1
|
Chernov AS, Telegin GB, Minakov AN, Kazakov VA, Rodionov MV, Palikov VA, Kudriaeva AA, Belogurov AA. Synthetic Amphipathic Helical Peptide L-37pA Ameliorates the Development of Acute Respiratory Distress Syndrome (ARDS) and ARDS-Induced Pulmonary Fibrosis in Mice. Int J Mol Sci 2024; 25:8384. [PMID: 39125954 PMCID: PMC11312864 DOI: 10.3390/ijms25158384] [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: 06/20/2024] [Revised: 07/16/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
In this study, we evaluated the ability of the synthetic amphipathic helical peptide (SAHP), L-37pA, which mediates pathogen recognition and innate immune responses, to treat acute respiratory distress syndrome (ARDS) accompanied by diffuse alveolar damage (DAD) and chronic pulmonary fibrosis (PF). For the modeling of ARDS/DAD, male ICR mice were used. Intrabronchial instillation (IB) of 200 µL of inflammatory agents was performed by an intravenous catheter 20 G into the left lung lobe only, leaving the right lobe unaffected. Intravenous injections (IVs) of L-37pA, dexamethasone (DEX) and physiological saline (saline) were used as therapies for ARDS/DAD. L37pA inhibited the circulating levels of inflammatory cytokines, such as IL-8, TNFα, IL1α, IL4, IL5, IL6, IL9 and IL10, by 75-95%. In all cases, the computed tomography (CT) data indicate that L-37pA reduced lung density faster to -335 ± 23 Hounsfield units (HU) on day 7 than with DEX and saline, to -105 ± 29 HU and -23 ± 11 HU, respectively. The results of functional tests showed that L-37pA treatment 6 h after ARDS/DAD initiation resulted in a more rapid improvement in the physiological respiratory lung by 30-45% functions compared with the comparison drugs. Our data suggest that synthetic amphipathic helical peptide L-37pA blocked a cytokine storm, inhibited acute and chronic pulmonary inflammation, prevented fibrosis development and improved physiological respiratory lung function in the ARDS/DAD mouse model. We concluded that a therapeutic strategy using SAHPs targeting SR-B receptors is a potential novel effective treatment for inflammation-induced ARDS, DAD and lung fibrosis of various etiologies.
Collapse
Affiliation(s)
- Aleksandr S. Chernov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow 117997, Russia; (G.B.T.); (A.N.M.); (V.A.K.); (V.A.P.); (A.A.K.); (A.A.B.J.)
| | - Georgii B. Telegin
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow 117997, Russia; (G.B.T.); (A.N.M.); (V.A.K.); (V.A.P.); (A.A.K.); (A.A.B.J.)
| | - Alexey N. Minakov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow 117997, Russia; (G.B.T.); (A.N.M.); (V.A.K.); (V.A.P.); (A.A.K.); (A.A.B.J.)
| | - Vitaly A. Kazakov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow 117997, Russia; (G.B.T.); (A.N.M.); (V.A.K.); (V.A.P.); (A.A.K.); (A.A.B.J.)
| | - Maksim V. Rodionov
- Medical Radiological Research Center (MRRC) Named after A.F. Tsyb-Branch of the National Medical Radiological Research Center of the Ministry of Health of the Russian Federation, Obninsk 249031, Russia;
| | - Viktor A. Palikov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow 117997, Russia; (G.B.T.); (A.N.M.); (V.A.K.); (V.A.P.); (A.A.K.); (A.A.B.J.)
| | - Anna A. Kudriaeva
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow 117997, Russia; (G.B.T.); (A.N.M.); (V.A.K.); (V.A.P.); (A.A.K.); (A.A.B.J.)
| | - Alexey A. Belogurov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow 117997, Russia; (G.B.T.); (A.N.M.); (V.A.K.); (V.A.P.); (A.A.K.); (A.A.B.J.)
