1
|
Li X, Liu Y, Liu J, Ma W, Ti R, Warshel A, Ye RD, Zhu L. CXC Chemokine Ligand 12 Facilitates Gi Protein Binding to CXC Chemokine Receptor 4 by Stabilizing Packing of the Proline-Isoleucine-Phenylalanine Motif: Insights from Automated Path Searching. J Am Chem Soc 2025; 147:10129-10138. [PMID: 40096846 PMCID: PMC11951141 DOI: 10.1021/jacs.4c14293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 02/15/2025] [Accepted: 02/19/2025] [Indexed: 03/19/2025]
Abstract
The activation of G protein-coupled receptors (GPCRs) is a complex multibody multievent process involving agonist binding, receptor activation, G protein coupling, and subsequent G protein activation. The order and energetics of these events, though crucial for the rational design of selective GPCR drugs, are challenging to characterize and remain largely underexplored. Here, we employed molecular dynamics simulations and our recently developed traveling salesman-based automated path searching (TAPS) algorithm to efficiently locate the minimum free-energy paths for the coupling events of the CXC chemokine receptor 4 (CXCR4) with its endogenous ligand CXC chemokine ligand 12 (CXCL12) and Gi protein. We show that, after overcoming three low energy barriers (3.24-6.89 kcal/mol), Gi alone can precouple with CXCR4 even without CXCL12, consistent with previous reports on the existence of the apo CXCR4-Gi complex and our NanoBiT experiments. The highest barrier of 6.89 kcal/mol in this process corresponds to the packing of the proline-isoleucine-phenylalanine (PIF) motif of CXCR4. Interestingly, without Gi, CXCL12 alone cannot activate CXCR4 (high barrier of 18.89 kcal/mol). Instead, it can enhance Gi coupling by circumventing the energy barrier of PIF packing. Shedding new light on the activation mechanism of CXCR4, these results present TAPS as a promising tool for uncovering complete activation pathways of GPCRs and the corresponding agonist design.
Collapse
Affiliation(s)
- Xinyu Li
- School of
Medicine and Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China
| | - Yezhou Liu
- Kobilka
Institute
of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China
| | - Jinchu Liu
- School of
Medicine and Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China
| | - Wenzhuo Ma
- School of
Medicine and Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China
| | - Rujuan Ti
- School of
Medicine and Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China
| | - Arieh Warshel
- Department
of Chemistry, University of Southern California, Los Angeles, California 90089, United States
| | - Richard D. Ye
- Kobilka
Institute
of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China
- The
Chinese
University of Hong Kong, Shenzhen Futian Biomedical Innovation R&D
Center, Shenzhen, Guangdong 518048, China
| | - Lizhe Zhu
- School of
Medicine and Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China
| |
Collapse
|
2
|
Rizatdinova SN, Ershova AE, Astrakhantseva IV. Pseudotyped Viruses: A Useful Platform for Pre-Clinical Studies Conducted in a BSL-2 Laboratory Setting. Biomolecules 2025; 15:135. [PMID: 39858529 PMCID: PMC11763035 DOI: 10.3390/biom15010135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/12/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
The study of pathogenic viruses has always posed significant biosafety challenges. In particular, the study of highly pathogenic viruses requires methods with low biological risk but relatively high sensitivity and convenience in detection. In recent years, pseudoviruses, which consist of a backbone of one virus and envelope proteins of another virus, have become one of the most widely used tools for exploring the mechanisms of viruses binding to cells, membrane fusion and viral entry, as well as for screening the libraries of antiviral substances, evaluating the potential of neutralizing monoclonal antibodies, developing neutralization tests, and therapeutic platforms. During the outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), pseudotyped virus-based assays played a pivotal role in advancing our understanding of virus-cell interactions and the role of its proteins in disease pathogenesis. Such tools facilitated the search for potential therapeutic agents and accelerated epidemiological studies on post-infection and post-vaccination humoral immunity. This review focuses on the use of pseudoviruses as a model for large-scale applications to study enveloped viruses.
Collapse
Affiliation(s)
| | | | - Irina V. Astrakhantseva
- Department of Immunobiology and Biomedicine, Sirius University of Science and Technology, 354349 Sirius, Krasnodarsky Krai, Russia; (S.N.R.); (A.E.E.)
| |
Collapse
|
3
|
Huang S, Liu M, Zhang H, Song W, Guo W, Feng Y, Ma X, Shi X, Liu J, Liu L, Qi T, Wang Z, Yan B, Shen Y. HIV-MTB Co-Infection Reduces CD4+ T Cells and Affects Granuloma Integrity. Viruses 2024; 16:1335. [PMID: 39205309 PMCID: PMC11360352 DOI: 10.3390/v16081335] [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: 06/22/2024] [Revised: 08/08/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024] Open
Abstract
Granuloma is a crucial pathological feature of tuberculosis (TB). The relationship between CD4+ T cells in both peripheral blood and granulomatous tissue, and the integrity of granulomas in Human Immunodeficiency Virus (HIV)-MTB co-infection, remains unexplored. This study collected biopsy specimens from 102 TB patients (53 with HIV-MTB co-infection and 49 only with TB). Hematoxylin and eosin (HE) staining and immunohistochemical staining were performed, followed by microscopic examination of the integrity of tuberculous granulomas. Through statistical analysis of peripheral blood CD4+ T cell counts, tissue CD4+ T cell proportion, and the integrity of granulomas, it was observed that HIV infection leads to poor formation of tuberculous granulomas. Peripheral blood CD4+ T cell counts were positively correlated with granuloma integrity, and there was a similar positive correlation between tissue CD4+ T cell proportions and granuloma integrity. Additionally, a positive correlation was found between peripheral blood CD4+ T cell counts and the proportion of CD4+ T cells in granuloma tissues. Therefore, HIV infection could impact the morphology and structure of tuberculous granulomas, with a reduced proportion of both peripheral blood and tissue CD4+ T lymphocytes.
Collapse
Affiliation(s)
- Suyue Huang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; (S.H.); (W.S.)
| | - Maoying Liu
- Department of Microbiology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Hui Zhang
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Wei Song
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; (S.H.); (W.S.)
| | - Wenjuan Guo
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; (S.H.); (W.S.)
| | - Yanling Feng
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; (S.H.); (W.S.)
| | - Xin Ma
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; (S.H.); (W.S.)
| | - Xia Shi
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; (S.H.); (W.S.)
| | - Jianjian Liu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; (S.H.); (W.S.)
| | - Li Liu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; (S.H.); (W.S.)
| | - Tangkai Qi
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; (S.H.); (W.S.)
| | - Zhenyan Wang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; (S.H.); (W.S.)
| | - Bo Yan
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; (S.H.); (W.S.)
| | - Yinzhong Shen
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China; (S.H.); (W.S.)
| |
Collapse
|
4
|
Suwattananuruk P, Yaset S, Chotipanich C, Moldes-Anaya A, Sundset R, Berzaghi R, Figenschau S, Claes S, Schols D, Rojsitthisak P, Kranz M, Vajragupta O. Radiosynthesis and preclinical evaluation of a 68Ga-labeled tetrahydroisoquinoline-based ligand for PET imaging of C-X-C chemokine receptor type 4 in an animal model of glioblastoma. EJNMMI Radiopharm Chem 2024; 9:61. [PMID: 39162901 PMCID: PMC11335985 DOI: 10.1186/s41181-024-00290-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 07/30/2024] [Indexed: 08/21/2024] Open
Abstract
BACKGROUND This study aimed to develop a novel positron emission tomography (PET) tracer, [68Ga]Ga-TD-01, for CXCR4 imaging. To achieve this goal, the molecular scaffold of TIQ15 was tuned by conjugation with the DOTA chelator to make it suitable for 68Ga radiolabeling. METHODS A bifunctional chelator was prepared by conjugating the amine group of TIQ15 with p-NCS-Bz-DOTA, yielding TD-01, with a high yield (68.92%). TD-01 was then radiolabeled with 68Ga using 0.1 M ammonium acetate at 60 °C for 10 min. A 1-h dynamic small animal PET/MRI study of the labeled compound in GL261-luc2 tumor-bearing mice was performed, and brain tumor uptake was assessed. Blocking studies involved pre-administration of TIQ15 (10 mg/kg) 10 min before the PET procedure started. RESULTS [68Ga]Ga-TD-01 exhibited a radiochemical yield (RCY) of 36.33 ± 1.50% (EOS), with a radiochemical purity > 99% and a molar activity of 55.79 ± 1.96 GBq/µmol (EOS). The radiotracer showed in vitro stability in PBS and human plasma for over 4 h. Biodistribution studies in healthy animals revealed favorable kinetics for subsequent PET pharmacokinetic modeling with low uptake in the brain and moderate uptake in lungs, intestines and spleen. Elimination could be assigned to a renal-hepatic pathway as showed by high uptake in kidneys, liver, and urinary bladder. Importantly, [68Ga]Ga-TD-01 uptake in glioblastoma (GBM)-bearing mice significantly decreased upon competition with TIQ15, with a baseline tumor-to-background ratios > 2.5 (20 min p.i.), indicating high specificity. CONCLUSION The newly developed CXCR4 PET tracer, [68Ga]Ga-TD-01, exhibited a high binding inhibition for CXCR4, excellent in vitro stability, and favorable pharmacokinetics, suggesting that the compound is a promising candidate for full in vivo characterization of CXCR4 expression in GBM, with potential for further development as a tool in cancer diagnosis.
Collapse
Affiliation(s)
- Piyapan Suwattananuruk
- Department of Food and Pharmaceutical Chemistry and Center of Excellence in Natural Products for Ageing and Chronic Diseases, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
- Molecular Probes for Imaging Research Network, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Sukanya Yaset
- National Cyclotron and PET Centre, Chulabhorn Hospital, Bangkok, Thailand
| | | | | | - Rune Sundset
- PET Imaging Center, University Hospital of North Norway, Tromsø, Norway
- Department of Clinical Medicine, Nuclear Medicine and Radiation Biology Research Group, UiT The Arctic University of Norway, Tromsø, Norway
| | - Rodrigo Berzaghi
- Department of Clinical Medicine, Nuclear Medicine and Radiation Biology Research Group, UiT The Arctic University of Norway, Tromsø, Norway
| | - Stine Figenschau
- Department of Clinical Medicine, Nuclear Medicine and Radiation Biology Research Group, UiT The Arctic University of Norway, Tromsø, Norway
| | - Sandra Claes
- Department of Microbiology, Immunology and Transplantation, Laboratory of Virology and Chemotherapy, KU Leuven, Louvain, Belgium
| | - Dominique Schols
- Department of Microbiology, Immunology and Transplantation, Laboratory of Virology and Chemotherapy, KU Leuven, Louvain, Belgium
| | - Pornchai Rojsitthisak
- Department of Food and Pharmaceutical Chemistry and Center of Excellence in Natural Products for Ageing and Chronic Diseases, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
- Molecular Probes for Imaging Research Network, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Mathias Kranz
- PET Imaging Center, University Hospital of North Norway, Tromsø, Norway.
- Department of Clinical Medicine, Nuclear Medicine and Radiation Biology Research Group, UiT The Arctic University of Norway, Tromsø, Norway.
| | - Opa Vajragupta
- Department of Food and Pharmaceutical Chemistry and Center of Excellence in Natural Products for Ageing and Chronic Diseases, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand.
- Molecular Probes for Imaging Research Network, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
5
|
Moezpoor MR, Stevenson M. Help or Hinder: Protein Host Factors That Impact HIV-1 Replication. Viruses 2024; 16:1281. [PMID: 39205255 PMCID: PMC11360189 DOI: 10.3390/v16081281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/05/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024] Open
Abstract
Interactions between human immunodeficiency virus type 1 (HIV-1) and the host factors or restriction factors of its target cells determine the cell's susceptibility to, and outcome of, infection. Factors intrinsic to the cell are involved at every step of the HIV-1 replication cycle, contributing to productive infection and replication, or severely attenuating the chances of success. Furthermore, factors unique to certain cell types contribute to the differences in infection between these cell types. Understanding the involvement of these factors in HIV-1 infection is a key requirement for the development of anti-HIV-1 therapies. As the list of factors grows, and the dynamic interactions between these factors and the virus are elucidated, comprehensive and up-to-date summaries that recount the knowledge gathered after decades of research are beneficial to the field, displaying what is known so that researchers can build off the groundwork of others to investigate what is unknown. Herein, we aim to provide a review focusing on protein host factors, both well-known and relatively new, that impact HIV-1 replication in a positive or negative manner at each stage of the replication cycle, highlighting factors unique to the various HIV-1 target cell types where appropriate.
Collapse
Affiliation(s)
- Michael Rameen Moezpoor
- Department of Microbiology and Immunology, University of Miami Leonard M. Miller School of Medicine, Miami, FL 33136, USA
| | - Mario Stevenson
- Raymond F. Schinazi and Family Endowed Chair in Biomedicine; Professor of Medicine; Director, Institute of AIDS and Emerging Infectious Diseases; Department of Microbiology and Immunology, University of Miami Leonard M. Miller School of Medicine, Life Science Technology Park, 1951 NW 7th Avenue, Room 2331B, Suite 200, Miami, FL 33136, USA;
| |
Collapse
|
6
|
Zhou Z, Guo J, Hetrick B, Tiwari S, Haikerwal A, Han Y, Bond VC, Huang MB, Mankowski MK, Snyder BA, Hogan PA, Sharma SK, Liotta DC, Reid TE, Wilson LJ, Wu Y. Characterization of a CXCR4 antagonist TIQ-15 with dual tropic HIV entry inhibition properties. PLoS Pathog 2024; 20:e1012448. [PMID: 39146384 PMCID: PMC11349218 DOI: 10.1371/journal.ppat.1012448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 08/27/2024] [Accepted: 07/25/2024] [Indexed: 08/17/2024] Open
Abstract
The chemokine co-receptors CXCR4 and CCR5 mediate HIV entry and signal transduction necessary for viral infection. However, to date only the CCR5 antagonist maraviroc is approved for treating HIV-1 infection. Given that approximately 50% of late-stage HIV patients also develop CXCR4-tropic virus, clinical anti-HIV CXCR4 antagonists are needed. Here, we describe a novel allosteric CXCR4 antagonist TIQ-15 which inhibits CXCR4-tropic HIV-1 infection of primary and transformed CD4 T cells. TIQ-15 blocks HIV entry with an IC50 of 13 nM. TIQ-15 also inhibits SDF-1α/CXCR4-mediated cAMP production, cofilin activation, and chemotactic signaling. In addition, TIQ-15 induces CXCR4 receptor internalization without affecting the levels of the CD4 receptor, suggesting that TIQ-15 may act through a novel allosteric site on CXCR4 for blocking HIV entry. Furthermore, TIQ-15 did not inhibit VSV-G pseudotyped HIV-1 infection, demonstrating its specificity in blocking CXCR4-tropic virus entry, but not CXCR4-independent endocytosis or post-entry steps. When tested against a panel of clinical isolates, TIQ-15 showed potent inhibition against CXCR4-tropic and dual-tropic viruses, and moderate inhibition against CCR5-tropic isolates. This observation was followed by a co-dosing study with maraviroc, and TIQ-15 demonstrated synergistic activity. In summary, here we describe a novel HIV-1 entry inhibitor, TIQ-15, which potently inhibits CXCR4-tropic viruses while possessing low-level synergistic activities against CCR5-tropic viruses. TIQ-15 could potentially be co-dosed with the CCR5 inhibitor maraviroc to block viruses of mixed tropisms.
