1
|
Wu R, Zhu W, Shao F, Wang J, Li D, Tuo Z, Yoo KH, Wusiman D, Shu Z, Ge W, Yang Y, Ke M, Wei W, Heavey S, Cho WC, Feng D. Expanding horizons in theragnostics: from oncology to multidisciplinary applications. LA RADIOLOGIA MEDICA 2025:10.1007/s11547-025-01971-7. [PMID: 40042756 DOI: 10.1007/s11547-025-01971-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 02/05/2025] [Indexed: 03/17/2025]
Abstract
Theragnostics is the integration of treatment and diagnosis, involving a drug or technology that combines diagnostic imaging with targeted therapy. This approach utilizes imaging to identify specific biological targets, which are then used to deliver therapeutic effects for the benefit of patients. The effectiveness and potential of theragnostics in improving patient outcomes are supported by significant clinical trials and technological innovations. Theragnostics has demonstrated its capacity to deliver targeted and real-time interventions, making it adaptable to diverse clinical domains. Its applications range from visualizing and eradicating tumors to addressing complex neurological disorders and cardiovascular diseases. The integration of nanomaterials and advancements in molecular biology further enhance the capabilities of theragnostics, promising a future where treatments are highly personalized, and diseases are understood and managed at a molecular level previously unattainable. Our comprehensive overview focuses on the current advancements in theragnostics applications across different disease domains. We highlight the role of molecular imaging technologies, such as PET/CT scans, in early diagnosis and treatment. Additionally, we explore the potential of chemokines as molecular imaging targets in systemic inflammatory diseases and central nervous system pathologies. In conclusion, the progression of theragnostics represents a transformative phase in medical practice, providing new avenues for precise treatment and improved patient outcomes. Its multidisciplinary nature and continuous innovation have the potential to profoundly impact the future of medical research and clinical practice, as well as revolutionizing the treatment and management of a wide array of diseases.
Collapse
Affiliation(s)
- Ruicheng Wu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Weizhen Zhu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Fanglin Shao
- Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Jie Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Dengxiong Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhouting Tuo
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Koo Han Yoo
- Department of Urology, Kyung Hee University, Seoul, South Korea
| | - Dilinaer Wusiman
- Department of Comparative Pathobiology, College of Veterinary Medicine, West Lafayette, USA
- Purdue Institute for Cancer Research, Purdue University, Westlll Lafayette, IN, USA
| | - Ziyu Shu
- Department of Earth Science and Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Wenjing Ge
- Department of Clinical Neurosciences, University of Cambridge, R3, Box 83, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Yubo Yang
- Department of Urology, Three Gorges Hospital, Chongqing University, Wanzhou, Chongqing, 404000, China
| | - Mang Ke
- Department of Urology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| | - Wuran Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Susan Heavey
- Division of Surgery & Interventional Science, University College London, London, W1W 7TS, UK.
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong SAR, China.
| | - Dechao Feng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Department of Urology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China.
- Division of Surgery & Interventional Science, University College London, London, W1W 7TS, UK.
| |
Collapse
|
2
|
Xu X, Jané P, Taelman V, Jané E, Dumont RA, Garama Y, Kim F, Del Val Gómez M, Gariani K, Walter MA. The Theranostic Genome. Nat Commun 2024; 15:10904. [PMID: 39738156 PMCID: PMC11686231 DOI: 10.1038/s41467-024-55291-x] [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: 01/25/2024] [Accepted: 12/05/2024] [Indexed: 01/01/2025] Open
Abstract
Theranostic drugs represent an emerging path to deliver on the promise of precision medicine. However, bottlenecks remain in characterizing theranostic targets, identifying theranostic lead compounds, and tailoring theranostic drugs. To overcome these bottlenecks, we present the Theranostic Genome, the part of the human genome whose expression can be utilized to combine therapeutic and diagnostic applications. Using a deep learning-based hybrid human-AI pipeline that cross-references PubMed, the Gene Expression Omnibus, DisGeNET, The Cancer Genome Atlas and the NIH Molecular Imaging and Contrast Agent Database, we bridge individual genes in human cancers with respective theranostic compounds. Cross-referencing the Theranostic Genome with RNAseq data from over 17'000 human tissues identifies theranostic targets and lead compounds for various human cancers, and allows tailoring targeted theranostics to relevant cancer subpopulations. We expect the Theranostic Genome to facilitate the development of new targeted theranostics to better diagnose, understand, treat, and monitor a variety of human cancers.