- Department of Biological Chemistry, Russian University of Medicine of the Ministry of Health of the Russian Federation, Moscow 127473, Russia
| |
Collapse
|
2
|
Sarkar Lotfabadi A, Abadi B, Rezaei N. Biomimetic nanotechnology for cancer immunotherapy: State of the art and future perspective. Int J Pharm 2024; 654:123923. [PMID: 38403091 DOI: 10.1016/j.ijpharm.2024.123923] [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: 12/10/2023] [Revised: 02/11/2024] [Accepted: 02/18/2024] [Indexed: 02/27/2024]
Abstract
Cancer continues to be a significant worldwide cause of mortality. This underscores the urgent need for novel strategies to complement and overcome the limitations of conventional therapies, such as imprecise targeting and drug resistance. Cancer Immunotherapy utilizes the body's immune system to target malignant cells, reducing harm to healthy tissue. Nevertheless, the efficacy of immunotherapy exhibits variation across individuals and has the potential to induce autoimmune responses. Biomimetic nanoparticles (bNPs) have transformative potential in cancer immunotherapy, promising improved accurate targeting, immune system activation, and resistance mechanisms, while also reducing the occurrence of systemic autoimmune side effects. This integration offers opportunities for personalized medicine and better therapeutic outcomes. Despite considerable potential, bNPs face barriers like insufficient targeting, restricted biological stability, and interactions within the tumor microenvironment. The resolution of these concerns is crucial in order to expedite the integration of bNPs from the research setting into clinical therapeutic uses. In addition, optimizing manufacturing processes and reducing bNP-related costs are essential for practical implementation. The present research introduces comprehensive classifications of bNPs as well as recent achievements in their application in cancer immunotherapies, emphasizing the need to address barriers for swift clinical integration.
Collapse
Affiliation(s)
- Alireza Sarkar Lotfabadi
- Department of Cellular and Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Banafshe Abadi
- Herbal and Traditional Medicines Research Center, Kerman University of Medical Sciences, Kerman, Iran; Brain Cancer Research Core (BCRC), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Rui M, Zhang W, Mi K, Ni H, Ji W, Yu X, Qin J, Feng C. Design and evaluation of α-helix-based peptide inhibitors for blocking PD-1/PD-L1 interaction. Int J Biol Macromol 2023; 253:126811. [PMID: 37690647 DOI: 10.1016/j.ijbiomac.2023.126811] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 09/12/2023]
Abstract
The current research in tumor immunotherapy indicates that blocking the protein-protein interaction (PPI) between PD-1 and its ligand, PD-L1, may be one of the most effective treatments for cancer patients. The α-helix is a common elements of protein secondary structure and is often involved in protein interaction. Thus, α-helix-based peptides could mimic proteins involved in such interactions and are also capable of modulating PPI in vivo. In this study, starting from a potential α-helix-rich protein, we designed a series of α-helix-based peptide candidates to block PD-1/PD-L1 interaction. These candidates were first screened using molecular docking and molecular dynamics simulations, and then their capacities to inhibit PD-1/PD-L1 interactions and to restore antitumor immune activities were investigated using the HTRF assay, SPR assay, cellular co-culture experiments and animal model experiments. Two peptides exhibited the best anti-tumor effects and the strong ability to restore the immunity of tumor-infiltrating T-cells. Further D-amino acid substitution was employed to improve the serum stability of peptide candidate, making the intravenous administration easier while maintaining the therapeutic efficacy. The resultant peptides showed promise as checkpoint inhibitors for application in tumor immunotherapy. These findings suggested that our strategy for developing peptides starting from an α-helical structure could be used in the design of bioactive inhibitors to potential block protein-protein interactions.