Collapse
Affiliation(s)
- Zheng Zhou
- Center for Infectious Disease Research, George Mason University, Manassas, Virginia, United States of America
| | - Jia Guo
- Center for Infectious Disease Research, George Mason University, Manassas, Virginia, United States of America
| | - Brian Hetrick
- Center for Infectious Disease Research, George Mason University, Manassas, Virginia, United States of America
| | - Sameer Tiwari
- Center for Infectious Disease Research, George Mason University, Manassas, Virginia, United States of America
| | - Amrita Haikerwal
- Center for Infectious Disease Research, George Mason University, Manassas, Virginia, United States of America
| | - Yang Han
- Center for Infectious Disease Research, George Mason University, Manassas, Virginia, United States of America
| | - Vincent C. Bond
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| | - Ming B. Huang
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| | - Marie K. Mankowski
- Department of Infectious Disease Research, Southern Research Institute, Frederick, Maryland, United States of America
| | - Beth A. Snyder
- Department of Infectious Disease Research, Southern Research Institute, Frederick, Maryland, United States of America
| | - Priscilla A. Hogan
- Department of Infectious Disease Research, Southern Research Institute, Frederick, Maryland, United States of America
| | - Savita K. Sharma
- Department of Chemistry, Emory University, Atlanta, Georgia, United States of America
| | - Dennis C. Liotta
- Department of Chemistry, Emory University, Atlanta, Georgia, United States of America
| | - Terry-Elinor Reid
- Center for Infectious Disease Research, George Mason University, Manassas, Virginia, United States of America
| | - Lawrence J. Wilson
- Department of Chemistry, Emory University, Atlanta, Georgia, United States of America
| | - Yuntao Wu
- Center for Infectious Disease Research, George Mason University, Manassas, Virginia, United States of America
| |
Collapse
|
7
|
Chou TC, Maggirwar NS, Marsden MD. HIV Persistence, Latency, and Cure Approaches: Where Are We Now? Viruses 2024; 16:1163. [PMID: 39066325 PMCID: PMC11281696 DOI: 10.3390/v16071163] [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: 06/25/2024] [Revised: 07/13/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
The latent reservoir remains a major roadblock to curing human immunodeficiency virus (HIV) infection. Currently available antiretroviral therapy (ART) can suppress active HIV replication, reduce viral loads to undetectable levels, and halt disease progression. However, antiretroviral drugs are unable to target cells that are latently infected with HIV, which can seed viral rebound if ART is stopped. Consequently, a major focus of the field is to study the latent viral reservoir and develop safe and effective methods to eliminate it. Here, we provide an overview of the major mechanisms governing the establishment and maintenance of HIV latency, the key challenges posed by latent reservoirs, small animal models utilized to study HIV latency, and contemporary cure approaches. We also discuss ongoing efforts to apply these approaches in combination, with the goal of achieving a safe, effective, and scalable cure for HIV that can be extended to the tens of millions of people with HIV worldwide.
Collapse
Affiliation(s)
- Tessa C. Chou
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, CA 92617, USA; (T.C.C.); (N.S.M.)
| | - Nishad S. Maggirwar
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, CA 92617, USA; (T.C.C.); (N.S.M.)
| | - Matthew D. Marsden
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, CA 92617, USA; (T.C.C.); (N.S.M.)
- Department of Medicine, Division of Infectious Disease, School of Medicine, University of California, Irvine, CA 92617, USA
| |
Collapse
|
8
|
Yi HB, Lee S, Seo K, Kim H, Kim M, Lee HS. Cellular and Biophysical Applications of Genetic Code Expansion. Chem Rev 2024; 124:7465-7530. [PMID: 38753805 DOI: 10.1021/acs.chemrev.4c00112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
Despite their diverse functions, proteins are inherently constructed from a limited set of building blocks. These compositional constraints pose significant challenges to protein research and its practical applications. Strategically manipulating the cellular protein synthesis system to incorporate novel building blocks has emerged as a critical approach for overcoming these constraints in protein research and application. In the past two decades, the field of genetic code expansion (GCE) has achieved significant advancements, enabling the integration of numerous novel functionalities into proteins across a variety of organisms. This technological evolution has paved the way for the extensive application of genetic code expansion across multiple domains, including protein imaging, the introduction of probes for protein research, analysis of protein-protein interactions, spatiotemporal control of protein function, exploration of proteome changes induced by external stimuli, and the synthesis of proteins endowed with novel functions. In this comprehensive Review, we aim to provide an overview of cellular and biophysical applications that have employed GCE technology over the past two decades.
Collapse
Affiliation(s)
- Han Bin Yi
- Department of Chemistry, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul 04107, Republic of Korea
| | - Seungeun Lee
- Department of Chemistry, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul 04107, Republic of Korea
| | - Kyungdeok Seo
- Department of Chemistry, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul 04107, Republic of Korea
| | - Hyeongjo Kim
- Department of Chemistry, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul 04107, Republic of Korea
| | - Minah Kim
- Department of Chemistry, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul 04107, Republic of Korea
| | - Hyun Soo Lee
- Department of Chemistry, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul 04107, Republic of Korea
| |
Collapse
|
9
|
Liu J, Ding C, Shi Y, Wang Y, Zhang X, Huang L, Fang Q, Shuai C, Gao Y, Wu J. Advances in Mechanism of HIV-1 Immune Reconstitution Failure: Understanding Lymphocyte Subpopulations and Interventions for Immunological Nonresponders. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1609-1620. [PMID: 38768409 DOI: 10.4049/jimmunol.2300777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 04/08/2024] [Indexed: 05/22/2024]
Abstract
In individuals diagnosed with AIDS, the primary method of sustained suppression of HIV-1 replication is antiretroviral therapy, which systematically increases CD4+ T cell levels and restores immune function. However, there is still a subset of 10-40% of people living with HIV who not only fail to reach normal CD4+ T cell counts but also experience severe immune dysfunction. These individuals are referred to as immunological nonresponders (INRs). INRs have a higher susceptibility to opportunistic infections and non-AIDS-related illnesses, resulting in increased morbidity and mortality rates. Therefore, it is crucial to gain new insights into the primary mechanisms of immune reconstitution failure to enable early and effective treatment for individuals at risk. This review provides an overview of the dynamics of key lymphocyte subpopulations, the main molecular mechanisms of INRs, clinical diagnosis, and intervention strategies during immune reconstitution failure, primarily from a multiomics perspective.
Collapse
Affiliation(s)
- Jiamin Liu
- School of Public Health, Anhui Medical University, Hefei, China
| | - Chengchao Ding
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yu Shi
- School of Public Health, Anhui Medical University, Hefei, China
| | - Yiyu Wang
- School of Public Health, Anhui Medical University, Hefei, China
| | - Xiangyu Zhang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Lina Huang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qin Fang
- Central Laboratory of HIV Molecular and Immunology, Anhui Provincial Center for Disease Control and Prevention, Hefei, China
| | - Chenxi Shuai
- Central Laboratory of HIV Molecular and Immunology, Anhui Provincial Center for Disease Control and Prevention, Hefei, China
| | - Yong Gao
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jianjun Wu
- School of Public Health, Anhui Medical University, Hefei, China
- Central Laboratory of HIV Molecular and Immunology, Anhui Provincial Center for Disease Control and Prevention, Hefei, China
| |
Collapse
|
10
|
Bauckneht M, Filippi L. Pentixather: paving the way for radioligand therapy in oncohematology. Expert Rev Anticancer Ther 2024; 24:205-209. [PMID: 38593347 DOI: 10.1080/14737140.2024.2341728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 04/08/2024] [Indexed: 04/11/2024]
Affiliation(s)
- Matteo Bauckneht
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Health Sciences (DISSAL), University of Genova, Genova, Italy
| | - Luca Filippi
- Nuclear Medicine Unit, Department of Oncohaematology, Fondazione PTV Policlinico Tor Vergata University Hospital, Rome, Italy
| |
Collapse
|
11
|
Dudek AM, Feist WN, Sasu EJ, Luna SE, Ben-Efraim K, Bak RO, Cepika AM, Porteus MH. A simultaneous knockout knockin genome editing strategy in HSPCs potently inhibits CCR5- and CXCR4-tropic HIV-1 infection. Cell Stem Cell 2024; 31:499-518.e6. [PMID: 38579682 PMCID: PMC11212398 DOI: 10.1016/j.stem.2024.03.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 12/29/2023] [Accepted: 03/06/2024] [Indexed: 04/07/2024]
Abstract
Allogeneic hematopoietic stem and progenitor cell transplant (HSCT) of CCR5 null (CCR5Δ32) cells can be curative for HIV-1-infected patients. However, because allogeneic HSCT poses significant risk, CCR5Δ32 matched bone marrow donors are rare, and CCR5Δ32 transplant does not confer resistance to the CXCR4-tropic virus, it is not a viable option for most patients. We describe a targeted Cas9/AAV6-based genome editing strategy for autologous HSCT resulting in both CCR5- and CXCR4-tropic HIV-1 resistance. Edited human hematopoietic stem and progenitor cells (HSPCs) maintain multi-lineage repopulation capacity in vivo, and edited primary human T cells potently inhibit infection by both CCR5-tropic and CXCR4-tropic HIV-1. Modification rates facilitated complete loss of CCR5-tropic replication and up to a 2,000-fold decrease in CXCR4-tropic replication without CXCR4 locus disruption. This multi-factor editing strategy in HSPCs could provide a broad approach for autologous HSCT as a functional cure for both CCR5-tropic and CXCR4-tropic HIV-1 infections.
Collapse
Affiliation(s)
- Amanda M Dudek
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - William N Feist
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Elena J Sasu
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sofia E Luna
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kaya Ben-Efraim
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Rasmus O Bak
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark; Aarhus Institute of Advanced Studies (AIAS), Aarhus University, 8000 Aarhus, Denmark
| | - Alma-Martina Cepika
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Matthew H Porteus
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
12
|
Alzahrani SO, McRobbie G, Khan A, D'huys T, Van Loy T, Walker AN, Renard I, Hubin TJ, Schols D, Burke BP, Archibald SJ. trans-IV restriction: a new configuration for metal bis-cyclam complexes as potent CXCR4 inhibitors. Dalton Trans 2024; 53:5616-5623. [PMID: 38439632 PMCID: PMC10949960 DOI: 10.1039/d3dt01729j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 02/26/2024] [Indexed: 03/06/2024]
Abstract
The chemokine receptor CXCR4 is implicated in multiple diseases including inflammatory disorders, cancer growth and metastasis, and HIV/AIDS. CXCR4 targeting has been evaluated in treating cancer metastasis and therapy resistance. Cyclam derivatives, most notably AMD3100 (Plerixafor™), are a common motif in small molecule CXCR4 antagonists. However, AMD3100 has not been shown to be effective in cancer treatment as an individual agent. Configurational restriction and transition metal complex formation increases receptor binding affinity and residence time. In the present study, we have synthesized novel trans-IV locked cyclam-based CXCR4 inhibitors, a previously unexploited configuration, and demonstrated their higher affinity for CXCR4 binding and CXCL12-mediated signaling inhibition compared to AMD3100. These results pave the way for even more potent CXCR4 inhibitors that may provide significant efficacy in cancer therapy.
Collapse
Affiliation(s)
- Seraj O Alzahrani
- Centre for Biomedicine and Positron Emission Tomography Research Centre, Hull York Medical School and University of Hull, Cottingham Road, Hull, HU6 7RX, UK.
| | - Graeme McRobbie
- Centre for Biomedicine and Positron Emission Tomography Research Centre, Hull York Medical School and University of Hull, Cottingham Road, Hull, HU6 7RX, UK.
| | - Abid Khan
- Centre for Biomedicine and Positron Emission Tomography Research Centre, Hull York Medical School and University of Hull, Cottingham Road, Hull, HU6 7RX, UK.
- The University of Manchester, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, Manchester, UK
| | - Thomas D'huys
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Tom Van Loy
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Ashlie N Walker
- Department of Chemistry and Physics, Southwestern Oklahoma State University, Weatherford, OK 73096, USA
| | - Isaline Renard
- Centre for Biomedicine and Positron Emission Tomography Research Centre, Hull York Medical School and University of Hull, Cottingham Road, Hull, HU6 7RX, UK.
- School of Biomedical Engineering and Imaging Sciences, King's College London, 4th Floor Lambeth Wing, St Thomas' Hospital, London, SE1 7EH, UK
| | - Timothy J Hubin
- Department of Chemistry and Physics, Southwestern Oklahoma State University, Weatherford, OK 73096, USA
| | - Dominique Schols
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Benjamin P Burke
- Centre for Biomedicine and Positron Emission Tomography Research Centre, Hull York Medical School and University of Hull, Cottingham Road, Hull, HU6 7RX, UK.
| | - Stephen J Archibald
- Centre for Biomedicine and Positron Emission Tomography Research Centre, Hull York Medical School and University of Hull, Cottingham Road, Hull, HU6 7RX, UK.
