1
|
Deng XC, Liang JL, Zhang SM, Wang YZ, Lin YT, Meng R, Wang JW, Feng J, Chen WH, Zhang XZ. Interference of ATP-Adenosine Axis by Engineered Biohybrid for Amplifying Immunogenic Cell Death-Mediated Antitumor Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2405673. [PMID: 39022876 DOI: 10.1002/adma.202405673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 07/07/2024] [Indexed: 07/20/2024]
Abstract
Immunogenic cell death (ICD) often results in the production and accumulation of adenosine (ADO), a byproduct that negatively impacts the therapeutic effect as well as facilitates tumor development and metastasis. Here, an innovative strategy is elaborately developed to effectively activate ICD while avoiding the generation of immunosuppressive adenosine. Specifically, ZIF-90, an ATP-responsive consumer, is synthesized as the core carrier to encapsulate AB680 (CD73 inhibitor) and then coated with an iron-polyphenol layer to prepare the ICD inducer (AZTF), which is further grafted onto prebiotic bacteria via the esterification reaction to obtain the engineered biohybrid (Bc@AZTF). Particularly, the designed Bc@AZTF can actively enrich in tumor sites and respond to the acidic tumor microenvironment to offload AZTF nanoparticles, which can consume intracellular ATP (iATP) content and simultaneously inhibit the ATP-adenosine axis to reduce the accumulation of adenosine, thereby alleviating adenosine-mediated immunosuppression and strikingly amplifying ICD effect. Importantly, the synergy of anti-PD-1 (αPD-1) with Bc@AZTF not only establishes a collaborative antitumor immune network to potentiate effective tumoricidal immunity but also activates long-lasting immune memory effects to manage tumor recurrence and rechallenge, presenting a new paradigm for ICD treatment combined with adenosine metabolism.
Collapse
Affiliation(s)
- Xin-Chen Deng
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
- Department of Cardiology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, P. R. China
| | - Jun-Long Liang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
- Department of Cardiology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, P. R. China
| | - Shi-Man Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
- Department of Cardiology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, P. R. China
| | - Yu-Zhang Wang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
- Department of Cardiology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, P. R. China
| | - Yan-Tong Lin
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
- Department of Cardiology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, P. R. China
| | - Ran Meng
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
- Department of Cardiology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, P. R. China
| | - Jia-Wei Wang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
- Department of Cardiology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, P. R. China
| | - Jun Feng
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
- Department of Cardiology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, P. R. China
| | - Wei-Hai Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
- Department of Cardiology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, P. R. China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
- Department of Cardiology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, P. R. China
| |
Collapse
|
2
|
Romano E, Rosa I, Fioretto BS, Manetti M. Recent Insights into Cellular and Molecular Mechanisms of Defective Angiogenesis in Systemic Sclerosis. Biomedicines 2024; 12:1331. [PMID: 38927538 PMCID: PMC11201654 DOI: 10.3390/biomedicines12061331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/10/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
In systemic sclerosis (SSc, or scleroderma), defective angiogenesis, clinically manifesting with abnormal capillary architecture and severe capillary reduction, represents a hallmark of early-stage disease, usually preceding the onset of tissue fibrosis, and is caused by several cellular and molecular mechanisms affecting microvascular endothelial cells with different outcomes. Indeed, once damaged, endothelial cells can be dysfunctionally activated, thus becoming unable to undergo angiogenesis and promoting perivascular inflammation. They can also undergo apoptosis, transdifferentiate into profibrotic myofibroblasts, or acquire a senescence-associated secretory phenotype characterized by the release of exosomes and several profibrotic and proinflammatory mediators. In this narrative review, we aimed to give a comprehensive overview of recent studies dealing with the cellular and molecular mechanisms underlying SSc defective angiogenesis and the related endothelial cell dysfunctions, mainly the endothelial-to-mesenchymal transition process. We also discussed potential novel vascular treatment strategies able to restore the angiogenic process and reduce the endothelial-to-mesenchymal transition in this complex disease.
Collapse
Affiliation(s)
- Eloisa Romano
- Section of Internal Medicine, Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy;
| | - Irene Rosa
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy; (I.R.); (B.S.F.)
| | - Bianca Saveria Fioretto
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy; (I.R.); (B.S.F.)
| | - Mirko Manetti
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy; (I.R.); (B.S.F.)