Collapse
Affiliation(s)
- Mengjie Rui
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, PR China
| | - Wen Zhang
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, PR China
| | - Ke Mi
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, PR China
| | - Hairong Ni
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, PR China
| | - Wei Ji
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, PR China
| | - Xuefei Yu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou, PR China
| | - Jiangjiang Qin
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, PR China
| | - Chunlai Feng
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, PR China; Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Tongji University, Ministry of Education, Shanghai, PR China.
| |
Collapse
|
4
|
LaLone V, Smith D, Diaz-Espinosa J, Rosania GR. Quantitative Raman chemical imaging of intracellular drug-membrane aggregates and small molecule drug precipitates in cytoplasmic organelles. Adv Drug Deliv Rev 2023; 202:115107. [PMID: 37769851 PMCID: PMC10841539 DOI: 10.1016/j.addr.2023.115107] [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: 07/16/2023] [Revised: 09/20/2023] [Accepted: 09/25/2023] [Indexed: 10/02/2023]
Abstract
Raman confocal microscopes have been used to visualize the distribution of small molecule drugs within different subcellular compartments. This visualization allows the discovery, characterization, and detailed analysis of the molecular transport phenomena underpinning the Volume of Distribution - a key parameter governing the systemic pharmacokinetics of small molecule drugs. In the specific case of lipophilic small molecules with large Volumes of Distribution, chemical imaging studies using Raman confocal microscopes have revealed how weakly basic, poorly soluble drug molecules can accumulate inside cells by forming stable, supramolecular complexes in association with cytoplasmic membranes or by precipitating out within organelles. To study the self-assembly and function of the resulting intracellular drug inclusions, Raman chemical imaging methods have been developed to measure and map the mass, concentration, and ionization state of drug molecules at a microscopic, subcellular level. Beyond the field of drug delivery, Raman chemical imaging techniques relevant to the study of microscopic drug precipitates and drug-lipid complexes which form inside cells are also being developed by researchers with seemingly unrelated scientific interests. Highlighting advances in data acquisition, calibration methods, and computational data management and analysis tools, this review will cover a decade of technological developments that enable the conversion of spectral signals obtained from Raman confocal microscopes into new discoveries and information about previously unknown, concentrative drug transport pathways driven by soluble-to-insoluble phase transitions occurring within the cytoplasmic organelles of eukaryotic cells.
Collapse
Affiliation(s)
- Vernon LaLone
- Cambium Analytica Research Laboratories, Traverse City, MI, United States
| | - Doug Smith
- Cambium Analytica Research Laboratories, Traverse City, MI, United States
| | - Jennifer Diaz-Espinosa
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, United States
| | - Gus R Rosania
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, United States.
| |
Collapse
|
5
|
Oriá RB, Freitas RS, Roque CR, Nascimento JCR, Silva AP, Malva JO, Guerrant RL, Vitek MP. ApoE Mimetic Peptides to Improve the Vicious Cycle of Malnutrition and Enteric Infections by Targeting the Intestinal and Blood-Brain Barriers. Pharmaceutics 2023; 15:pharmaceutics15041086. [PMID: 37111572 PMCID: PMC10141726 DOI: 10.3390/pharmaceutics15041086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/21/2023] [Accepted: 03/24/2023] [Indexed: 03/30/2023] Open
Abstract
Apolipoprotein E (apoE) mimetic peptides are engineered fragments of the native apoE protein’s LDL-receptor binding site that improve the outcomes following a brain injury and intestinal inflammation in a variety of models. The vicious cycle of enteric infections and malnutrition is closely related to environmental-driven enteric dysfunction early in life, and such chronic inflammatory conditions may blunt the developmental trajectories of children with worrisome and often irreversible physical and cognitive faltering. This window of time for microbiota maturation and brain plasticity is key to protecting cognitive domains, brain health, and achieving optimal/full developmental potential. This review summarizes the potential role of promising apoE mimetic peptides to improve the function of the gut-brain axis, including targeting the blood-brain barrier in children afflicted with malnutrition and enteric infections.