Collapse
Affiliation(s)
- Xiaoying Xu
- University of Lucerne, Lucerne, LU, Switzerland
| | - Pablo Jané
- University of Geneva, Geneva, GE, Switzerland
- Nuclear Medicine and Molecular Imaging Division, Geneva University Hospitals, Geneva, GE, Switzerland
| | | | - Eduardo Jané
- Departamento de Matemática Aplicada a la Ingeniería Aeroespacial - ETSIAE, Universidad Politécnica de Madrid, 28040, Madrid, Spain
| | | | | | | | - María Del Val Gómez
- Servicio de Medicina Nuclear, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Karim Gariani
- Division of Endocrinology, Diabetes, Nutrition and Patient Therapeutic Education, Geneva University Hospitals, Geneva, GE, Switzerland
| | - Martin A Walter
- University of Lucerne, Lucerne, LU, Switzerland.
- St. Anna Hospital, University of Lucerne, Lucerne, LU, Switzerland.
| |
Collapse
|
3
|
Ruddy TD, Davies RA, Kiess MC. Development and evolution of nuclear cardiology and cardiac PET in Canada. J Med Imaging Radiat Sci 2024; 55:S3-S9. [PMID: 38637261 DOI: 10.1016/j.jmir.2024.03.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/20/2024]
Abstract
Gated radionuclide angiography and myocardial perfusion imaging were developed in the United States and Europe in the 1970's and soon adopted in Canadian centers. Much of the early development of nuclear cardiology in Canada was in Toronto, Ontario and was quickly followed by new programs across the country. Clinical research in Canada contributed to the further development of nuclear cardiology and cardiac PET. The Canadian Nuclear Cardiology Society (CNCS) was formed in 1995 and became the Canadian Society of Cardiovascular Nuclear and CT Imaging (CNCT) in 2014. The CNCS had a major role in education and advocacy for cardiovascular nuclear medicine testing. The CNCS established the Dr Robert Burns Lecture and CNCT named the Canadian Society of Cardiovascular Nuclear and CT Imaging Annual Achievement Award for Dr Michael Freeman in memoriam of these two outstanding Canadian leaders in nuclear cardiology. The future of nuclear cardiology in Canada is exciting with the expanding use of SPECT imaging to include Tc-99m-pyrophosphate for diagnosis of transthyretin cardiac amyloidosis and the ongoing introduction of cardiac PET imaging.
Collapse
Affiliation(s)
- Terrence D Ruddy
- Division of Cardiology, University of Ottawa Heart Institute, Ottawa, Ontario, Canada.
| | - Ross A Davies
- Division of Cardiology, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Marla C Kiess
- Division of Cardiology, University of British Columbia, St. Paul's Hospital, Vancouver, British Columbia, Canada
| |
Collapse
|
4
|
Yi H, Qin L, Ye X, Song J, Ji J, Ye T, Li J, Li L. Progression of radio-labeled molecular imaging probes targeting chemokine receptors. Crit Rev Oncol Hematol 2024; 195:104266. [PMID: 38232861 DOI: 10.1016/j.critrevonc.2024.104266] [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: 03/13/2023] [Revised: 12/31/2023] [Accepted: 01/11/2024] [Indexed: 01/19/2024] Open
Abstract
Chemokine receptors are significantly expressed in the surface of most inflammatory cells and tumor cells. Guided by chemokines, inflammatory cells which express the relevant chemokine receptors migrate to inflammatory lesions and participate in the evolution of inflammation diseases. Similarly, driven by chemokines, immune cells infiltrate into tumor lesions not only induces alterations in the tumor microenvironment, disrupting the efficacy of tumor therapies, but also has the potential to selectively target tumoral cells and diminish tumor progression. Chemokine receptors, which are significantly expressed on the surface of tumor cell membranes, are regulated by chemokines and initiate tumor-associated signaling pathways within tumor cells, playing a complex role in tumor progression. Based on the antagonists targeting chemokine receptors, radionuclide-labeled molecular imaging probes have been developed for the emerging application of molecular imaging in diseases such as tumors and inflammation. The value and limitations of molecular probes in disease imaging are worth reviewing.
Collapse
Affiliation(s)
- Heqing Yi
- Department of Nuclear Medicine, Zhejiang Cancer Hospital, Banshan Street 1, Hangzhou, Zhejiang 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China
| | - Lilin Qin
- Second Clinical Medical College of Zhejiang Chinese Medical University, Banshan Street 1, Hangzhou, Zhejiang 310022, China
| | - Xuemei Ye
- Department of Nuclear Medicine, Zhejiang Cancer Hospital, Banshan Street 1, Hangzhou, Zhejiang 310022, China
| | - Jinling Song
- Department of Nuclear Medicine, Zhejiang Cancer Hospital, Banshan Street 1, Hangzhou, Zhejiang 310022, China
| | - Jianfeng Ji
- Department of Nuclear Medicine, Zhejiang Cancer Hospital, Banshan Street 1, Hangzhou, Zhejiang 310022, China
| | - Ting Ye
- Department of Nuclear Medicine, Zhejiang Cancer Hospital, Banshan Street 1, Hangzhou, Zhejiang 310022, China
| | - Juan Li
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Dongfang Street 150, Hangzhou, Zhejiang 310022, China.
| | - Linfa Li
- Department of Nuclear Medicine, Zhejiang Cancer Hospital, Banshan Street 1, Hangzhou, Zhejiang 310022, China.
| |
Collapse
|
5
|
Yang P, Wu Q, Sun L, Fang P, Liu L, Ji Y, Park JY, Qin X, Yang X, Wang H. Adaptive Immune Response Signaling Is Suppressed in Ly6C high Monocyte but Upregulated in Monocyte Subsets of ApoE-/- Mice - Functional Implication in Atherosclerosis. Front Immunol 2021; 12:809208. [PMID: 34987524 PMCID: PMC8721109 DOI: 10.3389/fimmu.2021.809208] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 11/23/2021] [Indexed: 11/26/2022] Open
Abstract
Rationale Inflammatory monocyte (MC) subset differentiation is a major feature in tissue inflammatory and atherosclerosis. The underlying molecular mechanism remains unclear. Objective This study aims to explore molecule targets and signaling which determinate immunological features in MC subsets. Methods and Results Blood Ly6Chigh and Ly6Clow MC subsets from control and ApoE-/- mice were isolated by flow cytometry sorting and subjected for bulk high-throughput RNA-sequencing. Intensive bioinformatic studies were performed by analyzing transcriptome through four pairs of comparisons: A) Ly6Chigh vs Ly6Clow in control mice; B) Ly6Chigh vs Ly6Clow in ApoE-/- mice; C) ApoE-/- Ly6Chigh vs control Ly6Chigh MC; D) ApoE-/- Ly6Clow vs control Ly6Clow MC. A total of 80 canonical pathways and 16 enriched pathways were recognized by top-down analysis using IPA and GSEA software, and further used for overlapping analysis. Immunological features and signaling were assessed on four selected functional groups, including MHCII, immune checkpoint, cytokine, and transcription factor (TF). Among the total 14578 significantly differentially expressed (SDE) genes identified though above four comparison, 1051 TF and 348 immunological genes were discovered. SDE immunological genes were matched with corresponding upstream SDE TF by IPA upstream analysis. Fourteen potential transcriptional axes were recognized to modulate immunological features in the Ly6C MC subset. Based on an intensive literature search, we found that the identified SDE immune checkpoint genes in Ly6Chigh MC are associated with pro-inflammatory/atherogenic balance function. Immune checkpoint genes GITR, CTLA4, and CD96 were upregulated in Ly6Clow MC from all mice and presented anti-inflammatory/atherogenic features. Six cytokine genes, including Ccl2, Tnfsf14, Il1rn, Cxcl10, Ccl9, and Cxcl2, were upregulated in Ly6Chigh MC from all mice and associated with pro-inflammatory/atherogenic feature. Cytokine receptor gene Il12rb2, Il1r1, Il27ra, Il5ra, Ngfr, Ccr7, and Cxcr5 were upregulated in Ly6Clow MC from all mice and presented anti-inflammatory/atherogenic features. MHCII genes (H2-Oa, H2-DMb2, H2-Ob, H2-Eb2, H2-Eb1, H2-Aa, and Cd74) were elevated in Ly6Clow MC from all mice. ApoE-/- augmented pro-atherogenic/inflammatory and antigen-presenting cells (APC) feature in both subsets due to elevated expression of cytokine genes (Cxcl11, Cntf, Il24, Xcl, Ccr5, Mpl, and Acvr2a) and MHCII gene (H2-Aa and H2-Ea-ps). Finally, we modeled immunological gene expression changes and functional implications in MC differentiation and adaptive immune response for MC subsets from control and ApoE-/- mice. Conclusions Ly6Chigh MC presented pro-inflammatory/atherogenic features and lower APC potential. Ly6Clow MC displayed anti-inflammatory/atherogenic features and higher APC potential. ApoE-/- confers upon both subsets with augmented pro-atherogenic/inflammatory function and APC potential.
Collapse
Affiliation(s)
- Pingping Yang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Center for Metabolic Disease Research, Department of Cardiovascular Science, Lewis Kats School of Medicine, Temple University, Philadelphia, PA, United States
- Department of Endocrinology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Qinghua Wu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Lizhe Sun
- Center for Metabolic Disease Research, Department of Cardiovascular Science, Lewis Kats School of Medicine, Temple University, Philadelphia, PA, United States
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Pu Fang
- Center for Metabolic Disease Research, Department of Cardiovascular Science, Lewis Kats School of Medicine, Temple University, Philadelphia, PA, United States
| | - Lu Liu
- Department of Endocrinology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yong Ji
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, China
| | - Joon-Young Park
- Center for Metabolic Disease Research, Department of Cardiovascular Science, Lewis Kats School of Medicine, Temple University, Philadelphia, PA, United States
| | - Xuebin Qin
- Tulane National Primate Research Center, School of Medicine, Tulane University, Covington, LA, United States
| | - Xiaofeng Yang
- Center for Metabolic Disease Research, Department of Cardiovascular Science, Lewis Kats School of Medicine, Temple University, Philadelphia, PA, United States
| | - Hong Wang
- Center for Metabolic Disease Research, Department of Cardiovascular Science, Lewis Kats School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
6
|
Márquez AB, van der Vorst EPC, Maas SL. Key Chemokine Pathways in Atherosclerosis and Their Therapeutic Potential. J Clin Med 2021; 10:3825. [PMID: 34501271 PMCID: PMC8432216 DOI: 10.3390/jcm10173825] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/20/2021] [Accepted: 08/20/2021] [Indexed: 12/24/2022] Open
Abstract
The search to improve therapies to prevent or treat cardiovascular diseases (CVDs) rages on, as CVDs remain a leading cause of death worldwide. Here, the main cause of CVDs, atherosclerosis, and its prevention, take center stage. Chemokines and their receptors have long been known to play an important role in the pathophysiological development of atherosclerosis. Their role extends from the initiation to the progression, and even the potential regression of atherosclerotic lesions. These important regulators in atherosclerosis are therefore an obvious target in the development of therapeutic strategies. A plethora of preclinical studies have assessed various possibilities for targeting chemokine signaling via various approaches, including competitive ligands and microRNAs, which have shown promising results in ameliorating atherosclerosis. Developments in the field also include detailed imaging with tracers that target specific chemokine receptors. Lastly, clinical trials revealed the potential of various therapies but still require further investigation before commencing clinical use. Although there is still a lot to be learned and investigated, it is clear that chemokines and their receptors present attractive yet extremely complex therapeutic targets. Therefore, this review will serve to provide a general overview of the connection between various chemokines and their receptors with atherosclerosis. The different developments, including mouse models and clinical trials that tackle this complex interplay will also be explored.
Collapse
Affiliation(s)
- Andrea Bonnin Márquez
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany;
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands
| | - Emiel P. C. van der Vorst
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany;
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, 80336 Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, 80336 Munich, Germany
| | - Sanne L. Maas
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany;
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany
| |
Collapse
|
7
|
Targeting the chemokine network in atherosclerosis. Atherosclerosis 2021; 330:95-106. [PMID: 34247863 DOI: 10.1016/j.atherosclerosis.2021.06.912] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/07/2021] [Accepted: 06/24/2021] [Indexed: 01/31/2023]
Abstract
Chemokines and their receptors represent a potential target for immunotherapy in chronic inflammation. They comprise a large family of cytokines with chemotactic activity, and their cognate receptors are expressed on all cells of the body. This network dictates leukocyte recruitment and activation, angiogenesis, cell proliferation and maturation. Dysregulation of chemokine and chemokine receptor expression as well as function participates in many pathologies including cancer, autoimmune diseases and chronic inflammation. In atherosclerosis, a lipid-driven chronic inflammation of middle-sized and large arteries, chemokines and their receptors participates in almost all stages of the disease from initiation of fatty streaks to mature atherosclerotic plaque formation. Atherosclerosis and its complications are the main driver of mortality and morbidity in cardiovascular diseases (CVD). Hence, exploring new fields of therapeutic targeting of atherosclerosis is of key importance. This review gives an overview of the recent advances on the role of key chemokines and chemokine receptors in atherosclerosis, addresses chemokine-based biomarkers at biochemical, imaging and genetic level in human studies, and highlights the clinial trials targeting atherosclerosis.
Collapse
|
8
|
Osborn EA, Albaghdadi M, Libby P, Jaffer FA. Molecular Imaging of Atherosclerosis. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00086-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
9
|
Overexpression of miR-29a-3p Suppresses Proliferation, Migration, and Invasion of Vascular Smooth Muscle Cells in Atherosclerosis via Targeting TNFRSF1A. BIOMED RESEARCH INTERNATIONAL 2020; 2020:9627974. [PMID: 32964047 PMCID: PMC7492923 DOI: 10.1155/2020/9627974] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 08/03/2020] [Indexed: 12/13/2022]
Abstract
Objective Increasing evidence highlights the significance of microRNAs (miRNAs) in the progression of atherosclerosis (AS). Our aim was to probe out the role and regulatory mechanism of miR-29a-3p in AS. Methods An in vivo model of AS was conducted by high-fat diet ApoE-/- mice. Oxidized low-density lipoprotein- (ox-LDL-) exposed vascular smooth muscle cells (VSMCs) were utilized as an in vitro of AS. Serum levels of total cholesterol (TC), triglyceride (TG), low-density lipoprotein cholesterol (LDL-C), and high-density lipoprotein cholesterol (HDL-C) were detected. Hematoxylin and eosin (H&E) and Masson's staining was presented to investigate the pathological changes. miR-29a-3p and TNFRSF1A expression was detected by RT-qPCR. Proliferative, migrated, and invaded abilities of VSMCs were determined via a series of assays. The interaction between miR-29a-3p and TNFRSF1A was verified through luciferase reporter assay. Results Upregulated miR-29a-3p and downregulated TNFRSF1A were found both in vitro and in vivo models of AS. miR-29a-3p mimic distinctly decreased the serum levels of TC, TG, and LDL-C and increased serum HDL-C levels. Moreover, its overexpression could ameliorate plaque formation of AS mice. In ox-LDL-induced VSMCs, miR-29a-3p overexpression notably decreased cell proliferation, migration, and invasion, which was reversed by TNFRSF1A overexpression. Also, miR-29a-3p could directly target the 3'UTR of TNFRSF1A. Conclusion miR-29a-3p overexpression ameliorated plaque formation of AS and suppressed proliferation, migration, and invasion of ox-LDL-induced VSMCs via TNFRSF1A, which offered novel insights into the progression of AS.
Collapse
|
10
|
Poret B, Desrues L, Bonin MA, Pedard M, Dubois M, Leduc R, Modzelewski R, Decazes P, Morin F, Vera P, Castel H, Bohn P, Gandolfo P. Development of Novel 111-In-Labelled DOTA Urotensin II Analogues for Targeting the UT Receptor Overexpressed in Solid Tumours. Biomolecules 2020; 10:E471. [PMID: 32204509 PMCID: PMC7175314 DOI: 10.3390/biom10030471] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 03/17/2020] [Accepted: 03/18/2020] [Indexed: 12/11/2022] Open
Abstract
Overexpression of G protein-coupled receptors (GPCRs) in tumours is widely used to develop GPCR-targeting radioligands for solid tumour imaging in the context of diagnosis and even treatment. The human vasoactive neuropeptide urotensin II (hUII), which shares structural analogies with somatostatin, interacts with a single high affinity GPCR named UT. High expression of UT has been reported in several types of human solid tumours from lung, gut, prostate, or breast, suggesting that UT is a valuable novel target to design radiolabelled hUII analogues for cancer diagnosis. In this study, two original urotensinergic analogues were first conjugated to a DOTA chelator via an aminohexanoic acid (Ahx) hydrocarbon linker and then -hUII and DOTA-urantide, complexed to the radioactive metal indium isotope to successfully lead to radiolabelled DOTA-Ahx-hUII and DOTA-Ahx-urantide. The 111In-DOTA-hUII in human plasma revealed that only 30% of the radioligand was degraded after a 3-h period. DOTA-hUII and DOTA-urantide exhibited similar binding affinities as native peptides and relayed calcium mobilization in HEK293 cells expressing recombinant human UT. DOTA-hUII, not DOTA-urantide, was able to promote UT internalization in UT-expressing HEK293 cells, thus indicating that radiolabelled 111In-DOTA-hUII would allow sufficient retention of radioactivity within tumour cells or radiolabelled DOTA-urantide may lead to a persistent binding on UT at the plasma membrane. The potential of these radioligands as candidates to target UT was investigated in adenocarcinoma. We showed that hUII stimulated the migration and proliferation of both human lung A549 and colorectal DLD-1 adenocarcinoma cell lines endogenously expressing UT. In vivo intravenous injection of 111In-DOTA-hUII in C57BL/6 mice revealed modest organ signals, with important retention in kidney. 111In-DOTA-hUII or 111In-DOTA-urantide were also injected in nude mice bearing heterotopic xenografts of lung A549 cells or colorectal DLD-1 cells both expressing UT. The observed significant renal uptake and low tumour/muscle ratio (around 2.5) suggest fast tracer clearance from the organism. Together, DOTA-hUII and DOTA-urantide were successfully radiolabelled with 111Indium, the first one functioning as a UT agonist and the second one as a UT-biased ligand/antagonist. To allow tumour-specific targeting and prolong body distribution in preclinical models bearing some solid tumours, these radiolabelled urotensinergic analogues should be optimized for being used as potential molecular tools for diagnosis imaging or even treatment tools.
Collapse
Affiliation(s)
- Benjamin Poret
- Institute for Research and Innovation in Biomedicine (IRIB), University of Rouen Normandy, INSERM U1239, DC2N, 76000 Rouen, France; (B.P.); (L.D.); (M.P.); (M.D.); (F.M.); (P.G.)
- EA 4108, Laboratory of Computer Science, Information Processing and Systems (LITIS), team “QuantIF”, Centre Henri Becquerel, 76000 Rouen, France; (R.M.); (P.D.); (P.V.); (P.B.)
- Department of Physiology & Pharmacology, Institute of Sherbrooke, Faculty of Medicine and Health Sciences, Sherbrooke University, Sherbrooke, QC J1H 5N4, Canada; (M.-A.B.); (R.L.)
| | - Laurence Desrues
- Institute for Research and Innovation in Biomedicine (IRIB), University of Rouen Normandy, INSERM U1239, DC2N, 76000 Rouen, France; (B.P.); (L.D.); (M.P.); (M.D.); (F.M.); (P.G.)
- EA 4108, Laboratory of Computer Science, Information Processing and Systems (LITIS), team “QuantIF”, Centre Henri Becquerel, 76000 Rouen, France; (R.M.); (P.D.); (P.V.); (P.B.)
- Institute for Research and Innovation in Biomedicine (IRIB), 76000 Rouen, France
| | - Marc-André Bonin
- Department of Physiology & Pharmacology, Institute of Sherbrooke, Faculty of Medicine and Health Sciences, Sherbrooke University, Sherbrooke, QC J1H 5N4, Canada; (M.-A.B.); (R.L.)
| | - Martin Pedard
- Institute for Research and Innovation in Biomedicine (IRIB), University of Rouen Normandy, INSERM U1239, DC2N, 76000 Rouen, France; (B.P.); (L.D.); (M.P.); (M.D.); (F.M.); (P.G.)
- Institute for Research and Innovation in Biomedicine (IRIB), 76000 Rouen, France
| | - Martine Dubois
- Institute for Research and Innovation in Biomedicine (IRIB), University of Rouen Normandy, INSERM U1239, DC2N, 76000 Rouen, France; (B.P.); (L.D.); (M.P.); (M.D.); (F.M.); (P.G.)
- Institute for Research and Innovation in Biomedicine (IRIB), 76000 Rouen, France
| | - Richard Leduc
- Department of Physiology & Pharmacology, Institute of Sherbrooke, Faculty of Medicine and Health Sciences, Sherbrooke University, Sherbrooke, QC J1H 5N4, Canada; (M.-A.B.); (R.L.)
| | - Romain Modzelewski
- EA 4108, Laboratory of Computer Science, Information Processing and Systems (LITIS), team “QuantIF”, Centre Henri Becquerel, 76000 Rouen, France; (R.M.); (P.D.); (P.V.); (P.B.)
- Institute for Research and Innovation in Biomedicine (IRIB), 76000 Rouen, France
| | - Pierre Decazes
- EA 4108, Laboratory of Computer Science, Information Processing and Systems (LITIS), team “QuantIF”, Centre Henri Becquerel, 76000 Rouen, France; (R.M.); (P.D.); (P.V.); (P.B.)
- Institute for Research and Innovation in Biomedicine (IRIB), 76000 Rouen, France
| | - Fabrice Morin
- Institute for Research and Innovation in Biomedicine (IRIB), University of Rouen Normandy, INSERM U1239, DC2N, 76000 Rouen, France; (B.P.); (L.D.); (M.P.); (M.D.); (F.M.); (P.G.)
- EA 4108, Laboratory of Computer Science, Information Processing and Systems (LITIS), team “QuantIF”, Centre Henri Becquerel, 76000 Rouen, France; (R.M.); (P.D.); (P.V.); (P.B.)
- Institute for Research and Innovation in Biomedicine (IRIB), 76000 Rouen, France
| | - Pierre Vera
- EA 4108, Laboratory of Computer Science, Information Processing and Systems (LITIS), team “QuantIF”, Centre Henri Becquerel, 76000 Rouen, France; (R.M.); (P.D.); (P.V.); (P.B.)
- Institute for Research and Innovation in Biomedicine (IRIB), 76000 Rouen, France
| | - Hélène Castel
- Institute for Research and Innovation in Biomedicine (IRIB), University of Rouen Normandy, INSERM U1239, DC2N, 76000 Rouen, France; (B.P.); (L.D.); (M.P.); (M.D.); (F.M.); (P.G.)
- Institute for Research and Innovation in Biomedicine (IRIB), 76000 Rouen, France
| | - Pierre Bohn
- EA 4108, Laboratory of Computer Science, Information Processing and Systems (LITIS), team “QuantIF”, Centre Henri Becquerel, 76000 Rouen, France; (R.M.); (P.D.); (P.V.); (P.B.)
- Institute for Research and Innovation in Biomedicine (IRIB), 76000 Rouen, France
| | - Pierrick Gandolfo
- Institute for Research and Innovation in Biomedicine (IRIB), University of Rouen Normandy, INSERM U1239, DC2N, 76000 Rouen, France; (B.P.); (L.D.); (M.P.); (M.D.); (F.M.); (P.G.)
- Institute for Research and Innovation in Biomedicine (IRIB), 76000 Rouen, France
| |
Collapse
|
11
|
Mota RI, Morgan SE, Bahnson EM. Diabetic vasculopathy: macro and microvascular injury. CURRENT PATHOBIOLOGY REPORTS 2020; 8:1-14. [PMID: 32655983 PMCID: PMC7351096 DOI: 10.1007/s40139-020-00205-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Diabetes is a common and prevalent medical condition as it affects many lives around the globe. Specifically, type-2 Diabetes (T2D) is characterized by chronic systemic inflammation alongside hyperglycemia and insulin resistance in the body, which can result in atherosclerotic legion formation in the arteries and thus progression of related conditions called diabetic vasculopathies. T2D patients are especially at risk for vascular injury; adjunct in many of these patients heir cholesterol and triglyceride levels reach dangerously high levels and accumulate in the lumen of their vascular system. RECENT FINDINGS Microvascular and macrovascular vasculopathies as complications of diabetes can accentuate the onset of organ illnesses, thus it is imperative that research efforts help identify more effective methods for prevention and diagnosis of early vascular injuries. Current research into vasculopathy identification/treatment will aid in the amelioration of diabetes-related symptoms and thus reduce the large number of deaths that this disease accounts annually. SUMMARY This review aims to showcase the evolution and effects of diabetic vasculopathy from development to clinical disease as macrovascular and microvascular complications with a concerted reference to sex-specific disease progression as well.
Collapse
Affiliation(s)
- Roberto I. Mota
- Department of Surgery, Division of Vascular Surgery; University of North Carolina at Chapel Hill, NC 27599
- Center for Nanotechnology in Drug Delivery; University of North Carolina at Chapel Hill, NC 27599
- McAllister Heart Institute, University of North Carolina at Chapel Hill, NC 27599
| | - Samuel E. Morgan
- Department of Surgery, Division of Vascular Surgery; University of North Carolina at Chapel Hill, NC 27599
- Center for Nanotechnology in Drug Delivery; University of North Carolina at Chapel Hill, NC 27599
| | - Edward M. Bahnson
- Department of Surgery, Division of Vascular Surgery; University of North Carolina at Chapel Hill, NC 27599
- Center for Nanotechnology in Drug Delivery; University of North Carolina at Chapel Hill, NC 27599
- McAllister Heart Institute, University of North Carolina at Chapel Hill, NC 27599
- Department of Cell Biology and Physiology. University of North Carolina at Chapel Hill, NC 27599
| |
Collapse
|
12
|
Heo GS, Sultan D, Liu Y. Current and novel radiopharmaceuticals for imaging cardiovascular inflammation. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF RADIOPHARMACEUTICAL CHEMISTRY AND BIOLOGY 2020; 64:4-20. [PMID: 32077667 DOI: 10.23736/s1824-4785.20.03230-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Cardiovascular disease (CVD) remains the leading cause of death worldwide despite advances in diagnostic technologies and treatment strategies. The underlying cause of most CVD is atherosclerosis, a chronic disease driven by inflammatory reactions. Atherosclerotic plaque rupture could cause arterial occlusion leading to ischemic tissue injuries such as myocardial infarction (MI) and stroke. Clinically, most imaging modalities are based on anatomy and provide limited information about the on-going molecular activities affecting the vulnerability of atherosclerotic lesion for risk stratification of patients. Thus, the ability to differentiate stable plaques from those that are vulnerable is an unmet clinical need. Of various imaging techniques, the radionuclide-based molecular imaging modalities including positron emission tomography and single-photon emission computerized tomography provide superior ability to noninvasively visualize molecular activities in vivo and may serve as a useful tool in tackling this challenge. Moreover, the well-established translational pathway of radiopharmaceuticals may also facilitate the translation of discoveries from benchtop to clinical investigation in contrast to other imaging modalities to fulfill the goal of precision medicine. The relationship between inflammation occurring within the plaque and its proneness to rupture has been well documented. Therefore, an active effort has been significantly devoted to develop radiopharmaceuticals specifically to measure CVD inflammatory status, and potentially elucidate those plaques which are prone to rupture. In the following review, molecular imaging of inflammatory biomarkers will be briefly discussed.
Collapse
Affiliation(s)
- Gyu S Heo
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, MO, USA
| | - Deborah Sultan
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, MO, USA
| | - Yongjian Liu
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, MO, USA -
| |
Collapse
|
13
|
Liu Y, Woodard PK. Chemokine receptors: Key for molecular imaging of inflammation in atherosclerosis. J Nucl Cardiol 2019; 26:1179-1181. [PMID: 29516368 PMCID: PMC6128785 DOI: 10.1007/s12350-018-1248-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 02/21/2018] [Indexed: 12/16/2022]
Affiliation(s)
- Yongjian Liu
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Pamela K Woodard
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
14
|
Lawal IO, Ankrah AO, Stoltz AC, Sathekge MM. Radionuclide imaging of inflammation in atherosclerotic vascular disease among people living with HIV infection: current practice and future perspective. Eur J Hybrid Imaging 2019; 3:5. [PMID: 34191183 PMCID: PMC8218042 DOI: 10.1186/s41824-019-0053-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 03/11/2019] [Indexed: 01/03/2023] Open
Abstract
People living with human immunodeficiency virus (HIV) infection have twice the risk of atherosclerotic vascular disease compared with non-infected individuals. Inflammation plays a critical role in the development and progression of atherosclerotic vascular disease. Therapies targeting inflammation irrespective of serum lipid levels have been shown to be effective in preventing the occurrence of CVD. Radionuclide imaging is a viable method for evaluating arterial inflammation. This evaluation is useful in quantifying CVD risk and for assessing the effectiveness of anti-inflammatory treatment. The most tested radionuclide method for quantifying arterial inflammation among people living with HIV infection has been with F-18 FDG PET/CT. The level of arterial uptake of F-18 FDG correlates with vascular inflammation and with the risk of development and progression of atherosclerotic disease. Several limitations exist to the use of F-18 FDG for PET quantification of arterial inflammation. Many targets expressed on macrophage, a significant player in arterial inflammation, have the potential for use in evaluating arterial inflammation among people living with HIV infection. The review describes the clinical utility of F-18 FDG PET/CT in assessing arterial inflammation as a risk for atherosclerotic disease among people living with HIV infection. It also outlines potential newer probes that may quantify arterial inflammation in the HIV-infected population by targeting different proteins expressed on macrophages.
Collapse
Affiliation(s)
- Ismaheel O. Lawal
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Private Bag X169, Pretoria, 0001 South Africa
| | - Alfred O. Ankrah
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Private Bag X169, Pretoria, 0001 South Africa
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen & University of Groningen, Groningen, The Netherlands
| | - Anton C. Stoltz
- Infectious Disease Unit, Department of Internal Medicine, University of Pretoria & Steve Biko Academic Hospital, Pretoria, South Africa
| | - Mike M. Sathekge
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Private Bag X169, Pretoria, 0001 South Africa
| |
Collapse
|
15
|
|