- School of Biomedical Engineering and Imaging Sciences, King's College London, 4th Floor Lambeth Wing, St Thomas' Hospital, London, SE1 7EH, UK
| |
Collapse
|
13
|
Yang S, Chen Y, Gu J, Harris A, Su RC, Ho EA. pH-sensitive dual-preventive siRNA-based nanomicrobicide reactivates autophagy and inhibits HIV infection in vaginal CD4+ cells. J Control Release 2024; 366:849-863. [PMID: 38176469 DOI: 10.1016/j.jconrel.2023.12.043] [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: 07/18/2023] [Revised: 12/13/2023] [Accepted: 12/24/2023] [Indexed: 01/06/2024]
Abstract
Women are more susceptible to HIV transmission through unprotected heterosexual intercourse due to biological and social vulnerabilities. Intravaginal delivery of siRNAs targeting viral genes, host genes, or in combination has shown promising outcomes against HSV, HPV and HIV. Therefore, in this study, we designed, developed and evaluated a pH-sensitive RNAi-based combination nanomicrobide for the prevention/reduction of vaginal transmission of HIV. The nanomicrobide was composed of siRNA-PEI encapsulated PLGA-PEG nanoparticles (siRNA NP) loaded in a HEC gel dosage form with siRNA targeting host gene CCR5 and the viral gene Nef as a dual preventive strategy. Knocking down CCR5, a co-receptor for HIV could prevent HIV from attaching to and entering host cells and knocking down Nef could reactivate autophagy that was inhibited by Nef to improve the elimination of intracellular virus that escaped the first line of defense. The siRNA NP showed a desirable particle size and zeta potential for intravaginal delivery and a pH-dependent release profile whereby low amounts of siRNA was released under acidic vaginal conditions (vaginal fluid simulant; VFS, pH 4.2) (6.0 ± 0.4% released over 15 days) but significantly higher amounts of siRNA was released under neutral pH conditions (phosphate buffered saline; PBS, pH 7.4) (22.9 ± 0.4% released over 15 days). The CCR5-Nef-specific siRNA NP efficiently knocked down CCR5 and Nef protein expression by 43% and 63%, respectively, reactivated Nef-blocked autophagy and inhibited the replication of HIV in vitro (71.8% reduction in p24 expression). After being formulated into a gel dosage form, siRNA NP could be readily released from the gel, penetrate the vaginal epithelial layer, get taken up into the target cells and knockdown Nef and CCR5 without causing cytotoxicity in a vaginal mucosal co-culture model. Functionalization of siRNA NP with anti-CD4 antibody and loaded into a 0.5% HEC gel improved vaginal distribution and uptake of siRNA in a mouse model with distribution of siRNA restricted to the reproductive tract without any unwanted systemic uptake. The 0.5% HEC gel loaded with siRNA NP-(m)CD4 significantly downregulated approximately 40% of CCR5 protein in the lower vagina and 36% of CCR5 protein in the upper vaginal and cervical region. In contrast, 0.5% HEC gel loaded with siRNA NP-IgG did not result in significant gene knockdown.
Collapse
Affiliation(s)
- Sidi Yang
- Laboratory for Drug Delivery and Biomaterials, School of Pharmacy, Faculty of Science, University of Waterloo, Canada; College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Canada
| | - Yufei Chen
- Laboratory for Drug Delivery and Biomaterials, School of Pharmacy, Faculty of Science, University of Waterloo, Canada; College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Canada
| | - Jijin Gu
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Canada
| | - Angela Harris
- Department of Medical Microbiology & Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Canada; National HIV and Retrovirology Laboratory, JC Wilt Infectious Disease Research Centre, Public Health Agency of Canada, Canada
| | - Ruey-Chyi Su
- Department of Medical Microbiology & Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Canada; National HIV and Retrovirology Laboratory, JC Wilt Infectious Disease Research Centre, Public Health Agency of Canada, Canada
| | - Emmanuel A Ho
- Laboratory for Drug Delivery and Biomaterials, School of Pharmacy, Faculty of Science, University of Waterloo, Canada; Waterloo Institute for Nanotechnology, University of Waterloo, Canada.
| |
Collapse
|
14
|
Faivre N, Verollet C, Dumas F. The chemokine receptor CCR5: multi-faceted hook for HIV-1. Retrovirology 2024; 21:2. [PMID: 38263120 PMCID: PMC10807162 DOI: 10.1186/s12977-024-00634-1] [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: 09/27/2023] [Accepted: 01/13/2024] [Indexed: 01/25/2024] Open
Abstract
Chemokines are cytokines whose primary role is cellular activation and stimulation of leukocyte migration. They perform their various functions by interacting with G protein-coupled cell surface receptors (GPCRs) and are involved in the regulation of many biological processes such as apoptosis, proliferation, angiogenesis, hematopoiesis or organogenesis. They contribute to the maintenance of the homeostasis of lymphocytes and coordinate the function of the immune system. However, chemokines and their receptors are sometimes hijacked by some pathogens to infect the host organism. For a given chemokine receptor, there is a wide structural, organizational and conformational diversity. In this review, we describe the evidence for structural variety reported for the chemokine receptor CCR5, how this variability can be exploited by HIV-1 to infect its target cells and what therapeutic solutions are currently being developed to overcome this problem.
Collapse
Affiliation(s)
- Natacha Faivre
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UPS), Toulouse, France
- International Research Laboratory (IRP) CNRS "IM-TB/HIV", Toulouse, France
- International Research Laboratory (IRP) CNRS "IM-TB/HIV", Buenos Aires, Argentina
| | - Christel Verollet
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UPS), Toulouse, France
- International Research Laboratory (IRP) CNRS "IM-TB/HIV", Toulouse, France
- International Research Laboratory (IRP) CNRS "IM-TB/HIV", Buenos Aires, Argentina
| | - Fabrice Dumas
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UPS), Toulouse, France.
| |
Collapse
|
15
|
Ray M, Sayeed A, Ganshert M, Saha A. Direct Binding Methods to Measure Receptor-Ligand Interactions. J Phys Chem B 2024; 128:3-19. [PMID: 38134048 DOI: 10.1021/acs.jpcb.3c05041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2023]
Abstract
G-protein-coupled receptors (GPCRs) contribute to numerous physiological processes via complex network mechanisms. While indirect signaling assays (Ca2+ mobilization, cAMP production, and GTPγS binding) have been useful in identifying and characterizing downstream signaling mechanisms of GPCRs, these methods lack measurements of direct binding affinities, kinetics, binding specificity, and selectivity that are important parameters in GPCR drug discovery. In comparison to existing direct methods that use radio- or fluorescent labels, label-free techniques can closely emulate the native interactions around binding partners. Surface plasmon resonance (SPR) is a label-free technique that utilizes the refractive index (RI) property and is applied widely in quantitative GPCR-ligand binding kinetics measurement including small molecules screening. However, purified GPCRs are further embedded in a synthetic lipid environment which is immobilized through different tags to the SPR sensor surface, resulting in a non-native environment. Here, we introduced a methodology that also uses the RI property to measure binding interactions in a label-free, immobilization-free arrangement. The free-solution technique is successfully applied in quantifying the interaction of bioactive lipids to cognate lipid GPCRs, which is not purified but rather present in near-native conditions, i.e., in milieu of other cytoplasmic lipids and proteins. To further consider the wide applicability of these free-solution approaches in biomolecular interaction research, additional applications on a variety of receptor-ligand pairs are imperative.
Collapse
Affiliation(s)
- Manisha Ray
- Department of Chemistry and Biochemistry, Loyola University Chicago, 1068 W. Sheridan Road, Chicago, Illinois 60660, United States
| | - Aryana Sayeed
- Department of Chemistry and Biochemistry, Loyola University Chicago, 1068 W. Sheridan Road, Chicago, Illinois 60660, United States
| | - Madeline Ganshert
- Department of Chemistry and Biochemistry, Loyola University Chicago, 1068 W. Sheridan Road, Chicago, Illinois 60660, United States
| | - Arjun Saha
- Department of Chemistry and Biochemistry, University of Wisconsin, Milwaukee Chemistry Bldg, 144, 3210 N Cramer Street, Milwaukee, Wisconsin 53211, United States
| |
Collapse
|
16
|
Matsui Y, Miura Y. Advancements in Cell-Based Therapies for HIV Cure. Cells 2023; 13:64. [PMID: 38201268 PMCID: PMC10778010 DOI: 10.3390/cells13010064] [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: 11/30/2023] [Revised: 12/21/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
The treatment of human immunodeficiency virus (HIV-1) has evolved since the establishment of combination antiretroviral therapy (ART) in the 1990s, providing HIV-infected individuals with approaches that suppress viral replication, prevent acquired immunodeficiency syndrome (AIDS) throughout their lifetime with continuous therapy, and halt HIV transmission. However, despite the success of these regimens, the global HIV epidemic persists, prompting a comprehensive exploration of potential strategies for an HIV cure. Here, we offer a consolidated overview of cell-based therapies for HIV-1, focusing on CAR-T cell approaches, gene editing, and immune modulation. Persistent challenges, including CAR-T cell susceptibility to HIV infection, stability, and viral reservoir control, underscore the need for continued research. This review synthesizes current knowledge, highlighting the potential of cellular therapies to address persistent challenges in the pursuit of an HIV cure.
Collapse
Affiliation(s)
- Yusuke Matsui
- Gladstone Institute of Virology, Gladstone Institutes, 1650 Owens St., San Francisco, CA 941578, USA
| | - Yasuo Miura
- Department of Transfusion Medicine and Cell Therapy, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake, Toyoake 470-1192, Aichi, Japan
| |
Collapse
|
17
|
Cossarini F, Aberg JA, Chen BK, Mehandru S. Viral Persistence in the Gut-Associated Lymphoid Tissue and Barriers to HIV Cure. AIDS Res Hum Retroviruses 2023; 40:54-65. [PMID: 37450338 PMCID: PMC10790554 DOI: 10.1089/aid.2022.0180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023] Open
Abstract
More than 40 years after the first reported cases of what then became known as acquired immunodeficiency syndrome (AIDS), tremendous progress has been achieved in transforming the disease from almost universally fatal to a chronic manageable condition. Nonetheless, the efforts to find a preventative vaccine or a cure for the underlying infection with Human Immunodeficiency Virus (HIV) remain largely unsuccessful. Many challenges intrinsic to the virus characteristics and host response need to be overcome for either goal to be achieved. This article will review the obstacles to an effective HIV cure, specifically the steps involved in the generation of HIV latency, focusing on the role of the gut-associated lymphoid tissue, which has received less attention compared with the peripheral blood, despite being the largest repository of lymphoid tissue in the human body, and a large site for HIV persistence.
Collapse
Affiliation(s)
- Francesca Cossarini
- Division of Infectious Diseases, Department of Medicine, Icahn School at Mount Sinai, New York, New York, USA
- Precision Immunology Institute, Icahn School at Mount Sinai, New York, New York, USA
| | - Judith A. Aberg
- Division of Infectious Diseases, Department of Medicine, Icahn School at Mount Sinai, New York, New York, USA
| | - Benjamin K. Chen
- Division of Infectious Diseases, Department of Medicine, Icahn School at Mount Sinai, New York, New York, USA
- Precision Immunology Institute, Icahn School at Mount Sinai, New York, New York, USA
| | - Saurabh Mehandru
- Precision Immunology Institute, Icahn School at Mount Sinai, New York, New York, USA
- Division of Gastroenterology, Department of Medicine, Icahn School at Mount Sinai, New York, New York, USA
| |
Collapse
|
18
|
Zhou S, Long N, Swanstrom R. Evolution Driven By A Varying Host Environment Selects For Distinct HIV-1 Entry Phenotypes and Other Informative Variants. FRONTIERS IN VIROLOGY (LAUSANNE, SWITZERLAND) 2023; 3:1291996. [PMID: 38239974 PMCID: PMC10795538 DOI: 10.3389/fviro.2023.1291996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2024]
Abstract
HIV-1 generates remarkable intra- and inter-host viral diversity during infection. In response to dynamic selective pressures of the host environment, HIV-1 will evolve distinct phenotypes - biological features that provide fitness advantages. The transmitted form of HIV-1 has been shown to require a high density of CD4 on the target cell surface (as found on CD4+ T cells) and typically uses CCR5 as a co-receptor during entry. This phenotype is referred to as R5 T cell-tropic (or R5 T-tropic); however, HIV-1 can switch to a secondary co-receptor, CXCR4, resulting in a X4 T cell-tropic phenotype. Macrophage-tropic (or M-tropic) HIV-1 can evolve to efficiently enter cells expressing low densities of CD4 on their surface (such as macrophages/microglia). So far only CCR5-using M-tropic viruses have been found. M-tropic HIV-1 is most frequently found within the central nervous system, and infection of the CNS has been associated with neurological impairment. It has been shown that interferon resistance phenotypes have a selective advantage during transmission, but the underlying mechanism of this is still unclear. During untreated infection, HIV-1 evolves under selective pressure from both the humoral/antibody response and CD8+ T cell killing. Sufficiently potent antiviral therapy will suppress viral replication, but if the antiviral drugs are not sufficiently potent to stop replication then the replicating virus will evolve drug resistance. HIV-1 phenotypes are highly relevant to treatment efforts, clinical outcomes, vaccine studies, and cure strategies. Therefore, it is critical to understand the dynamics of the host environment that drive these phenotypes and how they affect HIV-1 pathogenesis. This review will provide a comprehensive discussion of HIV-1 entry, transmission, and drug resistance phenotypes. Finally, we will assess the methods used in previous and current research to characterize these phenotypes.
Collapse
Affiliation(s)
- Shuntai Zhou
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Nathan Long
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ronald Swanstrom
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
19
|
Abstract
For many years, antibody drug conjugates (ADC) have teased with the promise of targeted payload delivery to diseased cells, embracing the targeting of the antibody to which a cytotoxic payload is conjugated. During the past decade this promise has started to be realised with the approval of more than a dozen ADCs for the treatment of various cancers. Of these ADCs, brentuximab vedotin really laid the foundations of a template for a successful ADC with lysosomal payload release from a cleavable dipeptide linker, measured DAR by conjugation to the Cys-Cys interchain bonds of the antibody and a cytotoxic payload. Using this ADC design model oncology has now expanded their repertoire of payloads to include non-cytotoxic compounds. These new payload classes have their origins in prior medicinal chemistry programmes aiming to design selective oral small molecule drugs. While this may not have been achieved, the resulting compounds provide excellent starting points for ADC programmes with some compounds amenable to immediate linker attachment while for others extensive SAR and structural information offer invaluable design insights. Many of these new oncology payload classes are of interest to other therapeutic areas facilitating rapid access to drug-linkers for exploration as non-oncology ADCs. Other therapeutic areas have also pursued unique payload classes with glucocorticoid receptor modulators (GRM) being the most clinically advanced in immunology. Here, ADC payloads come full circle, as oncology is now investigating GRM payloads for the treatment of cancer. This chapter aims to cover all these new ADC approaches while describing the medicinal chemistry origins of the new non-cytotoxic payloads.
Collapse
Affiliation(s)
- Adrian D Hobson
- Small Molecule Therapeutics & Platform Technologies, AbbVie Bioresearch Center, Worcester, MA, United States.
| |
Collapse
|
20
|
Hong J, Choi Y, Lee G, Kim J, Jang Y, Yoon CH, Seo HW, Park IK, Kang SH, Choi J. Nanosome-Mediated Delivery Of Hdac Inhibitors and Oxygen Molecules for the Transcriptional Reactivation of Latent Hiv-Infected Cd4 + T Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2301730. [PMID: 37118849 DOI: 10.1002/smll.202301730] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/11/2023] [Indexed: 06/19/2023]
Abstract
The treatment of human immunodeficiency virus (HIV) infection is notoriously difficult due to the ability of this virus to remain latent in the host's CD4+ T cells. Histone deacetylases (HDACs) interfere with DNA transcription in HIV-infected hosts, resulting in viral latency. Therefore, HDAC inhibitors can be used to activate viral transcription in latently infected cells, after which the virus can be eliminated through a shock-and-kill strategy. Here, a drug delivery system is developed to effectively deliver HDAC inhibitors to latent HIV-infected cells. Given that the efficacy of HDAC inhibitors is reduced under hypoxic conditions, oxygen-containing nanosomes are used as drug carriers. Oxygen-containing nanosomes can improve the efficiency of chemotherapy by delivering essential oxygen to cells. Additionally, their phospholipid bilayer structure makes them uniquely well-suited for drug delivery. In this study, a novel drug delivery system is developed by taking advantage of the oxygen carriers in these oxygen nanosomes, incorporating a multi-drug strategy consisting of HDAC inhibitors and PKA activators, and introducing CXCR4 binding peptides to specifically target CD4+ T cells. Oxygen nanosomes with enhanced targeting capability through the introduction of the CXCR4 binding peptide mitigate drug toxicity and slow down drug release. The observed changes in the expression of p24, a capsid protein of HIV, indirectly confirm that the proposed drug delivery system can effectively induce transcriptional reactivation of HIV in latent HIV-infected cells.