Collapse
Affiliation(s)
- Reinaldo B. Oriá
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology, School of Medicine, Institute of Biomedicine, Federal University of Ceara, Fortaleza 60430-270, Brazil
- Correspondence: ; Tel.: +55-85-3366-8239
| | - Raul S. Freitas
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology, School of Medicine, Institute of Biomedicine, Federal University of Ceara, Fortaleza 60430-270, Brazil
| | - Cássia R. Roque
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology, School of Medicine, Institute of Biomedicine, Federal University of Ceara, Fortaleza 60430-270, Brazil
| | - José Carlos R. Nascimento
- Laboratory of Tissue Healing, Ontogeny and Nutrition, Department of Morphology, School of Medicine, Institute of Biomedicine, Federal University of Ceara, Fortaleza 60430-270, Brazil
- Institute of Health Sciences, Medicine, University of International Integration of Afro-Brazilian Lusofonia, Redenção 62790-970, Brazil
| | - Ana Paula Silva
- Institute of Pharmacology and Experimental Therapeutics and Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine and Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
| | - João O. Malva
- Institute of Pharmacology and Experimental Therapeutics and Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine and Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Richard L. Guerrant
- Division of Infectious Diseases and International Health, Department of Medicine, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Michael P. Vitek
- Division of Neurology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
6
|
Xu D, Chen X, Li Y, Chen Z, Xu W, Wang X, Lv Y, Wang Z, Wu M, Liu G, Wang J. Reconfigurable Peptide Analogs of Apolipoprotein A-I Reveal Tunable Features of Nanodisc Assembly. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:1262-1276. [PMID: 36626237 DOI: 10.1021/acs.langmuir.2c03082] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Nanodisc (ND)-forming membrane scaffold proteins or peptides developed from apolipoprotein A-I (apoA-I) have led to considerable promise in structural biology and therapeutic applications. However, the rationale and regularity characteristics in peptide sequence design remain inconclusive. Here, we proposed a consensus-based normalization approach through the reversed engineering of apoA-IΔ1-45 to design reconfigurable apoA-I peptide analogs (APAs) for tunable ND assembly. We present extensive morphological validations and computational simulation analyses on divergent APA-NDs that are generated by our method. Fifteen divergent APAs were generated accordingly to study the assembly machinery of NDs. We show that APA designs exhibit multifactorial influence in terms of varying APA tandem repeats, sequence composition, and lipid-to-APA ratio to form tunable diameters of NDs. There is a strong positive correlation between DMPC-to-APA ratios and ND diameters. Longer APA with more tandem repeats tends to yield higher particle size homogeneity. Our results also suggest proline is a dispensable residue for the APA-ND formation. Interestingly, proline-rich substitution not only provides an inward-bending effect in forming smaller NDs but also induces the cumulative chain flexibility that enables larger ND formation at higher lipid ratios. Additionally, proline-tryptophan residues in APAs play a dominant role in forming larger NDs. Molecular simulation shows that enriched basic and acidic residues in APAs evoke abundant hydrogen-bond and salt bridge networks to reinforce the structural stability of APA-NDs. Together, our findings provide a rational basis for understanding APA design. The proposed model could be extended to other apolipoproteins for desired ND engineering.
Collapse
Affiliation(s)
- Daiyun Xu
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen518107, China
| | - Xu Chen
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen518107, China
| | - Yongxiao Li
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen518107, China
| | - Zhidong Chen
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen518107, China
| | - Wanting Xu
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen518107, China
| | - Xinpei Wang
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen518107, China
| | - Yonghui Lv
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen518107, China
| | - Zhe Wang
- Department of Pathology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen518033, China
| | - Meiying Wu
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen518107, China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen361102, China
| | - Junqing Wang
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen518107, China
| |
Collapse
|
7
|
Yu L, Jin Y, Song M, Zhao Y, Zhang H. When Natural Compounds Meet Nanotechnology: Nature-Inspired Nanomedicines for Cancer Immunotherapy. Pharmaceutics 2022; 14:pharmaceutics14081589. [PMID: 36015215 PMCID: PMC9412684 DOI: 10.3390/pharmaceutics14081589] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/25/2022] [Accepted: 07/28/2022] [Indexed: 02/01/2023] Open
Abstract
Recent significant strides of natural compounds in immunomodulation have highlighted their great potential against cancer. Despite many attempts being made for cancer immunotherapy, the biomedical application of natural compounds encounters a bottleneck because of their unclear mechanisms, low solubility and bioavailability, and limited efficacy. Herein, we summarize the immune regulatory mechanisms of different natural compounds at each step of the cancer-immunity cycle and highlight their anti-tumor potential and current limitations. We then propose and present various drug delivery strategies based on nanotechnology, including traditional nanoparticles (NPs)-based delivery strategies (lipid-based NPs, micelles, and polysaccharide/peptide/protein-based NPs) and novel delivery strategies (cell-derived NPs and carrier-free NPs), thus providing solutions to break through existing bottlenecks. Furthermore, representative applications of nature-inspired nanomedicines are also emphasized in detail with the advantages and disadvantages discussed. Finally, the challenges and prospects of natural compounds for cancer immunotherapy are provided, hopefully, to facilitate their far-reaching development toward clinical translation.
