterm birth. STUDY DESIGN Pregnant CD-1 mice on gestational day 17 received either an intrauterine injection of GM-CSF or vehicle control. A second series of mice were administered an intrauterine injection of Lipopolysaccharide along with either anti-mouse GM-CSF or control antibody. Mice were evaluated for rupture of membranes and/or preterm birth and the uterus, amniotic fluid, and serum were collected for analysis. RESULTS 87.5% of GM-CSF mice exhibited evidence of membrane rupture or preterm birth, compared with 0% in control mice (p < .001). Treatment with GM-CSF decreased the expression of TNFα (p < .05) while increasing the ratio of MMP2:TIMP1 (p < .05), MMP2:TIMP2 (p < .05), MMP2:TIMP3 (p < .001), MMP9:TIMP1 (p < .01), MMP9:TIMP2 (p < .05), MMP9:TIMP3 (p < .001), and MMP10:TIMP1 (p < .05). Mice treated with LPS and the GM-CSF antibody resulted in a decrease in the ratio of MMP2:TIMP1 (p < .0001) compared with controls. CONCLUSION These studies demonstrate GM-CSF will result in membrane rupture and preterm birth by increasing the ratio MMPs:TIMPs in our animal model. By increasing our understanding of the molecular pathways associated with GM-CSF, we may be able to develop future therapies to prevent preterm birth and reduce neonatal morbidity.
Collapse
Affiliation(s)
- Christopher Nold
- Department of Women's Health, Hartford Hospital, Hartford, CT, USA.,Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Kristyn Esteves
- Department of Obstetrics and Gynecology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Todd Jensen
- Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Anthony T Vella
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, USA
| |
Collapse
|
140
|
Smail SW, Saeed M, Twana Alkasalias, Khudhur ZO, Younus DA, Rajab MF, Abdulahad WH, Hussain HI, Niaz K, Safdar M. Inflammation, immunity and potential target therapy of SARS-COV-2: A total scale analysis review. Food Chem Toxicol 2021; 150:112087. [PMID: 33640537 PMCID: PMC7905385 DOI: 10.1016/j.fct.2021.112087] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 01/28/2021] [Accepted: 02/16/2021] [Indexed: 12/18/2022]
Abstract
Coronavirus disease-19 (COVID-19) is a complex disease that causes illness ranging from mild to severe respiratory problems. It is caused by a novel coronavirus SARS-CoV-2 (Severe acute respiratory syndrome coronavirus-2) that is an enveloped positive-sense single-stranded RNA (+ssRNA) virus belongs to coronavirus CoV family. It has a fast-spreading potential worldwide, which leads to high mortality regardless of lows death rates. Now some vaccines or a specific drug are approved but not available for every country for disease prevention and/or treatment. Therefore, it is a high demand to identify the known drugs and test them as a possible therapeutic approach. In this critical situation, one or more of these drugs may represent the only option to treat or reduce the severity of the disease, until some specific drugs or vaccines will be developed and/or approved for everyone in this pandemic. In this updated review, the available repurpose immunotherapeutic treatment strategies are highlighted, elucidating the crosstalk between the immune system and SARS-CoV-2. Despite the reasonable data availability, the effectiveness and safety of these drugs against SARS-CoV-2 needs further studies and validations aiming for a better clinical outcome.
