1
|
Shi K, Fu W, Farhadi Sabet Z, Ye J, Liang S, Liu T, Liu Q, Guo M, You M, Wu J, Bai R, Liu Y, Hu B, Cui X, Li J, Chen C. Hydrogel-Mediated Jamming of Exosome Communications That Counter Tumor Adaption in the Tumor Immune Microenvironment. ACS NANO 2024. [PMID: 39441690 DOI: 10.1021/acsnano.4c07603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Hypoxia, a common occurrence within solid tumors, can stimulate the dissemination of deceptive tumor exosomes, which function as communicative bridges and orchestrate the recruitment of various supportive cell types for enhanced tumor adaptability in a tumor immune microenvironment. Current nanotechnology provides us intelligent strategies to combat the hypoxic tumor microenvironment. However, once exposed to external stimuli, such as chemotherapy, tumor cells simultaneously release malignant signals to develop tumor migration and immunosuppression, posing challenges to clinical practice. Taking advantage of the membrane-targeting therapeutic strategy, the application of a self-assembled short peptide (PepABS-py), affording hydrogels on tumor cell surfaces, can block exosome dissemination with fiber-like nanostructures and effectively limit the systemic adverse effects of traditional therapeutics. Moreover, PepABS-py can attenuate the hypoxic tumor microenvironment in vivo by carrying an inhibitor of the hypoxic tumor-overexpressed CA IX enzyme, where hypoxia is also a crucial regulator to induce tumor exosomes and mediate intercellular communications within the immune system. Herein, its application on jamming exosome communications can target the T cell-related signaling pathway by regulating microRNAs in exosome cargoes and ultimately enhances CD8+ T cell infiltration and alleviates inflammatory monocytes at metastasis sites. Collectively, with the capability of blocking exosome dissemination, PepABS-py can be applied as a promising tumor membrane-targeting therapeutic tool to counter tumor adaption within an immune microenvironment and further advance traditional chemotherapy.
Collapse
Affiliation(s)
- Kejian Shi
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Wenjiao Fu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
- Sino-Danish College, Sino-Danish Center for Education and Research, University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Zeinab Farhadi Sabet
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Jinmin Ye
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Shijian Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Tao Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Qiaolin Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Mengyu Guo
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Min You
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Junguang Wu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Ru Bai
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Ying Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Bin Hu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Xuejing Cui
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Jiayang Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, New Cornerstone Science Laboratory, National Center for Nanoscience and Technology of China, Beijing 100190, People's Republic of China
- Sino-Danish College, Sino-Danish Center for Education and Research, University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
- Research Unit of Nanoscience and Technology, Chinese Academy of Medical Sciences, Beijing 100021, People's Republic of China
| |
Collapse
|
2
|
Mickael C, Sanders LA, Lee MH, Kumar R, Fonseca-Balladares D, Gandjeva A, Cautivo-Reyes K, Kassa B, Kumar S, Irwin D, Swindle D, Phang T, Stearman RS, Molofsky AB, McKee AS, Stenmark KR, Graham BB, Tuder RM. Classical dendritic cells contribute to hypoxia-induced pulmonary hypertension. FASEB J 2024; 38:e70015. [PMID: 39212294 PMCID: PMC11462638 DOI: 10.1096/fj.202400338rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 08/06/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024]
Abstract
Pulmonary hypertension (PH) is a chronic and progressive disease with significant morbidity and mortality. It is characterized by remodeled pulmonary vessels associated with perivascular and intravascular accumulation of inflammatory cells. Although there is compelling evidence that bone marrow-derived cells, such as macrophages and T cells, cluster in the vicinity of pulmonary vascular lesions in humans and contribute to PH development in different animal models, the role of dendritic cells in PH is less clear. Dendritic cells' involvement in PH is likely since they are responsible for coordinating innate and adaptive immune responses. We hypothesized that dendritic cells drive hypoxic PH. We demonstrate that a classical dendritic cell (cDC) subset (cDC2) is increased and activated in wild-type mouse lungs after hypoxia exposure. We observe significant protection after the depletion of cDCs in ZBTB46 DTR chimera mice before hypoxia exposure and after established hypoxic PH. In addition, we find that cDC depletion is associated with a reduced number of two macrophage subsets in the lung (FolR2+ MHCII+ CCR2+ and FolR2+ MHCII+ CCR2-). We found that depleting cDC2s, but not cDC1s, was protective against hypoxic PH. Finally, proof-of-concept studies in human lungs show increased perivascular cDC2s in patients with Idiopathic Pulmonary Arterial Hypertension (IPAH). Our data points to an essential role of cDCs, particularly cDC2s, in the pathophysiology of experimental PH.
Collapse
Grants
- R01 HL142701 NHLBI NIH HHS
- R01 HL161004 NHLBI NIH HHS
- R01 AI162806 NIAID NIH HHS
- R01HL142701 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01AI162806 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- American Thoracic Society (ATS)
- K01 HL161024 NHLBI NIH HHS
- K08HL168310 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- K01HL161024 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL135872 NHLBI NIH HHS
- W81XWH2210457 U.S. Department of Defense (DOD)
- Cardiovascular Medical Research and Education Fund (CMREF)
- Actelion Pharmaceuticals (Actelion Pharmaceuticals Ltd)
- R25HL146166 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R25 HL146166 NHLBI NIH HHS
- R01NS126765 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- K08 HL168310 NHLBI NIH HHS
- United Therapeutics Corporation (Uni Ther)
- R01 NS126765 NINDS NIH HHS
- 19CDA34730030 American Heart Association (AHA)
- R01HL135872 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- PO1HL152961 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL158076 NHLBI NIH HHS
- R01 H161004 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- P01 HL152961 NHLBI NIH HHS
Collapse
Affiliation(s)
- Claudia Mickael
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Aurora, Colorado, USA
- Cardiovascular Research Laboratories, University of Colorado, Aurora, Colorado, USA
| | - Linda A. Sanders
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Aurora, Colorado, USA
- Cardiovascular Research Laboratories, University of Colorado, Aurora, Colorado, USA
| | - Michael H. Lee
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
- Lung Biology Center, Zuckerberg San Francisco General Hospital, San Francisco, California, USA
| | - Rahul Kumar
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
- Lung Biology Center, Zuckerberg San Francisco General Hospital, San Francisco, California, USA
| | - Dara Fonseca-Balladares
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
- Lung Biology Center, Zuckerberg San Francisco General Hospital, San Francisco, California, USA
| | - Aneta Gandjeva
- Cardiovascular Research Laboratories, University of Colorado, Aurora, Colorado, USA
| | - Kelly Cautivo-Reyes
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA
- Gilead Sciences, Foster City, California, USA
| | - Biruk Kassa
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
- Lung Biology Center, Zuckerberg San Francisco General Hospital, San Francisco, California, USA
| | - Sushil Kumar
- Cardiovascular Research Laboratories, University of Colorado, Aurora, Colorado, USA
| | - David Irwin
- Cardiovascular Research Laboratories, University of Colorado, Aurora, Colorado, USA
| | - Delaney Swindle
- Cardiovascular Research Laboratories, University of Colorado, Aurora, Colorado, USA
| | - Tzu Phang
- Section of Hematology, Oncology, and Bone Marrow Transplantation-Cellular Therapeutics (BMT-CT), Department of Pediatrics, School of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Robert S. Stearman
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep, and Occupational Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Ari B. Molofsky
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA
| | - Amy S. McKee
- Department of Medicine, Division of Clinical Immunology, University of Colorado, Aurora, Colorado, USA
- Department of Microbiology and Immunology and ClinImmune Cell and Gene Therapy, University of Colorado, Aurora, Colorado, USA
| | - Kurt R. Stenmark
- Cardiovascular Research Laboratories, University of Colorado, Aurora, Colorado, USA
| | - Brian B. Graham
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
- Lung Biology Center, Zuckerberg San Francisco General Hospital, San Francisco, California, USA
| | - Rubin M. Tuder
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Aurora, Colorado, USA
- Cardiovascular Research Laboratories, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
3
|
Marallano VJ, Ughetta ME, Tejero R, Nanda S, Ramalingam R, Stalbow L, Sattiraju A, Huang Y, Ramakrishnan A, Shen L, Wojcinski A, Kesari S, Zou H, Tsankov AM, Friedel RH. Hypoxia drives shared and distinct transcriptomic changes in two invasive glioma stem cell lines. Sci Rep 2024; 14:7246. [PMID: 38538643 PMCID: PMC10973515 DOI: 10.1038/s41598-024-56102-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 03/01/2024] [Indexed: 07/12/2024] Open
Abstract
Glioblastoma (GBM) is the most common primary malignant cancer of the central nervous system. Insufficient oxygenation (hypoxia) has been linked to GBM invasion and aggression, leading to poor patient outcomes. Hypoxia induces gene expression for cellular adaptations. However, GBM is characterized by high intertumoral (molecular subtypes) and intratumoral heterogeneity (cell states), and it is not well understood to what extent hypoxia triggers patient-specific gene responses and cellular diversity in GBM. Here, we surveyed eight patient-derived GBM stem cell lines for invasion phenotypes in 3D culture, which identified two GBM lines showing increased invasiveness in response to hypoxia. RNA-seq analysis of the two patient GBM lines revealed a set of shared hypoxia response genes concerning glucose metabolism, angiogenesis, and autophagy, but also a large set of patient-specific hypoxia-induced genes featuring cell migration and anti-inflammation, highlighting intertumoral diversity of hypoxia responses in GBM. We further applied the Shared GBM Hypoxia gene signature to single cell RNA-seq datasets of glioma patients, which showed that hypoxic cells displayed a shift towards mesenchymal-like (MES) and astrocyte-like (AC) states. Interestingly, in response to hypoxia, tumor cells in IDH-mutant gliomas displayed a strong shift to the AC state, whereas tumor cells in IDH-wildtype gliomas mainly shifted to the MES state. This distinct hypoxia response of IDH-mutant gliomas may contribute to its more favorable prognosis. Our transcriptomic studies provide a basis for future approaches to better understand the diversity of hypoxic niches in gliomas.
Collapse
Affiliation(s)
- Valerie J Marallano
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Mary E Ughetta
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Rut Tejero
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sidhanta Nanda
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Rohana Ramalingam
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Lauren Stalbow
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Anirudh Sattiraju
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Yong Huang
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Aarthi Ramakrishnan
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Li Shen
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Alexandre Wojcinski
- Pacific Neuroscience Institute and Saint John's Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA, 90404, USA
| | - Santosh Kesari
- Pacific Neuroscience Institute and Saint John's Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA, 90404, USA
| | - Hongyan Zou
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Alexander M Tsankov
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Roland H Friedel
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
4
|
Gornostaeva AN, Bobyleva PI, Andreeva ER, Gogiya BS, Buravkova LB. Alteration of PBMC transcriptome profile after interaction with multipotent mesenchymal stromal cells under "physiological" hypoxia. Immunobiology 2024; 229:152766. [PMID: 38091798 DOI: 10.1016/j.imbio.2023.152766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/17/2023] [Accepted: 11/22/2023] [Indexed: 01/21/2024]
Abstract
Multipotent mesenchymal stromal cells (MSCs) have demonstrated a pronounced immunosuppressive activity, the manifestation of which depends on the microenvironmental factors, including O2 level. Here we examined the effects of MSCs on transcriptomic profile of allogeneic phytohemagglutinin-stimulated peripheral blood mononuclear cells (PBMCs) after interaction at ambient (20%) or "physiological" hypoxia (5%) O2. As revealed with microarray analysis, PBMC transcriptome at 20% O2 was more affected, which was manifested as differential expression of more than 300 genes, whereas under 5% O2 220 genes were changed. Most of genes at 20% O2 were downregulated, while at hypoxia most of genes were upregulated. Altered gene patterns were only partly overlapped at different O2 levels. A set of altered genes at hypoxia only was of particular interest. According to Gene Ontology a part of above genes was responsible for adhesion, cell communication, and immune response. At both oxygen concentrations, MSCs demonstrated effective immunosuppression manifested as attenuation of T cell activation and proliferation as well as anti-inflammatory shift of cytokine profile. Thus, MSC-mediated immunosuppression is executed with greater efficacy at a "physiological" hypoxia, since the same result has been achieved through a change in the expression of a fewer genes in target PBMCs.
Collapse
Affiliation(s)
- A N Gornostaeva
- Cell Physiology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences, Khoroshevskoye shosse 76a, 123007 Moscow, Russia.
| | - P I Bobyleva
- Cell Physiology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences, Khoroshevskoye shosse 76a, 123007 Moscow, Russia
| | - E R Andreeva
- Cell Physiology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences, Khoroshevskoye shosse 76a, 123007 Moscow, Russia
| | - B Sh Gogiya
- Department of Herniology and Plastic Surgery, A. V. Vishnevsky Institute of Surgery, Bolshaya Serpukhovskaya Str, 27, 117997 Moscow, Russia
| | - L B Buravkova
- Cell Physiology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences, Khoroshevskoye shosse 76a, 123007 Moscow, Russia
| |
Collapse
|
5
|
Bhatt K, Nukovic A, Colombani T, Bencherif SA. Biomaterial-assisted local oxygenation safeguards the prostimulatory phenotype and functions of human dendritic cells in hypoxia. Front Immunol 2023; 14:1278397. [PMID: 38169677 PMCID: PMC10758617 DOI: 10.3389/fimmu.2023.1278397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 11/08/2023] [Indexed: 01/05/2024] Open
Abstract
Dendritic cells (DCs), professional antigen-presenting cells, function as sentinels of the immune system. DCs initiate and fine-tune adaptive immune responses by presenting antigenic peptides to B and T lymphocytes to mount an effective immune response against cancer and pathogens. However, hypoxia, a condition characterized by low oxygen (O2) tension in different tissues, significantly impacts DC functions, including antigen uptake, activation and maturation, migration, as well as T-cell priming and proliferation. In this study, we employed O2-releasing biomaterials (O2-cryogels) to study the effect of localized O2 supply on human DC phenotype and functions. Our results indicate that O2-cryogels effectively mitigate DC exposure to hypoxia under hypoxic conditions. Additionally, O2-cryogels counteract hypoxia-induced inhibition of antigen uptake and migratory activity in DCs through O2 release and hyaluronic acid (HA) mediated mechanisms. Furthermore, O2-cryogels preserve and restore DC maturation and co-stimulation markers, including HLA-DR, CD86, and CD40, along with the secretion of proinflammatory cytokines in hypoxic conditions. Finally, our findings demonstrate that the supplemental O2 released from the cryogels preserves DC-mediated T-cell priming, ultimately leading to the activation and proliferation of allogeneic CD3+ T cells. This work emphasizes the potential of local oxygenation as a powerful immunomodulatory agent to improve DC activation and functions in hypoxia, offering new approaches for cancer and infectious disease treatments.
Collapse
Affiliation(s)
- Khushbu Bhatt
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, United States
| | - Alexandra Nukovic
- Department of Chemical Engineering, Northeastern University, Boston, MA, United States
| | - Thibault Colombani
- Department of Chemical Engineering, Northeastern University, Boston, MA, United States
| | - Sidi A. Bencherif
- Department of Chemical Engineering, Northeastern University, Boston, MA, United States
- Department of Bioengineering, Northeastern University, Boston, MA, United States
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, United States
| |
Collapse
|
6
|
Jia Q, Huang Z, Wang G, Sun X, Wu Y, Yang B, Yang T, Liu J, Li P, Li J. Osteopontin: An important protein in the formation of kidney stones. Front Pharmacol 2022; 13:1036423. [PMID: 36452224 PMCID: PMC9703462 DOI: 10.3389/fphar.2022.1036423] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 10/26/2022] [Indexed: 09/10/2024] Open
Abstract
The incidence of kidney stones averages 10%, and the recurrence rate of kidney stones is approximately 10% at 1 year, 35% at 5 years, 50% at 10 years, and 75% at 20 years. However, there is currently a lack of good medicines for the prevention and treatment of kidney stones. Osteopontin (OPN) is an important protein in kidney stone formation, but its role is controversial, with some studies suggesting that it inhibits stone formation, while other studies suggest that it can promote stone formation. OPN is a highly phosphorylated protein, and with the deepening of research, there is growing evidence that it promotes stone formation, and the phosphorylated protein is believed to have adhesion effect, promote stone aggregation and nucleation. In addition, OPN is closely related to immune cell infiltration, such as OPN as a pro-inflammatory factor, which can activate mast cells (degranulate to release various inflammatory factors), macrophages (differentiated into M1 macrophages), and T cells (differentiated into T1 cells) etc., and these inflammatory cells play a role in kidney damage and stone formation. In short, OPN mainly exists in the phosphorylated form in kidney stones, plays an important role in the formation of stones, and may be an important target for drug therapy of kidney stones.
Collapse
Affiliation(s)
- Qingxia Jia
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
| | - Ziye Huang
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Guang Wang
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xia Sun
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yuyun Wu
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Bowei Yang
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Tongxin Yang
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jianhe Liu
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Pei Li
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jiongming Li
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
7
|
CCL18 Expression Is Higher in a Glioblastoma Multiforme Tumor than in the Peritumoral Area and Causes the Migration of Tumor Cells Sensitized by Hypoxia. Int J Mol Sci 2022; 23:ijms23158536. [PMID: 35955670 PMCID: PMC9369326 DOI: 10.3390/ijms23158536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 07/28/2022] [Accepted: 07/28/2022] [Indexed: 12/10/2022] Open
Abstract
Glioblastoma multiforme (GBM) is a brain tumor with a very poor prognosis. For this reason, researchers worldwide study the impact of the tumor microenvironment in GBM, such as the effect of chemokines. In the present study, we focus on the role of the chemokine CCL18 and its receptors in the GBM tumor. We measured the expression of CCL18, CCR8 and PITPNM3 in the GMB tumor from patients (16 men and 12 women) using quantitative real-time polymerase chain reaction. To investigate the effect of CCL18 on the proliferation and migration of GBM cells, experiments were performed using U-87 MG cells. The results showed that CCL18 expression was higher in the GBM tumor than in the peritumoral area. The women had a decreased expression of PITPNM3 receptor in the GBM tumor, while in the men a lower expression of CCR8 was observed. The hypoxia-mimetic agent, cobalt chloride (CoCl2), increased the expression of CCL18 and PITPNM3 and thereby sensitized U-87 MG cells to CCL18, which did not affect the proliferation of U-87 MG cells but increased the migration of the test cells. The results indicate that GBM cells migrate from hypoxic areas, which may be important in understanding the mechanisms of tumorigenesis.
Collapse
|
8
|
Davuluri KS, Singh AK, Kumar V, Singh SV, Singh AV, Kumar S, Yadav R, Kushwaha S, Chauhan DS. Stimulated expression of ELR+ chemokines, VEGFA and TNF-AIP3 promote mycobacterial dissemination in extrapulmonary tuberculosis patients and Cavia porcellus model of tuberculosis. Tuberculosis (Edinb) 2022; 135:102224. [PMID: 35763913 DOI: 10.1016/j.tube.2022.102224] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 06/06/2022] [Accepted: 06/12/2022] [Indexed: 11/16/2022]
Abstract
Pathogenic mycobacteria induce and accelerate blood vessel formation driven by extensive inflammation during granuloma formation, which is a central feature of mycobacterial pathogenesis. Tumor necrosis factor-alpha (TNF-α) enhances the expression of vascular endothelial growth factor (VEGF) and glutamic acid-leucine-arginine (ELR+) chemokines, which are potent inducers of vascularization. Most of the reported research work contends that VEGF growth factor induces neovascularization in human tuberculosis (TB) patients, but the evidence is inconclusive. Considerable ambiguity exists concerning the factors responsible for miliary tuberculosis. To identify such factors, we proposed an alternative explanation that could be found in miliary tuberculosis (MTB) cases. We performed a comparative analysis of angiogenic factors TNF-α, VEGF, and angiogenic ELR+ CXC and CC chemokine ligands in extrapulmonary tuberculosis (EPTB) and pulmonary tuberculosis (PTB) patients. To observe the relationship of these factors with the severity of bacterial burden, guinea pigs were infected with Mycobacterium tuberculosis (M.tb) and levels of the angiogenic factors were examined at different time intervals. Expression of these factors also exhibited a significant positive correlation with bacterial burden in other organs like the spleen, liver, and lymph nodes. We demonstrated statistical data on bacterial burden at different time points following the dissemination of infection in guinea pigs. In this study, we observed that there was a stimulated increase in the expression of ELR+ chemokines and VEGF in EPTB patients as compared to PTB patients. Following increased dissemination, the host immune response clears bacteria from the lungs during disease progression in guinea pigs.
Collapse
Affiliation(s)
- Kusuma Sai Davuluri
- Department of Microbiology and Molecular Biology, National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Tajganj, Agra, 282001, India.
| | - Amit Kumar Singh
- Department of Animal Experimentation and Facility, National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Tajganj, Agra, 281406, India.
| | - Vimal Kumar
- Department of Animal Experimentation and Facility, National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Tajganj, Agra, 281406, India.
| | - Shoor Vir Singh
- Department of Biotechnology, GLA University, Mathura, 281406, India.
| | - Ajay Vir Singh
- Department of Microbiology and Molecular Biology, National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Tajganj, Agra, 282001, India.
| | - Santhosh Kumar
- Department of Pulmonary Medicine, SNMC, Agra, 282001, India
| | - Rajbala Yadav
- Department of Microbiology and Molecular Biology, National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Tajganj, Agra, 282001, India
| | - Shweta Kushwaha
- Department of Microbiology and Molecular Biology, National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Tajganj, Agra, 282001, India
| | - Devendra Singh Chauhan
- Department of Microbiology and Molecular Biology, National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Tajganj, Agra, 282001, India.
| |
Collapse
|
9
|
Shiezadeh F, Azami N, Arab HR, Rezaee SAR, Moeintaghavi A, Banihashemrad A. Evaluation of Neutrophilic Receptors; CXCL8 and CXCR2 in Patients with Chronic Periodontitis Compared to Healthy Subjects by Real Time PCR Method. Open Dent J 2022. [DOI: 10.2174/18742106-v16-e2202241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Objective:
This study aimed to evaluate the levels of CXCL8 and its receptor (CXCR2) in gingival tissue neutrophils from patients with chronic periodontitis compared to periodontal healthy subjects using Real Time PCR method.
Methods:
The test group consisted of 21 patients with chronic periodontitis and the control group consisted of 18 healthy individuals. In both groups, gingival tissue samples were obtained during periodontal surgery.CXCL8 and CXCR2 RNA in tissue samples were examined by PCR method, and then the levels of genes expression were measured. Mann-Whitney U nonparametric test was used for statistical analysis.
Results:
CXCL8 gene expression in the gingival tissue of the test group with chronic periodontitis was significantly higher than the control group (p=0.028). CXCR2 gene expression in the gingival tissue of the test group with chronic periodontitis was significantly lower than the control group (p=0.043). In both test and control groups, there was a negative correlation between CXCL8 and CXCR2 gene expression. This correlation was statistically significant in the test group (p=0.001), but there was no significant correlation in the control group (p=0.431).
Conclusion:
The results of this present study suggested that the level of gene expression for CXCL8 was greater in patients with chronic periodontitis and CXCR2 was greater in healthy individuals. Although in people with chronic periodontitis, CXCR2 decreases slightly as CXCL8 levels increase.
Collapse
|
10
|
Jiang Y, Xi Y, Li Y, Zuo Z, Zeng C, Fan J, Zhang D, Tao H, Guo Y. Ethanol promoting the upregulation of C-X-C Motif Chemokine Ligand 1(CXCL1) and C-X-C Motif Chemokine Ligand 6(CXCL6) in models of early alcoholic liver disease. Bioengineered 2022; 13:4688-4701. [PMID: 35156518 PMCID: PMC8973977 DOI: 10.1080/21655979.2022.2030557] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Alcoholic liver disease (ALD) denotes a series of liver diseases caused by ethanol. Recently, immune-related genes (IRGs) play increasingly crucial role in diseases. However, it’s unclear the role of IRGs in ALD. Bioinformatic analysis was used to discern the core immune-related differential genes (IRDGs) in the present study. Subsequently, Cell Counting Kit-8 say, oil red O staining, and triglyceride detection were employed to explore optimal experimental conditions of establishing hepatocellular models of early ALD. Ultimately, real-time reverse transcription-PCR and immunohistochemistry/immunocytochemistry methods were adopted to verify the expressions of mRNA and proteins of core IRDGs, respectively. C-X-C Motif Chemokine Ligand 1 (Cxcl1) and Cxcl6 were regarded as core IRDGs via integrated bioinformatics analysis. Besides, Lieber Decarli Ethanol feeding and 200 mM and 300 mM ethanol stimulating L02 cells for 36 h can both successfully hepatocellular model. In ethanol groups, the levels of CXCL1 and CXCL6 mRNA were significantly upregulated than pair-fed groups (P < 0.0001). Also, immunohistochemistry revealed that positive particles of CXCL1 and CXCL6 in mice model of early ALD were obviously more than control groups (P < 0.0001). Besides, in L02 hepatocytes stimulated by ethanol, CXCL1 and CXCL6 mRNA were over-expressed, compared with normal L02 cells (P < 0.0001). Meanwhile, immunocytochemistry indicated that CXCL1 and CXCL6 proteins in hepatocellular model of early ALD were higher than normal L02 hepatocytes stimulus (P < 0.0001). Ethanol promoted the upregulation of Cxcl1 and Cxcl6 mRNA and proteins in models of early ALD, denoting their potentiality of acting as biomarkers of ALD.
Collapse
Affiliation(s)
- Yao Jiang
- Clinical Laboratory, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Yuge Xi
- Clinical Laboratory, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Yiqin Li
- Clinical Laboratory, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Zhihua Zuo
- Department of Clinical Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Chuyi Zeng
- Department of Clinical Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jia Fan
- Clinical Laboratory, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Dan Zhang
- Clinical Laboratory, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Hualin Tao
- Department of Clinical Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yongcan Guo
- Clinical Laboratory, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
11
|
Ge S, Yang W, Chen H, Yuan Q, Liu S, Zhao Y, Zhang J. MyD88 in Macrophages Enhances Liver Fibrosis by Activation of NLRP3 Inflammasome in HSCs. Int J Mol Sci 2021; 22:ijms222212413. [PMID: 34830293 PMCID: PMC8622429 DOI: 10.3390/ijms222212413] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/04/2021] [Accepted: 11/15/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic liver disease mediated by the activation of hepatic stellate cells (HSCs) leads to liver fibrosis. The signal adaptor MyD88 of Toll-like receptor (TLR) signaling is involved during the progression of liver fibrosis. However, the specific role of MyD88 in myeloid cells in liver fibrosis has not been thoroughly investigated. In this study, we used a carbon tetrachloride (CCl4)-induced mouse fibrosis model in which MyD88 was selectively depleted in myeloid cells. MyD88 deficiency in myeloid cells attenuated liver fibrosis in mice and decreased inflammatory cell infiltration. Furthermore, deficiency of MyD88 in macrophages inhibits the secretion of CXC motif chemokine 2 (CXCL2), which restrains the activation of HSCs characterized by NLR Family Pyrin Domain Containing 3 (NLRP3) inflammasome activation. Moreover, targeting CXCL2 by CXCR2 inhibitors attenuated the activation of HSCs and reduced liver fibrosis. Thus, MyD88 may represent a potential candidate target for the prevention and treatment of liver fibrosis.
Collapse
Affiliation(s)
- Shuang Ge
- National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-Targeting Theranostics, Guangxi Medical University, Nanning 530021, China; (S.G.); (W.Y.)
| | - Wei Yang
- National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-Targeting Theranostics, Guangxi Medical University, Nanning 530021, China; (S.G.); (W.Y.)
| | - Haiqiang Chen
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China; (H.C.); (Q.Y.); (S.L.)
| | - Qi Yuan
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China; (H.C.); (Q.Y.); (S.L.)
| | - Shi Liu
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China; (H.C.); (Q.Y.); (S.L.)
| | - Yongxiang Zhao
- National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-Targeting Theranostics, Guangxi Medical University, Nanning 530021, China; (S.G.); (W.Y.)
- Correspondence: (Y.Z.); (J.Z.)
| | - Jinhua Zhang
- National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-Targeting Theranostics, Guangxi Medical University, Nanning 530021, China; (S.G.); (W.Y.)
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China; (H.C.); (Q.Y.); (S.L.)
- Correspondence: (Y.Z.); (J.Z.)
| |
Collapse
|
12
|
Blocking HIF to Enhance NK Cells: Hints for New Anti-Tumor Therapeutic Strategies? Vaccines (Basel) 2021; 9:vaccines9101144. [PMID: 34696251 PMCID: PMC8539190 DOI: 10.3390/vaccines9101144] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 09/30/2021] [Accepted: 10/01/2021] [Indexed: 12/18/2022] Open
Abstract
Natural Killer (NK) cells are becoming an ever more promising tool to design new anti-tumor strategies. However, two major issues are still a challenge to obtain versatile and effective NK-based therapies: the way to maximize the persistency of powerful NK effectors in the patient, and the way to overcome the multiple escape mechanisms that keep away or suppress NK cells at the tumor site. In this regard, targeting the hypoxia-inducible factors (HIFs), which is important for both tumor progression and immune suppression, may be an opportunity. Especially, in the context of the ongoing studies focused on more effective NK-based therapeutic products.
Collapse
|
13
|
The Role of Dendritic Cells during Physiological and Pathological Dentinogenesis. J Clin Med 2021; 10:jcm10153348. [PMID: 34362130 PMCID: PMC8348392 DOI: 10.3390/jcm10153348] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/25/2021] [Accepted: 07/26/2021] [Indexed: 12/13/2022] Open
Abstract
The dental pulp is a soft connective tissue of ectomesenchymal origin that harbors distinct cell populations, capable of interacting with each other to maintain the vitality of the tooth. After tooth injuries, a sequence of complex biological events takes place in the pulpal tissue to restore its homeostasis. The pulpal response begins with establishing an inflammatory reaction that leads to the formation of a matrix of reactionary or reparative dentin, according to the nature of the exogenous stimuli. Using several in vivo designs, antigen-presenting cells, including macrophages and dendritic cells (DCs), are identified in the pulpal tissue before tertiary dentin deposition under the afflicted area. However, the precise nature of this phenomenon and its relationship to inherent pulp cells are not yet clarified. This literature review aims to discuss the role of pulpal DCs and their relationship to progenitor/stem cells, odontoblasts or odontoblast-like cells, and other immunocompetent cells during physiological and pathological dentinogenesis. The concept of “dentin-pulp immunology” is proposed for understanding the crosstalk among these cell types after tooth injuries, and the possibility of immune-based therapies is introduced to accelerate pulpal healing after exogenous stimuli.
Collapse
|
14
|
Bhattacharya S, Agarwal S, Shrimali NM, Guchhait P. Interplay between hypoxia and inflammation contributes to the progression and severity of respiratory viral diseases. Mol Aspects Med 2021; 81:101000. [PMID: 34294412 PMCID: PMC8287505 DOI: 10.1016/j.mam.2021.101000] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/07/2021] [Accepted: 07/16/2021] [Indexed: 02/07/2023]
Abstract
History of pandemics is dominated by viral infections and specifically respiratory viral diseases like influenza and COVID-19. Lower respiratory tract infection is the fourth leading cause of death worldwide. Crosstalk between resultant inflammation and hypoxic microenvironment may impair ventilatory response of lungs. This reduces arterial partial pressure of oxygen, termed as hypoxemia, which is observed in a section of patients with respiratory virus infections including SARS-CoV-2 (COVID-19). In this review, we describe the interplay between inflammation and hypoxic microenvironment in respiratory viral infection and its contribution to disease pathogenesis.
Collapse
Affiliation(s)
- Sulagna Bhattacharya
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India; School of Biotechnology, Kalinga Institute of Industrial Technology, Orissa, India
| | - Sakshi Agarwal
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India
| | - Nishith M Shrimali
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India
| | - Prasenjit Guchhait
- Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India.
| |
Collapse
|
15
|
Korbecki J, Kojder K, Kapczuk P, Kupnicka P, Gawrońska-Szklarz B, Gutowska I, Chlubek D, Baranowska-Bosiacka I. The Effect of Hypoxia on the Expression of CXC Chemokines and CXC Chemokine Receptors-A Review of Literature. Int J Mol Sci 2021; 22:ijms22020843. [PMID: 33467722 PMCID: PMC7830156 DOI: 10.3390/ijms22020843] [Citation(s) in RCA: 129] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/06/2021] [Accepted: 01/12/2021] [Indexed: 12/26/2022] Open
Abstract
Hypoxia is an integral component of the tumor microenvironment. Either as chronic or cycling hypoxia, it exerts a similar effect on cancer processes by activating hypoxia-inducible factor-1 (HIF-1) and nuclear factor (NF-κB), with cycling hypoxia showing a stronger proinflammatory influence. One of the systems affected by hypoxia is the CXC chemokine system. This paper reviews all available information on hypoxia-induced changes in the expression of all CXC chemokines (CXCL1, CXCL2, CXCL3, CXCL4, CXCL5, CXCL6, CXCL7, CXCL8 (IL-8), CXCL9, CXCL10, CXCL11, CXCL12 (SDF-1), CXCL13, CXCL14, CXCL15, CXCL16, CXCL17) as well as CXC chemokine receptors—CXCR1, CXCR2, CXCR3, CXCR4, CXCR5, CXCR6, CXCR7 and CXCR8. First, we present basic information on the effect of these chemoattractant cytokines on cancer processes. We then discuss the effect of hypoxia-induced changes on CXC chemokine expression on the angiogenesis, lymphangiogenesis and recruitment of various cells to the tumor niche, including myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs), regulatory T cells (Tregs) and tumor-infiltrating lymphocytes (TILs). Finally, the review summarizes data on the use of drugs targeting the CXC chemokine system in cancer therapies.
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (P.K.); (P.K.); (D.C.)
| | - Klaudyna Kojder
- Department of Anaesthesiology and Intensive Care, Pomeranian Medical University in Szczecin, Unii Lubelskiej 1, 71-281 Szczecin, Poland;
| | - Patrycja Kapczuk
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (P.K.); (P.K.); (D.C.)
| | - Patrycja Kupnicka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (P.K.); (P.K.); (D.C.)
| | - Barbara Gawrońska-Szklarz
- Department of Pharmacokinetics and Therapeutic Drug Monitoring, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland;
| | - Izabela Gutowska
- Department of Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72 Av., 70-111 Szczecin, Poland;
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (P.K.); (P.K.); (D.C.)
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 Av., 70-111 Szczecin, Poland; (J.K.); (P.K.); (P.K.); (D.C.)
- Correspondence: ; Tel.: +48-914661515
| |
Collapse
|
16
|
Wang B, Zhao Q, Zhang Y, Liu Z, Zheng Z, Liu S, Meng L, Xin Y, Jiang X. Targeting hypoxia in the tumor microenvironment: a potential strategy to improve cancer immunotherapy. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:24. [PMID: 33422072 PMCID: PMC7796640 DOI: 10.1186/s13046-020-01820-7] [Citation(s) in RCA: 134] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 12/20/2020] [Indexed: 12/14/2022]
Abstract
With the success of immune checkpoint inhibitors (ICIs), significant progress has been made in the field of cancer immunotherapy. Despite the long-lasting outcomes in responders, the majority of patients with cancer still do not benefit from this revolutionary therapy. Increasing evidence suggests that one of the major barriers limiting the efficacy of immunotherapy seems to coalesce with the hypoxic tumor microenvironment (TME), which is an intrinsic property of all solid tumors. In addition to its impact on shaping tumor invasion and metastasis, the hypoxic TME plays an essential role in inducing immune suppression and resistance though fostering diverse changes in stromal cell biology. Therefore, targeting hypoxia may provide a means to enhance the efficacy of immunotherapy. In this review, the potential impact of hypoxia within the TME, in terms of key immune cell populations, and the contribution to immune suppression are discussed. In addition, we outline how hypoxia can be manipulated to tailor the immune response and provide a promising combinational therapeutic strategy to improve immunotherapy.
Collapse
Affiliation(s)
- Bin Wang
- Department of Radiation Oncology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China.,Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, 130021, China.,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China
| | - Qin Zhao
- Department of Radiation Oncology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China.,Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, 130021, China.,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China
| | - Yuyu Zhang
- Department of Radiation Oncology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China.,Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, 130021, China.,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China
| | - Zijing Liu
- Department of Radiation Oncology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China.,Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, 130021, China.,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China
| | - Zhuangzhuang Zheng
- Department of Radiation Oncology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China.,Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, 130021, China.,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China
| | - Shiyu Liu
- Department of Radiation Oncology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China.,Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, 130021, China.,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China
| | - Lingbin Meng
- Department of Hematology and Medical Oncology, Moffitt Cancer Center, Tampa, FL, 33612, USA
| | - Ying Xin
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, 126 Xinmin Street, Changchun, 130021, China.
| | - Xin Jiang
- Department of Radiation Oncology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China. .,Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, 130021, China. .,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China.
| |
Collapse
|
17
|
Armitage JD, Newnes HV, McDonnell A, Bosco A, Waithman J. Fine-Tuning the Tumour Microenvironment: Current Perspectives on the Mechanisms of Tumour Immunosuppression. Cells 2021; 10:E56. [PMID: 33401460 PMCID: PMC7823446 DOI: 10.3390/cells10010056] [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: 12/08/2020] [Revised: 12/28/2020] [Accepted: 12/30/2020] [Indexed: 02/07/2023] Open
Abstract
Immunotherapy has revolutionised the treatment of cancers by harnessing the power of the immune system to eradicate malignant tissue. However, it is well recognised that some cancers are highly resistant to these therapies, which is in part attributed to the immunosuppressive landscape of the tumour microenvironment (TME). The contexture of the TME is highly heterogeneous and contains a complex architecture of immune, stromal, vascular and tumour cells in addition to acellular components such as the extracellular matrix. While understanding the dynamics of the TME has been instrumental in predicting durable responses to immunotherapy and developing new treatment strategies, recent evidence challenges the fundamental paradigms of how tumours can effectively subvert immunosurveillance. Here, we discuss the various immunosuppressive features of the TME and how fine-tuning these mechanisms, rather than ablating them completely, may result in a more comprehensive and balanced anti-tumour response.
Collapse
Affiliation(s)
- Jesse D. Armitage
- Telethon Kids Institute, The University of Western Australia, Nedlands, WA 6009, Australia; (J.D.A.); (H.V.N.); (A.M.)
| | - Hannah V. Newnes
- Telethon Kids Institute, The University of Western Australia, Nedlands, WA 6009, Australia; (J.D.A.); (H.V.N.); (A.M.)
| | - Alison McDonnell
- Telethon Kids Institute, The University of Western Australia, Nedlands, WA 6009, Australia; (J.D.A.); (H.V.N.); (A.M.)
- National Centre for Asbestos Related Diseases, QEII Medical Centre, The University of Western Australia, Nedlands, WA 6009, Australia
| | - Anthony Bosco
- Telethon Kids Institute, The University of Western Australia, Nedlands, WA 6009, Australia; (J.D.A.); (H.V.N.); (A.M.)
| | - Jason Waithman
- Telethon Kids Institute, The University of Western Australia, Nedlands, WA 6009, Australia; (J.D.A.); (H.V.N.); (A.M.)
| |
Collapse
|
18
|
Han Y, Ding L, Cheng X, Zhao M, Zhao T, Guo L, Li X, Geng Y, Fan M, Liao H, Zhu L. Hypoxia Augments Cerebral Inflammation in a Dextran Sulfate Sodium-Induced Colitis Mouse Model. Front Cell Neurosci 2020; 14:611764. [PMID: 33362475 PMCID: PMC7756107 DOI: 10.3389/fncel.2020.611764] [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: 09/30/2020] [Accepted: 11/18/2020] [Indexed: 11/23/2022] Open
Abstract
The importance of hypoxia in the pathophysiology of inflammatory bowel disease (IBD) is increasingly being realized; also, hypoxia seems to be an important accelerator of brain inflammation, as has been reported by our group and others. IBD is a chronic intestinal disorder that leads to the development of inflammation, which is related to brain dysfunction. However, no studies have reported whether hypoxia is associated with IBD-induced neuroinflammation. Therefore, the objective of the present study was to determine whether hypoxia augments cerebral inflammation in a DSS-induced colitis mouse model. The mouse model was developed using 3% DSS for five days combined with exposure to hypoxic conditions (6,000 m) for two days. Mice were randomly divided into four groups: control group, DSS group, hypoxia group, and DSS plus hypoxia group. The results demonstrated that DSS combined with hypoxia resulted in up-regulation of colonic and plasmatic proinflammatory cytokines. Meanwhile, DSS plus hypoxia increased expression of Iba1, which is a marker of activated microglia, accompanied by increased expression of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6) in the brain. Moreover, the expression of tight junction proteins, such as zonula occludens-1 (ZO-1), occludin, and claudin-5, was markedly downregulated. The current study provides new insight into how hypoxia exposure induces excessive inflammatory responses andpathophysiological consequences in the brain in a DSS-induced colitis model.
Collapse
Affiliation(s)
- Ying Han
- Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, China.,Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Liping Ding
- National Nanjing Center for Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Xiang Cheng
- Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Ming Zhao
- Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Tong Zhao
- Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Liang Guo
- Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Xinyang Li
- Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Yanan Geng
- Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, China.,Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Ming Fan
- Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, China.,Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Hong Liao
- National Nanjing Center for Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Lingling Zhu
- Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
19
|
Monaci S, Aldinucci C, Rossi D, Giuntini G, Filippi I, Ulivieri C, Marotta G, Sozzani S, Carraro F, Naldini A. Hypoxia Shapes Autophagy in LPS-Activated Dendritic Cells. Front Immunol 2020; 11:573646. [PMID: 33329536 PMCID: PMC7734254 DOI: 10.3389/fimmu.2020.573646] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 10/28/2020] [Indexed: 01/07/2023] Open
Abstract
During their lifespan, dendritic cells (DCs) are exposed to different pO2 levels that affect their differentiation and functions. Autophagy is one of the adaptive responses to hypoxia with important implications for cell survival. While the autophagic machinery in DCs was shown to impact signaling of TLRs, its regulation by the MD-2/TLR4 ligand LPS is still unclear. The aim of this study was to evaluate whether LPS can induce autophagy in DCs exposed to either aerobic or hypoxic conditions. Using human monocyte-derived DCs and the combination of immunofluorescence confocal analysis, measure of mitochondrial membrane potential, Western blotting, and RT-qPCR, we showed that the ability of LPS to modulate autophagy was strictly dependent upon pO2 levels. Indeed, LPS inhibited autophagy in aerobic conditions whereas the autophagic process was induced in a hypoxic environment. Under hypoxia, LPS treatment caused a significant increase of functional lysosomes, LC3B and Atg protein upregulation, and reduction of SQSTM1/p62 protein levels. This selective regulation was accompanied by activation of signalling pathways and expression of cytokines typically associated with DC survival. Bafilomycin A1 and chloroquine, which are recognized as autophagic inhibitors, confirmed the induction of autophagy by LPS under hypoxia and its impact on DC survival. In conclusion, our results show that autophagy represents one of the mechanisms by which the activation of the MD-2/TLR4 ligand LPS promotes DC survival under hypoxic conditions.
Collapse
Affiliation(s)
- Sara Monaci
- Cellular and Molecular Physiology Unit, Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Carlo Aldinucci
- Cellular and Molecular Physiology Unit, Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Daniela Rossi
- Cellular and Molecular Physiology Unit, Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Gaia Giuntini
- Cellular and Molecular Physiology Unit, Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Irene Filippi
- Cellular and Molecular Physiology Unit, Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | | | - Giuseppe Marotta
- Cellular Therapy Unit and South-East Tuscany Blood Establishment, University Hospital, Siena, Italy
| | - Silvano Sozzani
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Fabio Carraro
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Antonella Naldini
- Cellular and Molecular Physiology Unit, Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy,*Correspondence: Antonella Naldini,
| |
Collapse
|
20
|
Korbecki J, Olbromski M, Dzięgiel P. CCL18 in the Progression of Cancer. Int J Mol Sci 2020; 21:ijms21217955. [PMID: 33114763 PMCID: PMC7663205 DOI: 10.3390/ijms21217955] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/21/2020] [Accepted: 10/24/2020] [Indexed: 02/07/2023] Open
Abstract
A neoplastic tumor consists of cancer cells that interact with each other and non-cancerous cells that support the development of the cancer. One such cell are tumor-associated macrophages (TAMs). These cells secrete many chemokines into the tumor microenvironment, including especially a large amount of CCL18. This chemokine is a marker of the M2 macrophage subset; this is the reason why an increase in the production of CCL18 is associated with the immunosuppressive nature of the tumor microenvironment and an important element of cancer immune evasion. Consequently, elevated levels of CCL18 in the serum and the tumor are connected with a worse prognosis for the patient. This paper shows the importance of CCL18 in neoplastic processes. It includes a description of the signal transduction from PITPNM3 in CCL18-dependent migration, invasion, and epithelial-to-mesenchymal transition (EMT) cancer cells. The importance of CCL18 in angiogenesis has also been described. The paper also describes the effect of CCL18 on the recruitment to the cancer niche and the functioning of cells such as TAMs, regulatory T cells (Treg), cancer-associated fibroblasts (CAFs) and tumor-associated dendritic cells (TADCs). The last part of the paper describes the possibility of using CCL18 as a therapeutic target during anti-cancer therapy.
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, Chałubińskiego 6a St, 50-368 Wrocław, Poland; (M.O.); (P.D.)
- Correspondence: ; Tel.: +48-717-841-354
| | - Mateusz Olbromski
- Department of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, Chałubińskiego 6a St, 50-368 Wrocław, Poland; (M.O.); (P.D.)
| | - Piotr Dzięgiel
- Department of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, Chałubińskiego 6a St, 50-368 Wrocław, Poland; (M.O.); (P.D.)
- Department of Physiotherapy, Wroclaw University School of Physical Education, Ignacego Jana Paderewskiego 35 Av., 51-612 Wroclaw, Poland
| |
Collapse
|
21
|
Korbecki J, Kojder K, Barczak K, Simińska D, Gutowska I, Chlubek D, Baranowska-Bosiacka I. Hypoxia Alters the Expression of CC Chemokines and CC Chemokine Receptors in a Tumor-A Literature Review. Int J Mol Sci 2020; 21:ijms21165647. [PMID: 32781743 PMCID: PMC7460668 DOI: 10.3390/ijms21165647] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/03/2020] [Accepted: 08/04/2020] [Indexed: 02/06/2023] Open
Abstract
Hypoxia, i.e., oxygen deficiency condition, is one of the most important factors promoting the growth of tumors. Since its effect on the chemokine system is crucial in understanding the changes in the recruitment of cells to a tumor niche, in this review we have gathered all the available data about the impact of hypoxia on β chemokines. In the introduction, we present the chronic (continuous, non-interrupted) and cycling (intermittent, transient) hypoxia together with the mechanisms of activation of hypoxia inducible factors (HIF-1 and HIF-2) and NF-κB. Then we describe the effect of hypoxia on the expression of chemokines with the CC motif: CCL1, CCL2, CCL3, CCL4, CCL5, CCL7, CCL8, CCL11, CCL13, CCL15, CCL16, CCL17, CCL18, CCL19, CCL20, CCL21, CCL22, CCL24, CCL25, CCL26, CCL27, CCL28 together with CC chemokine receptors: CCR1, CCR2, CCR3, CCR4, CCR5, CCR6, CCR7, CCR8, CCR9, and CCR10. To better understand the effect of hypoxia on neoplastic processes and changes in the expression of the described proteins, we summarize the available data in a table which shows the effect of individual chemokines on angiogenesis, lymphangiogenesis, and recruitment of eosinophils, myeloid-derived suppressor cells (MDSC), regulatory T cells (Treg), and tumor-associated macrophages (TAM) to a tumor niche.
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland; (J.K.); (D.S.); (D.C.)
| | - Klaudyna Kojder
- Department of Anaesthesiology and Intensive Care, Pomeranian Medical University in Szczecin, Unii Lubelskiej 1, 71-281 Szczecin, Poland;
| | - Katarzyna Barczak
- Department of Conservative Dentistry and Endodontics, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland;
| | - Donata Simińska
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland; (J.K.); (D.S.); (D.C.)
| | - Izabela Gutowska
- Department of Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland;
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland; (J.K.); (D.S.); (D.C.)
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland; (J.K.); (D.S.); (D.C.)
- Correspondence: ; Tel.: +48-914661515; Fax: +48-914661516
| |
Collapse
|
22
|
Safley SA, Graham ML, Weegman BP, Einstein SA, Barber GF, Janecek JJ, Mutch LA, Singh A, Ramachandran S, Garwood M, Sambanis A, Papas KK, Hering BJ, Weber CJ. Noninvasive Fluorine-19 Magnetic Resonance Relaxometry Measurement of the Partial Pressure of Oxygen in Acellular Perfluorochemical-loaded Alginate Microcapsules Implanted in the Peritoneal Cavity of Nonhuman Primates. Transplantation 2020; 104:259-269. [PMID: 31385927 PMCID: PMC6994361 DOI: 10.1097/tp.0000000000002896] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND We have utilized a noninvasive technique for measuring the partial pressure of oxygen (pO2) in alginate microcapsules implanted intraperitoneally in healthy nonhuman primates (NHPs). Average pO2 is important for determining if a transplant site and capsules with certain passive diffusion characteristics can support the islet viability, metabolic activity, and dose necessary to reverse diabetes. METHODS Perfluoro-15-crown-5-ether alginate capsules were infused intraperitoneally into 3 healthy NHPs. Peritoneal pO2 levels were measured on days 0 and 7 using fluorine-19 magnetic resonance relaxometry and a fiber-optic probe. Fluorine-19 MRI was used to determine the locations of capsules within the peritoneal space on days 0 and 7. Gross and histologic evaluations of the capsules were used to assess their biocompatibility postmortem. RESULTS At day 0 immediately after infusion of capsules equilibrated to room air, capsules were concentrated near the infusion site, and the pO2 measurement using magnetic resonance relaxometry was 147 ± 9 mm Hg. On day 7 after capsules were dispersed throughout the peritoneal cavity, the pO2 level was 61 ± 11 mm Hg. Measurements using the fiber-optic oxygen sensor were 132 ± 7.5 mm Hg (day 0) and 89 ± 6.1 mm Hg (day 7). Perfluoro-15-crown-5-ether capsules retrieved on day 7 were intact and free-floating without host cell attachment, although the numbers of peritoneal CD20 B cells, CD4 and CD8 T cells, and CD14 macrophages increased consistent with a mild foreign body reaction. CONCLUSIONS The peritoneal pO2 of normal NHPs is relatively low and we predict would decrease further when encapsulated islets are transplanted intraperitoneally.
Collapse
Affiliation(s)
| | - Melanie L. Graham
- Preclinical Research Center, Department of Surgery, University of Minnesota, St. Paul, MN
| | - Bradley P. Weegman
- Sylvatica Biotech, Inc., Charleston, SC
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN
| | - Samuel A. Einstein
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Jody J. Janecek
- Preclinical Research Center, Department of Surgery, University of Minnesota, St. Paul, MN
| | - Lucas A. Mutch
- Preclinical Research Center, Department of Surgery, University of Minnesota, St. Paul, MN
| | - Amar Singh
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN
| | | | - Michael Garwood
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN
| | | | | | - Bernhard J. Hering
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN
| | | |
Collapse
|
23
|
Zhang HW, Wang Q, Mei HX, Zheng SX, Ali AM, Wu QX, Ye Y, Xu HR, Xiang SY, Jin SW. RvD1 ameliorates LPS-induced acute lung injury via the suppression of neutrophil infiltration by reducing CXCL2 expression and release from resident alveolar macrophages. Int Immunopharmacol 2019; 76:105877. [DOI: 10.1016/j.intimp.2019.105877] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 08/26/2019] [Accepted: 09/03/2019] [Indexed: 02/08/2023]
|
24
|
Dai H, Thomson AW, Rogers NM. Dendritic Cells as Sensors, Mediators, and Regulators of Ischemic Injury. Front Immunol 2019; 10:2418. [PMID: 31681306 PMCID: PMC6803430 DOI: 10.3389/fimmu.2019.02418] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 09/27/2019] [Indexed: 12/18/2022] Open
Abstract
Dendritic cells (DCs) are highly specialized, bone marrow (BM)-derived antigen-processing and -presenting cells crucial to the induction, integration and regulation of innate, and adaptive immunity. They are stimulated by damage-associated molecular patterns (DAMPS) via pattern recognition receptors to promote inflammation and initiate immune responses. In addition to residing within the parenchyma of all organs as part of the heterogeneous mononuclear phagocyte system, DCs are an abundant component of the inflammatory cell infiltrate that appears in response to ischemia reperfusion injury (IRI). They can play disparate roles in the pathogenesis of IRI since their selective depletion has been found to be protective, deleterious, or of no benefit in mouse models of IRI. In addition, administration of DC generated and manipulated ex vivo can protect organs from IRI by suppressing inflammatory cytokine production, limiting the capacity of DCs to activate NKT cells, or enhancing regulatory T cell function. Few studies however have investigated specific signal transduction mechanisms underlying DC function and how these affect IRI. Here, we address current knowledge of the role of DCs in regulation of IRI, current gaps in understanding and prospects for innovative therapeutic intervention at the biological and pharmacological levels.
Collapse
Affiliation(s)
- Helong Dai
- Department of Urological Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, China
- Clinical Research Center for Organ Transplantation of Hunan Province, Changsha, China
| | - Angus W. Thomson
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Natasha M. Rogers
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Center for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, NSW, Australia
- Renal Division, Westmead Hospital, Westmead, NSW, Australia
- Westmead Clinical School, University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
25
|
Comparison of crevicular fluid cytokine levels after the application of surface sealants. J Orofac Orthop 2019; 80:242-253. [DOI: 10.1007/s00056-019-00184-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 06/05/2019] [Indexed: 10/26/2022]
|
26
|
Del Prete A, Scutera S, Sozzani S, Musso T. Role of osteopontin in dendritic cell shaping of immune responses. Cytokine Growth Factor Rev 2019; 50:19-28. [PMID: 31126876 DOI: 10.1016/j.cytogfr.2019.05.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 05/09/2019] [Indexed: 02/07/2023]
Abstract
Osteopontin (OPN) is a pleiotropic cytokine produced both by immune and non-immune cells and active on different cellular targets. OPN production has been associated with several pathological conditions, including autoimmune diseases (e.g. lupus, multiple sclerosis and rheumatoid arthritis) and cancer. Emerging evidence suggests that the role of OPN has been underestimated, as it seems to be working at multiple levels of immune regulation, such as the shaping of T cell effector responses, the regulation of the tumor microenvironment, and the functional interaction with mesenchymal stromal cells. In this context, dendritic cells (DCs) play a crucial role being both an important source and a cellular target for OPN action. DC family is composed by several cell subsets endowed with specific immune functions. OPN exerts its biological functions through multiple receptors and is produced in different intracellular and secreted forms. OPN production by DC subsets is emerging as a crucial mechanism of regulation in normal and pathological conditions and starts to be exploited as a therapeutic target. This review will focus on the role of DC-derived OPN in shaping immune response and on the complex role of this cytokines in the regulation in immune response.
Collapse
Affiliation(s)
- Annalisa Del Prete
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy; Humanitas Clinical and Research Center-IRCCS Rozzano-Milano, Italy
| | - Sara Scutera
- Microbiology section, Department of Public Health and Pediatric Sciences, University of Torino, Turin, Italy
| | - Silvano Sozzani
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| | - Tiziana Musso
- Microbiology section, Department of Public Health and Pediatric Sciences, University of Torino, Turin, Italy
| |
Collapse
|
27
|
Paardekooper LM, Vos W, van den Bogaart G. Oxygen in the tumor microenvironment: effects on dendritic cell function. Oncotarget 2019; 10:883-896. [PMID: 30783517 PMCID: PMC6368231 DOI: 10.18632/oncotarget.26608] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 01/09/2019] [Indexed: 12/13/2022] Open
Abstract
Solid tumors grow at a high speed leading to insufficient blood supply to tumor cells. This makes the tumor hypoxic, resulting in the Warburg effect and an increased generation of reactive oxygen species (ROS). Hypoxia and ROS affect immune cells in the tumor micro-environment, thereby affecting their immune function. Here, we review the known effects of hypoxia and ROS on the function and physiology of dendritic cells (DCs). DCs can (cross-)present tumor antigen to activate naive T cells, which play a pivotal role in anti-tumor immunity. ROS might enter DCs via aquaporins in the plasma membrane, diffusion across the plasma membrane or via extracellular vesicles (EVs) released by tumor cells. Hypoxia and ROS exert complex effects on DCs, and can both inhibit and activate maturation of immature DCs. Furthermore, ROS transferred by EVs and/or produced by the DC can both promote antigen (cross-)presentation through phagosomal alkalinization, which preserves antigens by inhibiting proteases, and by direct oxidative modification of proteases. Hypoxia leads to a more migratory and inflammatory DC phenotype. Lastly, hypoxia alters DCs to shift the T- cell response towards a tumor suppressive Th17 phenotype. From numerous studies, the concept is emerging that hypoxia and ROS are mutually dependent effectors on DC function in the tumor micro-environment. Understanding their precise roles and interplay is important given that an adaptive immune response is required to clear tumor cells.
Collapse
Affiliation(s)
- Laurent M Paardekooper
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Willemijn Vos
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Geert van den Bogaart
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
28
|
Subat S, Mogushi K, Yasen M, Kohda T, Ishikawa Y, Tanaka H. Identification of genes and pathways, including the CXCL2 axis, altered by DNA methylation in hepatocellular carcinoma. J Cancer Res Clin Oncol 2018; 145:675-684. [PMID: 30564899 DOI: 10.1007/s00432-018-2824-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 12/12/2018] [Indexed: 12/12/2022]
Abstract
PURPOSE Recent genetic studies have suggested that tumor suppressor genes are often silenced during carcinogenesis via epigenetic modification caused by methylation of promoter CpG islands. Here, we characterized genes inactivated by DNA methylation in human hepatocellular carcinoma (HCC) to identify the genes and pathways involved in DNA methylation in hepatocellular carcinoma. METHODS Eight HCC-derived cell lines were treated with a DNA demethylating agent, 5-aza-2'-deoxycytidine. Additionally, 100 pairs of primary HCC and adjacent non-cancerous tissues as well as 15 normal liver tissues were analyzed by comprehensive gene expression analysis using microarrays. Moreover, gene set enrichment analysis identified the major molecular pathways associated with DNA methylation. Validation of gene expression and DNA methylation status was performed by real-time PCR after bisulfite modification. RESULTS We showed that CXCL2, an immune-related chemokine, expression was significantly downregulated in tumor tissues and also significantly upregulated by DAC treatment in cell lines. Furthermore, we observed a statistically significant difference in methylation status between normal liver tissues and tumor tissues (P < 0.05). In addition, tumors with higher CXCL2 expression included significantly more numbers of multiple tumors than the lower expression group. CONCLUSIONS We identified CXCL2, an immune-related chemokine, decreased in hepatocellular carcinoma and the regulation mechanism may be controlled by methylation. Further studies should be warranted to examine if and to what extent the gene is actually suppressed by methylation and if there is a possibility that the CXCL2 axis plays a role for diagnosis and treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Sophia Subat
- Department of Systems Biology, Graduate School of Biomedical Science, Tokyo Medical and Dental University, Tokyo, Japan
- Department of Epigenetics, Graduate School of Biomedical Science, Tokyo Medical and Dental University, Tokyo, Japan
- Division of Pathology, The Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ward, Tokyo, 135-8550, Japan
| | - Kaoru Mogushi
- Department of Systems Biology, Graduate School of Biomedical Science, Tokyo Medical and Dental University, Tokyo, Japan
- Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Mahmut Yasen
- Department of Systems Biology, Graduate School of Biomedical Science, Tokyo Medical and Dental University, Tokyo, Japan
- Division of Pathology, The Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ward, Tokyo, 135-8550, Japan
- Department of Surgery, Xinjiang Medical University Affiliated Tumor Hospital, Urumqi, Xinjiang Uyghur Autonomous Region, China
| | - Takashi Kohda
- Department of Epigenetics, Graduate School of Biomedical Science, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yuichi Ishikawa
- Division of Pathology, The Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ward, Tokyo, 135-8550, Japan.
| | - Hiroshi Tanaka
- Department of Systems Biology, Graduate School of Biomedical Science, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
29
|
Alshahrani I. Biomolecular phases in transverse palatal distraction: A review. Saudi J Biol Sci 2018; 25:1322-1325. [PMID: 30505176 PMCID: PMC6252022 DOI: 10.1016/j.sjbs.2018.05.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 05/05/2018] [Accepted: 05/06/2018] [Indexed: 11/17/2022] Open
Abstract
Transverse palatal distraction is a biological process of regenerating new bone and enveloping soft tissues in the maxillary palate region. This technique is similar to Osteo-distraction (OD) procedure for bone lengthening in which gradual and controlled traction forces are applied on the osteotomy gaps to produce new bone in between the surgically separated bone segments. This review describes the different phases after osteotomy and the biological process involved during the new bone and soft tissue formation. The mechanical environment formed in the distraction area is due to the traction forces by the distractor appliance. This environment stimulates differentiation of pluripotent cells, neovascularization, osteogenesis and remodeling of newly formed bone. The role of different pro-inflammatory cytokines, interleukins, bone morphogenic proteins, transforming growth factors, fibroblast growth factors-2) and extracellular matrix proteins (osteonectin, osteopontin) during the distraction phases has been described in detail. Also, an important note on the nutritional aspect during Osteo-distraction will benefit the clinicians to guide their patients after osteotomy throughout the distraction process.
Collapse
|
30
|
Parodi M, Raggi F, Cangelosi D, Manzini C, Balsamo M, Blengio F, Eva A, Varesio L, Pietra G, Moretta L, Mingari MC, Vitale M, Bosco MC. Hypoxia Modifies the Transcriptome of Human NK Cells, Modulates Their Immunoregulatory Profile, and Influences NK Cell Subset Migration. Front Immunol 2018; 9:2358. [PMID: 30459756 PMCID: PMC6232835 DOI: 10.3389/fimmu.2018.02358] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 09/24/2018] [Indexed: 12/29/2022] Open
Abstract
Hypoxia, which characterizes most tumor tissues, can alter the function of different immune cell types, favoring tumor escape mechanisms. In this study, we show that hypoxia profoundly acts on NK cells by influencing their transcriptome, affecting their immunoregulatory functions, and changing the chemotactic responses of different NK cell subsets. Exposure of human peripheral blood NK cells to hypoxia for 16 or 96 h caused significant changes in the expression of 729 or 1,100 genes, respectively. Gene Set Enrichment Analysis demonstrated that these changes followed a consensus hypoxia transcriptional profile. As assessed by Gene Ontology annotation, hypoxia-targeted genes were implicated in several biological processes: metabolism, cell cycle, differentiation, apoptosis, cell stress, and cytoskeleton organization. The hypoxic transcriptome also showed changes in genes with immunological relevance including those coding for proinflammatory cytokines, chemokines, and chemokine-receptors. Quantitative RT-PCR analysis confirmed the modulation of several immune-related genes, prompting further immunophenotypic and functional studies. Multiplex ELISA demonstrated that hypoxia could variably reduce NK cell ability to release IFNγ, TNFα, GM-CSF, CCL3, and CCL5 following PMA+Ionomycin or IL15+IL18 stimulation, while it poorly affected the response to IL12+IL18. Cytofluorimetric analysis showed that hypoxia could influence NK chemokine receptor pattern by sustaining the expression of CCR7 and CXCR4. Remarkably, this effect occurred selectively (CCR7) or preferentially (CXCR4) on CD56bright NK cells, which indeed showed higher chemotaxis to CCL19, CCL21, or CXCL12. Collectively, our data suggest that the hypoxic environment may profoundly influence the nature of the NK cell infiltrate and its effects on immune-mediated responses within tumor tissues.
Collapse
Affiliation(s)
- Monica Parodi
- UOC Immunologia, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Federica Raggi
- Laboratorio di Biologia Molecolare, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Davide Cangelosi
- Laboratorio di Biologia Molecolare, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Claudia Manzini
- Laboratorio di Immunologia Clinica e Sperimentale, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Mirna Balsamo
- Dipartimento di Medicina Sperimentale, Università di Genova, Genova, Italy
| | - Fabiola Blengio
- Laboratorio di Biologia Molecolare, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Alessandra Eva
- Laboratorio di Biologia Molecolare, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Luigi Varesio
- Laboratorio di Biologia Molecolare, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Gabriella Pietra
- UOC Immunologia, IRCCS Ospedale Policlinico San Martino, Genova, Italy.,Dipartimento di Medicina Sperimentale, Università di Genova, Genova, Italy
| | - Lorenzo Moretta
- Immunology Area, Ospedale Pediatrico Bambin Gesù, Rome, Italy
| | - Maria Cristina Mingari
- UOC Immunologia, IRCCS Ospedale Policlinico San Martino, Genova, Italy.,Dipartimento di Medicina Sperimentale, Università di Genova, Genova, Italy.,Center of Excellence for Biomedical Research, University of Genoa, Genova, Italy
| | - Massimo Vitale
- UOC Immunologia, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Maria Carla Bosco
- Laboratorio di Biologia Molecolare, IRCCS Istituto Giannina Gaslini, Genova, Italy
| |
Collapse
|
31
|
Cellular Stress Responses and Gut Microbiota in Inflammatory Bowel Disease. Gastroenterol Res Pract 2018; 2018:7192646. [PMID: 30026758 PMCID: PMC6031203 DOI: 10.1155/2018/7192646] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 05/08/2018] [Indexed: 12/11/2022] Open
Abstract
Progresses in the past two decades have greatly expanded our understanding of inflammatory bowel disease (IBD), an incurable disease with multifaceted and challenging clinical manifestations. The pathogenesis of IBD involves multiple processes on the cellular level, which include the stress response signaling such as endoplasmic reticulum (ER) stress, oxidative stress, and hypoxia. Under physiological conditions, the stress responses play key roles in cell survival, mucosal barrier integrity, and immunomodulation. However, they can also cause energy depletion, trigger cell death and tissue injury, promote inflammatory response, and drive the progression of clinical disease. In recent years, gut microflora has emerged as an essential pathogenic factor and therapeutic target for IBD. Altered compositional and metabolic profiles of gut microbiota, termed dysbiosis, are associated with IBD. Recent studies, although limited, have shed light on how ER stress, oxidative stress, and hypoxic stress interact with gut microorganisms, a potential source of stress in the microenvironment of gastrointestinal tract. Our knowledge of cellular stress responses in intestinal homeostasis as well as their cross-talks with gut microbiome will further our understanding of the pathogenesis of inflammatory bowel disease and probably open avenues for new therapies.
Collapse
|
32
|
Van Welden S, Selfridge AC, Hindryckx P. Intestinal hypoxia and hypoxia-induced signalling as therapeutic targets for IBD. Nat Rev Gastroenterol Hepatol 2017; 14:596-611. [PMID: 28853446 DOI: 10.1038/nrgastro.2017.101] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Tissue hypoxia occurs when local oxygen demand exceeds oxygen supply. In chronic inflammatory conditions such as IBD, the increased oxygen demand by resident and gut-infiltrating immune cells coupled with vascular dysfunction brings about a marked reduction in mucosal oxygen concentrations. To counter the hypoxic challenge and ensure their survival, mucosal cells induce adaptive responses, including the activation of hypoxia-inducible factors (HIFs) and modulation of nuclear factor-κB (NF-κB). Both pathways are tightly regulated by oxygen-sensitive prolyl hydroxylases (PHDs), which therefore represent promising therapeutic targets for IBD. In this Review, we discuss the involvement of mucosal hypoxia and hypoxia-induced signalling in the pathogenesis of IBD and elaborate in detail on the role of HIFs, NF-κB and PHDs in different cell types during intestinal inflammation. We also provide an update on the development of PHD inhibitors and discuss their therapeutic potential in IBD.
Collapse
Affiliation(s)
- Sophie Van Welden
- Department of Gastroenterology, Ghent University, De Pintelaan 185, 1K12-E, 9000 Ghent, Belgium
| | - Andrew C Selfridge
- Robarts Clinical Trials West, 4350 Executive Drive 210, San Diego, California 92121, USA
| | - Pieter Hindryckx
- Department of Gastroenterology, Ghent University, De Pintelaan 185, 1K12-E, 9000 Ghent, Belgium
| |
Collapse
|
33
|
Pyfferoen L, Brabants E, Everaert C, De Cabooter N, Heyns K, Deswarte K, Vanheerswynghels M, De Prijck S, Waegemans G, Dullaers M, Hammad H, De Wever O, Mestdagh P, Vandesompele J, Lambrecht BN, Vermaelen KY. The transcriptome of lung tumor-infiltrating dendritic cells reveals a tumor-supporting phenotype and a microRNA signature with negative impact on clinical outcome. Oncoimmunology 2016; 6:e1253655. [PMID: 28197369 PMCID: PMC5283643 DOI: 10.1080/2162402x.2016.1253655] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 09/23/2016] [Accepted: 10/24/2016] [Indexed: 01/06/2023] Open
Abstract
Targeting immunomodulatory pathways has ushered a new era in lung cancer therapy. Further progress requires deeper insights into the biology of immune cells in the lung cancer micro-environment. Dendritic cells (DCs) represent a heterogeneous and highly plastic immune cell system with a central role in controlling immune responses. The intratumoral infiltration and activation status of DCs are emerging as clinically relevant parameters in lung cancer. In this study, we used an orthotopic preclinical model of lung cancer to dissect how the lung tumor micro-environment affects tissue-resident DCs and extract novel biologically and clinically relevant information. Lung tumor-infiltrating leukocytes expressing generic DC markers were found to predominantly consist of CD11b+ cells that, compare with peritumoral lung DC counterparts, strongly overexpress the T-cell inhibitory molecule PD-L1 and acquire classical surface markers of tumor-associated macrophages (TAMs). Transcriptome analysis of these CD11b+ tumor-infiltrating DCs (TIDCs) indicates impaired antitumoral immunogenicity, confirms the skewing toward TAM-related features, and indicates exposure to a hypoxic environment. In parallel, TIDCs display a specific microRNA (miRNA) signature dominated by the prototypical lung cancer oncomir miR-31. In vitro, hypoxia drives intrinsic miR-31 expression in CD11b+ DCs. Conditioned medium of miR-31 overexpressing CD11b+ DCs induces pro-invasive lung cancer cell shape changes and is enriched with pro-metastatic soluble factors. Finally, analysis of TCGA datasets reveals that the TIDC-associated miRNA signature has a negative prognostic impact in non-small cell lung cancer. Together, these data suggest a novel mechanism through which the lung cancer micro-environment exploits the plasticity of the DC system to support tumoral progression.
Collapse
Affiliation(s)
- Lotte Pyfferoen
- Tumor Immunology Laboratory, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium; VIB Inflammation Research Center, Ghent, Belgium
| | - Elisabeth Brabants
- Tumor Immunology Laboratory, Department of Respiratory Medicine, Ghent University Hospital , Ghent, Belgium
| | - Celine Everaert
- Center for Medical Genetics, Ghent University , Ghent, Belgium
| | - Nancy De Cabooter
- Tumor Immunology Laboratory, Department of Respiratory Medicine, Ghent University Hospital , Ghent, Belgium
| | - Kelly Heyns
- Tumor Immunology Laboratory, Department of Respiratory Medicine, Ghent University Hospital , Ghent, Belgium
| | - Kim Deswarte
- VIB Inflammation Research Center , Ghent, Belgium
| | | | | | - Glenn Waegemans
- Laboratory of Experimental Cancer Research, Ghent University , Ghent, Belgium
| | - Melissa Dullaers
- Tumor Immunology Laboratory, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium; VIB Inflammation Research Center, Ghent, Belgium
| | | | - Olivier De Wever
- Laboratory of Experimental Cancer Research, Ghent University , Ghent, Belgium
| | - Pieter Mestdagh
- Center for Medical Genetics, Ghent University , Ghent, Belgium
| | - Jo Vandesompele
- Center for Medical Genetics, Ghent University , Ghent, Belgium
| | - Bart N Lambrecht
- VIB Inflammation Research Center, Ghent, Belgium; Department of Pulmonary Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Karim Y Vermaelen
- Tumor Immunology Laboratory, Department of Respiratory Medicine, Ghent University Hospital , Ghent, Belgium
| |
Collapse
|
34
|
Huang G, Tao L, Shen S, Chen L. Hypoxia induced CCL28 promotes angiogenesis in lung adenocarcinoma by targeting CCR3 on endothelial cells. Sci Rep 2016; 6:27152. [PMID: 27250766 PMCID: PMC4890017 DOI: 10.1038/srep27152] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 05/16/2016] [Indexed: 02/06/2023] Open
Abstract
Tumor hypoxia is one of the important features of lung adenocarcinoma. Chemokines might mediate the effects caused by tumor hypoxia. As confirmed in tumor tissue and serum of patients, CC chemokine 28 (CCL28) was the only hypoxia induced chemokine in lung adenocarcinoma cells. CCL28 could promote tube formation, migration and proliferation of endothelial cells. In addition, angiogenesis was promoted by CCL28 in the chick chorioallantoic membrane and matrigel implanted in dorsal back of athymic nude mice (CByJ.Cg-Foxn1nu/J). Tumors formed by lung adenocarcinoma cells with high expression of CCL28 grew faster and had a higher vascular density, whereas tumor formation rate of lung adenocarcinoma cells with CCL28 expression knockdown was quite low and had a lower vascular density. CCR3, receptor of CCL28, was highly expressed in vascular endothelial cells in lung adenocarcinoma when examining by immunohistochemistry. Further signaling pathways in endothelial cells, modulated by CCL28, were analyzed by Phosphorylation Antibody Array. CCL28/CCR3 signaling pathway could bypass that of VEGF/VEGFR on the levels of PI3K-Akt, p38 MAPK and PLC gamma. The effects could be neutralized by antibody against CCR3. In conclusion, CCL28, as a chemokine induced by tumor hypoxia, could promote angiogenesis in lung adenocarcinoma through targeting CCR3 on microvascular endothelial cells.
Collapse
Affiliation(s)
- Guichun Huang
- Medical Oncology Department of Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Leilei Tao
- Medical Oncology Department of Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Sunan Shen
- Medical School of Nanjing University, Nanjing, China
| | - Longbang Chen
- Medical Oncology Department of Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
35
|
Hypoxia, mitochondrial dysfunction and synovial invasiveness in rheumatoid arthritis. Nat Rev Rheumatol 2016; 12:385-97. [DOI: 10.1038/nrrheum.2016.69] [Citation(s) in RCA: 173] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
36
|
Dendritic Cells under Hypoxia: How Oxygen Shortage Affects the Linkage between Innate and Adaptive Immunity. J Immunol Res 2016; 2016:5134329. [PMID: 26966693 PMCID: PMC4757696 DOI: 10.1155/2016/5134329] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 01/07/2016] [Accepted: 01/13/2016] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells (DCs) are considered as one of the main regulators of immune responses. They collect antigens, process them, and present typical antigenic structures to lymphocytes, thereby inducing an adaptive immune response. All these processes take place under conditions of oxygen shortage (hypoxia) which is often not considered in experimental settings. This review highlights how deeply hypoxia modulates human as well as mouse immature and mature dendritic cell functions. It tries to link in vitro results to actual in vivo studies and outlines how hypoxia-mediated shaping of dendritic cells affects the activation of (innate) immunity.
Collapse
|
37
|
Yilmaz A, Ratka J, Rohm I, Pistulli R, Goebel B, Asadi Y, Petri A, Kiehntopf M, Figulla HR, Jung C. Decrease in circulating plasmacytoid dendritic cells during short-term systemic normobaric hypoxia. Eur J Clin Invest 2016; 46:115-22. [PMID: 25652640 DOI: 10.1111/eci.12416] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 01/31/2015] [Indexed: 12/23/2022]
Abstract
BACKGROUND During exposure to high altitude, the immune system is altered. During hypoxia, an increase in interleukin (IL)-6 and high sensitivity C-reactive protein (hs-CRP), and an increase in natural killer cells and decrease in T cells in blood was shown. However, the impact of hypoxia on dendritic cells has not been investigated yet. MATERIAL AND METHODS Twelve healthy volunteers were subjected to a transient normobaric hypoxia for 6·5 h simulating an oxygen concentration at 5500 m. During exposure to hypoxia, blood samples were collected and analysed by flow cytometrical cell sorting (FACS) for circulating myeloid (mDCs) and plasmacytoid (pDCs) DCs. Serum levels of IL-6 and tumour necrosis factor (TNF)-α were analysed. In a cell culture hypoxia chamber, blood samples were subjected to the same hypoxia and analysed regarding DCs. RESULTS Exposure to normobaric hypoxia induced a significant decrease in circulating pDCs about 45% (P = 0·001) but not of mDC compared to baseline normoxia. Furthermore, we observed a significant increase of TNF-α about 340% (P = 0·03) and of IL-6 about 286% (P = 0·002). In cell culture experiments exposure of blood to hypoxia led to no significant changes in DCs, so that a direct cytotoxic effect was excluded. During hypoxia, we observed a transient increase in stromal-derived factor 1 (SDF-1) which is important for pDC tissue recruitment. CONCLUSIONS We show a significant decrease in circulating pDCs during hypoxia in parallel to a pro-inflammatory response. Further studies are necessary to evaluate whether the decrease in circulating pDCs might be the result of an enhanced tissue recruitment.
Collapse
Affiliation(s)
- Atilla Yilmaz
- Internal Medicine II, Elisabeth Hospital Schmalkalden, Schmalkalden, Germany
| | - Josi Ratka
- Clinic for Internal Medicine I, University Hospital of Jena, Jena, Germany
| | - Ilonka Rohm
- Clinic for Internal Medicine I, University Hospital of Jena, Jena, Germany
| | - Rudin Pistulli
- Clinic for Internal Medicine I, University Hospital of Jena, Jena, Germany
| | - Bjorn Goebel
- Clinic for Internal Medicine I, University Hospital of Jena, Jena, Germany
| | - Yahya Asadi
- Clinic for Internal Medicine I, University Hospital of Jena, Jena, Germany
| | - Alexander Petri
- Clinic for Internal Medicine I, University Hospital of Jena, Jena, Germany
| | - Michael Kiehntopf
- Department of Clinical Chemistry & Laboratory Medicine, University Hospital of Jena, Jena, Germany
| | - Hans R Figulla
- Clinic for Internal Medicine I, University Hospital of Jena, Jena, Germany
| | - Christian Jung
- Clinic for Internal Medicine I, University Hospital of Jena, Jena, Germany
| |
Collapse
|
38
|
Chen D, Wang Y, Wang H, Wu Y, Xia S, Zhang M. CD8(+) T activation attenuates CD4(+) T proliferation through dendritic cells modification. Cell Immunol 2015; 296:138-48. [PMID: 26022412 DOI: 10.1016/j.cellimm.2015.05.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 05/09/2015] [Accepted: 05/09/2015] [Indexed: 11/28/2022]
Abstract
Emerging evidence has suggested that CD8(+) T had modulatory function on CD4(+) T mediated autoimmune and inflammatory diseases. However, the underlying mechanisms remain unclear. In this study, we found that CD8(+) T activation inhibited OVA(323-339) antigen specific CD4(+) T cells proliferation in vitro and in vivo. Further investigation demonstrated that this immunosuppression largely depended on the soluble factor from activated CD8(+) T to modify the phenotype and functions of DCs. Moreover, not only the inhibitors for IDO or iNOS, but also IFN-γ neutralization markedly reversed this immunosuppression on OVA(323-339) antigen specific CD4(+) T cells proliferation. Interestingly, CD8(+) T cells absence aggravated the pathological damage in lung in OVA-induced asthma model, but alleviated by CD8(+) T transfer and activation. Thus, these findings suggested that activated CD8(+) T population exerted feedback regulation in DCs modification, and then attenuated CD4(+) T mediated immune response.
Collapse
Affiliation(s)
- Dongwei Chen
- Institute of Immunology, Medical Center, Tsinghua University, Beijing, People's Republic of China
| | - Ying Wang
- Institute of Immunology, Medical Center, Tsinghua University, Beijing, People's Republic of China
| | - Huan Wang
- Institute of Immunology, Medical Center, Tsinghua University, Beijing, People's Republic of China
| | - Yiqing Wu
- Institute of Immunology, Medical Center, Tsinghua University, Beijing, People's Republic of China
| | - Sheng Xia
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China.
| | - Minghui Zhang
- Institute of Immunology, Medical Center, Tsinghua University, Beijing, People's Republic of China.
| |
Collapse
|
39
|
Miragliotta V, Pirone A, Donadio E, Abramo F, Ricciardi MP, Theoret CL. Osteopontin expression in healing wounds of horses and in human keloids. Equine Vet J 2014; 48:72-7. [PMID: 25290989 DOI: 10.1111/evj.12372] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 09/30/2014] [Indexed: 01/20/2023]
Abstract
REASONS FOR PERFORMING STUDY Convincing evidence shows that persistent or excessive expression of osteopontin (OPN) is linked to fibroproliferation of various organs in laboratory animals and in man, such that its downregulation is a logical therapeutic objective. OBJECTIVES To investigate OPN expression in an equine model of wound healing and in clinical specimens of equine exuberant granulation tissue and human keloids in an effort to better understand the contribution of this protein to inflammation-associated skin fibrosis. STUDY DESIGN Description of gene and protein expression in an experimental equine model of wound healing and clinical specimens in horse and man. METHODS Osteopontin gene expression was evaluated by quantitative PCR, while protein expression was investigated by means of immunohistochemical staining. RESULTS Quantitative PCR showed that the OPN gene is expressed in normal intact skin of horses and continues to be expressed during the wound-healing process. An increase in gene expression was observed throughout the phases of wound healing, with a final decrease at wound closure. The protein was not detected in normal skin. Keratinocytes in wound-edge samples did not express the protein, whereas dermal immunoreactivity was confined to inflammatory cells. Healed wounds were devoid of staining. Equine exuberant granulation tissue showed immunoreactivity of the surrounding epidermis, infiltrating neutrophils, mononuclear cells, endothelial cells and fibroblasts. Human keloids showed OPN immunoreactivity throughout the epidermis as well as in mononuclear cells and scattered fibroblasts. CONCLUSIONS Immunohistochemical data show a different pattern of expression between normally healing and fibrotic wounds (exuberant granulation tissue and keloids), thus suggesting a role in fibroproliferation in horses and man.
Collapse
Affiliation(s)
- V Miragliotta
- Department of Veterinary Sciences, University of Pisa, Pisa, Italy
| | - A Pirone
- Department of Veterinary Sciences, University of Pisa, Pisa, Italy
| | - E Donadio
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | - F Abramo
- Department of Veterinary Sciences, University of Pisa, Pisa, Italy
| | - M P Ricciardi
- Department of Veterinary Sciences, University of Pisa, Pisa, Italy
| | - C L Theoret
- Department of Veterinary Biomedicine, University of Montreal, Quebec, Canada
| |
Collapse
|
40
|
Filippi I, Morena E, Aldinucci C, Carraro F, Sozzani S, Naldini A. Short-term hypoxia enhances the migratory capability of dendritic cell through HIF-1α and PI3K/Akt pathway. J Cell Physiol 2014; 229:2067-76. [PMID: 24818793 DOI: 10.1002/jcp.24666] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 05/05/2014] [Accepted: 05/06/2014] [Indexed: 01/01/2023]
Abstract
Hypoxia represents an inadequate oxygen supply to tissues, which can modulate cell functions, primarily through the hypoxia-inducible transcription factor HIF-1α. Dendritic cells (DC) are professional antigen-presenting cells and their migration maybe affected by hypoxia, since the local microenvironment in lymphoid organs, as well as in inflamed and tumor tissues, is characterized by low oxygen tensions. In this study we observed an enhanced migratory capability of human monocyte-derived DC, using in vitro migration assays performed under hypoxic conditions. Such enhancement was independent on either the chemoattractants involved or the maturation level of DC. However, HIF-1α appeared to be crucial for the migration only of immature DC and not for mature DC under hypoxia, as indicated by HIF-1α siRNA approaches. Furthermore, we observed that while Akt phosphorylation was enhanced in both immature and mature DC exposed to hypoxia, other signaling pathways, such as p38 and p42/p44 MAPK, were differently affected during hypoxic treatment. More interestingly, aspecific and specific inhibition of PI3K/Akt indicated that such pathway was relevant for the migration of both immature and matured DC under hypoxia, even when DC were transfected with HIF-1α siRNA. Our results indicate that, besides HIF-1α, several other pathways, including PI3K/Akt, may be involved in the response to hypoxia of immature and, more specifically, of mature DC to sustain their trafficking and functions within hypoxic microenvironments.
Collapse
Affiliation(s)
- Irene Filippi
- Unit of Cellular and Molecular Physiology, Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | | | | | | | | | | |
Collapse
|
41
|
Souto GR, Queiroz CM, Costa FO, Mesquita RA. Relationship between chemokines and dendritic cells in human chronic periodontitis. J Periodontol 2014; 85:1416-23. [PMID: 24605873 DOI: 10.1902/jop.2014.130662] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND The purpose of this study was to evaluate the relationship between chemokines and dendritic cells (DCs) in human chronic periodontitis (CP). METHODS Gingival samples were obtained from 23 individuals with CP, and six samples of normal mucosa (NM) overlapping the third molar were used to control for the chemokine levels. Periodontal examination was conducted. Immunohistochemistry was performed for Factor XIIIa(+) and cluster of differentiation (CD)1a(+) immature DCs and CD83(+) mature DCs. Levels of the CC chemokine ligand (CCL)2, CCL3, CCL5, CCL19, CCL20, and CXC chemokine ligand (CXCL)8 were measured in gingival tissues using enzyme-linked immunosorbent assay. Inflammatory infiltrate, DCs, chemokines, classification of human CP, and clinical parameters were correlated and compared. RESULTS The expression of CCL2 and CCL20 was positively correlated with increased densities of CD1a(+) DCs. CCL3 and CXCL8 were positively related to the clinical attachment level. CCL3, CCL5, CCL19, and CXCL8 levels increased in the gingival samples of patients with CP compared with NM, whereas CCL20 levels increased in advanced CP compared with mild-moderate CP. CONCLUSIONS More CD1a(+) immature DCs are related to CCL2 and CCL20. CCL3 and CXCL8 chemokines are related to a greater severity of human CP.
Collapse
Affiliation(s)
- Giovanna Ribeiro Souto
- Department of Oral Surgery and Pathology, School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | | | | |
Collapse
|
42
|
Bhandari T, Nizet V. Hypoxia-Inducible Factor (HIF) as a Pharmacological Target for Prevention and Treatment of Infectious Diseases. Infect Dis Ther 2014; 3:159-74. [PMID: 25134687 PMCID: PMC4269623 DOI: 10.1007/s40121-014-0030-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Indexed: 02/07/2023] Open
Abstract
In the present era of ever-increasing antibiotic resistance and increasingly complex and immunosuppressed patient populations, physicians and scientists are seeking novel approaches to battle difficult infectious disease conditions. Development of a serious infection implies a failure of innate immune capabilities in the patient, and one may consider whether pharmacological strategies exist to correct and enhance innate immune cell function. Hypoxia-inducible factor-1 (HIF-1), the central regulator of the cellular response to hypoxic stress, has recently been recognized to control the activation state and key microbicidal functions of immune cells. HIF-1 boosting drugs are in clinical development for anemia and other indications, and could be repositioned as infectious disease therapeutics. With equal attention to opportunities and complexities, we review our current understanding of HIF-1 regulation of microbial host-pathogen interactions with an eye toward future drug development.
Collapse
Affiliation(s)
- Tamara Bhandari
- Center for Immunity, Infection and Inflammation, Department of Pediatrics and Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, USA
| | - Victor Nizet
- Center for Immunity, Infection and Inflammation, Department of Pediatrics and Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, USA.
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, USA.
- Center for Immunity, Infection and Inflammation, Medical Sciences Research 4113, University of California, San Diego, 9500 Gilman Drive, MC 0760, La Jolla, CA, 92093-0760, USA.
| |
Collapse
|
43
|
Lourenço AG, Komesu MC, Machado AA, Quintana SM, Bourlet T, Pozzetto B, Delézay O. Semen lactoferrin promotes CCL20 production by epithelial cells: Involvement in HIV transmission. World J Virol 2014; 3:11-17. [PMID: 25019057 PMCID: PMC4087152 DOI: 10.5501/wjv.v3.i2.11] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 04/15/2014] [Accepted: 05/29/2014] [Indexed: 02/05/2023] Open
Abstract
AIM: To study the effect of seminal plasma on Chemokine (C-C motif) ligand 20 (CCL20) production by epithelial cells and its relationship with lactoferrin.
METHODS: HEC-1A cells, a cell line derived from a monostratified endocervical epithelium, were incubated with samples of seminal plasma (diluted 1:10 in culture medium) recovered from human immunodeficiency virus (HIV) seronegative (HIV-) or HIV seropositive (HIV+) subjects. Recombinant human interleukin 1 beta (IL-1β) was used as positive control, and culture medium only as negative control. The measurement of CCL20 production in the supernatants of HEC-1A cells and of lactoferrin in seminal plasma was determined by enzyme-linked immunosorbent assay techniques. A fractionation of seminal plasma proteins was performed by ion exchange chromatography on a pool of seminal plasma specimens from HIV- subjects. Each fraction was tested for its ability to stimulate the production of CCL20 by HEC-1A cells and for its lactoferrin concentration. The HIV viral load in seminal plasma samples from HIV+ patients was measured using the HIV-Monitor kit (Roche Diagnostic Systems, Branchburg, NJ, United States).
RESULTS: The positive control IL-1β was responsible for an increase of 11.36 ± 3.36 times in the production of CCL20. Stimulation of HEC-1A cells was performed in 34 seminal plasma samples (22 from HIV+ subjects and 12 from HIV- subjects). The mean production of CCL20 by HEC-1A in presence of seminal plasma from HIV- and HIV+ subjects was respectively 5.38 ± 0.91 and 7.57 ± 3.26 times higher than that obtained with the untreated cells (P < 0.05 between the two groups). Using the same 34 specimens of seminal plasma, no correlation was observed between the concentration of total proteins in seminal plasma and their ability to stimulate the secretion of CCL20 by HEC-1 cells. In contrast, the ability to produce CCL20 by HEC-1A cells correlated to the concentration of lactoferrin in the seminal plasma samples (r coefficient = 0.56; CI: 0.26-0.76; P < 0.001). After fractionation by ion exchange chromatography, the seminal plasma fractions exhibiting the highest concentrations of lactoferrin were responsible for the greatest stimulation of CCL20 production by HEC-1A cells (r coefficient = 0.89; CI: 0.78-0.95; P < 0.0001).
CONCLUSION: Lactoferrin present in seminal plasma correlated with an increased production of CCL20 by HEC-1A cells and therefore could facilitate HIV entry through the genital mucosa.
Collapse
|
44
|
Tan CW, Lee YH, Tan HH, Lau MSK, Choolani M, Griffith L, Chan JKY. CD26/DPPIV down-regulation in endometrial stromal cell migration in endometriosis. Fertil Steril 2014; 102:167-177.e9. [PMID: 24825423 DOI: 10.1016/j.fertnstert.2014.04.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 04/01/2014] [Accepted: 04/03/2014] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To test the hypothesis that endometrial stromal cells (ESCs) in endometriosis exhibit increased cell motility under hypoxia. DESIGN Prospective case-control study. SETTING University research laboratory. PATIENT(S) Women with endometriosis (n = 18) or benign gynecological disease (n=19). INTERVENTION(S) Eutopic ESCs were cultured under normoxia (20% O2) or hypoxia (6.5% O2), and migration and invasion capacity assayed, with pathway-focused polymerase chain reaction (PCR) array and ELISAs performed. CD26/dipeptidyl peptidase IV (DPPIV) expression was determined by flow cytometric analysis and enzymatic activity assay. The ESCs supplemented with Diprotin A (CD26 inhibitor), stromal cell-derived factor-1α, or AMD3100 (C-X-C motif receptor 4; CXCR4 blocker) were assayed for their migratory potential. MAIN OUTCOME MEASURE(S) Endometrial stromal cell migration and invasion under hypoxia. RESULT(S) Endometriotic ESCs showed significantly higher migration and invasion through collagen gels under hypoxia compared with nonendometriotic ESCs. The PCR array revealed down-regulation of the migration inhibitor CD26/DPPIV and up-regulation of angiogenic factors (vascular endothelial growth factor A, C-X-C motif Ligand 6; CXCL6) in endometriotic ESCs under hypoxia. The CD26/DPPIV surface expression and activity as well as angiogenic protein secretions suggested that the molecular mechanisms underlying aberrant migratory and angiogenic behavior in endometriotic ESCs. A combinatorial treatment with diprotin A and stromal cell-derived factor-1α effectively enhanced migration and invasion preferentially in endometriotic ESCs cultured hypoxically. CONCLUSION(S) Loss of CD26/DPPIV under hypoxia and the subsequent increase in migratory and angiogenic factors may favor conditions for lesion development in endometriosis.
Collapse
Affiliation(s)
- Chin Wen Tan
- BioSystems and Micromechanics, Singapore-MIT Alliance for Research & Technology, Singapore; Department of Obstetrics and Gynaecology, National University of Singapore, Singapore
| | - Yie Hou Lee
- BioSystems and Micromechanics, Singapore-MIT Alliance for Research & Technology, Singapore
| | - Heng Hao Tan
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore
| | - Matthew Sie Kuei Lau
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore
| | - Mahesh Choolani
- Department of Obstetrics and Gynaecology, National University of Singapore, Singapore
| | - Linda Griffith
- BioSystems and Micromechanics, Singapore-MIT Alliance for Research & Technology, Singapore; Department of Biological and Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts; Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Jerry Kok Yen Chan
- BioSystems and Micromechanics, Singapore-MIT Alliance for Research & Technology, Singapore; Department of Obstetrics and Gynaecology, National University of Singapore, Singapore; Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore; Cancer & Stem Cell Biology Program, Duke NUS Graduate Medical School, Singapore.
| |
Collapse
|
45
|
|
46
|
Gross J, Olze H, Mazurek B. Differential expression of transcription factors and inflammation-, ROS-, and cell death-related genes in organotypic cultures in the modiolus, the organ of Corti and the stria vascularis of newborn rats. Cell Mol Neurobiol 2014; 34:523-38. [PMID: 24595552 DOI: 10.1007/s10571-014-0036-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 02/14/2014] [Indexed: 12/22/2022]
Abstract
Cells respond to injury and hypoxia by changing gene expression. To study how the main compartments of the cochlea, the stria vascularis (SV), the organ of Corti (OC), and the modiolus (MOD), respond to such stress, we analyzed the expression of selected genes using microarray analysis. Organotypic cultures of SV, OC, and MOD from newborn rats were used as an experimental model. In the present study, we compare the expression of a total of 50 genes involved in apoptosis and necrosis, reactive oxygen species (ROS) metabolism, inflammation as well as selected transcription factors (TF) and analyze their role for the different cell death patterns observed in the three regions. MOD, OC, and SV differ not only in their basal gene profiles but also in their ability to respond to injury and hypoxia. The results provide two coexpression clusters across the three regions, a Hif-1a coexpression cluster and a cluster around the cell death-associated transcripts Casp3, Capn1, Capn2, and Capns1. These clusters include the TF Jun, Bmyc, Nfyc, Foxd3, Hes1, the ROS-associated molecules Sod3, and Nos2, and the inflammatory chemokine Ccl20. The evidence of both clusters indicates the complex and regulated character of gene expression following injury and hypoxia across the three regions SV, OC, and MOD. The high vulnerability of spiral ganglion neurons in the MOD region, previously explained on the basis of the availability of neuro-trophic factors, is associated with the increased endogenous production of ROS and nitric oxide and inadequate activation of protective acting genes.
Collapse
Affiliation(s)
- Johann Gross
- Molecular Biology Research Laboratory, Department of Otorhinolaryngology, Charité-Universitätsmedizin Berlin, Campus Charité Mitte, Berlin, Germany,
| | | | | |
Collapse
|
47
|
Souto GR, Queiroz-Junior CM, Costa FO, Mesquita RA. Smoking effect on chemokines of the human chronic periodontitis. Immunobiology 2014; 219:633-6. [PMID: 24780137 DOI: 10.1016/j.imbio.2014.03.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 03/21/2014] [Accepted: 03/22/2014] [Indexed: 11/30/2022]
Abstract
AIM Evaluate the effects of smoking on chemokines of the human chronic periodontitis (CP). MATERIALS AND METHODS Gingival samples were obtained from 23 smokers (S) and 20 non-smokers (NS) diagnosed with CP. Periodontal examination was performed. The CCL2, CCL3, CCL5, CCL19, CCL20, and CXCL8 chemokine levels were measured in gingival tissues using enzyme-linked immunosorbent assay. Chemokines were compared between S and NS, and were correlated with the number of cigarettes per day (C/day) and time of the smoking habit in years (SH/years). RESULTS CCL3 and CXCL8 of S were significantly smaller than that found in NS subjects, whereas the CCL5 levels increased in the S group. Negative correlations could be observed between CCL19 levels and SH/year. CONCLUSION Smoking suppresses the immune response which may contribute to an increased susceptibility to periodontal disease in smokers.
Collapse
Affiliation(s)
- Giovanna Ribeiro Souto
- Department of Oral Surgery and Pathology. School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| | - Celso Martins Queiroz-Junior
- Department of Oral Surgery and Pathology. School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fernando Oliveira Costa
- Department of Oral Surgery and Pathology. School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ricardo Alves Mesquita
- Department of Oral Surgery and Pathology. School of Dentistry, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
48
|
Mattox ML, D'Angelo JA, Dickinson BL. Redox control of indoleamine 2,3-dioxygenase expression and activity in human monocyte-derived dendritic cells is independent of changes in oxygen tension. Scand J Immunol 2014; 79:325-32. [PMID: 24612287 DOI: 10.1111/sji.12164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 02/26/2014] [Indexed: 11/28/2022]
Abstract
Dendritic cells (DCs) initiate adaptive immune responses to pathogens and tumours and maintain tolerance to self and innocuous antigens. These functions occur in organs and tissues exhibiting wide variations in nutrients, growth factors, redox and oxygen tension. Understanding how these microenvironmental factors influence DCs to affect immunological outcomes is of increasing relevance with the emerging success of DC-based cellular vaccines. In a previous study, we examined whether redox, an important environmental cue, could influence DC expression of the immunosuppressive enzyme indoleamine 2,3-dioxygenase (IDO). IDO-competent DCs promote long-term immune homoeostasis by limiting exaggerated inflammatory responses and directing regulatory T-cell effector function. To alter redox, we manipulated the activity of the cystine/glutamate antiporter, which functions to maintain intracellular and extracellular redox. The results of that study showed that redox perturbation strongly induced IDO expression and activity in DCs. While this study was performed using standard cell culture techniques with DCs cultured under 5% CO₂ and 20% O₂, it is clear that DCs capture and present antigens in inflamed tissues and secondary lymphoid organs which exhibit low oxygen tension (1-5% O₂). Therefore, here we investigated whether oxygen tension influences DC expression of IDO in the context of homoeostatic and altered redox.
Collapse
Affiliation(s)
- M L Mattox
- The West Virginia School of Osteopathic Medicine, Lewisburg, WV, USA
| | | | | |
Collapse
|
49
|
The Molecular and Cellular Events That Take Place during Craniofacial Distraction Osteogenesis. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2014; 2:e98. [PMID: 25289295 PMCID: PMC4174219 DOI: 10.1097/gox.0000000000000043] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Accepted: 12/04/2013] [Indexed: 12/26/2022]
Abstract
Summary: Gradual bone lengthening using distraction osteogenesis principles is the gold standard for the treatment of hypoplastic facial bones. However, the long treatment time is a major disadvantage of the lengthening procedures. The aim of this study is to review the current literature and summarize the cellular and molecular events occurring during membranous craniofacial distraction osteogenesis. Mechanical stimulation by distraction induces biological responses of skeletal regeneration that is accomplished by a cascade of biological processes that may include differentiation of pluripotential tissue, angiogenesis, osteogenesis, mineralization, and remodeling. There are complex interactions between bone-forming osteoblasts and other cells present within the bone microenvironment, particularly vascular endothelial cells that may be pivotal members of a complex interactive communication network in bone. Studies have implicated number of cytokines that are intimately involved in the regulation of bone synthesis and turnover. The gene regulation of numerous cytokines (transforming growth factor-β, bone morphogenetic proteins, insulin-like growth factor-1, and fibroblast growth factor-2) and extracellular matrix proteins (osteonectin, osteopontin) during distraction osteogenesis has been best characterized and discussed. Understanding the biomolecular mechanisms that mediate membranous distraction osteogenesis may guide the development of targeted strategies designed to improve distraction osteogenesis and accelerate bone regeneration that may lead to shorten the treatment duration.
Collapse
|
50
|
Gallo PM, Gallucci S. The dendritic cell response to classic, emerging, and homeostatic danger signals. Implications for autoimmunity. Front Immunol 2013; 4:138. [PMID: 23772226 PMCID: PMC3677085 DOI: 10.3389/fimmu.2013.00138] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 05/23/2013] [Indexed: 12/18/2022] Open
Abstract
Dendritic cells (DCs) initiate and control immune responses, participate in the maintenance of immunological tolerance and are pivotal players in the pathogenesis of autoimmunity. In patients with autoimmune disease and in experimental animal models of autoimmunity, DCs show abnormalities in both numbers and activation state, expressing immunogenic levels of costimulatory molecules and pro-inflammatory cytokines. Exogenous and endogenous danger signals activate DCs to stimulate the immune response. Classic endogenous danger signals are released, activated, or secreted by host cells and tissues experiencing stress, damage, and non-physiologic cell death; and are therefore referred to as damage-associated molecular patterns (DAMPs). Some DAMPs are released from cells, where they are normally sequestered, during necrosis (e.g., heat shock proteins, uric acid, ATP, HMGB1, mitochondria-derived molecules). Others are actively secreted, like Type I Interferons. Here we discuss important DAMPs in the context of autoimmunity. For some, there is a clear pathogenic link (e.g., nucleic acids and lupus). For others, there is less evidence. Additionally, we explore emerging danger signals. These include inorganic materials and man-made technologies (e.g., nanomaterials) developed as novel therapeutic approaches. Some nanomaterials can activate DCs and may trigger unintended inflammatory responses. Finally, we will review “homeostatic danger signals,” danger signals that do not derive directly from pathogens or dying cells but are associated with perturbations of tissue/cell homeostasis and may signal pathological stress. These signals, like acidosis, hypoxia, and changes in osmolarity, also play a role in inflammation and autoimmunity.
Collapse
Affiliation(s)
- Paul M Gallo
- Laboratory of Dendritic Cell Biology, Department of Microbiology and Immunology, Temple Autoimmunity Center, Temple University School of Medicine , Philadelphia, PA , USA
| | | |
Collapse
|