Collapse
Affiliation(s)
- Linna Yu
- People’s Hospital of Qianxinan Buyi and Miao Minority Autonomous Prefecture, Xingyi 562400, China;
| | - Yi Jin
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicines, Department of Pharmaceutics, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, China; (Y.J.); (M.S.)
| | - Mingjie Song
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicines, Department of Pharmaceutics, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, China; (Y.J.); (M.S.)
| | - Yu Zhao
- People’s Hospital of Qianxinan Buyi and Miao Minority Autonomous Prefecture, Xingyi 562400, China;
- Correspondence: (Y.Z.); (H.Z.)
| | - Huaqing Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicines, Department of Pharmaceutics, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, China; (Y.J.); (M.S.)
- Correspondence: (Y.Z.); (H.Z.)
| |
Collapse
|
8
|
HDL, ApoA-I and ApoE-Mimetic Peptides: Potential Broad Spectrum Agent for Clinical Use? Int J Pept Res Ther 2022. [DOI: 10.1007/s10989-021-10352-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
9
|
Daskou M, Mu W, Sharma M, Vasilopoulos H, Heymans R, Ritou E, Rezek V, Hamid P, Kossyvakis A, Sen Roy S, Grijalva V, Chattopadhyay A, Kitchen SG, Fogelman AM, Reddy ST, Kelesidis T. ApoA-I mimetics reduce systemic and gut inflammation in chronic treated HIV. PLoS Pathog 2022; 18:e1010160. [PMID: 34995311 PMCID: PMC8740974 DOI: 10.1371/journal.ppat.1010160] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 11/30/2021] [Indexed: 12/31/2022] Open
Abstract
Novel therapeutic strategies are needed to attenuate increased systemic and gut inflammation that contribute to morbidity and mortality in chronic HIV infection despite potent antiretroviral therapy (ART). The goal of this study is to use preclinical models of chronic treated HIV to determine whether the antioxidant and anti-inflammatory apoA-I mimetic peptides 6F and 4F attenuate systemic and gut inflammation in chronic HIV. We used two humanized murine models of HIV infection and gut explants from 10 uninfected and 10 HIV infected persons on potent ART, to determine the in vivo and ex vivo impact of apoA-I mimetics on systemic and intestinal inflammation in HIV. When compared to HIV infected humanized mice treated with ART alone, mice on oral apoA-I mimetic peptide 6F with ART had consistently reduced plasma and gut tissue cytokines (TNF-α, IL-6) and chemokines (CX3CL1) that are products of ADAM17 sheddase activity. Oral 6F attenuated gut protein levels of ADAM17 that were increased in HIV-1 infected mice on potent ART compared to uninfected mice. Adding oxidized lipoproteins and endotoxin (LPS) ex vivo to gut explants from HIV infected persons increased levels of ADAM17 in myeloid and intestinal cells, which increased TNF-α and CX3CL1. Both 4F and 6F attenuated these changes. Our preclinical data suggest that apoA-I mimetic peptides provide a novel therapeutic strategy that can target increased protein levels of ADAM17 and its sheddase activity that contribute to intestinal and systemic inflammation in treated HIV. The large repertoire of inflammatory mediators involved in ADAM17 sheddase activity places it as a pivotal orchestrator of several inflammatory pathways associated with morbidity in chronic treated HIV that make it an attractive therapeutic target.