Collapse
Affiliation(s)
- Shukur Wasman Smail
- Department of Biology, College of Science, Salahaddin University-Erbil, Iraq; Department of Biology, College of Science, Cihan University-Erbil, Kurdistan Region, Iraq
| | - Muhammad Saeed
- Faculty of Animal Production and Technology, Cholistan University of Veterinary and Animal Sciences-63100, Bahawalpur, Pakistan
| | - Twana Alkasalias
- Department of Pathological Analysis, College of Science, Knowledge University, Erbil, Kurdistan Region, Iraq; General Directorate for Scientific Research Center, Salahaddin University- Erbil, Erbil, Kurdistan Region, Iraq; Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Zhikal Omar Khudhur
- Department of Medical Analysis, Faculty of Science, Tishk International University - Erbil, Kurdistan Region, Iraq
| | - Delan Ameen Younus
- General Directorate for Scientific Research Center, Salahaddin University- Erbil, Erbil, Kurdistan Region, Iraq
| | - Mustafa Fahmi Rajab
- Department of Biology, College of Science, Salahaddin University-Erbil, Iraq
| | - Wayel Habib Abdulahad
- Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen 9713 GZ, the Netherlands; Department of Pathology and Medical Biology, University of Groningen, Hanzeplein 1, Groningen 9713 GZ, the Netherlands
| | - Hafiz Iftikhar Hussain
- Department of Pathology, Faculty of Veterinary Sciences, Cholistan University of Veterinary and Animal Sciences-63100, Bahawalpur, Pakistan
| | - Kamal Niaz
- Department of Pharmacology & Toxicology, Faculty of Bio-Sciences, Cholistan University of Veterinary and Animal Sciences-63100, Bahawalpur, Pakistan
| | - Muhammad Safdar
- Department of Breeding and Genetics, Faculty of Animal Production and Technology, Cholistan University of Veterinary and Animal Sciences-63100, Bahawalpur, Pakistan.
| |
Collapse
|
141
|
Targeting Pin1 for Modulation of Cell Motility and Cancer Therapy. Biomedicines 2021; 9:biomedicines9040359. [PMID: 33807199 PMCID: PMC8065645 DOI: 10.3390/biomedicines9040359] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/25/2021] [Accepted: 03/27/2021] [Indexed: 01/09/2023] Open
Abstract
Peptidyl-prolyl cis-trans isomerase NIMA-interacting 1 (Pin1) specifically binds and isomerizes the phosphorylated serine/threonine-proline (pSer/Thr-Pro) motif, which leads to changes in protein conformation and function. Pin1 is widely overexpressed in cancers and plays an important role in tumorigenesis. Mounting evidence has revealed that targeting Pin1 is a potential therapeutic approach for various cancers by inhibiting cell proliferation, reducing metastasis, and maintaining genome stability. In this review, we summarize the underlying mechanisms of Pin1-mediated upregulation of oncogenes and downregulation of tumor suppressors in cancer development. Furthermore, we also discuss the multiple roles of Pin1 in cancer hallmarks and examine Pin1 as a desirable pharmaceutical target for cancer therapy. We also summarize the recent progress of Pin1-targeted small-molecule compounds for anticancer activity.
Collapse
|
142
|
Borgers JSW, Haanen JBAG. Cellular Therapy and Cytokine Treatments for Melanoma. Hematol Oncol Clin North Am 2021; 35:129-144. [PMID: 33759770 DOI: 10.1016/j.hoc.2020.08.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cancer immunotherapy plays an important role in the treatment of patients with advanced stage melanoma. Recombinant cytokines were the first tested and approved treatments; however, due to disappointing response rates and severe toxicities, their use has significantly decreased. More recently, adoptive cell transfer therapies have shown to be a promising new treatment strategy able to induce complete and durable remissions in patients with melanoma progressive on first-line treatment. This review provides an overview of the cellular therapies (tumor-infiltrating lymphocytes, T-cell receptor T cells, chimeric antigen receptor T cells) and cytokine treatments (interleukin-2 [IL-2], IL-15, IL-7, IL-10, IL-21, interferon alpha, granulocyte-macrophage colony-stimulating factor) for melanoma.