Collapse
Affiliation(s)
- Joohye Hong
- School of Integrative Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Yonghyun Choi
- School of Integrative Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
- Feynman Institute of Technology, Nanomedicine Corporation, Seoul, 06974, Republic of Korea
| | - Gahyun Lee
- School of Integrative Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Jiwon Kim
- School of Integrative Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Yeonwoo Jang
- School of Integrative Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Cheol-Hee Yoon
- Division of Chronic Viral Disease, Center for Emerging Virus Research, National Institute of Health, Cheongju, 28159, Republic of Korea
| | - Hyun Wook Seo
- Division of Chronic Viral Disease, Center for Emerging Virus Research, National Institute of Health, Cheongju, 28159, Republic of Korea
| | - In-Kyu Park
- Department of Biomedical Sciences, Chonnam National University Medical School, 160 Baekseo-ro, Gwangju, 58128, Republic of Korea
| | - Shin Hyuk Kang
- Departments of Plastic and Reconstructive Surgery, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, 06973, Republic of Korea
| | - Jonghoon Choi
- School of Integrative Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
- Feynman Institute of Technology, Nanomedicine Corporation, Seoul, 06974, Republic of Korea
| |
Collapse
|
21
|
Chvatal-Medina M, Lopez-Guzman C, Diaz FJ, Gallego S, Rugeles MT, Taborda NA. Molecular mechanisms by which the HIV-1 latent reservoir is established and therapeutic strategies for its elimination. Arch Virol 2023; 168:218. [PMID: 37530901 DOI: 10.1007/s00705-023-05800-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 04/12/2023] [Indexed: 08/03/2023]
Abstract
The human immunodeficiency virus type 1 (HIV-1) reservoir, composed of cells harboring the latent, integrated virus, is not eliminated by antiretroviral therapy. It therefore represents a significant barrier to curing the infection. The biology of HIV-1 reservoirs, the mechanisms of their persistence, and effective strategies for their eradication are not entirely understood. Here, we review the molecular mechanisms by which HIV-1 reservoirs develop, the cells and compartments where the latent virus resides, and advancements in curative therapeutic strategies. We first introduce statistics and relevant data on HIV-1 infection, aspects of pathogenesis, the role of antiretroviral therapy, and the general features of the latent HIV reservoir. Then, the article is built on three main pillars: The molecular mechanisms related to latency, the different strategies for targeting the reservoir to obtain a cure, and the current progress in immunotherapy to counteract said reservoirs.
Collapse
Affiliation(s)
- Mateo Chvatal-Medina
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Carolina Lopez-Guzman
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Francisco J Diaz
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Salomon Gallego
- Grupo de Investigaciones Biomédicas Uniremington, Programa de Medicina, Facultad de Ciencias de la Salud, Corporación Universitaria Remington, Medellín, Colombia
| | - Maria T Rugeles
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Natalia A Taborda
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia.
- Grupo de Investigaciones Biomédicas Uniremington, Programa de Medicina, Facultad de Ciencias de la Salud, Corporación Universitaria Remington, Medellín, Colombia.
- Universidad Cooperativa de Colombia, Campus Medellin, Envigado, Colombia.
| |
Collapse
|
22
|
Van Zandt AR, MacLean AG. Advances in HIV therapeutics and cure strategies: findings obtained through non-human primate studies. J Neurovirol 2023; 29:389-399. [PMID: 37635184 PMCID: PMC11636591 DOI: 10.1007/s13365-023-01162-y] [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: 02/24/2023] [Revised: 07/07/2023] [Accepted: 07/24/2023] [Indexed: 08/29/2023]
Abstract
Human immunodeficiency virus (HIV), the main contributor of the ongoing AIDS epidemic, remains one of the most challenging and complex viruses to target and eradicate due to frequent genome mutation and immune evasion. Despite the development of potent antiretroviral therapies, HIV remains an incurable infection as the virus persists in latent reservoirs throughout the body. To innovate a safe and effective cure strategy for HIV in humans, animal models are needed to better understand viral proliferation, disease progression, and therapeutic response. Nonhuman primates infected with simian immunodeficiency virus (SIV) provide an ideal model to study HIV infection and pathogenesis as they are closely related to humans genetically and express phenotypically similar immune systems. Examining the clinical outcomes of novel treatment strategies within nonhuman primates facilitates our understanding of HIV latency and advances the development of a true cure to HIV.
Collapse
Affiliation(s)
- Alison R Van Zandt
- Tulane National Primate Research Center, Covington, LA, USA
- Biomedical Sciences Training Program, Tulane University School of Medicine, New Orleans, LA, USA
| | - Andrew G MacLean
- Tulane National Primate Research Center, Covington, LA, USA.
- Biomedical Sciences Training Program, Tulane University School of Medicine, New Orleans, LA, USA.
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA.
- Tulane Brain Institute, New Orleans, LA, USA.
- Tulane Center for Aging, New Orleans, LA, USA.
| |
Collapse
|
23
|
Rashidfarrokhi A, Pillai R, Hao Y, Wu WL, Karadal-Ferrena B, Dimitriadoy SG, Cross M, Yeaton AH, Huang SM, Bhutkar AJ, Herrera A, Rajalingam S, Hayashi M, Huang KL, Bartnicki E, Zavitsanou AM, Wohlhieter CA, Leboeuf SE, Chen T, Loomis C, Mezzano V, Kulicke R, Davis FP, Stransky N, Smolen GA, Rudin CM, Moreira AL, Khanna KM, Pass HI, Wong KK, Koide S, Tsirigos A, Koralov SB, Papagiannakopoulos T. Tumor-intrinsic LKB1-LIF signaling axis establishes a myeloid niche to promote immune evasion and tumor growth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.15.549147. [PMID: 37502974 PMCID: PMC10370066 DOI: 10.1101/2023.07.15.549147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Tumor mutations can influence the surrounding microenvironment leading to suppression of anti-tumor immune responses and thereby contributing to tumor progression and failure of cancer therapies. Here we use genetically engineered lung cancer mouse models and patient samples to dissect how LKB1 mutations accelerate tumor growth by reshaping the immune microenvironment. Comprehensive immune profiling of LKB1 -mutant vs wildtype tumors revealed dramatic changes in myeloid cells, specifically enrichment of Arg1 + interstitial macrophages and SiglecF Hi neutrophils. We discovered a novel mechanism whereby autocrine LIF signaling in Lkb1 -mutant tumors drives tumorigenesis by reprogramming myeloid cells in the immune microenvironment. Inhibiting LIF signaling in Lkb1 -mutant tumors, via gene targeting or with a neutralizing antibody, resulted in a striking reduction in Arg1 + interstitial macrophages and SiglecF Hi neutrophils, expansion of antigen specific T cells, and inhibition of tumor progression. Thus, targeting LIF signaling provides a new therapeutic approach to reverse the immunosuppressive microenvironment of LKB1 -mutant tumors.
Collapse
|
24
|
Chaudhuri D, Lu T, Jacob B, Abraham S, Shankar P, Poss MA, Neamati N, Camarero JA. Lipidation of a bioactive cyclotide-based CXCR4 antagonist greatly improves its pharmacokinetic profile in vivo. J Control Release 2023; 359:26-32. [PMID: 37236320 PMCID: PMC10527528 DOI: 10.1016/j.jconrel.2023.05.026] [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: 03/08/2023] [Revised: 04/28/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023]
Abstract
The CXCR4 chemokine is a key molecular regulator of many biological functions controlling leukocyte functions during inflammation and immunity, and during embryonic development. Overexpression of CXCR4 is also associated with many types of cancer where its activation promotes angiogenesis, tumor growth/survival, and metastasis. In addition, CXCR4 is involved in HIV replication, working as a co-receptor for viral entry, making CXCR4 a very attractive target for developing novel therapeutic agents. Here we report the pharmacokinetic profile in rats of a potent CXCR4 antagonist cyclotide, MCo-CVX-5c, previously developed in our group that displayed a remarkable in vivo resistance to biological degradation in serum. This bioactive cyclotide, however, was rapidly eliminated through renal clearance. Several lipidated versions of cyclotide MCo-CVX-5c showed a significant increase in the half-life when compared to the unlipidated form. The palmitoylated version of cyclotide MCo-CVX-5c displayed similar CXCR4 antagonistic activity as the unlipidated cyclotide, while the cyclotide modified with octadecanedioic (18-oxo-octadecanoic) acid exhibited a remarkable decrease in its ability to antagonize CXCR4. Similar results were also obtained when tested for its ability to inhibit growth in two cancer cell lines and HIV infection in cells. These results show that the half-life of cyclotides can be improved by lipidation although it can also affect their biological activity depending on the lipid employed.
Collapse
Affiliation(s)
- Dipankar Chaudhuri
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Tiangong Lu
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109-2800, USA
| | - Binu Jacob
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Sojan Abraham
- Department of Biomedical Sciences, Center of Excellence in Infectious Disease, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX 79430, USA
| | - Premlata Shankar
- Department of Biomedical Sciences, Center of Excellence in Infectious Disease, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX 79430, USA
| | - Michael A Poss
- Bristol Myers Squibb Research and Development, P.O. Box 4000, Princeton, NJ 08543, USA
| | - Nouri Neamati
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109-2800, USA
| | - Julio A Camarero
- Department of Pharmacology and Pharmaceutical Sciences, Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA; Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA9033, USA.
| |
Collapse
|
25
|
Huang T, Cai J, Wang P, Zhou J, Zhang H, Wu Z, Zhao J, Huang Z, Deng K. Ponatinib Represses Latent HIV-1 by Inhibiting AKT-mTOR. Antimicrob Agents Chemother 2023; 67:e0006723. [PMID: 37212670 PMCID: PMC10269114 DOI: 10.1128/aac.00067-23] [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: 01/19/2023] [Accepted: 04/11/2023] [Indexed: 05/23/2023] Open
Abstract
Although antiretroviral therapy (ART) is effective in suppressing viral replication, it does not cure HIV-1 infection due to the presence of the viral latent reservoir. Rather than reactivating the latent viruses, the "block and lock" strategy aims to shift the viral reservoir to a deeper state of transcriptional silencing, thus preventing viral rebound after ART interruption. Although some latency-promoting agents (LPAs) have been reported, none of them have been approved for clinical application due to cytotoxicity and limited efficacy; therefore, it is important to search for novel and effective LPAs. Here, we report an FDA-approved drug, ponatinib, that can broadly repress latent HIV-1 reactivation in different cell models of HIV-1 latency and in primary CD4+ T cells from ART-suppressed individuals ex vivo. Ponatinib does not change the expression of activation or exhaustion markers on primary CD4+ T cells and does not induce severe cytotoxicity and cell dysfunction. Mechanistically, ponatinib suppresses proviral HIV-1 transcription by inhibiting the activation of the AKT-mTOR pathway, which subsequently blocks the interaction between key transcriptional factors and the HIV-1 long terminal repeat (LTR). In summary, we discovered a novel latency-promoting agent, ponatinib, which could have promising significance for future applications of HIV-1 functional cure.
Collapse
Affiliation(s)
- Ting Huang
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- School of Medicine, Sun Yat-sen University, Shenzhen, China
| | - Jinfeng Cai
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Peipei Wang
- Department of Infectious Diseases, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiasheng Zhou
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Haitao Zhang
- Department of Infectious Diseases, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Ziqi Wu
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jiacong Zhao
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhanlian Huang
- Department of Infectious Diseases, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kai Deng
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
26
|
Eddy J, Pham F, Chee R, Park E, Dapprich N, DeRuiter SL, Shen A. Intestinal endothelial cells increase HIV infection and latency in resting and activated CD4 + T cells, particularly affecting CCR6 + CD4 + T cells. Retrovirology 2023; 20:7. [PMID: 37202790 PMCID: PMC10197447 DOI: 10.1186/s12977-023-00621-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 04/29/2023] [Indexed: 05/20/2023] Open
Abstract
BACKGROUND With suppressive antiretroviral therapy, HIV infection is well-managed in most patients. However, eradication and cure are still beyond reach due to latent viral reservoirs in CD4 + T cells, particularly in lymphoid tissue environments including the gut associated lymphatic tissues. In HIV patients, there is extensive depletion of T helper cells, particularly T helper 17 cells from the intestinal mucosal area, and the gut is one of the largest viral reservoir sites. Endothelial cells line lymphatic and blood vessels and were found to promote HIV infection and latency in previous studies. In this study, we examined endothelial cells specific to the gut mucosal area-intestinal endothelial cells-for their impact on HIV infection and latency in T helper cells. RESULTS We found that intestinal endothelial cells dramatically increased productive and latent HIV infection in resting CD4 + T helper cells. In activated CD4 + T cells, endothelial cells enabled the formation of latent infection in addition to the increase of productive infection. Endothelial-cell-mediated HIV infection was more prominent in memory T cells than naïve T cells, and it involved the cytokine IL-6 but did not involve the co-stimulatory molecule CD2. The CCR6 + T helper 17 subpopulation was particularly susceptible to such endothelial-cell-promoted infection. CONCLUSION Endothelial cells, which are widely present in lymphoid tissues including the intestinal mucosal area and interact regularly with T cells physiologically, significantly increase HIV infection and latent reservoir formation in CD4 + T cells, particularly in CCR6 + T helper 17 cells. Our study highlighted the importance of endothelial cells and the lymphoid tissue environment in HIV pathology and persistence.
Collapse
Affiliation(s)
- Jessica Eddy
- Department of Biology, Calvin University, 3201 Burton St. SE, Grand Rapids, MI 49546 USA
| | - Fisher Pham
- Department of Biology, Calvin University, 3201 Burton St. SE, Grand Rapids, MI 49546 USA
| | - Rachel Chee
- Department of Biology, Calvin University, 3201 Burton St. SE, Grand Rapids, MI 49546 USA
| | - Esther Park
- Department of Biology, Calvin University, 3201 Burton St. SE, Grand Rapids, MI 49546 USA
| | - Nathan Dapprich
- Department of Biology, Calvin University, 3201 Burton St. SE, Grand Rapids, MI 49546 USA
| | - Stacy L. DeRuiter
- Department of Mathematics & Statistics, Calvin University, 3201 Burton St. SE, Grand Rapids, MI 49546 USA
| | - Anding Shen
- Department of Biology, Calvin University, 3201 Burton St. SE, Grand Rapids, MI 49546 USA
| |
Collapse
|
27
|
Zhang H, Cai J, Li C, Deng L, Zhu H, Huang T, Zhao J, Zhou J, Deng K, Hong Z, Xia J. Wogonin inhibits latent HIV-1 reactivation by downregulating histone crotonylation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 116:154855. [PMID: 37172478 DOI: 10.1016/j.phymed.2023.154855] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/27/2023] [Accepted: 05/02/2023] [Indexed: 05/15/2023]
Abstract
BACKGROUND Wogonin, a flavone isolated from Scutellaria baicalensis Georgi, is a commonly used phytochemical with anti-inflammatory and antitumor properties. However, the antiviral activity of wogonin against human immunodeficiency virus type 1 (HIV-1) has not been reported. PURPOSE The current study aimed to explore whether wogonin can suppress latent HIV-1 reactivation and the mechanism of wogonin in inhibiting proviral HIV-1 transcription. METHODS We assessed the effects of wogonin on HIV-1 reactivation using flow cytometry, cytotoxicity assay, quantitative PCR (qPCR), viral quality assurance (VQA), and western blot analysis. RESULTS Wogonin, a flavone isolated from S. baicalensis, significantly inhibited the reactivation of latent HIV-1 in cellular models and in primary CD4+ T cells from antiretroviral therapy (ART)-suppressed individuals ex vivo. Wogonin exhibited low cytotoxicity and long-lasting inhibition of HIV-1 transcription. Triptolide is a latency-promoting agent (LPA) that inhibits HIV-1 transcription and replication; wogonin had a stronger ability to inhibit HIV-1 latent reactivation than triptolide. Mechanistically, wogonin inhibited the reactivation of latent HIV-1 by inhibiting the expression of p300, a histone acetyltransferase, and decreasing the crotonylation of histone H3/H4 in the HIV-1 promoter region. CONCLUSION Our study found that wogonin is a novel LPA that can inhibit HIV-1 transcription by HIV-1 epigenetic silencing, which could bear promising significance for future applications of HIV-1 functional cure.
Collapse
Affiliation(s)
- Haitao Zhang
- Department of Infectious Diseases, Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China; Ward 1 of infection Department, Shenzhen Third People's Hospital, The Second Hospital Affiliated with the School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Jinfeng Cai
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| | - Chunna Li
- Department of Infectious Diseases, Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Lisi Deng
- Department of Infectious Diseases, Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Hongqiong Zhu
- Department of Infectious Diseases, Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Ting Huang
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jiacong Zhao
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jiasheng Zhou
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Kai Deng
- Institute of Human Virology, Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhongsi Hong
- Department of Infectious Diseases, Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China.
| | - Jinyu Xia
- Department of Infectious Diseases, Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China.
| |
Collapse
|
28
|
Deere JD, Merriam D, Leggat KM, Chang WLW, Méndez-Lagares G, Kieu H, Dutra J, Fontaine J, Lu W, Chin N, Chen C, Tran BCT, Salinas J, Miller CN, Deeks SG, Lifson JD, Engelman K, Magnani D, Reimann K, Stevenson M, Hartigan-O'Connor DJ. SIV clearance from neonatal macaques following transient CCR5 depletion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.01.533682. [PMID: 37205470 PMCID: PMC10187202 DOI: 10.1101/2023.05.01.533682] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Treatment of people with HIV (PWH) with antiretroviral therapy (ART) results in sustained suppression of viremia, but HIV persists indefinitely as integrated provirus in CD4-expressing cells. Intact persistent provirus, the "rebound competent viral reservoir" (RCVR), is the primary obstacle to achieving a cure. Most variants of HIV enter CD4 + T cells by binding to the chemokine receptor, CCR5. The RCVR has been successfully depleted only in a handful of PWH following cytotoxic chemotherapy and bone marrow transplantation from donors with a mutation in CCR5 . Here we show that long-term SIV remission and apparent cure can be achieved for infant macaques via targeted depletion of potential reservoir cells that express CCR5. Neonatal rhesus macaques were infected with virulent SIVmac251, then treated with ART beginning one week after infection, followed by treatment with either a CCR5/CD3-bispecific or a CD4-specific antibody, both of which depleted target cells and increased the rate of plasma viremia decrease. Upon subsequent cessation of ART, three of seven animals treated with CCR5/CD3-bispecific antibody rebounded quickly and two rebounded 3 or 6 months later. Remarkably, the other two animals remained aviremic and efforts to detect replication-competent virus were unsuccessful. Our results show that bispecific antibody treatment can achieve meaningful SIV reservoir depletion and suggest that functional HIV cure might be achievable for recently infected individuals having a restricted reservoir.
Collapse
|
29
|
Znaidia M, de Souza-Angelo Y, Létoffé S, Staropoli I, Grzelak L, Ghigo JM, Schwartz O, Casartelli N. Exposure to Secreted Bacterial Factors Promotes HIV-1 Replication in CD4 + T Cells. Microbiol Spectr 2023; 11:e0431322. [PMID: 36853052 PMCID: PMC10100953 DOI: 10.1128/spectrum.04313-22] [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: 10/21/2022] [Accepted: 02/08/2023] [Indexed: 03/01/2023] Open
Abstract
Microbial translocation is associated with systemic immune activation in HIV-1 disease. Circulating T cells can encounter microbial products in the bloodstream and lymph nodes, where viral replication takes place. The mechanisms by which bacteria contribute to HIV-associated pathogenesis are not completely deciphered. Here, we examined how bacteria may impact T cell function and viral replication. We established cocultures between a panel of live bacteria and uninfected or HIV-1-infected activated peripheral blood CD4-positive (CD4+) T cells. We show that some bacteria, such as Escherichia coli and Acinetobacter baumannii, sustain lymphocyte activation and enhance HIV-1 replication. Bacteria secrete soluble factors that upregulate CD25 and ICAM-1 cell surface levels and activate NF-κB nuclear translocation. Our data also demonstrate that CD25 polarizes at the virological synapse, suggesting a previously unappreciated role of CD25 during viral replication. These findings highlight how interactions between bacterial factors and T cells may promote T cell activation and HIV-1 replication. IMPORTANCE People living with HIV suffer from chronic immune activation despite effective antiretroviral therapy. Early after infection, HIV-1 actively replicates in the gut, causing the breakage of the intestinal epithelial barrier and microbial translocation. Microbial translocation and chronic immune activation have been proven linked; however, gaps in our knowledge on how bacteria contribute to the development of HIV-related diseases remain. Whether T cells in the peripheral blood react to bacterial products and how this affects viral replication are unknown. We show that some bacteria enriched in people living with HIV activate T cells and favor HIV-1's spread. Bacteria release soluble factors that cause the overexpression of cellular molecules related to their activation state. T cells overexpressing these molecules also replicate HIV-1 more efficiently. These results help us learn more about how HIV-1, T cells, and bacteria interact with each other, as well as the mechanisms behind chronic immune activation.
Collapse
Affiliation(s)
- M. Znaidia
- Institut Pasteur, Université Paris-Cité, UMR CNRS 3569, Virus and Immunity Unit, Paris, France
| | - Y. de Souza-Angelo
- Institut Pasteur, Université Paris-Cité, UMR CNRS 3569, Virus and Immunity Unit, Paris, France
| | - S. Létoffé
- Institut Pasteur, Université Paris-Cité, UMR CNRS 6047, Genetics of Biofilms Laboratory, Paris, France
| | - I. Staropoli
- Institut Pasteur, Université Paris-Cité, UMR CNRS 3569, Virus and Immunity Unit, Paris, France
| | - L. Grzelak
- Institut Pasteur, Université Paris-Cité, UMR CNRS 3569, Virus and Immunity Unit, Paris, France
| | - J. M. Ghigo
- Institut Pasteur, Université Paris-Cité, UMR CNRS 6047, Genetics of Biofilms Laboratory, Paris, France
| | - O. Schwartz
- Institut Pasteur, Université Paris-Cité, UMR CNRS 3569, Virus and Immunity Unit, Paris, France
- Vaccine Research Institute, Créteil, France
| | - N. Casartelli
- Institut Pasteur, Université Paris-Cité, UMR CNRS 3569, Virus and Immunity Unit, Paris, France
| |
Collapse
|
30
|
Chameettachal A, Mustafa F, Rizvi TA. Understanding Retroviral Life Cycle and its Genomic RNA Packaging. J Mol Biol 2023; 435:167924. [PMID: 36535429 DOI: 10.1016/j.jmb.2022.167924] [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: 10/17/2022] [Revised: 12/12/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
Members of the family Retroviridae are important animal and human pathogens. Being obligate parasites, their replication involves a series of steps during which the virus hijacks the cellular machinery. Additionally, many of the steps of retrovirus replication are unique among viruses, including reverse transcription, integration, and specific packaging of their genomic RNA (gRNA) as a dimer. Progress in retrovirology has helped identify several molecular mechanisms involved in each of these steps, but many are still unknown or remain controversial. This review summarizes our present understanding of the molecular mechanisms involved in various stages of retrovirus replication. Furthermore, it provides a comprehensive analysis of our current understanding of how different retroviruses package their gRNA into the assembling virions. RNA packaging in retroviruses holds a special interest because of the uniqueness of packaging a dimeric genome. Dimerization and packaging are highly regulated and interlinked events, critical for the virus to decide whether its unspliced RNA will be packaged as a "genome" or translated into proteins. Finally, some of the outstanding areas of exploration in the field of RNA packaging are highlighted, such as the role of epitranscriptomics, heterogeneity of transcript start sites, and the necessity of functional polyA sequences. An in-depth knowledge of mechanisms that interplay between viral and cellular factors during virus replication is critical in understanding not only the virus life cycle, but also its pathogenesis, and development of new antiretroviral compounds, vaccines, as well as retroviral-based vectors for human gene therapy.
Collapse
Affiliation(s)
- Akhil Chameettachal
- Department of Microbiology & Immunology, College of Medicine and Health Sciences (CMHS), United Arab Emirates University, Al Ain, United Arab Emirates. https://twitter.com/chameettachal
| | - Farah Mustafa
- Department of Biochemistry, College of Medicine and Health Sciences (CMHS), United Arab Emirates University, Al Ain, United Arab Emirates; Zayed bin Sultan Center for Health Sciences (ZCHS), United Arab Emirates University, Al Ain, United Arab Emirates.
| | - Tahir A Rizvi
- Department of Microbiology & Immunology, College of Medicine and Health Sciences (CMHS), United Arab Emirates University, Al Ain, United Arab Emirates; Zayed bin Sultan Center for Health Sciences (ZCHS), United Arab Emirates University, Al Ain, United Arab Emirates.
| |
Collapse
|
31
|
Mardi N, Haiaty S, Rahbarghazi R, Mobarak H, Milani M, Zarebkohan A, Nouri M. Exosomal transmission of viruses, a two-edged biological sword. Cell Commun Signal 2023; 21:19. [PMID: 36691072 PMCID: PMC9868521 DOI: 10.1186/s12964-022-01037-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 12/28/2022] [Indexed: 01/24/2023] Open
Abstract
As a common belief, most viruses can egress from the host cells as single particles and transmit to uninfected cells. Emerging data have revealed en bloc viral transmission as lipid bilayer-cloaked particles via extracellular vesicles especially exosomes (Exo). The supporting membrane can be originated from multivesicular bodies during intra-luminal vesicle formation and autophagic response. Exo are nano-sized particles, ranging from 40-200 nm, with the ability to harbor several types of signaling molecules from donor to acceptor cells in a paracrine manner, resulting in the modulation of specific signaling reactions in target cells. The phenomenon of Exo biogenesis consists of multiple and complex biological steps with the participation of diverse constituents and molecular pathways. Due to similarities between Exo biogenesis and virus replication and the existence of shared pathways, it is thought that viruses can hijack the Exo biogenesis machinery to spread and evade immune cells. To this end, Exo can transmit complete virions (as single units or aggregates), separate viral components, and naked genetic materials. The current review article aims to scrutinize challenges and opportunities related to the exosomal delivery of viruses in terms of viral infections and public health. Video Abstract.
Collapse
Affiliation(s)
- Narges Mardi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sanya Haiaty
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St., Tabriz, Iran
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Halimeh Mobarak
- Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St., Tabriz, Iran
| | - Morteza Milani
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Zarebkohan
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
32
|
Marichannegowda MH, Zemil M, Wieczorek L, Sanders-Buell E, Bose M, O'Sullivan AM, King D, Francisco L, Diaz-Mendez F, Setua S, Chomont N, Phanuphak N, Ananworanich J, Hsu D, Vasan S, Michael NL, Eller LA, Tovanabutra S, Tagaya Y, Robb ML, Polonis VR, Song H. Tracking coreceptor switch of the transmitted/founder HIV-1 identifies co-evolution of HIV-1 antigenicity, coreceptor usage and CD4 subset targeting. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.21.525033. [PMID: 36712089 PMCID: PMC9882280 DOI: 10.1101/2023.01.21.525033] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The CCR5 (R5) to CXCR4 (X4) coreceptor switch in natural HIV-1 infection is associated with faster progression to AIDS, but the underlying mechanisms remain unclear. The difficulty in capturing the earliest moment of coreceptor switch in vivo limits our understanding of this phenomenon. Here, by tracking the evolution of the transmitted/founder (T/F) HIV-1 in a prospective cohort of individuals at risk for HIV-1 infection identified very early in acute infection, we investigated this process with high resolution. The earliest X4 variants evolved from the R5 tropic T/F strains. Strong X4 usage can be conferred by a single mutation. The mutations responsible for coreceptor switch can confer escape to neutralization and drive X4 variants to replicate mainly in the central memory and naïve CD4+ T cells. We propose a novel concept to explain the co-evolution of virus antigenicity and entry tropism termed "escape by shifting". This concept posits that for viruses with receptor or coreceptor flexibility, entry tropism alteration represents a mechanism of immune evasion in vivo .
Collapse
|
33
|
Hölken JM, Teusch N. The Monocytic Cell Line THP-1 as a Validated and Robust Surrogate Model for Human Dendritic Cells. Int J Mol Sci 2023; 24:1452. [PMID: 36674966 PMCID: PMC9866978 DOI: 10.3390/ijms24021452] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/05/2023] [Accepted: 01/07/2023] [Indexed: 01/13/2023] Open
Abstract
We have implemented an improved, cost-effective, and highly reproducible protocol for a simple and rapid differentiation of the human leukemia monocytic cell line THP-1 into surrogates for immature dendritic cells (iDCs) or mature dendritic cells (mDCs). The successful differentiation of THP-1 cells into iDCs was determined by high numbers of cells expressing the DC activation markers CD54 (88%) and CD86 (61%), and the absence of the maturation marker CD83. The THP-1-derived mDCs are characterized by high numbers of cells expressing CD54 (99%), CD86 (73%), and the phagocytosis marker CD11b (49%) and, in contrast to THP-1-derived iDCs, CD83 (35%) and the migration marker CXCR4 (70%). Treatment of iDCs with sensitizers, such as NiSO4 and DNCB, led to high expression of CD54 (97%/98%; GMFI, 3.0/3.2-fold induction) and CD86 (64%/96%; GMFI, 4.3/3.2-fold induction) compared to undifferentiated sensitizer-treated THP-1 (CD54, 98%/98%; CD86, 55%/96%). Thus, our iDCs are highly suitable for toxicological studies identifying potential sensitizing or inflammatory compounds. Furthermore, the expression of CD11b, CD83, and CXCR4 on our iDC and mDC surrogates could allow studies investigating the molecular mechanisms of dendritic cell maturation, phagocytosis, migration, and their use as therapeutic targets in various disorders, such as sensitization, inflammation, and cancer.
Collapse
Affiliation(s)
| | - Nicole Teusch
- Institute of Pharmaceutical Biology and Biotechnology, Heinrich-Heine University Düsseldorf, Universitätsstraße 1, 40225 Düsseldorf, Germany
| |
Collapse
|
34
|
Hoerter A, Arnett E, Schlesinger LS, Pienaar E. Systems biology approaches to investigate the role of granulomas in TB-HIV coinfection. Front Immunol 2022; 13:1014515. [PMID: 36405707 PMCID: PMC9670175 DOI: 10.3389/fimmu.2022.1014515] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 09/20/2022] [Indexed: 09/29/2023] Open
Abstract
The risk of active tuberculosis disease is 15-21 times higher in those coinfected with human immunodeficiency virus-1 (HIV) compared to tuberculosis alone, and tuberculosis is the leading cause of death in HIV+ individuals. Mechanisms driving synergy between Mycobacterium tuberculosis (Mtb) and HIV during coinfection include: disruption of cytokine balances, impairment of innate and adaptive immune cell functionality, and Mtb-induced increase in HIV viral loads. Tuberculosis granulomas are the interface of host-pathogen interactions. Thus, granuloma-based research elucidating the role and relative impact of coinfection mechanisms within Mtb granulomas could inform cohesive treatments that target both pathogens simultaneously. We review known interactions between Mtb and HIV, and discuss how the structure, function and development of the granuloma microenvironment create a positive feedback loop favoring pathogen expansion and interaction. We also identify key outstanding questions and highlight how coupling computational modeling with in vitro and in vivo efforts could accelerate Mtb-HIV coinfection discoveries.
Collapse
Affiliation(s)
- Alexis Hoerter
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States
| | - Eusondia Arnett
- Host-Pathogen Interactions Program, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Larry S. Schlesinger
- Host-Pathogen Interactions Program, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Elsje Pienaar
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States
- Regenstrief Center for Healthcare Engineering, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
35
|
Targeting CXCR4 and CD47 Receptors: An Overview of New and Old Molecules for a Biological Personalized Anticancer Therapy. Int J Mol Sci 2022; 23:ijms232012499. [PMID: 36293358 PMCID: PMC9604048 DOI: 10.3390/ijms232012499] [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: 09/02/2022] [Revised: 10/14/2022] [Accepted: 10/14/2022] [Indexed: 11/26/2022] Open
Abstract
Biological therapy, with its multifaceted applications, has revolutionized the treatment of tumors, mainly due to its ability to exclusively target cancer cells and reduce the adverse effects on normal tissues. This review focuses on the therapies targeting the CXCR4 and CD47 receptors. We surveyed the results of early clinical trials testing compounds classified as nonpeptides, small peptides, CXCR4 antagonists or specific antibodies whose activity reduces or completely blocks the intracellular signaling pathways and cell proliferation. We then examined antibodies and fusion proteins against CD47, the receptor that acts as a “do not eat me” signal to phagocytes escaping immune surveillance. Despite these molecules being tested in early clinical trials, some drawbacks are emerging that impair their use in practice. Finally, we examined the ImmunoGenic Surrender mechanism that involves crosstalk and co-internalization of CXCR4 and CD47 upon engagement of CXCR4 by ligands or other molecules. The favorable effect of such compounds is dual as CD47 surface reduction impact on the immune response adds to the block of CXCR4 proliferative potential. These results suggest that a combination of different therapeutic approaches has more beneficial effects on patients’ survival and may pave the way for new accomplishments in personalized anticancer therapy.
Collapse
|
36
|
Chigorimbo-Murefu NTL, Potgieter M, Dzanibe S, Gabazana Z, Buri G, Chawla A, Nleya B, Olivier AJ, Harryparsad R, Calder B, Garnett S, Maziya L, Lewis DA, Jaspan H, Wilson D, Passmore JAS, Mulder N, Blackburn J, Bekker LG, Gray CM. A pilot study to show that asymptomatic sexually transmitted infections alter the foreskin epithelial proteome. Front Microbiol 2022; 13:928317. [PMID: 36325020 PMCID: PMC9618803 DOI: 10.3389/fmicb.2022.928317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
There is limited data on the role of asymptomatic STIs (aSTIs) on the risk of human immunodeficiency virus (HIV) acquisition in the male genital tract (MGT). The impact of foreskin removal on lowering HIV acquisition is well described, but molecular events leading to HIV acquisition are unclear. Here, in this pilot study, we show that asymptomatic urethral infection with Chlamydia trachomatis (CT) significantly impacts the foreskin proteome composition. We developed and optimized a shotgun liquid chromatography coupled tandem mass spectrometry (MS)-based proteomics approach and utilized this on foreskins collected at medical male circumcision (MMC) from 16 aSTI+ men and 10 age-matched STI- controls. We used a novel bioinformatic metaproteomic pipeline to detect differentially expressed (DE) proteins. Gene enrichment ontology analysis revealed proteins associated with inflammatory and immune activation function in both inner and outer foreskin from men with an aSTI. Neutrophil activation/degranulation and viral-evasion proteins were significantly enriched in foreskins from men with aSTI, whereas homotypic cell-cell adhesion proteins were enriched in foreskin tissue from men without an aSTI. Collectively, our data show that asymptomatic urethral sexually transmitted infections result in profound alterations in epithelial tissue that are associated with depletion of barrier integrity and immune activation.
Collapse
Affiliation(s)
- Nyaradzo T. L. Chigorimbo-Murefu
- Divisions of Medical Virology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Matthys Potgieter
- Division of Computational Biology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Division of Chemical and Systems Biology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Sonwabile Dzanibe
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Zikhona Gabazana
- Division of Computational Biology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Gershom Buri
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Aditya Chawla
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Bokani Nleya
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Abraham J. Olivier
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Rushil Harryparsad
- Divisions of Medical Virology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Bridget Calder
- Division of Chemical and Systems Biology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Shaun Garnett
- Division of Chemical and Systems Biology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Lungile Maziya
- Department of Medicine, Edendale Hospital, Pietermaritzburg, South Africa
| | - David A. Lewis
- Western Sydney Sexual Health Centre, Western Sydney Local Health District, Parramatta, NSW, Australia
- Westmead Clinical School and Sydney Institute for Infectious Diseases, University of Sydney, Sydney, NSW, Australia
| | - Heather Jaspan
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Seattle Children’s Research Institute, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
| | - Doug Wilson
- Department of Medicine, Edendale Hospital, Pietermaritzburg, South Africa
| | - Jo-Ann S. Passmore
- Divisions of Medical Virology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Nicola Mulder
- Division of Computational Biology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Jonathan Blackburn
- Division of Chemical and Systems Biology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | | | - Clive M. Gray
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Division of Molecular Biology and Human Genetics, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
37
|
Jasmer KJ, Muñoz Forti K, Woods LT, Cha S, Weisman GA. Therapeutic potential for P2Y 2 receptor antagonism. Purinergic Signal 2022:10.1007/s11302-022-09900-3. [PMID: 36219327 DOI: 10.1007/s11302-022-09900-3] [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: 08/26/2022] [Accepted: 09/22/2022] [Indexed: 10/17/2022] Open
Abstract
G protein-coupled receptors are the target of more than 30% of all FDA-approved drug therapies. Though the purinergic P2 receptors have been an attractive target for therapeutic intervention with successes such as the P2Y12 receptor antagonist, clopidogrel, P2Y2 receptor (P2Y2R) antagonism remains relatively unexplored as a therapeutic strategy. Due to a lack of selective antagonists to modify P2Y2R activity, studies using primarily genetic manipulation have revealed roles for P2Y2R in a multitude of diseases. These include inflammatory and autoimmune diseases, fibrotic diseases, renal diseases, cancer, and pathogenic infections. With the advent of AR-C118925, a selective and potent P2Y2R antagonist that became commercially available only a few years ago, new opportunities exist to gain a more robust understanding of P2Y2R function and assess therapeutic effects of P2Y2R antagonism. This review discusses the characteristics of P2Y2R that make it unique among P2 receptors, namely its involvement in five distinct signaling pathways including canonical Gαq protein signaling. We also discuss the effects of other P2Y2R antagonists and the pivotal development of AR-C118925. The remainder of this review concerns the mounting evidence implicating P2Y2Rs in disease pathogenesis, focusing on those studies that have evaluated AR-C118925 in pre-clinical disease models.
Collapse
Affiliation(s)
- Kimberly J Jasmer
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA.,Department of Biochemistry, University of Missouri, Columbia, MO, USA
| | - Kevin Muñoz Forti
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA.,Department of Biochemistry, University of Missouri, Columbia, MO, USA
| | - Lucas T Woods
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA.,Department of Biochemistry, University of Missouri, Columbia, MO, USA
| | - Seunghee Cha
- Department of Oral and Maxillofacial Diagnostic Sciences, Center for Orphaned Autoimmune Disorders, University of Florida College of Dentistry, Gainesville, FL, USA
| | - Gary A Weisman
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA. .,Department of Biochemistry, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
38
|
Mulherkar TH, Gómez DJ, Sandel G, Jain P. Co-Infection and Cancer: Host–Pathogen Interaction between Dendritic Cells and HIV-1, HTLV-1, and Other Oncogenic Viruses. Viruses 2022; 14:v14092037. [PMID: 36146843 PMCID: PMC9503663 DOI: 10.3390/v14092037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/08/2022] [Accepted: 09/12/2022] [Indexed: 11/16/2022] Open
Abstract
Dendritic cells (DCs) function as a link between innate and adaptive immune responses. Retroviruses HIV-1 and HTLV-1 modulate DCs to their advantage and utilize them to propagate infection. Coinfection of HTLV-1 and HIV-1 has implications for cancer malignancies. Both viruses initially infect DCs and propagate the infection to CD4+ T cells through cell-to-cell transmission using mechanisms including the formation of virologic synapses, viral biofilms, and conduits. These retroviruses are both neurotrophic with neurovirulence determinants. The neuropathogenesis of HIV-1 and HTLV-1 results in neurodegenerative diseases such as HIV-associated neurocognitive disorders (HAND) and HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). Infected DCs are known to traffic to the brain (CNS) and periphery (PNS, lymphatics) to induce neurodegeneration in HAND and HAM/TSP patients. Elevated levels of neuroinflammation have been correlated with cognitive decline and impairment of motor control performance. Current vaccinations and therapeutics for HIV-1 and HTLV-1 are assessed and can be applied to patients with HIV-1-associated cancers and adult T cell leukemia/lymphoma (ATL). These diseases caused by co-infections can result in both neurodegeneration and cancer. There are associations with cancer malignancies and HIV-1 and HTLV-1 as well as other human oncogenic viruses (EBV, HBV, HCV, HDV, and HPV). This review contains current knowledge on DC sensing of HIV-1 and HTLV-1 including DC-SIGN, Tat, Tax, and current viral therapies. An overview of DC interaction with oncogenic viruses including EBV, Hepatitis viruses, and HPV is also provided. Vaccines and therapeutics targeting host–pathogen interactions can provide a solution to co-infections, neurodegeneration, and cancer.
Collapse
Affiliation(s)
- Tania H. Mulherkar
- Department of Microbiology and Immunology, Drexel University, College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
| | - Daniel Joseph Gómez
- Department of Microbiology and Immunology, Drexel University, College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
- Department of Biological Sciences, California State University, 25800 Carlos Bee Blvd, Hayward, CA 94542, USA
| | - Grace Sandel
- Department of Microbiology and Immunology, Drexel University, College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
| | - Pooja Jain
- Department of Microbiology and Immunology, Drexel University, College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
- Correspondence:
| |
Collapse
|
39
|
Current Status of 68Ga-Pentixafor in Solid Tumours. Diagnostics (Basel) 2022; 12:diagnostics12092135. [PMID: 36140541 PMCID: PMC9497673 DOI: 10.3390/diagnostics12092135] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 08/26/2022] [Indexed: 12/24/2022] Open
Abstract
Chemokine receptor CXCR4 is overexpressed in neoplasms and its expression is related to tumour invasion, metastasis and aggressiveness. 68Ga-Pentixafor is used to non-invasively image the expression of CXCR4 in tumours and has been widely used in haematological malignancies. Recent evidence shows that therapies targeting CXCR4 can increase the chemosensitivity of the tumour as well as inhibit tumour metastasis and aggressiveness. 68Ga-Pentixafor has shown promise as an elegant radiotracer to aid in the selection of patients whose tumours demonstrate CXCR4 overexpression and who therefore may benefit from novel therapies targeting CXCR4. In addition, its therapeutic partners 177Lu- and 90Y-Pentixather have been investigated in the treatment of patients with advanced haematological malignancies, and initial studies have shown a good treatment response in metabolically active lesions. 68Ga-Pentixafor in solid tumours complements 18F-FDG by providing prognostic information and selecting patients who may benefit from therapies targeting CXCR4. This review summarises the available literature on the potential applications of 68Ga-Pentixafor in solid tumours.
Collapse
|
40
|
Evolution of Multiple Domains of the HIV-1 Envelope Glycoprotein during Coreceptor Switch with CCR5 Antagonist Therapy. Microbiol Spectr 2022; 10:e0072522. [PMID: 35727047 PMCID: PMC9431240 DOI: 10.1128/spectrum.00725-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
HIV-1 uses CD4 as a receptor and chemokine receptors CCR5 and/or CXCR4 as coreceptors. CCR5 antagonists are a class of antiretrovirals used to inhibit viral entry. Phenotypic prediction algorithms such as Geno2Pheno are used to assess CCR5 antagonist eligibility, for which the V3 region is screened. However, there exist scenarios where the algorithm cannot give an accurate prediction of tropism. The current study examined coreceptor shift of HIV-1 from CCR5-tropic strains to CXCR4-tropic or dual-tropic strains among five subjects in a clinical trial of the CCR5 antagonist vicriviroc. Envelope gene amplicon libraries were constructed and subjected to next-generation sequencing, as well as single-clone sequencing and functional analyses. Approximately half of the amplified full-length single envelope-encoding clones had no significant activity for infection of cells expressing high levels of CD4 and CCR5 or CXCR4. Functional analysis of 9 to 21 individual infectious clones at baseline and at the time of VF were used to construct phylogenetic trees and sequence alignments. These studies confirmed that specific residues and the overall charge of the V3 loop were the major determinants of coreceptor use, in addition to specific residues in other domains of the envelope protein in V1/V2, V4, C3, and C4 domains that may be important for coreceptor shift. These results provide greater insight into the viral genetic determinants of coreceptor shift. IMPORTANCE This study is novel in combining single-genome sequence analysis and next-generation sequencing to characterize HIV-1 quasispecies. The work highlights the importance of mutants present at frequencies of 1% or less in development of drug resistance. This study highlights a critical role of specific amino acid substitutions outside V3 that contribute to coreceptor shift as well as important roles of the V1/V2, V4, C3, and C4 domain residues.
Collapse
|
41
|
Aggarwal A, Stella AO, Walker G, Akerman A, Esneau C, Milogiannakis V, Burnett DL, McAllery S, Silva MR, Lu Y, Foster CSP, Brilot F, Pillay A, Van Hal S, Mathivanan V, Fichter C, Kindinger A, Hoppe AC, Munier ML, Amatayakul-Chantler S, Roth N, Coppola G, Symonds GP, Schofield P, Jackson J, Lenthall H, Henry JY, Mazigi O, Jäck HM, Davenport MP, Darley DR, Matthews GV, Khoury DS, Cromer D, Goodnow CC, Christ D, Robosa R, Starck DJ, Bartlett NW, Rawlinson WD, Kelleher AD, Turville SG. Platform for isolation and characterization of SARS-CoV-2 variants enables rapid characterization of Omicron in Australia. Nat Microbiol 2022; 7:896-908. [PMID: 35637329 PMCID: PMC9159941 DOI: 10.1038/s41564-022-01135-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 04/26/2022] [Indexed: 01/31/2023]
Abstract
Genetically distinct variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have emerged since the start of the COVID-19 pandemic. Over this period, we developed a rapid platform (R-20) for viral isolation and characterization using primary remnant diagnostic swabs. This, combined with quarantine testing and genomics surveillance, enabled the rapid isolation and characterization of all major SARS-CoV-2 variants circulating in Australia in 2021. Our platform facilitated viral variant isolation, rapid resolution of variant fitness using nasopharyngeal swabs and ranking of evasion of neutralizing antibodies. In late 2021, variant of concern Omicron (B1.1.529) emerged. Using our platform, we detected and characterized SARS-CoV-2 VOC Omicron. We show that Omicron effectively evades neutralization antibodies and has a different entry route that is TMPRSS2-independent. Our low-cost platform is available to all and can detect all variants of SARS-CoV-2 studied so far, with the main limitation being that our platform still requires appropriate biocontainment.
Collapse
Affiliation(s)
- Anupriya Aggarwal
- The Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - Alberto Ospina Stella
- The Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - Gregory Walker
- Serology and Virology Division (SAViD), NSW Health Pathology, Sydney, New South Wales, Australia
| | - Anouschka Akerman
- The Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - Camille Esneau
- Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales, Australia
| | - Vanessa Milogiannakis
- The Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - Deborah L Burnett
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Samantha McAllery
- The Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - Mariana Ruiz Silva
- The Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - Yonghui Lu
- Serology and Virology Division (SAViD), NSW Health Pathology, Sydney, New South Wales, Australia
| | - Charles S P Foster
- Serology and Virology Division (SAViD), NSW Health Pathology, Sydney, New South Wales, Australia
| | - Fabienne Brilot
- Brain Autoimmunity Group, Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, School of Medical Sciences, Sydney University of Sydney, Sydney Institute for Infectious Diseases, Sydney, New South Wales, Australia
| | - Aleha Pillay
- Brain Autoimmunity Group, Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, School of Medical Sciences, Sydney University of Sydney, Sydney Institute for Infectious Diseases, Sydney, New South Wales, Australia
| | | | - Vennila Mathivanan
- The Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - Christina Fichter
- The Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - Andrea Kindinger
- The Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - Alexandra Carey Hoppe
- The Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - Mee Ling Munier
- The Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - Supavadee Amatayakul-Chantler
- Department of Bioanalytical Sciences, Plasma Product Development, Research and Development, CSL Behring, Broadmeadows, Melbourne, Victoria, Australia
| | - Nathan Roth
- Department of Bioanalytical Sciences, Plasma Product Development, Research and Development, CSL Behring AG, Bern, Switzerland
| | - Germano Coppola
- Department of Bioanalytical Sciences, Plasma Product Development, Research and Development, CSL Behring, Broadmeadows, Melbourne, Victoria, Australia
| | | | - Peter Schofield
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Jennifer Jackson
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Helen Lenthall
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Jake Y Henry
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Ohan Mazigi
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | | | - Miles P Davenport
- The Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - David R Darley
- St Vincent's Hospital, Sydney, New South Wales, Australia
| | - Gail V Matthews
- The Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
- St Vincent's Hospital, Sydney, New South Wales, Australia
| | - David S Khoury
- The Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - Deborah Cromer
- The Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
| | | | - Daniel Christ
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Roselle Robosa
- Molecular Diagnostic Medicine Laboratory, Sydpath, St Vincent's Hospital, Sydney, New South Wales, Australia
| | - Damien J Starck
- Molecular Diagnostic Medicine Laboratory, Sydpath, St Vincent's Hospital, Sydney, New South Wales, Australia
| | - Nathan W Bartlett
- Serology and Virology Division (SAViD), NSW Health Pathology, Sydney, New South Wales, Australia
| | - William D Rawlinson
- Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales, Australia
| | - Anthony D Kelleher
- The Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
- St Vincent's Hospital, Sydney, New South Wales, Australia
| | - Stuart G Turville
- The Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia.
| |
Collapse
|
42
|
Heterogeneity of Latency Establishment in the Different Human CD4
+
T Cell Subsets Stimulated with IL-15. J Virol 2022; 96:e0037922. [PMID: 35499323 PMCID: PMC9131862 DOI: 10.1128/jvi.00379-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
HIV integrates into the host genome, creating a viral reservoir of latently infected cells that persists despite effective antiretroviral treatment. CD4-positive (CD4+) T cells are the main contributors to the HIV reservoir. CD4+ T cells are a heterogeneous population, and the mechanisms of latency establishment in the different subsets, as well as their contribution to the reservoir, are still unclear. In this study, we analyzed HIV latency establishment in different CD4+ T cell subsets stimulated with interleukin 15 (IL-15), a cytokine that increases both susceptibility to infection and reactivation from latency. Using a dual-reporter virus that allows discrimination between latent and productive infection at the single-cell level, we found that IL-15-treated primary human CD4+ T naive and CD4+ T stem cell memory (TSCM) cells are less susceptible to HIV infection than CD4+ central memory (TCM), effector memory (TEM), and transitional memory (TTM) cells but are also more likely to harbor transcriptionally silent provirus. The propensity of these subsets to harbor latent provirus compared to the more differentiated memory subsets was independent of differential expression of pTEFb components. Microscopy analysis of NF-κB suggested that CD4+ T naive cells express smaller amounts of nuclear NF-κB than the other subsets, partially explaining the inefficient long terminal repeat (LTR)-driven transcription. On the other hand, CD4+ TSCM cells display similar levels of nuclear NF-κB to CD4+ TCM, CD4+ TEM, and CD4+ TTM cells, indicating the availability of transcription initiation and elongation factors is not solely responsible for the inefficient HIV gene expression in the CD4+ TSCM subset. IMPORTANCE The formation of a latent reservoir is the main barrier to HIV cure. Here, we investigated how HIV latency is established in different CD4+ T cell subsets in the presence of IL-15, a cytokine that has been shown to efficiently induce latency reversal. We observed that, even in the presence of IL-15, the less differentiated subsets display lower levels of productive HIV infection than the more differentiated subsets. These differences were not related to different expression of pTEFb, and modest differences in NF-κB were observed for CD4+ T naive cells only, implying the involvement of other mechanisms. Understanding the molecular basis of latency establishment in different CD4+ T cell subsets might be important for tailoring specific strategies to reactivate HIV transcription in all the CD4+ T subsets that compose the latent reservoir.
Collapse
|
43
|
Kwon D, Takata K, Zhang Z, Chong L, Fraser B, Zeisler J, Miyata-Takata T, Merkens H, Rousseau J, Aoki T, Kuo HT, Tan R, Zhang C, Lau J, Villa D, Uribe CF, Lin KS, Steidl C, Benard F. Targeting Refractory Mantle Cell Lymphoma for Imaging and Therapy Using C-X-C Chemokine Receptor Type 4 Radioligands. Clin Cancer Res 2022; 28:1628-1639. [PMID: 35078860 PMCID: PMC9365342 DOI: 10.1158/1078-0432.ccr-21-3284] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/30/2021] [Accepted: 01/21/2022] [Indexed: 01/07/2023]
Abstract
PURPOSE Mantle cell lymphoma (MCL) is associated with poor survival. The purpose of this study was to assess whether the C-X-C chemokine receptor type 4 (CXCR4) is a useful target for imaging and radioligand therapy of MCL, using a novel pair of radioligands, [68Ga]Ga and [177Lu]Lu-BL02. EXPERIMENTAL DESIGN We performed a retrospective analysis of 146 patients with MCL to evaluate CXCR4 expression and its correlation with outcomes. Guided by in silico methods, we designed BL02, a new radioligand labelled with 68Ga or 177Lu for PET imaging and therapy, respectively. We performed imaging and biodistribution studies in xenograft models with varying CXCR4 expression. We evaluated [177Lu]Lu-BL02 in MCL models, and evaluated its potential for therapy in Z138 MCL xenografts. RESULTS Phosphorylated and nonphosphorylated CXCR4 expression were correlated with poor survival in patients with MCL and characterized by unique underlying molecular signatures. [68Ga]Ga-BL02 uptake correlated with CXCR4 expression, and localized lesions in a metastatic xenograft model. [177Lu]Lu-BL02 showed high uptake in MCL xenografts. Therapy studies with a single dose in the Z138 model showed tumor regression and improved survival compared with a control group. Upon regrowth, the treated mice experienced concurrent metastasis alongside localized xenograft regrowth, and recurrent lesions showed enhanced CXCR4 signaling. CONCLUSIONS CXCR4 is an independent factor of poor prognosis for MCL and a promising target for imaging and radioligand therapy. [68Ga]Ga-BL02 showed high contrast to visualize CXCR4-expressing xenografts for PET imaging and [177Lu]Lu-BL02 induced rapid tumor regression in a preclinical model of MCL.
Collapse
Affiliation(s)
- Daniel Kwon
- Molecular Oncology, British Columbia Cancer, Vancouver, British Columbia, Canada
| | - Katsuyoshi Takata
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, British Columbia, Canada.,Division of Molecular and Cellular Pathology, Niigata University Graduate School of Medical and Dental Sciences, Asahimachi-dori, Chuo-ku, Niigata, Japan
| | - Zhengxing Zhang
- Molecular Oncology, British Columbia Cancer, Vancouver, British Columbia, Canada
| | - Lauren Chong
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, British Columbia, Canada
| | - Bryan Fraser
- Molecular Oncology, British Columbia Cancer, Vancouver, British Columbia, Canada
| | - Jutta Zeisler
- Molecular Oncology, British Columbia Cancer, Vancouver, British Columbia, Canada
| | - Tomoko Miyata-Takata
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, British Columbia, Canada
| | - Helen Merkens
- Molecular Oncology, British Columbia Cancer, Vancouver, British Columbia, Canada
| | - Julie Rousseau
- Molecular Oncology, British Columbia Cancer, Vancouver, British Columbia, Canada
| | - Tomohiro Aoki
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, British Columbia, Canada
| | - Hsiou-Ting Kuo
- Molecular Oncology, British Columbia Cancer, Vancouver, British Columbia, Canada
| | - Ruiyan Tan
- Molecular Oncology, British Columbia Cancer, Vancouver, British Columbia, Canada
| | - Chengcheng Zhang
- Molecular Oncology, British Columbia Cancer, Vancouver, British Columbia, Canada
| | - Joseph Lau
- Molecular Oncology, British Columbia Cancer, Vancouver, British Columbia, Canada
| | - Diego Villa
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, British Columbia, Canada
| | - Carlos F. Uribe
- Molecular Oncology, British Columbia Cancer, Vancouver, British Columbia, Canada.,Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kuo-Shyan Lin
- Molecular Oncology, British Columbia Cancer, Vancouver, British Columbia, Canada.,Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Christian Steidl
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Francois Benard
- Molecular Oncology, British Columbia Cancer, Vancouver, British Columbia, Canada.,Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada.,Corresponding Author: Francois Benard, Department of Functional Imaging, BC Cancer Agency, 600 West 10th Avenue, Vancouver, V5Z 4E6, Canada. E-mail:
| |
Collapse
|
44
|
Alsayed RKME, Khan AQ, Ahmad F, Ansari AW, Alam MA, Buddenkotte J, Steinhoff M, Uddin S, Ahmad A. Epigenetic Regulation of CXCR4 Signaling in Cancer Pathogenesis and Progression. Semin Cancer Biol 2022; 86:697-708. [PMID: 35346802 DOI: 10.1016/j.semcancer.2022.03.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/18/2022] [Accepted: 03/21/2022] [Indexed: 02/08/2023]
Abstract
Signaling involving chemokine receptor CXCR4 and its ligand SDF-1/CXL12 has been investigated for many years for its possible role in cancer progression and pathogenesis. Evidence emerging from clinical studies in recent years has further established diagnostic as well as prognostic importance of CXCR4 signaling. CXCR4 and SDF-1 are routinely reported to be elevated in tumors, distant metastases, which correlates with poor survival of patients. These findings have kindled interest in the mechanisms that regulate CXCR4/SDF-1 expression. Of note, there is a particular interest in the epigenetic regulation of CXCR4 signaling that may be responsible for upregulated CXCR4 in primary as well as metastatic cancers. This review first lists the clinical evidence supporting CXCR4 signaling as putative cancer diagnostic and/or prognostic biomarker, followed by a discussion on reported epigenetic mechanisms that affect CXCR4 expression. These mechanisms include regulation by non-coding RNAs, such as, microRNAs, long non-coding RNAs and circular RNAs. Additionally, we also discuss the regulation of CXCR4 expression through methylation and acetylation. Better understanding and appreciation of epigenetic regulation of CXCR4 signaling can invariably lead to identification of novel therapeutic targets as well as therapies to regulate this oncogenic signaling.
Collapse
Affiliation(s)
- Reem Khaled M E Alsayed
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, 3050, Qatar
| | - Abdul Q Khan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, 3050, Qatar
| | - Fareed Ahmad
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, 3050, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, 3050, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha, 3050, Qatar
| | - Abdul Wahid Ansari
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, 3050, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, 3050, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha, 3050, Qatar
| | - Majid Ali Alam
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, 3050, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, 3050, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha, 3050, Qatar
| | - Jorg Buddenkotte
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, 3050, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, 3050, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha, 3050, Qatar
| | - Martin Steinhoff
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, 3050, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, 3050, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha, 3050, Qatar; Weill Cornell Medicine-Qatar, Medical School, Doha, 24144, Qatar; Dept. of Dermatology, Weill Cornell Medicine, New York, 10065, NY, USA
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, 3050, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, 3050, Qatar; Laboratory Animal Research Center, Qatar University, Doha, 2713, Qatar
| | - Aamir Ahmad
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, 3050, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, 3050, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha, 3050, Qatar.
| |
Collapse
|
45
|
Russell AE, Liao Z, Tkach M, Tarwater PM, Ostrowski M, Théry C, Witwer KW. Cigarette smoke-induced extracellular vesicles from dendritic cells alter T-cell activation and HIV replication. Toxicol Lett 2022; 360:33-43. [PMID: 35181468 PMCID: PMC9014967 DOI: 10.1016/j.toxlet.2022.02.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 01/26/2022] [Accepted: 02/12/2022] [Indexed: 10/19/2022]
Abstract
Despite decreased rates of tobacco smoking in many areas, cigarette smoking remains a major contributor to many health problems. Cigarette smoking can reduce immune system functioning while concurrently increasing inflammation. Dendritic cells in the lung exposed to cigarette smoke become stimulated and go on to activate T-cells. Extracellular vesicles (EVs) are nano-sized particles released by cells. They participate in intercellular communication by transferring functional proteins and nucleic acids to recipient cells and have been implicated in immune responses. For example, they can display MHC-peptide complexes to activate T-cells. In the current study, we sought to understand the role of cigarette smoke extract (CSE) on dendritic cell-derived EVs and their capacity to activate and differentiate T-cells. Primary human dendritic cells (iDCs) were exposed to CSE and EVs were separated and characterized. We exposed autologous primary CD4 + T-cells to iDC-EVs and observed T helper cell populations skewing towards Th1 and Th17 phenotypes. As HIV + individuals are disproportionately likely to be current smokers, we also examined the effects of iDC-EVs on acutely infected T-cells as well as on a cell model of HIV latency (ACH-2). We found that in most cases, iDC-CSE EVs tended to reduce p24 release from the acutely infected primary T-cells, albeit with great variability. We did not observe large effects of iDC-EVs or direct CSE exposure on p24 release from the ACH-2 cell line. Together, these data suggest that iDC-CSE EVs have the capacity to modulate the immune responses, in part by pushing T-cells towards Th1 and Th17 phenotypes.
Collapse
Affiliation(s)
- Ashley E Russell
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Biology, School of Science, Penn State Erie, The Behrend College, Erie, PA, United States.
| | - Zhaohao Liao
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Mercedes Tkach
- INSERM U932, Institut Curie Centre de Recherche, PSL Research University, Paris, France
| | - Patrick M Tarwater
- Department of Epidemiology, The Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Matias Ostrowski
- Instituto INBIRS, Universidad de Buenos Aires-CONICET, Buenos Aires, Argentina
| | - Clotilde Théry
- INSERM U932, Institut Curie Centre de Recherche, PSL Research University, Paris, France
| | - Kenneth W Witwer
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; The Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease.
| |
Collapse
|
46
|
Pedersen JG, Egedal JH, Packard TA, Thavachelvam K, Xie G, van der Sluis RM, Greene WC, Roan NR, Jakobsen MR. Cell-Extrinsic Priming Increases Permissiveness of CD4+ T Cells to Human Immunodeficiency Virus Infection by Increasing C-C Chemokine Receptor Type 5 Co-receptor Expression and Cellular Activation Status. Front Microbiol 2021; 12:763030. [PMID: 34899645 PMCID: PMC8661899 DOI: 10.3389/fmicb.2021.763030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/05/2021] [Indexed: 11/13/2022] Open
Abstract
The chemokine receptor CCR5 is expressed on multiple cell types, including macrophages, dendritic cells, and T cells, and is the major co-receptor used during HIV transmission. Using a standard αCD3/CD28 in vitro stimulation protocol to render CD4+ T cells from PBMCs permissive to HIV infection, we discovered that the percentage of CCR5+ T cells was significantly elevated in CD4+ T cells when stimulated in the presence of peripheral blood mononuclear cells (PBMCs) as compared to when stimulated as purified CD4+ T cells. This indicated that environmental factors unique to the T-PBMCs condition affect surface expression of CCR5 on CD4+ T cells. Conditioned media from αCD3/CD28-stimulated PBMCs induced CCR5 expression in cultures of unstimulated cells. Cytokine profile analysis of these media suggests IL-12 as an inducer of CCR5 expression. Mass cytometric analysis showed that stimulated T-PBMCs exhibited a uniquely activated phenotype compared to T-Pure. In line with increased CCR5 expression and activation status in stimulated T-PBMCs, CD4+ T cells from these cultures were more susceptible to infection by CCR5-tropic HIV-1 as compared with T-Pure cells. These results suggest that in order to increase ex vivo infection rates of blood-derived CD4+ T cells, standard stimulation protocols used in HIV infection studies should implement T-PBMCs or purified CD4+ T cells should be supplemented with IL-12.
Collapse
Affiliation(s)
| | - Johanne H Egedal
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Gladstone Institute of Virology, San Francisco, CA, United States
| | - Thomas A Packard
- Gladstone Institute of Virology, San Francisco, CA, United States
| | | | - Guorui Xie
- Gladstone Institute of Virology, San Francisco, CA, United States.,Department of Urology, University of California, San Francisco, San Francisco, CA, United States
| | - Renée Marije van der Sluis
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Aarhus Institute of Advanced Studies, Aarhus University, Aarhus, Denmark
| | - Warner C Greene
- Gladstone Institute of Virology, San Francisco, CA, United States
| | - Nadia R Roan
- Gladstone Institute of Virology, San Francisco, CA, United States.,Department of Urology, University of California, San Francisco, San Francisco, CA, United States
| | | |
Collapse
|
47
|
Arumugam T, Ramphal U, Adimulam T, Chinniah R, Ramsuran V. Deciphering DNA Methylation in HIV Infection. Front Immunol 2021; 12:795121. [PMID: 34925380 PMCID: PMC8674454 DOI: 10.3389/fimmu.2021.795121] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 11/17/2021] [Indexed: 12/13/2022] Open
Abstract
With approximately 38 million people living with HIV/AIDS globally, and a further 1.5 million new global infections per year, it is imperative that we advance our understanding of all factors contributing to HIV infection. While most studies have focused on the influence of host genetic factors on HIV pathogenesis, epigenetic factors are gaining attention. Epigenetics involves alterations in gene expression without altering the DNA sequence. DNA methylation is a critical epigenetic mechanism that influences both viral and host factors. This review has five focal points, which examines (i) fluctuations in the expression of methylation modifying factors upon HIV infection (ii) the effect of DNA methylation on HIV viral genes and (iii) host genome (iv) inferences from other infectious and non-communicable diseases, we provide a list of HIV-associated host genes that are regulated by methylation in other disease models (v) the potential of DNA methylation as an epi-therapeutic strategy and biomarker. DNA methylation has also been shown to serve as a robust therapeutic strategy and precision medicine biomarker against diseases such as cancer and autoimmune conditions. Despite new drugs being discovered for HIV, drug resistance is a problem in high disease burden settings such as Sub-Saharan Africa. Furthermore, genetic therapies that are under investigation are irreversible and may have off target effects. Alternative therapies that are nongenetic are essential. In this review, we discuss the potential role of DNA methylation as a novel therapeutic intervention against HIV.
Collapse
Affiliation(s)
- Thilona Arumugam
- School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Upasana Ramphal
- School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, South Africa
- KwaZulu-Natal Research Innovation and Sequencing Platform (KRISP), School of Laboratory Medicine & Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Theolan Adimulam
- School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Romona Chinniah
- School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Veron Ramsuran
- School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, South Africa
- KwaZulu-Natal Research Innovation and Sequencing Platform (KRISP), School of Laboratory Medicine & Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
48
|
Cuesta-Mateos C, Terrón F, Herling M. CCR7 in Blood Cancers - Review of Its Pathophysiological Roles and the Potential as a Therapeutic Target. Front Oncol 2021; 11:736758. [PMID: 34778050 PMCID: PMC8589249 DOI: 10.3389/fonc.2021.736758] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 10/12/2021] [Indexed: 11/23/2022] Open
Abstract
According to the classical paradigm, CCR7 is a homing chemokine receptor that grants normal lymphocytes access to secondary lymphoid tissues such as lymph nodes or spleen. As such, in most lymphoproliferative disorders, CCR7 expression correlates with nodal or spleen involvement. Nonetheless, recent evidence suggests that CCR7 is more than a facilitator of lymphatic spread of tumor cells. Here, we review published data to catalogue CCR7 expression across blood cancers and appraise which classical and novel roles are attributed to this receptor in the pathogenesis of specific hematologic neoplasms. We outline why novel therapeutic strategies targeting CCR7 might provide clinical benefits to patients with CCR7-positive hematopoietic tumors.
Collapse
Affiliation(s)
- Carlos Cuesta-Mateos
- Immunology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria- Instituto la Princesa (IIS-IP), Madrid, Spain.,Immunological and Medicinal Products (IMMED S.L.), Madrid, Spain.,Catapult Therapeutics BV, Lelystad, Netherlands
| | - Fernando Terrón
- Immunological and Medicinal Products (IMMED S.L.), Madrid, Spain.,Catapult Therapeutics BV, Lelystad, Netherlands
| | - Marco Herling
- Clinic of Hematology and Cellular Therapy, University of Leipzig, Leipzig, Germany
| |
Collapse
|
49
|
Revskij D, Haubold S, Plinski C, Viergutz T, Tuchscherer A, Kröger-Koch C, Albrecht E, Günther J, Tröscher A, Hammon HM, Schuberth HJ, Mielenz M. Cellular detection of the chemokine receptor CXCR4 in bovine mammary glands and its distribution and regulation on bovine leukocytes. J Dairy Sci 2021; 105:866-876. [PMID: 34763920 DOI: 10.3168/jds.2021-20799] [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: 05/28/2021] [Accepted: 09/17/2021] [Indexed: 11/19/2022]
Abstract
Mastitis has a high incidence in dairy cows. Experimental infection with Escherichia coli increased the number of leukocytes in milk and the gene expression of the chemokine receptor CXCR4 in mammary gland tissues. A link between CXCR4 expression and lipopolysaccharide sensing was demonstrated in other species using in vitro models. The receptor that binds the chemokine stomal cell-derived factor 1 might be associated with the inflammatory response in bovine mammary glands. However, studies in cows are rare, and data on the localization of CXCR4 in bovine mammary glands and its distribution in bovine leukocytes are lacking. Fatty acids (FA) affect the inflammatory response. In human peripheral blood monocytes, exposure to conjugated linoleic acids (CLA) decreases the expression of CXCR4, leading to a decreased inflammatory response in these cells. In this study, we analyzed the expression of CXCR4 in the mammary glands of dairy cows by immunohistochemistry (n = 5) and laser capture microdissection followed by qualitative PCR (n = 3). We characterized the surface expression of CXCR4 on bovine leukocytes, including monocyte subpopulations, first by flow cytometry (n = 5) and then confirmed these results by Western blotting (n = 3). Rumen fistulated dairy cows (n = 4; 126 ± 4 d in milk) were fitted with abomasal infusion tubes, arranged in a 4 × 4 Latin square design, and supplemented for 6 wk twice daily with rising doses of FA followed by a 3-wk washout period. Then, CXCR4 expression on leukocytes was analyzed. The cows received a corn-based diet and were supplemented with coconut oil delivering medium-chain FA (38 g/d), linseed-safflower oil mix delivering n-3 FA (EFA, 39 g of linseed oil and 2 g of safflower oil per day), Lutalin (cis-9,trans-11 and trans-10,cis-12 CLA, 5 g/d; BASF), and EFA + CLA. In the bovine mammary gland, the epithelial cells of the lactiferous duct, but not alveolar epithelial cells, showed clear CXCR4 protein and mRNA signals. Among the leukocyte subsets, monocytes displayed the highest percentage of CXCR4-positive cells (87%), whereas circulating neutrophils showed almost no CXCR4 surface expression (3%) but stored the receptor intracellularly. The percentage of CXCR4-positive leukocytes was not affected by the different FA supplements, but FA supplementation reduced the receptor abundance per cell (40% on average). In conclusion, CXCR4 was clearly detected in the lactiferous duct cells of the mammary gland but not in the alveolar epithelial cells. Compared with other leukocytes, bovine monocytes showed the highest signal intensity of CXCR4 on their surface, whereas granulocytes stored CXCR4 intracellularly. Supplementation with all the FA reduced the surface expression of CXCR4 per leukocyte and could therefore potentially affect the inflammatory status associated with the surface expression of CXCR4. The importance of our observations should be verified in cows with mastitis in the future.
Collapse
Affiliation(s)
- Denis Revskij
- Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - Susanne Haubold
- Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - Christian Plinski
- Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - Torsten Viergutz
- Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - Armin Tuchscherer
- Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - Claudia Kröger-Koch
- Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - Elke Albrecht
- Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - Juliane Günther
- Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | | | - Harald M Hammon
- Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - Hans-Joachim Schuberth
- Institute of Immunology, University of Veterinary Medicine, Foundation, Buenteweg 2, 30559 Hannover, Germany
| | - Manfred Mielenz
- Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany.
| |
Collapse
|
50
|
Zhang Q, Friedman PA. Receptor-Loaded Virion Endangers GPCR Signaling: Mechanistic Exploration of SARS-CoV-2 Infections and Pharmacological Implications. Int J Mol Sci 2021; 22:ijms222010963. [PMID: 34681624 PMCID: PMC8535999 DOI: 10.3390/ijms222010963] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/07/2021] [Accepted: 10/09/2021] [Indexed: 01/14/2023] Open
Abstract
SARS-CoV-2 exploits the respiratory tract epithelium including lungs as the primary entry point and reaches other organs through hematogenous expansion, consequently causing multiorgan injury. Viral E protein interacts with cell junction-associated proteins PALS1 or ZO-1 to gain massive penetration by disrupting the inter-epithelial barrier. Conversely, receptor-mediated viral invasion ensures limited but targeted infections in multiple organs. The ACE2 receptor represents the major virion loading site by virtue of its wide tissue distribution as demonstrated in highly susceptible lung, intestine, and kidney. In brain, NRP1 mediates viral endocytosis in a similar manner to ACE2. Prominently, PDZ interaction involves the entire viral loading process either outside or inside the host cells, whereas E, ACE2, and NRP1 provide the PDZ binding motif required for interacting with PDZ domain-containing proteins PALS1, ZO-1, and NHERF1, respectively. Hijacking NHERF1 and β-arrestin by virion loading may impair specific sensory GPCR signalosome assembling and cause disordered cellular responses such as loss of smell and taste. PDZ interaction enhances SARS-CoV-2 invasion by supporting viral receptor membrane residence, implying that the disruption of these interactions could diminish SARS-CoV-2 infections and be another therapeutic strategy against COVID-19 along with antibody therapy. GPCR-targeted drugs are likely to alleviate pathogenic symptoms-associated with SARS-CoV-2 infection.
Collapse
|