Collapse
Affiliation(s)
- Maria Daskou
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - William Mu
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Madhav Sharma
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Hariclea Vasilopoulos
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Rachel Heymans
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Eleni Ritou
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Valerie Rezek
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Philip Hamid
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Athanasios Kossyvakis
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Shubhendu Sen Roy
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Victor Grijalva
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Arnab Chattopadhyay
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Scott G. Kitchen
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Alan M. Fogelman
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Srinivasa T. Reddy
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California, United States of America
- Molecular Toxicology Interdepartmental Degree Program, University of California Los Angeles, Los Angeles, California, United States of America
| | - Theodoros Kelesidis
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| |
Collapse
|
10
|
Theodoros K, Sharma M, Anton P, Hugo C, Ellen O, Hultgren NW, Ritou E, Williams DS, Orian S S, Srinivasa T R. The ApoA-I mimetic peptide 4F attenuates in vitro replication of SARS-CoV-2, associated apoptosis, oxidative stress and inflammation in epithelial cells. Virulence 2021; 12:2214-2227. [PMID: 34494942 PMCID: PMC8437485 DOI: 10.1080/21505594.2021.1964329] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 07/12/2021] [Accepted: 07/30/2021] [Indexed: 02/06/2023] Open
Abstract
An oral antiviral against SARS-CoV-2 that also attenuates inflammatory instigators of severe COVID-19 is not available to date. Herein, we show that the apoA-I mimetic peptide 4 F inhibits Spike mediated viral entry and has antiviral activity against SARS-CoV-2 in human lung epithelial Calu3 and Vero-E6 cells. In SARS-CoV-2 infected Calu3 cells, 4 F upregulated inducers of the interferon pathway such as MX-1 and Heme oxygenase 1 (HO-1) and downregulated mitochondrial reactive oxygen species (mito-ROS) and CD147, a host protein that mediates viral entry. 4 F also reduced associated cellular apoptosis and secretion of IL-6 in both SARS-CoV-2 infected Vero-E6 and Calu3 cells. Thus, 4 F attenuates in vitro SARS-CoV-2 replication, associated apoptosis in epithelial cells and secretion of IL-6, a major cytokine related to COVID-19 morbidity. Given established safety of 4 F in humans, clinical studies are warranted to establish 4 F as therapy for COVID-19.
Collapse
Affiliation(s)
- Kelesidis Theodoros
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Madhav Sharma
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Petcherski Anton
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Cristelle Hugo
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - O’Connor Ellen
- Molecular Toxicology Interdepartmental Degree Program, University of California Los Angeles, United States
| | - Nan W Hultgren
- Department of Ophthalmology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Eleni Ritou
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - David S Williams
- Department of Ophthalmology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Shirihai Orian S
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Reddy Srinivasa T
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
- Molecular Toxicology Interdepartmental Degree Program, University of California Los Angeles, United States
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| |
Collapse
|
11
|
Landowski M, Bowes Rickman C. Targeting Lipid Metabolism for the Treatment of Age-Related Macular Degeneration: Insights from Preclinical Mouse Models. J Ocul Pharmacol Ther 2021; 38:3-32. [PMID: 34788573 PMCID: PMC8817708 DOI: 10.1089/jop.2021.0067] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Age-related macular degeneration (AMD) is a major leading cause of irreversible visual impairment in the world with limited therapeutic interventions. Histological, biochemical, genetic, and epidemiological studies strongly implicate dysregulated lipid metabolism in the retinal pigmented epithelium (RPE) in AMD pathobiology. However, effective therapies targeting lipid metabolism still need to be identified and developed for this blinding disease. To test lipid metabolism-targeting therapies, preclinical AMD mouse models are needed to establish therapeutic efficacy and the role of lipid metabolism in the development of AMD-like pathology. In this review, we provide a comprehensive overview of current AMD mouse models available to researchers that could be used to provide preclinical evidence supporting therapies targeting lipid metabolism for AMD. Based on previous studies of AMD mouse models, we discuss strategies to modulate lipid metabolism as well as examples of studies evaluating lipid-targeting therapeutics to restore lipid processing in the RPE. The use of AMD mouse models may lead to worthy lipid-targeting candidate therapies for clinical trials to prevent the blindness caused by AMD.