Collapse
Affiliation(s)
- Jessica S W Borgers
- Department of Medical Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| | - John B A G Haanen
- Department of Medical Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands.
| |
Collapse
|
143
|
Ahmadian M, Jahanian-Najafabadi A, Akbari V. Optimization of Buffer Additives for Efficient Recovery of hGM-CSF from Inclusion Bodies Using Response Surface Methodology. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2021; 19:297-309. [PMID: 33680031 PMCID: PMC7758011 DOI: 10.22037/ijpr.2020.1101169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Overexpression of human granulocyte-macrophage colony-stimulating factor (hGM-CSF) by Escherichia coli leads to formation of insoluble and inactive proteins, inclusion bodies. The aim of this study was to improve recovery of biologically active hGM-CSF from inclusion bodies. The effect of types, concentrations and pHs of denaturing agents and addition of reducing agents on the yield of inclusion bodies solubilization was evaluated. Next, various conditions were evaluated for refolding hGM-CSF using a two-step design of experiment (DOE) including primary screening by factorial design, and then optimization by response surface design. It was found that hGM-CSF inclusion bodies can be efficiently solubilized with 4 M urea and 4 mM β-mercaptoethanol, pH = 9. A response surface quadratic model was employed to predict the optimum refolding conditions and the accuracy of this model was confirmed by high value of R2 (0.99) and F-value of 0.64. DOE results revealed that sorbitol (0.235 M), imidazole (97 mM), and SDS (0.09%) would be the optimum buffer additives for refolding of hGM-CSF. Following refolding studies, the obtained protein was subjected to circular dichroism which confirmed correct secondary structure of the refolded hGM-CSF. The refolded hGM-CSF exhibited reasonable biological activity compared with standard protein. The approach developed in this work can be important to improve the refolding of other proteins with similar structural features.
Collapse
Affiliation(s)
- Mina Ahmadian
- Department of Pharmaceutical Biotechnology and Isfahan Pharmaceutical Research Center, Faculty of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Jahanian-Najafabadi
- Department of Pharmaceutical Biotechnology and Isfahan Pharmaceutical Research Center, Faculty of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Vajihe Akbari
- Department of Pharmaceutical Biotechnology and Isfahan Pharmaceutical Research Center, Faculty of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
144
|
Ramelyte E, Tastanova A, Balázs Z, Ignatova D, Turko P, Menzel U, Guenova E, Beisel C, Krauthammer M, Levesque MP, Dummer R. Oncolytic virotherapy-mediated anti-tumor response: a single-cell perspective. Cancer Cell 2021; 39:394-406.e4. [PMID: 33482123 DOI: 10.1016/j.ccell.2020.12.022] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 11/05/2020] [Accepted: 12/21/2020] [Indexed: 01/09/2023]
Abstract
Talimogene laherparepvec (T-VEC) is a genetically modified herpes simplex 1 virus (HSV-1) approved for cancer therapy. We investigate its effect on the clinical, histological, single-cell transcriptomic, and immune repertoire level using repeated fine-needle aspirates (FNAs) of injected and noninjected lesions in primary cutaneous B cell lymphoma (pCBCL). Thirteen patients received intralesional T-VEC, 11 of which demonstrate tumor response in the injected lesions. Using single-cell sequencing of the FNAs, we identify the malignant population and separate three pCBCL subtypes. Twenty-four hours after the injection, we detect HSV-1T-VEC transcripts in malignant and nonmalignant cells of the injected lesion but not of the noninjected lesion. Oncolytic virotherapy results in a rapid eradication of malignant cells. It also leads to interferon pathway activation and early influx of natural killer cells, monocytes, and dendritic cells. These events are followed by enrichment in cytotoxic T cells and a decrease of regulatory T cells in injected and noninjected lesions.
Collapse
Affiliation(s)
- Egle Ramelyte
- Dermatology Department, University Hospital Zurich and Medical Faculty, University of Zurich, 8091 Zurich, Switzerland
| | - Aizhan Tastanova
- Dermatology Department, University Hospital Zurich and Medical Faculty, University of Zurich, 8091 Zurich, Switzerland
| | - Zsolt Balázs
- Department of Quantitative Biomedicine, University of Zurich, 8057 Zurich, Switzerland; Biomedical Informatics, University Hospital of Zurich, 8057 Zurich, Switzerland
| | - Desislava Ignatova
- Dermatology Department, University Hospital Zurich and Medical Faculty, University of Zurich, 8091 Zurich, Switzerland; Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| | - Patrick Turko
- Dermatology Department, University Hospital Zurich and Medical Faculty, University of Zurich, 8091 Zurich, Switzerland
| | - Ulrike Menzel
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland
| | - Emmanuella Guenova
- Dermatology Department, University Hospital Zurich and Medical Faculty, University of Zurich, 8091 Zurich, Switzerland; Department of Dermatology, Lausanne University Hospital (CHUV) and Faculty of Biology and Medicine, University of Lausanne, 1015 Lausanne, Switzerland
| | - Christian Beisel
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland
| | - Michael Krauthammer
- Department of Quantitative Biomedicine, University of Zurich, 8057 Zurich, Switzerland; Biomedical Informatics, University Hospital of Zurich, 8057 Zurich, Switzerland
| | - Mitchell Paul Levesque
- Dermatology Department, University Hospital Zurich and Medical Faculty, University of Zurich, 8091 Zurich, Switzerland
| | - Reinhard Dummer
- Dermatology Department, University Hospital Zurich and Medical Faculty, University of Zurich, 8091 Zurich, Switzerland.
| |
Collapse
|
145
|
Tabana Y, Okoye IS, Siraki A, Elahi S, Barakat KH. Tackling Immune Targets for Breast Cancer: Beyond PD-1/PD-L1 Axis. Front Oncol 2021; 11:628138. [PMID: 33747948 PMCID: PMC7973280 DOI: 10.3389/fonc.2021.628138] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 02/08/2021] [Indexed: 12/24/2022] Open
Abstract
The burden of breast cancer is imposing a huge global problem. Drug discovery research and novel approaches to treat breast cancer have been carried out extensively over the last decades. Although immune checkpoint inhibitors are showing promising preclinical and clinical results in treating breast cancer, they are facing multiple limitations. From an immunological perspective, a recent report highlighted breast cancer as an "inflamed tumor" with an immunosuppressive microenvironment. Consequently, researchers have been focusing on identifying novel immunological targets that can tune up the tumor immune microenvironment. In this context, several novel non-classical immune targets have been targeted to determine their ability to uncouple immunoregulatory pathways at play in the tumor microenvironment. This article will highlight strategies designed to increase the immunogenicity of the breast tumor microenvironment. It also addresses the latest studies on targets which can enhance immune responses to breast cancer and discusses examples of preclinical and clinical trial landscapes that utilize these targets.
Collapse
Affiliation(s)
- Yasser Tabana
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Isobel S. Okoye
- School of Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Arno Siraki
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Shokrollah Elahi
- School of Dentistry, University of Alberta, Edmonton, AB, Canada
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
| | - Khaled H. Barakat
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
146
|
Kwon B, Hong SY, Kim EY, Kim JH, Kim M, Park JH, Sohn Y, Jung HS. Effect of Cone of Pinus densiflora on DNCB-Induced Allergic Contact Dermatitis-Like Skin Lesion in Balb/c Mice. Nutrients 2021; 13:nu13030839. [PMID: 33806628 PMCID: PMC7998145 DOI: 10.3390/nu13030839] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/24/2021] [Accepted: 02/28/2021] [Indexed: 11/25/2022] Open
Abstract
Cone of Pinus densiflora (CP), or Korean red pinecone, is a cluster of Pinus densiflora fruit. CP has also been verified in several studies to have anti-oxidation, anti-fungal, anti-bacterial, and anti-melanogenic effects. However, anti-inflammatory effects have not yet been confirmed in the inflammatory responses of pinecones to allergic contact dermatitis. The purpose of this study is to prove the anti-inflammatory effect of CP on allergic contact dermatitis (ACD) in vitro and in vivo. CP inhibited the expression of TSLP, TARC, MCP-1, TNF-α, and IL-6 in TNF-α/IFN-γ-stimulated HaCaT cells and MCP-1, GM-CSF, TNF-α, IL-6, and IL-8 in PMACI (phorbol-12-myristate-13-acetate plus A23187)-stimulated HMC-1 cells. CP inhibited the phosphorylation of mitogen-activated protein kinase (MAPKs), as well as the translocation of NF-κB on TNF-α/IFN-γ stimulated in HaCaT cells. In vivo, CP decreased major symptoms of ACD, levels of IL-6 in skin lesion, thickening of the epidermis and dermis, infiltration of eosinophils and mast cells, and the infiltration of CD4+ T cells and CD8+ T cells. This result suggests that CP represents a potential alternative medicine to ACD for diseases such as chronic skin inflammation.
Collapse
Affiliation(s)
- Boguen Kwon
- Department of Anatomy, College of Korean Medicine, Kyung Hee University, 26, Kyunghee dae-ro, Dongdaemun-gu, Seoul 02447, Korea; (B.K.); (S.Y.H.); (E.-Y.K.); (J.-H.K.); (M.K.)
| | - Soo Yeon Hong
- Department of Anatomy, College of Korean Medicine, Kyung Hee University, 26, Kyunghee dae-ro, Dongdaemun-gu, Seoul 02447, Korea; (B.K.); (S.Y.H.); (E.-Y.K.); (J.-H.K.); (M.K.)
| | - Eun-Young Kim
- Department of Anatomy, College of Korean Medicine, Kyung Hee University, 26, Kyunghee dae-ro, Dongdaemun-gu, Seoul 02447, Korea; (B.K.); (S.Y.H.); (E.-Y.K.); (J.-H.K.); (M.K.)
| | - Jae-Hyun Kim
- Department of Anatomy, College of Korean Medicine, Kyung Hee University, 26, Kyunghee dae-ro, Dongdaemun-gu, Seoul 02447, Korea; (B.K.); (S.Y.H.); (E.-Y.K.); (J.-H.K.); (M.K.)
| | - Minsun Kim
- Department of Anatomy, College of Korean Medicine, Kyung Hee University, 26, Kyunghee dae-ro, Dongdaemun-gu, Seoul 02447, Korea; (B.K.); (S.Y.H.); (E.-Y.K.); (J.-H.K.); (M.K.)
| | - Jae Ho Park
- Department of Pharmaceutical Science, Jungwon University, 85, Munmu-ro, Goesan-eup, Goesan-gun, Chungbuk 28024, Korea;
| | - Youngjoo Sohn
- Department of Anatomy, College of Korean Medicine, Kyung Hee University, 26, Kyunghee dae-ro, Dongdaemun-gu, Seoul 02447, Korea; (B.K.); (S.Y.H.); (E.-Y.K.); (J.-H.K.); (M.K.)
- Correspondence: (Y.S.); (H.-S.J.); Tel.: +82-2-961-0327 (H.-S.J.); Fax: +82-2-961-9449 (H.-S.J.)
| | - Hyuk-Sang Jung
- Department of Anatomy, College of Korean Medicine, Kyung Hee University, 26, Kyunghee dae-ro, Dongdaemun-gu, Seoul 02447, Korea; (B.K.); (S.Y.H.); (E.-Y.K.); (J.-H.K.); (M.K.)
- Correspondence: (Y.S.); (H.-S.J.); Tel.: +82-2-961-0327 (H.-S.J.); Fax: +82-2-961-9449 (H.-S.J.)
| |
Collapse
|
147
|
Üçal M, Maurer C, Etschmaier V, Hamberger D, Grünbacher G, Tögl L, Roosen MJ, Molcanyi M, Vorholt D, Hatay FF, Hescheler J, Pallasch C, Schäfer U, Patz S. Inflammatory Pre-Conditioning of Adipose-Derived Stem Cells with Cerebrospinal Fluid from Traumatic Brain Injury Patients Alters the Immunomodulatory Potential of ADSC Secretomes. J Neurotrauma 2021; 38:2311-2322. [PMID: 33514282 DOI: 10.1089/neu.2020.7017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Immunomodulation by adipose-tissue-derived stem cells (ADSCs) is of special interest for the alleviation of damaging inflammatory responses in central nervous system injuries. The present study explored the effects of cerebrospinal fluid (CSF) from traumatic brain injury (TBI) patients on this immunomodulatory potential of ADSCs. CSF conditioning of ADSCs increased messenger RNA levels of both pro- and anti-inflammatory genes compared to controls. Exposure of phorbol-12-myristate-13-acetate-differentiated THP1 macrophages to the secretome of CSF-conditioned ADSCs downregulated both proinflammatory (cyclooxygenase-2, tumor necrosis factor alpha) and anti-inflammatory (suppressor of cytokine signaling 3, interleukin-1 receptor antagonist, and transforming growth factor beta) genes in these cells. Interleukin-10 expression was elevated in both naïve and conditioned secretomes. ADSC secretome treatment, further, induced macrophage maturation of THP1 cells and increased the percentage of CD11b+, CD14+, CD86+, and, to a lesser extent, CD206+ cells. This, moreover, enhanced the phagocytic activity of CD14+ and CD86+ cells, though independently of pre-conditioning. Secretome exposure, finally, also induced a reduction in the percentage of CD192+ adherent cells in cultures of peripheral blood mononuclear cells (PBMCs) from both healthy subjects and TBI patients. This limited efficacy (of both naïve and pre-conditioned secretomes) suggests that the effects of lymphocyte-monocyte paracrine signaling on the fate of cultured PBMCs are strongest upon adherent cell populations.
Collapse
Affiliation(s)
- Muammer Üçal
- Department of Neurosurgery, Medical University Graz, Graz, Austria
| | - Christa Maurer
- Department of Neurosurgery, Medical University Graz, Graz, Austria.,Ruprecht-Karls-University Heidelberg, Institute for Anatomy and Cell Biology, Division for Medical Cell Biology, Heidelberg, Germany
| | | | - Daniel Hamberger
- Department of Neurosurgery, Medical University Graz, Graz, Austria.,National Centre for Tumour Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| | - Gerda Grünbacher
- Department of Neurosurgery, Medical University Graz, Graz, Austria
| | - Lennart Tögl
- Department of Neurosurgery, Medical University Graz, Graz, Austria
| | - Marvin J Roosen
- Department of Neurosurgery, Medical University Graz, Graz, Austria
| | - Marek Molcanyi
- Department of Neurosurgery, Medical University Graz, Graz, Austria.,Institute of Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Daniela Vorholt
- Department of Internal Medicine, Centre for Integrated Oncology Aachen Bonn Cologne Düsseldorf, CECAD Centre of Excellence on Cellular Stress Responses in Aging-Associated Diseases, Centre for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - F Fulya Hatay
- Institute of Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Jürgen Hescheler
- Institute of Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Christian Pallasch
- Department of Internal Medicine, Centre for Integrated Oncology Aachen Bonn Cologne Düsseldorf, CECAD Centre of Excellence on Cellular Stress Responses in Aging-Associated Diseases, Centre for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Ute Schäfer
- Department of Neurosurgery, Medical University Graz, Graz, Austria
| | - Silke Patz
- Department of Neurosurgery, Medical University Graz, Graz, Austria
| |
Collapse
|
148
|
Banji D, Alqahtani SS, Banji OJ, Machanchery S, Shoaib A. Calming the inflammatory storm in severe COVID-19 infections: Role of biologics- A narrative review. Saudi Pharm J 2021; 29:213-222. [PMID: 33850422 PMCID: PMC8030716 DOI: 10.1016/j.jsps.2021.01.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 01/20/2021] [Indexed: 02/09/2023] Open
Abstract
The risk of Coronavirus infection continues, and the fear of resurgence indicates the lack of a successful therapeutic strategy. In severe COVID-19 infection, many immune cells and their products are involved, making management difficult. The abundant release of cytokines and chemokines in severe COVID-19 patients leads to profound hyper inflammation and the mobilization of immune cells, triggering the cytokine storm. The complications associated with the cytokine storm include severe respiratory distress, intravascular coagulation, multi-organ failure, and death. The enormous formation of interleukin (IL)-6 and hemopoietic factors such as granulocyte-macrophage colony-stimulating factor (GM-CSF) are implicated in the severity of the infection. Moreover, these inflammatory cytokines and factors signal through the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway causing the activation of cytokine-related genes. The neutralization of these proteins could be of therapeutic help in COVID-19 patients and could mitigate the risk of mortality. IL-6 antagonist, IL-6 receptor antagonists, GM-CSF receptor inhibitors, and JAK-STAT inhibitors are being investigated to prevent intense lung injury in COVID-19 patients and increase the chances of survival. The review focuses the role of IL-6, GM-CSF, and JAK-STAT inhibitors in regulating the immune response in severely affected COVID-19 patients.
Collapse
Affiliation(s)
- David Banji
- Department of Clinical Pharmacy, Pharmacy Practice Research Unit, College of Pharmacy, Jazan University, Saudi Arabia
| | - Saad S. Alqahtani
- Department of Clinical Pharmacy, Pharmacy Practice Research Unit, College of Pharmacy, Jazan University, Saudi Arabia
| | - Otilia J.F. Banji
- Department of Clinical Pharmacy, Pharmacy Practice Research Unit, College of Pharmacy, Jazan University, Saudi Arabia
| | - Shamna Machanchery
- Department of Clinical Pharmacy, Pharmacy Practice Research Unit, College of Pharmacy, Jazan University, Saudi Arabia
| | - Ambreen Shoaib
- Department of Clinical Pharmacy, Pharmacy Practice Research Unit, College of Pharmacy, Jazan University, Saudi Arabia
| |
Collapse
|
149
|
Lin D, He H, Sun J, He X, Long W, Cui X, Sun Y, Zhao S, Zheng X, Zeng Z, Zhang K, Wang H. Co-delivery of PSMA antigen epitope and mGM-CSF with a cholera toxin-like chimeric protein suppressed prostate tumor growth via activating dendritic cells and promoting CTL responses. Vaccine 2021; 39:1609-1620. [PMID: 33612342 DOI: 10.1016/j.vaccine.2021.02.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 01/09/2021] [Accepted: 02/02/2021] [Indexed: 01/22/2023]
Abstract
Subunit vaccines derived from tumor antigens play a role in tumor therapy because of their unique advantages. However, because of the weak immunogenicity of peptides in subunit vaccines, it is difficult to trigger an effective cytotoxic T lymphocyte (CTL) response, which is critical for cancer therapy. A requirement for the activation of CTL cells by exogenous antigens is the stimulation of antigen presenting cells (APC) with the help of adjuvants and cross-presentation to T lymphocytes. Standard nonconjugated adjuvant-peptide mixtures do not ensure co-targeting of the antigen and the adjuvant to the same APC, which limits the effects of adjuvants. In this study, a fusion protein consisting of murine granulocyte-macrophage colony stimulating factor (mGM-CSF) fused with CTA2 (A2 subunit of cholera toxin) was generated and assembled with CTB-PSMA624-632 (prostate specific membrane antigen (PSMA) peptide 624-632 fused to CTB) to obtain a cholera toxin-like protein. The chimeric protein retained the biological activity of mGM-CSF and had stronger GM1 binding activity than (CTB-PSMA624-632)5. C57BL/6J mice immunized with the CT-like chimeric protein exhibited delayed tumor growth following challenge with human PSMA-EGFP-expressing RM-1 cells. Experiment results showed that the CT-like chimeric protein could induce the maturation of DC cells and improve CTL responses. Overall, these results indicate that the nasal administration of a CT-like chimeric protein vaccine results in the development of effective immunity against prostate tumor cells and might be useful for future clinical anti-tumoral applications.
Collapse
Affiliation(s)
- Danmin Lin
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China
| | - Huafeng He
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China
| | - Jiajie Sun
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China
| | - Xianying He
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China
| | - Wei Long
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China
| | - Xiping Cui
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China
| | - Yunxiao Sun
- Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, PR China
| | - Suqing Zhao
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China
| | - Xi Zheng
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China
| | - Zheng Zeng
- The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, PR China
| | - Kun Zhang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China; School of Biotechnology and Health, Wuyi University, Jiangmen 529020, PR China
| | - Huaqian Wang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China.
| |
Collapse
|
150
|
Rösler B, Heinhuis B, Wang X, Silvestre R, Joosten LAB, Netea MG, Arts P, Mantere T, Lefeber DJ, Hoischen A, van de Veerdonk FL. Mimicking Behçet's disease: GM-CSF gain of function mutation in a family suffering from a Behçet's disease-like disorder marked by extreme pathergy. Clin Exp Immunol 2021; 204:189-198. [PMID: 33349924 DOI: 10.1111/cei.13568] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 11/10/2020] [Accepted: 11/10/2020] [Indexed: 12/15/2022] Open
Abstract
Behçet's disease (BD) is an inflammatory disease mainly affecting men along the ancient Silk Route. In the present study we describe a Dutch family suffering from BD-like disease with extreme pathergic responses, but without systemic inflammation. Genetic assessment revealed a combination of the human leukocyte antigen (HLA)-B*51 risk-allele together with a rare heterozygous variant in the CSF2 gene (c.130A>C, p.N44H) encoding for granulocyte-macrophage colony-stimulating factor (GM-CSF) found by whole exome sequencing. We utilized an over-expression vector system in a human hepatocyte cell line to produce the aberrant variant of GM-CSF. Biological activity of the protein was measured by signal transducer and activator of transcription 5 (STAT-5) phosphorylation, a downstream molecule of the GM-CSF receptor, in wild-type peripheral mononuclear cells (PBMCs) using flow cytometry. Increased STAT-5 phosphorylation was observed in response to mutated GM-CSF when compared to the wild-type or recombinant protein. CSF2 p.N44H results in disruption of one of the protein's two N-glycosylation sites. Enzymatically deglycosylated wild-type GM-CSF also enhanced STAT-5 phosphorylation. The patient responded well to anti-tumor necrosis factor (TNF)-α treatment, which may be linked to the capacity of TNF-α to induce GM-CSF in phorbol 12-myristate 13-acetate (PMA)-treated PBMCs, while GM-CSF itself only induced dose-dependent interleukin (IL)-1Ra production. The identified CSF2 pathway could provide novel insights into the pathergic response of BD-like disease and offer new opportunities for personalized treatment.
Collapse
Affiliation(s)
- B Rösler
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| | - B Heinhuis
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| | - X Wang
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| | - R Silvestre
- Microbiology and Infection Research Domain, Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's, PT Government Associate Laboratory, Guimarães, Portugal
| | - L A B Joosten
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| | - M G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands.,Department for Genomics and Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - P Arts
- Department of Human Genetics and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - T Mantere
- Department of Human Genetics and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - D J Lefeber
- Department of Neurology, Translational Metabolic Laboratory, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - A Hoischen
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands.,Department of Human Genetics and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - F L van de Veerdonk
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|