- Imaging Platform, Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy
| |
Collapse
|
3
|
Shi B, Amin A, Dalvi P, Wang W, Lukacs N, Kai L, Cheresh P, Peclat TR, Chini CC, Chini EN, van Schooten W, Varga J. Heavy-chain antibody targeting of CD38 NAD + hydrolase ectoenzyme to prevent fibrosis in multiple organs. Sci Rep 2023; 13:22085. [PMID: 38086958 PMCID: PMC10716202 DOI: 10.1038/s41598-023-49450-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 12/08/2023] [Indexed: 12/18/2023] Open
Abstract
The functionally pleiotropic ectoenzyme CD38 is a glycohydrolase widely expressed on immune and non-hematopoietic cells. By converting NAD+ to ADP-ribose and nicotinamide, CD38 governs organismal NAD+ homeostasis and the activity of NAD+-dependent cellular enzymes. CD38 has emerged as a major driver of age-related NAD+ decline underlying adverse metabolic states, frailty and reduced health span. CD38 is upregulated in systemic sclerosis (SSc), a chronic disease characterized by fibrosis in multiple organs. We sought to test the hypothesis that inhibition of the CD38 ecto-enzymatic activity using a heavy-chain monoclonal antibody Ab68 will, via augmenting organismal NAD+, prevent fibrosis in a mouse model of SSc characterized by NAD+ depletion. Here we show that treatment of mice with a non-cytotoxic heavy-chain antibody that selectively inhibits CD38 ectoenzyme resulted in NAD+ boosting that was associated with significant protection from fibrosis in multiple organs. These findings suggest that targeted inhibition of CD38 ecto-enzymatic activity could be a potential pharmacological approach for SSc fibrosis treatment.
Collapse
Affiliation(s)
- Bo Shi
- Northwestern Scleroderma Program, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Asif Amin
- Department of Internal Medicine, The University of Michigan, Ann Arbor, MI, 48109, USA
| | | | - Wenxia Wang
- Northwestern Scleroderma Program, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Nicholas Lukacs
- Department of Pathology, The University of Michigan, Ann Arbor, MI, 48109, USA
| | - Li Kai
- Northwestern Scleroderma Program, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Paul Cheresh
- Division of Pulmonary and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Thais R Peclat
- Department of Anesthesiology and Kogod Center on Aging, Mayo Clinic, Jacksonville, FL, USA
| | - Claudia C Chini
- Department of Anesthesiology and Kogod Center on Aging, Mayo Clinic, Jacksonville, FL, USA
| | - Eduardo N Chini
- Department of Anesthesiology and Kogod Center on Aging, Mayo Clinic, Jacksonville, FL, USA
| | | | - John Varga
- Department of Internal Medicine, The University of Michigan, Ann Arbor, MI, 48109, USA.
- Michigan Scleroderma Program, The University of Michigan, Ann Arbor, MI, 48104, USA.
| |
Collapse
|
4
|
Borek I, Birnhuber A, Voelkel NF, Marsh LM, Kwapiszewska G. The vascular perspective on acute and chronic lung disease. J Clin Invest 2023; 133:e170502. [PMID: 37581311 PMCID: PMC10425217 DOI: 10.1172/jci170502] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2023] Open
Abstract
The pulmonary vasculature has been frequently overlooked in acute and chronic lung diseases, such as acute respiratory distress syndrome (ARDS), pulmonary fibrosis (PF), and chronic obstructive pulmonary disease (COPD). The primary emphasis in the management of these parenchymal disorders has largely revolved around the injury and aberrant repair of epithelial cells. However, there is increasing evidence that the vascular endothelium plays an active role in the development of acute and chronic lung diseases. The endothelial cell network in the capillary bed and the arterial and venous vessels provides a metabolically highly active barrier that controls the migration of immune cells, regulates vascular tone and permeability, and participates in the remodeling processes. Phenotypically and functionally altered endothelial cells, and remodeled vessels, can be found in acute and chronic lung diseases, although to different degrees, likely because of disease-specific mechanisms. Since vascular remodeling is associated with pulmonary hypertension, which worsens patient outcomes and survival, it is crucial to understand the underlying vascular alterations. In this Review, we describe the current knowledge regarding the role of the pulmonary vasculature in the development and progression of ARDS, PF, and COPD; we also outline future research directions with the hope of facilitating the development of mechanism-based therapies.
Collapse
Affiliation(s)
- Izabela Borek
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Anna Birnhuber
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Otto Loewi Research Center, Division of Physiology and Pathophysiology, Medical University of Graz, Graz, Austria
| | - Norbert F. Voelkel
- Pulmonary Medicine Department, University of Amsterdam Medical Centers, Amsterdam, Netherlands
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Leigh M. Marsh
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Otto Loewi Research Center, Division of Physiology and Pathophysiology, Medical University of Graz, Graz, Austria
| | - Grazyna Kwapiszewska
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Otto Loewi Research Center, Division of Physiology and Pathophysiology, Medical University of Graz, Graz, Austria
- Institute for Lung Health, German Lung Center (DZL), Cardiopulmonary Institute, Giessen, Germany
| |
Collapse
|
5
|
Al-Adwi Y, Westra J, van Goor H, Burgess JK, Denton CP, Mulder DJ. Macrophages as determinants and regulators of fibrosis in systemic sclerosis. Rheumatology (Oxford) 2023; 62:535-545. [PMID: 35861385 PMCID: PMC9891414 DOI: 10.1093/rheumatology/keac410] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/24/2022] [Accepted: 07/07/2022] [Indexed: 02/04/2023] Open
Abstract
SSc is a multiphase autoimmune disease with a well-known triad of clinical manifestations including vasculopathy, inflammation and fibrosis. Although a plethora of drugs has been suggested as potential candidates to halt SSc progression, nothing has proven clinically efficient. In SSc, both innate and adaptive immune systems are abnormally activated fuelling fibrosis of the skin and other vital organs. Macrophages have been implicated in the pathogenesis of SSc and are thought to be a major source of immune dysregulation. Due to their plasticity, macrophages can initiate and sustain chronic inflammation when classically activated while, simultaneously or parallelly, when alternatively activated they are also capable of secreting fibrotic factors. Here, we briefly explain the polarization process of macrophages. Subsequently, we link the activation of macrophages and monocytes to the molecular pathology of SSc, and illustrate the interplay between macrophages and fibroblasts. Finally, we present recent/near-future clinical trials and discuss novel targets related to macrophages/monocytes activation in SSc.
Collapse
Affiliation(s)
- Yehya Al-Adwi
- University of Groningen, University Medical Centre Groningen, Department of Internal Medicine, Division of Vascular Medicine
| | - Johanna Westra
- University of Groningen, University Medical Centre Groningen, Department of Rheumatology and Clinical Immunology
| | - Harry van Goor
- University of Groningen, University Medical Centre Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
| | - Janette K Burgess
- University of Groningen, University Medical Centre Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
| | - Christopher P Denton
- UCL Division of Medicine, University College London
- UCL Centre for Rheumatology and Connective Tissue Diseases, Royal Free Hospital, London, UK
| | - Douwe J Mulder
- University of Groningen, University Medical Centre Groningen, Department of Internal Medicine, Division of Vascular Medicine
| |
Collapse
|
6
|
Yu J, Tang R, Ding K. Epigenetic Modifications in the Pathogenesis of Systemic Sclerosis. Int J Gen Med 2022; 15:3155-3166. [PMID: 35342304 PMCID: PMC8942200 DOI: 10.2147/ijgm.s356877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 03/04/2022] [Indexed: 11/23/2022] Open
Abstract
Systemic sclerosis is a rare chronic autoimmune disease, which mainly manifests as immune disorders, vascular damage, and progressive fibrosis. The etiology of SSc is complex and involves multiple factors. Both genetic and environmental factors are involved in its pathogenesis. As one of the molecular mechanisms of environmental factors, epigenetic regulation plays an important role in the occurrence and development of systemic sclerosis, which involves DNA methylation, histone modification and non-coding RNA regulation. This review summarizes research advances in epigenetics, including exosomes, lncRNA, and mentions possible biomarkers and therapeutic targets among them.
Collapse
Affiliation(s)
- Jiangfan Yu
- Department of Dermatology, Second Xiangya Hospital of Central South University, Changsha, 410011, People’s Republic of China
| | - Rui Tang
- Department of Rheumatology and Immunology, Second Xiangya Hospital of Central South University, Changsha, 410011, People’s Republic of China
| | - Ke Ding
- Department of Urology, Xiangya Hospital of Central South University, Changsha, 410008, People’s Republic of China
- Correspondence: Ke Ding, Department of Urology, Xiangya Hospital of Central South University, Changsha, 410008, People’s Republic of China, Email
| |
Collapse
|
7
|
Höppner J, Bruni C, Distler O, Robson SC, Burmester GR, Siegert E, Distler JHW. Purinergic signaling in systemic sclerosis. Rheumatology (Oxford) 2021; 61:2770-2782. [PMID: 34849624 DOI: 10.1093/rheumatology/keab859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 11/13/2022] Open
Abstract
Systemic sclerosis (SSc) is a chronic autoimmune rheumatic disease that involves numerous organs and presents major management challenges. The histopathologic hallmarks of SSc include vasculopathy, fibrosis and autoimmune phenomena involving both innate and adaptive immune systems. Purinergic signalling is a pathway that may be implicated in the pathophysiology of several of these disease manifestations. Extracellular purines are potent signalling mediators, which have been shown to be dysregulated in SSc. As examples, purines can exacerbate vasculopathy and provoke platelet dysfunction; as well as contributing to immune dysregulation. Elements of purinergic signalling further promote organ and tissue fibrosis in several disease models. Here, we provide an overview of extracellular purine metabolism in purinergic signalling and link disorders of these to the molecular pathology of SSc. We also discuss targeting the purinergic signalling and explore the translational applications for new therapeutic options in SSc.
Collapse
Affiliation(s)
- Jakob Höppner
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Cosimo Bruni
- Department of Experimental and Clinical Medicine, Division of Rheumatology, Careggi University Hospital, University of Florence, Florence, Italy.,Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Oliver Distler
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Simon C Robson
- Departments of Anesthesia and Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Gerd R Burmester
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Elise Siegert
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Jörg H W Distler
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany
| |
Collapse
|
8
|
Elyaspour Z, Zibaeenezhad MJ, Razmkhah M, Razeghian-Jahromi I. Is It All About Endothelial Dysfunction and Thrombosis Formation? The Secret of COVID-19. Clin Appl Thromb Hemost 2021; 27:10760296211042940. [PMID: 34693754 PMCID: PMC8543709 DOI: 10.1177/10760296211042940] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The world is in a hard battle against COVID-19. Endothelial cells are among the most critical targets of SARS-CoV-2. Dysfunction of endothelium leads to vascular injury following by coagulopathies and thrombotic conditions in the vital organs increasing the risk of life-threatening events. Growing evidences revealed that endothelial dysfunction and consequent thrombotic conditions are associated with the severity of outcomes. It is not yet fully clear that these devastating sequels originate directly from the virus or a side effect of virus-induced cytokine storm. Due to endothelial dysfunction, plasma levels of some biomarkers are changed and relevant clinical manifestations appear as well. Stabilization of endothelial integrity and supporting its function are among the promising therapeutic strategies. Other than respiratory, COVID-19 could be called a systemic vascular disease and this aspect should be scrutinized in more detail in order to reduce related mortality. In the present investigation, the effects of COVID-19 on endothelial function and thrombosis formation are discussed. In this regard, critical players, laboratory findings, clinical manifestation, and suggestive therapies are presented.
Collapse
Affiliation(s)
- Zahra Elyaspour
- 48435Cardiovascular Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Mahboobeh Razmkhah
- 48435Shiraz Institute for Cancer Research, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Iman Razeghian-Jahromi
- 48435Cardiovascular Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
9
|
Saini A, Patel R, Gaba S, Singh G, Gupta GD, Monga V. Adenosine receptor antagonists: Recent advances and therapeutic perspective. Eur J Med Chem 2021; 227:113907. [PMID: 34695776 DOI: 10.1016/j.ejmech.2021.113907] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 10/05/2021] [Accepted: 10/05/2021] [Indexed: 12/14/2022]
Abstract
Adenosine is an endogenous purine-based nucleoside expressed nearly in all body tissues. It regulates various body functions by activating four G-protein coupled receptors, A1, A2A, A2B, and A3. These receptors are widely acknowledged as drug targets for treating different neurological, metabolic, and inflammatory diseases. Although numerous adenosine receptor inhibitors have been developed worldwide, achieving target selectivity is still a big hurdle in drug development. However, the identification of specific radioligands-based affinity assay, fluorescent ligands, and MS-based ligand assay have contributed to the development of selective and potent adenosine ligands. In recent years various small heterocyclic-based molecules have shown some promising results. Istradefylline has been approved for treating Parkinson's in Japan, while preladenant, tozadenant, CVT-6883, MRS-1523, and many more are under different phases of clinical development. The present review is focused on the quest to develop potent and selective adenosine inhibitors from 2013 to early 2021 by various research groups. The review also highlights their biological activity, selectivity, structure-activity relationship, molecular docking, and mechanistic studies. A special emphsesis on drug designing strategies has been also given the manuscript. The comprehensive compilation of research work carried out in the field will provide inevitable scope for designing and developing novel adenosine inhibitors with improved selectivity and efficacy.
Collapse
Affiliation(s)
- Anjali Saini
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga, 142001, Punjab, India
| | - Rajiv Patel
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga, 142001, Punjab, India
| | - Sobhi Gaba
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga, 142001, Punjab, India
| | - Gurpreet Singh
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga, 142001, Punjab, India.
| | - G D Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga, 142001, Punjab, India
| | - Vikramdeep Monga
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga, 142001, Punjab, India.
| |
Collapse
|
10
|
Jana S, Heaven MR, Alayash AI. Cell-Free Hemoglobin Does Not Attenuate the Effects of SARS-CoV-2 Spike Protein S1 Subunit in Pulmonary Endothelial Cells. Int J Mol Sci 2021; 22:9041. [PMID: 34445747 PMCID: PMC8396564 DOI: 10.3390/ijms22169041] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 12/28/2022] Open
Abstract
SARS-CoV-2 primarily infects epithelial airway cells that express the host entry receptor angiotensin-converting enzyme 2 (ACE2), which binds to the S1 spike protein on the surface of the virus. To delineate the impact of S1 spike protein interaction with the ACE2 receptor, we incubated the S1 spike protein with human pulmonary arterial endothelial cells (HPAEC). HPAEC treatment with the S1 spike protein caused disruption of endothelial barrier function, increased levels of numerous inflammatory molecules (VCAM-1, ICAM-1, IL-1β, CCL5, CXCL10), elevated mitochondrial reactive oxygen species (ROS), and a mild rise in glycolytic reserve capacity. Because low oxygen tension (hypoxia) is associated with severe cases of COVID-19, we also evaluated treatment with hemoglobin (HbA) as a potential countermeasure in hypoxic and normal oxygen environments in analyses with the S1 spike protein. We found hypoxia downregulated the expression of the ACE2 receptor and increased the critical oxygen homeostatic signaling protein, hypoxia-inducible factor (HIF-1α); however, treatment of the cells with HbA yielded no apparent change in the levels of ACE2 or HIF-1α. Use of quantitative proteomics revealed that S1 spike protein-treated cells have few differentially regulated proteins in hypoxic conditions, consistent with the finding that ACE2 serves as the host viral receptor and is reduced in hypoxia. However, in normoxic conditions, we found perturbed abundance of proteins in signaling pathways related to lysosomes, extracellular matrix receptor interaction, focal adhesion, and pyrimidine metabolism. We conclude that the spike protein alone without the rest of the viral components is sufficient to elicit cell signaling in HPAEC, and that treatment with HbA failed to reverse the vast majority of these spike protein-induced changes.
Collapse
Affiliation(s)
| | | | - Abdu I. Alayash
- Laboratory of Biochemistry and Vascular Biology, Center for Biologics Evaluation and Research, Food and Drug Administration (FDA), Silver Spring, MD 20993, USA; (S.J.); (M.R.H.)
| |
Collapse
|
11
|
Li X, Berg NK, Mills T, Zhang K, Eltzschig HK, Yuan X. Adenosine at the Interphase of Hypoxia and Inflammation in Lung Injury. Front Immunol 2021; 11:604944. [PMID: 33519814 PMCID: PMC7840604 DOI: 10.3389/fimmu.2020.604944] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/25/2020] [Indexed: 12/19/2022] Open
Abstract
Hypoxia and inflammation often coincide in pathogenic conditions such as acute respiratory distress syndrome (ARDS) and chronic lung diseases, which are significant contributors to morbidity and mortality for the general population. For example, the recent global outbreak of Coronavirus disease 2019 (COVID-19) has placed viral infection-induced ARDS under the spotlight. Moreover, chronic lung disease ranks the third leading cause of death in the United States. Hypoxia signaling plays a diverse role in both acute and chronic lung inflammation, which could partially be explained by the divergent function of downstream target pathways such as adenosine signaling. Particularly, hypoxia signaling activates adenosine signaling to inhibit the inflammatory response in ARDS, while in chronic lung diseases, it promotes inflammation and tissue injury. In this review, we discuss the role of adenosine at the interphase of hypoxia and inflammation in ARDS and chronic lung diseases, as well as the current strategy for therapeutic targeting of the adenosine signaling pathway.
Collapse
Affiliation(s)
- Xiangyun Li
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
- Department of Anesthesiology, Tianjin Medical University NanKai Hospital, Tianjin, China
| | - Nathanial K. Berg
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Tingting Mills
- Department of Biochemistry, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Kaiying Zhang
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Holger K. Eltzschig
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Xiaoyi Yuan
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
12
|
Gao ZW, Wang X, Zhang HZ, Lin F, Liu C, Dong K. The roles of adenosine deaminase in autoimmune diseases. Autoimmun Rev 2020; 20:102709. [PMID: 33197575 DOI: 10.1016/j.autrev.2020.102709] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 08/11/2020] [Indexed: 02/07/2023]
Abstract
Autoimmune diseases patients are characterized by the autoimmune disorders, whose immune system can't distinguish between auto- and foreign- antigens. Thus, Immune homeostasis disorder is the key factor for autoimmune diseases development. Adenosine deaminase (ADA) is the degrading enzyme for an immunosuppressive signal - adenosine, and play an important role in immune homeostasis regulation. Increasing evidences have shown that ADA is involved in various autoimmune diseases. ADA activity were changed in multiple autoimmune diseases patients and could be served as a biomarker for clinical diagnosis. In this study, we analyze the change of ADA activity in patients with autoimmune diseases, and we underline its potential diagnostic value for autoimmune diseases patients.
Collapse
Affiliation(s)
- Zhao-Wei Gao
- Department of Clinical Diagnose, Tangdu Hospital, Airforce military medical university, Xi'an City, Shannxi Province, China
| | - Xi Wang
- Department of Clinical Diagnose, Tangdu Hospital, Airforce military medical university, Xi'an City, Shannxi Province, China
| | - Hui-Zhong Zhang
- Department of Clinical Diagnose, Tangdu Hospital, Airforce military medical university, Xi'an City, Shannxi Province, China
| | - Fang Lin
- Department of Clinical Diagnose, Tangdu Hospital, Airforce military medical university, Xi'an City, Shannxi Province, China
| | - Chong Liu
- Department of Clinical Diagnose, Tangdu Hospital, Airforce military medical university, Xi'an City, Shannxi Province, China
| | - Ke Dong
- Department of Clinical Diagnose, Tangdu Hospital, Airforce military medical university, Xi'an City, Shannxi Province, China.
| |
Collapse
|
13
|
|
14
|
Understanding Fibrosis in Systemic Sclerosis: Novel and Emerging Treatment Approaches. Curr Rheumatol Rep 2020; 22:77. [DOI: 10.1007/s11926-020-00953-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2020] [Indexed: 12/11/2022]
|
15
|
Marchetti M. COVID-19-driven endothelial damage: complement, HIF-1, and ABL2 are potential pathways of damage and targets for cure. Ann Hematol 2020; 99:1701-1707. [PMID: 32583086 PMCID: PMC7312112 DOI: 10.1007/s00277-020-04138-8] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 06/09/2020] [Indexed: 02/07/2023]
Abstract
COVID-19 pandemia is a major health emergency causing hundreds of deaths worldwide. The high reported morbidity has been related to hypoxia and inflammation leading to endothelial dysfunction and aberrant coagulation in small and large vessels. This review addresses some of the pathways leading to endothelial derangement, such as complement, HIF-1α, and ABL tyrosine kinases. This review also highlights potential targets for prevention and therapy of COVID-19-related organ damage and discusses the role of marketed drugs, such as eculizumab and imatinib, as suitable candidates for clinical trials.
Collapse
Affiliation(s)
- Monia Marchetti
- Hematology Department, Az Osp SS Antonio e Biagio e Cesare Arrigo, Alessandria, Italy.
| |
Collapse
|