Collapse
Affiliation(s)
- Michael Landowski
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, Wisconsin, USA.,McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Catherine Bowes Rickman
- Department of Ophthalmology, Duke University Medical Center, Durham, North Carolina, USA.,Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
12
|
Jin Y, Chifodya K, Han G, Jiang W, Chen Y, Shi Y, Xu Q, Xi Y, Wang J, Zhou J, Zhang H, Ding Y. High-density lipoprotein in Alzheimer's disease: From potential biomarkers to therapeutics. J Control Release 2021; 338:56-70. [PMID: 34391838 DOI: 10.1016/j.jconrel.2021.08.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 08/09/2021] [Accepted: 08/10/2021] [Indexed: 12/17/2022]
Abstract
The inverse correlation between high-density lipoprotein (HDL) levels in vivo and the risk of Alzheimer's disease (AD) has become an inspiration for HDL-inspired AD therapy, including plain HDL and various intelligent HDL-based drug delivery systems. In this review, we will focus on the two endogenous HDL subtypes in the central nervous system (CNS), apolipoprotein E-based HDL (apoE-HDL) and apolipoprotein A-I-based HDL (apoA-I-HDL), especially their influence on AD pathophysiology to reveal HDL's potential as biomarkers for risk prediction, and summarize the relevant therapeutic mechanisms to propose possible treatment strategies. We will emphasize the latest advances of HDL as therapeutics (plain HDL and HDL-based drug delivery systems) to discuss the potential for AD therapy and review innovative techniques in the preparation of HDL-based nanoplatforms to provide a basis for the rational design and future development of anti-AD drugs.
Collapse
Affiliation(s)
- Yi Jin
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing 210009, China; State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China; NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, Nanjing 210009, China
| | - Kudzai Chifodya
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Guochen Han
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing 210009, China; State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China; NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, Nanjing 210009, China
| | - Wenxin Jiang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Yun Chen
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Yang Shi
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Qiao Xu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Yilong Xi
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Jun Wang
- Department of Geriatrics, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Jianping Zhou
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing 210009, China; State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China; NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, Nanjing 210009, China.
| | - Huaqing Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing 210009, China; State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China; NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, Nanjing 210009, China.
| | - Yang Ding
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing 210009, China; State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China; NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, Nanjing 210009, China.
| |
Collapse
|
13
|
Groner J, Goepferich A, Breunig M. Atherosclerosis: Conventional intake of cardiovascular drugs versus delivery using nanotechnology - A new chance for causative therapy? J Control Release 2021; 333:536-559. [PMID: 33794270 DOI: 10.1016/j.jconrel.2021.03.034] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 03/24/2021] [Accepted: 03/25/2021] [Indexed: 02/06/2023]
Abstract
Atherosclerosis is the leading cause of death in developed countries. The pathogenetic mechanism relies on a macrophage-based immune reaction to low density lipoprotein (LDL) deposition in blood vessels with dysfunctional endothelia. Thus, atherosclerosis is defined as a chronic inflammatory disease. A plethora of cardiovascular drugs have been developed and are on the market, but the major shortcoming of standard medications is that they do not address the root cause of the disease. Statins and thiazolidinediones that have recently been recognized to exert specific anti-atherosclerotic effects represent a potential breakthrough on the horizon. But their whole potential cannot be realized due to insufficient availability at the pathological site and severe off-target effects. The focus of this review will be to elaborate how both groups of drugs could immensely profit from nanoparticulate carriers. This delivery principle would allow for their accumulation in target macrophages and endothelial cells of the atherosclerotic plaque, increasing bioavailability where it is needed most. Based on the analyzed literature we conclude design criteria for the delivery of statins and thiazolidinediones with nanoparticles for anti-atherosclerotic therapy. Nanoparticles need to be below a diameter of 100 nm to accumulate in the atherosclerotic plaque and should be fabricated using biodegradable materials. Further, the thiazolidinediones or statins must be encapsulated into the particle core, because especially for thiazolidindiones the uptake into cells is prerequisite for their mechanism of action. For optimal uptake into targeted macrophages and endothelial cells, the ideal particle should present ligands on its surface which bind specifically to scavenger receptors. The impact of statins on the lectin-type oxidized LDL receptor 1 (LOX1) seems particularly promising because of its outstanding role in the inflammatory process. Using this pioneering concept, it will be possible to promote the impact of statins and thiazolidinediones on macrophages and endothelial cells and significantly enhance their anti-atherosclerotic therapeutic potential.
Collapse
Affiliation(s)
- Jonas Groner
- Department of Pharmaceutical Technology, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany
| | - Achim Goepferich
- Department of Pharmaceutical Technology, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany
| | - Miriam Breunig
- Department of Pharmaceutical Technology, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany.
| |
Collapse
|
14
|
Mu W, Sharma M, Heymans R, Ritou E, Rezek V, Hamid P, Kossyvakis A, Sen Roy S, Grijalva V, Chattopadhyay A, Papesh J, Meriwether D, Kitchen SG, Fogelman AM, Reddy ST, Kelesidis T. Apolipoprotein A-I mimetics attenuate macrophage activation in chronic treated HIV. AIDS 2021; 35:543-553. [PMID: 33306550 PMCID: PMC8010648 DOI: 10.1097/qad.0000000000002785] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
OBJECTIVES Despite antiretroviral therapy (ART), there is an unmet need for therapies to mitigate immune activation in HIV infection. The goal of this study is to determine whether the apoA-I mimetics 6F and 4F attenuate macrophage activation in chronic HIV. DESIGN Preclinical assessment of the in-vivo impact of Tg6F and the ex-vivo impact of apoA-I mimetics on biomarkers of immune activation and gut barrier dysfunction in treated HIV. METHODS We used two humanized murine models of HIV infection to determine the impact of oral Tg6F with ART (HIV+ART+Tg6F+) on innate immune activation (plasma human sCD14, sCD163) and gut barrier dysfunction [murine I-FABP, endotoxin (LPS), LPS-binding protein (LBP), murine sCD14]. We also used gut explants from 10 uninfected and 10 HIV-infected men on potent ART and no morbidity, to determine the impact of ex-vivo treatment with 4F for 72 h on secretion of sCD14, sCD163, and I-FABP from gut explants. RESULTS When compared with mice treated with ART alone (HIV+ART+), HIV+ART+Tg6F+ mice attenuated macrophage activation (h-sCD14, h-sCD163), gut barrier dysfunction (m-IFABP, LPS, LBP, and m-sCD14), plasma and gut tissue oxidized lipoproteins. The results were consistent with independent mouse models and ART regimens. Both 4F and 6F attenuated shedding of I-FABP and sCD14 from gut explants from HIV-infected and uninfected participants. CONCLUSION Given that gut barrier dysfunction and macrophage activation are contributors to comorbidities like cardiovascular disease in HIV, apoA-I mimetics should be tested as therapy for morbidity in chronic treated HIV.
Collapse
Affiliation(s)
- William Mu
- Division of Infectious Diseases
- Division of Hematology and Oncology
| | | | | | | | | | - Philip Hamid
- Division of Infectious Diseases
- Division of Hematology and Oncology
| | | | | | - