1
|
Jung EK, Chu TH, Kim SA, Vo MC, Nguyen VT, Lee KH, Jung SH, Yoon M, Cho D, Lee JJ, Yoon TM. Efficacy of natural killer cell therapy combined with chemoradiotherapy in murine models of head and neck squamous cell carcinoma. Cytotherapy 2024; 26:242-251. [PMID: 38142382 DOI: 10.1016/j.jcyt.2023.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 10/24/2023] [Accepted: 11/08/2023] [Indexed: 12/25/2023]
Abstract
BACKGROUND AIMS Natural killer (NK) cell-based cancer immunotherapy is effective when combined with other treatment modalities such as irradiation and chemotherapy. NK cell's antitumor function to treat solid tumor, including head and neck squamous cell carcinoma (HNSCC), has been targeted recently. This study assessed NK cell recruitment in response to chemoradiation therapy (CRT) in HNSCC. METHODS Ex vivo expansion of NK cell, flow cytometry, cell viability assay, cytotoxicity assay, immunohistochemistry, and animal model were performed. RESULTS Mouse NK cells were recruited to the tumor site by CRT in a nude mouse model. Furthermore, expanded and activated human NK cells (eNKs) were recruited to the tumor site in response to CRT, and CRT enhanced the anti-tumor activity of eNK in an NOD/SCID IL-2Rγnull mouse model. Various HNSCC cancer cell lines exhibited different NK cell ligand activation patterns in response to CRT that correlated with NK cell-mediated cytotoxicity. CONCLUSIONS Identifying the activation patterns of NK cell ligands during CRT might improve patient selection for adjuvant NK cell immunotherapy combined with CRT. This is the first study to investigate the NK cell's antitumor function and recruitment with CRT in HNSCC mouse model.
Collapse
Affiliation(s)
- Eun Kyung Jung
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Jeollanamdo, Korea
| | - Tan-Huy Chu
- Department of Hematology-Oncology, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Jeollanamdo, Korea; Department of Hematology, Pham Ngoc Thach University of Medicine, Ho Chi Minh City, Vietnam
| | - Sun-Ae Kim
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Jeollanamdo, Korea
| | - Manh-Cuong Vo
- Department of Hematology-Oncology, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Jeollanamdo, Korea
| | - Van-Tan Nguyen
- Department of Hematology-Oncology, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Jeollanamdo, Korea
| | - Kyung-Hwa Lee
- Department of Pathology, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Jeollanamdo, Korea
| | - Sung-Hoon Jung
- Department of Hematology-Oncology, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Jeollanamdo, Korea
| | - Meesun Yoon
- Department of Radiation Oncology, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Jeollanamdo, Korea
| | - Duck Cho
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Je-Jung Lee
- Department of Hematology-Oncology, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Jeollanamdo, Korea.
| | - Tae Mi Yoon
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Jeollanamdo, Korea.
| |
Collapse
|
2
|
Li X, Qiao Q, Liu X, Hu Q, Yu Y, Qin X, Tian T, Tian Y, Ou X, Niu B, Yang C, Kong L, Zhang Z. Engineered Biomimetic Nanovesicles Based on Neutrophils for Hierarchical Targeting Therapy of Acute Respiratory Distress Syndrome. ACS NANO 2024; 18:1658-1677. [PMID: 38166370 DOI: 10.1021/acsnano.3c09848] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2024]
Abstract
Acute Respiratory Distress Syndrome (ARDS) is a clinically severe respiratory disease that causes severe medical and economic burden. To improve therapeutic efficacy, effectively targeting delivery to the inflamed lungs and inflamed cells remains an ongoing challenge. Herein, we designed engineered biomimetic nanovesicles (DHA@ANeu-DDAB) by fusion of lung-targeting functional lipid, neutrophil membrane containing activated β2 integrins, and the therapeutic lipid, docosahexaenoic acid (DHA). By the advantage of lung targeting lipid and β2 integrin targeting adhesion, DHA@ANeu-DDAB can first target lung tissue and further target inflammatory vascular endothelial cells, to achieve "tissue first, cell second" hierarchical delivery. In addition, the β2 integrins in DHA@ANeu-DDAB could bind to the intercellular cell adhesion molecule-1/2 (ICAM-1/2) ligand on the endothelium in the inflamed blood vessels, thus inhibiting neutrophils' infiltration in the blood circulation. DHA administration to inflamed lungs could effectively regulate macrophage phenotype and promote its anti-inflammatory activity via enhanced biosynthesis of specialized pro-resolving mediators. In the lipopolysaccharide-induced ARDS mouse model, DHA@ANeu-DDAB afforded a comprehensive and efficient inhibition of lung inflammation and promoted acute lung damage repair. Through mimicking physiological processes, these engineered biomimetic vesicles as a delivery system possess good potential in targeting therapy for ARDS.
Collapse
Affiliation(s)
- Xiaonan Li
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qi Qiao
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiong Liu
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qian Hu
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yulin Yu
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xianya Qin
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tianyi Tian
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yinmei Tian
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiangjun Ou
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Boning Niu
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Conglian Yang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Li Kong
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhiping Zhang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
- National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Engineering Research Centre for Novel Drug Delivery System, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
3
|
Marcinek A, Brauchle B, Rohrbacher L, Hänel G, Philipp N, Märkl F, Strzalkowski T, Lacher SM, Udiljak D, Spiekermann K, Theurich S, Kobold S, Kischel R, James JR, Bücklein VL, Subklewe M. CD33 BiTE ® molecule-mediated immune synapse formation and subsequent T-cell activation is determined by the expression profile of activating and inhibitory checkpoint molecules on AML cells. Cancer Immunol Immunother 2023; 72:2499-2512. [PMID: 37041225 PMCID: PMC10264534 DOI: 10.1007/s00262-023-03439-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 03/27/2023] [Indexed: 04/13/2023]
Abstract
Bispecific T-cell engager (BiTE®) molecules recruit T cells to cancer cells through CD3ε binding, independently of T-cell receptor (TCR) specificity. Whereas physiological T-cell activation is dependent on signal 1 (TCR engagement) and signal 2 (co-stimulation), BiTE molecule-mediated T-cell activation occurs without additional co-stimulation. As co-stimulatory and inhibitory molecules modulate the strength and nature of T-cell responses, we studied the impact of the expression profile of those molecules on target cells for BiTE molecule-mediated T-cell activation in the context of acute myeloid leukemia (AML). Accordingly, we created a novel in vitro model system using murine Ba/F3 cells transduced with human CD33 ± CD86 ± PD-L1. T-cell fitness was assessed by T-cell function assays in co-cultures and immune synapse formation by applying a CD33 BiTE molecule (AMG 330). Using our cell-based model platform, we found that the expression of positive co-stimulatory molecules on target cells markedly enhanced BiTE molecule-mediated T-cell activation. The initiation and stability of the immune synapse between T cells and target cells were significantly increased through the expression of CD86 on target cells. By contrast, the co-inhibitory molecule PD-L1 impaired the stability of BiTE molecule-induced immune synapses and subsequent T-cell responses. We validated our findings in primary T-cell-AML co-cultures, demonstrating a PD-L1-mediated reduction in redirected T-cell activation. The addition of the immunomodulatory drug (IMiD) lenalidomide to co-cultures led to stabilization of immune synapses and improved subsequent T-cell responses. We conclude that target cells modulate CD33 BiTE molecule-dependent T-cell activation and hence, combinatorial strategies might contribute to enhanced efficacy.
Collapse
Affiliation(s)
- Anetta Marcinek
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany
| | - Bettina Brauchle
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany
| | - Lisa Rohrbacher
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany
| | - Gerulf Hänel
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany
| | - Nora Philipp
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany
| | - Florian Märkl
- Division of Clinical Pharmacology, Department of Medicine IV; Member of the German Center for Lung Research (DZL), University Hospital, LMU, Munich, Germany
| | - Thaddäus Strzalkowski
- Division of Clinical Pharmacology, Department of Medicine IV; Member of the German Center for Lung Research (DZL), University Hospital, LMU, Munich, Germany
| | - Sonja M Lacher
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany
| | - Dragica Udiljak
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany
| | - Karsten Spiekermann
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Experimental Leukemia and Lymphoma Research (ELLF), Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Sebastian Theurich
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Cancer-and Immunometabolism Research Group, LMU Gene Center, Munich, Germany
| | - Sebastian Kobold
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Clinical Pharmacology, Department of Medicine IV; Member of the German Center for Lung Research (DZL), University Hospital, LMU, Munich, Germany
| | - Roman Kischel
- AMGEN Research Munich GmbH, Munich, Germany
- AMGEN Inc., Thousand Oaks, CA, USA
| | - John R James
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK
| | - Veit L Bücklein
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany
| | - Marion Subklewe
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany.
- Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany.
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
4
|
Martinez-Sanz P, Laurent ARG, Slot E, Hoogenboezem M, Bąbała N, van Bruggen R, Rongvaux A, Flavell RA, Tytgat GAM, Franke K, Matlung HL, Kuijpers TW, Amsen D, Karrich JJ. Humanized MISTRG as a preclinical in vivo model to study human neutrophil-mediated immune processes. Front Immunol 2023; 14:1105103. [PMID: 36969261 PMCID: PMC10032520 DOI: 10.3389/fimmu.2023.1105103] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 02/22/2023] [Indexed: 03/11/2023] Open
Abstract
IntroductionMISTRG mice have been genetically modified to allow development of a human myeloid compartment from engrafted human CD34+ haemopoietic stem cells, making them particularly suited to study the human innate immune system in vivo. Here, we characterized the human neutrophil population in these mice to establish a model that can be used to study the biology and contribution in immune processes of these cells in vivo.Methods and resultsWe could isolate human bone marrow neutrophils from humanized MISTRG mice and confirmed that all neutrophil maturation stages from promyelocytes (CD11b–CD16–) to end-stage segmented cells (CD11b+CD16+) were present. We documented that these cells possessed normal functional properties, including degranulation, reactive oxygen species production, adhesion, and antibody-dependent cellular cytotoxicity towards antibody-opsonized tumor cells ex vivo. The acquisition of functional capacities positively correlated with the maturation state of the cell. We found that human neutrophils were retained in the bone marrow of humanized MISTRG mice during steady state. However, the mature segmented CD11b+CD16+ human neutrophils were released from the bone marrow in response to two well-established neutrophil-mobilizing agents (i.e., G-CSF and/or CXCR4 antagonist Plerixafor). Moreover, the neutrophil population in the humanized MISTRG mice actively reacted to thioglycolate-induced peritonitis and could infiltrate implanted human tumors, as shown by flow cytometry and fluorescent microscopy.DiscussionThese results show that functional human neutrophils are generated and can be studied in vivo using the humanized MISTRG mice, providing a model to study the various functions of neutrophils in inflammation and in tumors.
Collapse
Affiliation(s)
- Paula Martinez-Sanz
- Sanquin Research and Landsteiner Laboratory, Department of Molecular Hematology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- *Correspondence: Paula Martinez-Sanz, ; Julien J. Karrich, ; Derk Amsen,
| | - Adrien R. G. Laurent
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Edith Slot
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Mark Hoogenboezem
- Sanquin Research and Landsteiner Laboratory, Department of Molecular Hematology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Nikolina Bąbała
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Robin van Bruggen
- Sanquin Research and Landsteiner Laboratory, Department of Molecular Hematology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Anthony Rongvaux
- Department of Immunology, University of Washington, Seattle, WA, United States
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, WA, United States
| | - Richard A. Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, United States
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, United States
| | - Godelieve A. M. Tytgat
- Princess Maxima Center for Pediatric Oncology, Department of Pediatric Oncology, Utrecht, Netherlands
| | - Katka Franke
- Sanquin Research and Landsteiner Laboratory, Department of Molecular Hematology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Hanke L. Matlung
- Sanquin Research and Landsteiner Laboratory, Department of Molecular Hematology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Taco W. Kuijpers
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Rheumatology and Infectious Diseases, Emma Children's Hospital, Department of Pediatric Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Derk Amsen
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- *Correspondence: Paula Martinez-Sanz, ; Julien J. Karrich, ; Derk Amsen,
| | - Julien J. Karrich
- Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- *Correspondence: Paula Martinez-Sanz, ; Julien J. Karrich, ; Derk Amsen,
| |
Collapse
|
5
|
Cheng RYH, Hung KL, Zhang T, Stoffers CM, Ott AR, Suchland ER, Camp ND, Khan IF, Singh S, Yang YJ, Rawlings DJ, James RG. Ex vivo engineered human plasma cells exhibit robust protein secretion and long-term engraftment in vivo. Nat Commun 2022; 13:6110. [PMID: 36245034 PMCID: PMC9573882 DOI: 10.1038/s41467-022-33787-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 10/04/2022] [Indexed: 12/24/2022] Open
Abstract
Due to their unique longevity and capacity to secrete high levels of protein, plasma B cells have the potential to be used as a cell therapy for protein replacement. Here, we show that ex vivo engineered human plasma cells exhibit single-cell RNA profiles, scanning electron micrograph ultrastructural features, and in vivo homing capacity of long-lived plasma cells. After transferring human plasma cells to immunodeficient mice in the presence of the human cytokines BAFF and IL-6, we observe increases in retention of plasma cells in the bone marrow, with engraftment exceeding a year. The most profound in vivo effects of human IL-6 are observed within 20 days of transfer and could be explained by decreased apoptosis in newly differentiated plasma cells. Collectively, these results show that ex vivo engineered and differentiated human plasma cells have the potential for long-lived in vivo protein secretion, which can be modeled in small animals.
Collapse
Affiliation(s)
- Rene Yu-Hong Cheng
- Center of immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA, 98101, USA
- Molecular Engineering and Science Institute, University of Washington, Seattle, WA, 98195, USA
| | - King L Hung
- Center of immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA, 98101, USA
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Tingting Zhang
- Center of immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA, 98101, USA
| | - Claire M Stoffers
- Center of immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA, 98101, USA
| | - Andee R Ott
- Center of immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA, 98101, USA
| | - Emmaline R Suchland
- Center of immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA, 98101, USA
| | - Nathan D Camp
- Center of immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA, 98101, USA
| | - Iram F Khan
- Center of immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA, 98101, USA
| | - Swati Singh
- Center of immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA, 98101, USA
| | - Ying-Jen Yang
- Department of Applied Mathematics, University of Washington, Seattle, WA, 98195, USA
| | - David J Rawlings
- Center of immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA, 98101, USA.
- Departments of Pediatrics, University of Washington, Seattle, WA, 98195, USA.
- Department of Immunology, University of Washington, Seattle, WA, 98195, USA.
| | - Richard G James
- Center of immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA, 98101, USA.
- Molecular Engineering and Science Institute, University of Washington, Seattle, WA, 98195, USA.
- Departments of Pediatrics, University of Washington, Seattle, WA, 98195, USA.
- Department of pharmacology, University of Washington, Seattle, WA, 98195, USA.
- Brotman-Baty Institute for Precision Medicine, Seattle, WA, 98195, USA.
| |
Collapse
|
6
|
The metabolism of cells regulates their sensitivity to NK cells depending on p53 status. Sci Rep 2022; 12:3234. [PMID: 35217717 PMCID: PMC8881467 DOI: 10.1038/s41598-022-07281-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 02/09/2022] [Indexed: 01/01/2023] Open
Abstract
Leukemic cells proliferate faster than non-transformed counterparts. This requires them to change their metabolism to adapt to their high growth. This change can stress cells and facilitate recognition by immune cells such as cytotoxic lymphocytes, which express the activating receptor Natural Killer G2-D (NKG2D). The tumor suppressor gene p53 regulates cell metabolism, but its role in the expression of metabolism-induced ligands, and subsequent recognition by cytotoxic lymphocytes, is unknown. We show here that dichloroacetate (DCA), which induces oxidative phosphorylation (OXPHOS) in tumor cells, induces the expression of such ligands, e.g. MICA/B, ULBP1 and ICAM-I, by a wtp53-dependent mechanism. Mutant or null p53 have the opposite effect. Conversely, DCA sensitizes only wtp53-expressing cells to cytotoxic lymphocytes, i.e. cytotoxic T lymphocytes and NK cells. In xenograft in vivo models, DCA slows down the growth of tumors with low proliferation. Treatment with DCA, monoclonal antibodies and NK cells also decreased tumors with high proliferation. Treatment of patients with DCA, or a biosimilar drug, could be a clinical option to increase the effectiveness of CAR T cell or allogeneic NK cell therapies.
Collapse
|
7
|
Allende-Vega N, Marco Brualla J, Falvo P, Alexia C, Constantinides M, Fayd'herbe de Maudave A, Coenon L, Gitenay D, Mitola G, Massa P, Orecchioni S, Bertolini F, Marzo I, Anel A, Villalba M. Metformin sensitizes leukemic cells to cytotoxic lymphocytes by increasing expression of intercellular adhesion molecule-1 (ICAM-1). Sci Rep 2022; 12:1341. [PMID: 35079096 PMCID: PMC8789909 DOI: 10.1038/s41598-022-05470-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 12/07/2021] [Indexed: 12/17/2022] Open
Abstract
Solid tumor cells have an altered metabolism that can protect them from cytotoxic lymphocytes. The anti-diabetic drug metformin modifies tumor cell metabolism and several clinical trials are testing its effectiveness for the treatment of solid cancers. The use of metformin in hematologic cancers has received much less attention, although allogeneic cytotoxic lymphocytes are very effective against these tumors. We show here that metformin induces expression of Natural Killer G2-D (NKG2D) ligands (NKG2DL) and intercellular adhesion molecule-1 (ICAM-1), a ligand of the lymphocyte function-associated antigen 1 (LFA-1). This leads to enhance sensitivity to cytotoxic lymphocytes. Overexpression of anti-apoptotic Bcl-2 family members decrease both metformin effects. The sensitization to activated cytotoxic lymphocytes is mainly mediated by the increase on ICAM-1 levels, which favors cytotoxic lymphocytes binding to tumor cells. Finally, metformin decreases the growth of human hematological tumor cells in xenograft models, mainly in presence of monoclonal antibodies that recognize tumor antigens. Our results suggest that metformin could improve cytotoxic lymphocyte-mediated therapy.
Collapse
Affiliation(s)
| | - Joaquin Marco Brualla
- Apoptosis, Immunity and Cancer Group, Department of Biochemistry and Molecular and Cell Biology, Faculty of Sciences, University of Zaragoza and Aragón Health Research Institute (IIS Aragón), Campus San Francisco Sq., 50009, Zaragoza, Spain
| | - Paolo Falvo
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | | | | | | | - Lois Coenon
- IRMB, Univ Montpellier, INSERM, Montpellier, France
| | | | - Giulia Mitola
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Paul Massa
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Stefania Orecchioni
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Francesco Bertolini
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Isabel Marzo
- Apoptosis, Immunity and Cancer Group, Department of Biochemistry and Molecular and Cell Biology, Faculty of Sciences, University of Zaragoza and Aragón Health Research Institute (IIS Aragón), Campus San Francisco Sq., 50009, Zaragoza, Spain
| | - Alberto Anel
- Apoptosis, Immunity and Cancer Group, Department of Biochemistry and Molecular and Cell Biology, Faculty of Sciences, University of Zaragoza and Aragón Health Research Institute (IIS Aragón), Campus San Francisco Sq., 50009, Zaragoza, Spain.
| | - Martin Villalba
- IRMB, Univ Montpellier, INSERM, Montpellier, France.
- CNRS, IRMB, INSERM, Univ Montpellier, CHU Montpellier, Montpellier, France.
- Institut Sainte Catherine, Avignon, France.
| |
Collapse
|
8
|
Liu Y, Li X, Zhao Y. Curcumin alleviated lipopolysaccharide-evoked H9c2 cells damage via suppression of intercellular adhesion molecule 1/CD40/NF-κB signaling. Hum Exp Toxicol 2022; 41:9603271211069043. [PMID: 35549587 DOI: 10.1177/09603271211069043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Curcumin has been reported to have many benefits, including anti-inflammatory, anti-cancer, and so on. In this research, we aimed to investigate the function of curcumin on lipopolysaccharide (LPS)-injured H9c2 cells. METHODS H9c2 cells stimulated by LPS mimic the in vitro model of myocarditis injury. Comparative Toxicogenomics Database (CTD) was applied to detect the genes associated with curcumin. GEO database was used to analyze Intercellular Adhesion Molecule 1 (ICAM1) and CD40 expression in myocarditis patients. KEGG enrichment analysis was employed to investigate the meaningful pathways related to differentially expressed genes. Cell proliferation, apoptosis, expression of ICAM1/CD40/P65- NF-κB, and level of TNF-α, IL-6, and IL-10 were observed by cell counting kit-8, flow cytometry and western blotting assays, ELISA assay, respectively. RESULTS After curcumin treatment, the decreased activity of H9c2 cells evoked by LPS was improved. ICAM1 and CD40, which highly expressed in myocarditis patients, were identified as targets of curcumin and negatively regulated by curcumin. Inhibition of ICAM1 or CD40 strengthened the protective effect of curcumin on LPS-evoked H9c2 cells damage, accompanied by increased cell viability and decreased cell apoptosis and inflammation. Additionally, addition of curcumin or depletion of ICAM1/CD40 suppressed p-P65 NF-κB expression. CONCLUSIONS Curcumin mitigated LPS-evoked H9c2 cells damage by suppression of ICAM1/CD40/NF-κB, providing a potential molecular mechanism for the clinical application of curcumin.
Collapse
Affiliation(s)
- Yi Liu
- Department of Cardiology, 159434Xuzhou Central Hospital, Xuzhou, China
| | - Xiaoli Li
- Department of Cardiology, 159434Xuzhou Central Hospital, Xuzhou, China
| | - Yan Zhao
- Department of Cardiology, 159434Xuzhou Central Hospital, Xuzhou, China
| |
Collapse
|
9
|
Jerebtsova M, Ahmad A, Kumari N, Rutagarama O, Nekhai S. Macrophage HIV-1 Gene Expression and Delay Resolution of Inflammation in HIV-Tg Mice. Viruses 2020; 12:v12030277. [PMID: 32121564 PMCID: PMC7150751 DOI: 10.3390/v12030277] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 02/25/2020] [Accepted: 02/27/2020] [Indexed: 02/06/2023] Open
Abstract
While antiretroviral therapy increases the longevity of people living with HIV (PLWH), about 30% of this population suffers from three or more concurrent comorbidities, whose mechanisms are not well understood. Chronic activation and dysfunction of the immune system could be one potential cause of these comorbidities. We recently demonstrated reduced macrophage infiltration and delayed resolution of inflammation in the lungs of HIV-transgenic mice. Additionally, trans-endothelial migration of HIV-positive macrophages was reduced in vitro. Here, we analyze macrophages’ response to LPS challenge in the kidney and peritoneum of HIV-Tg mice. In contrast to the lung infiltration, renal and peritoneal macrophage infiltrations were similar in WT and HIV-Tg mice. Higher levels of HIV-1 gene expression were detected in lung macrophages compared to peritoneal macrophages. In peritoneal macrophages, HIV-1 gene expression was increased when they were cultured at 21% O2 compared to 5% O2, inversely correlating with reduced trans-endothelial migration at higher oxygen levels in vitro. The resolution of macrophage infiltration was reduced in both the lung and the peritoneal cavity of HIV-Tg mice. Taken together, our study described the organ-specific alteration of macrophage dynamics in HIV-Tg mice. The delayed resolution of macrophage infiltration might constitute a risk factor for the development of multiple comorbidities in PLWH.
Collapse
Affiliation(s)
- Marina Jerebtsova
- Department of Microbiology, College of Medicine, Howard University, Washington, DC 20059, USA;
- Correspondence: (M.J.); (S.N.)
| | - Asrar Ahmad
- Center for Sickle Cell Disease, College of Medicine, Howard University, Washington, DC 20059, USA; (A.A.); (N.K.)
| | - Namita Kumari
- Center for Sickle Cell Disease, College of Medicine, Howard University, Washington, DC 20059, USA; (A.A.); (N.K.)
| | - Ornela Rutagarama
- Department of Microbiology, College of Medicine, Howard University, Washington, DC 20059, USA;
| | - Sergei Nekhai
- Department of Microbiology, College of Medicine, Howard University, Washington, DC 20059, USA;
- Center for Sickle Cell Disease, College of Medicine, Howard University, Washington, DC 20059, USA; (A.A.); (N.K.)
- Department of Medicine, College of Medicine, Howard University, Washington, DC 20059, USA
- Correspondence: (M.J.); (S.N.)
| |
Collapse
|
10
|
Cao XX, Yang JK, Wang L. Association between intercellular adhesion molecule 1 (ICAM1) polymorphisms and diabetic foot susceptibility: A case-control study. Medicine (Baltimore) 2020; 99:e18052. [PMID: 32176024 PMCID: PMC7440310 DOI: 10.1097/md.0000000000018052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The objective of the present study was to explore the association between intercellular adhesion molecule 1 (ICAM1) polymorphisms (rs5498 and rs3093030) and diabetic foot (DF) susceptibility in a Chinese Han population.128 type 2 diabetes mellitus (T2DM) patients with DF, 147 T2DM patients without DF, and 155 healthy individuals were enrolled in this study. ICAM1 polymorphisms were genotyped by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). The genotypes and alleles of the polymorphisms were compared by χ test between the 2 groups. Association between ICAM1 polymorphisms and DF susceptibility was expressed through odds ratio (OR) with corresponding 95% confidence interval (95%CI). Effects of ICAM1 polymorphisms on DF clinical characteristics were analyzed by t test.GG genotype of rs5498 polymorphism was distinctly correlated with decreased T2DM risk (OR = 0.369, 95%CI = 0.152-0.895) and reduced susceptibility to DF among healthy controls (OR = 0.316, 95%CI = 0.119-0.837). Similar results were discovered between rs5498 G allele and decreased risk of T2DM (OR = 0.676, 95%CI = 0.475-0.963) and DF (OR = 0.656, 95%CI = 0.453-0.950) among healthy controls. Individuals carrying rs3093030 T allele had low susceptibility to DF developed from T2DM (OR = 0.634, 95%CI = 0.412-0.974). DF patients carrying rs5498 AA genotype had significantly higher serum creatinine levels than GG genotype carriers (P = .003).ICAM1 rs3093030 polymorphism may act as a protective factor against DF developed from T2DM, moreover, rs5498 may be involved in onset of T2DM.Clinical trial number: ChiCTR-INR-18010231.
Collapse
Affiliation(s)
- Xue-Xia Cao
- Department of Endocrinology, Xuanwu Hospital of Capital Medical University, Beijing
| | - Jin-Kui Yang
- Department of Endocrinology, Beijing Tongren Hospital, Beijing, China
| | - Li Wang
- Department of Endocrinology, Xuanwu Hospital of Capital Medical University, Beijing
| |
Collapse
|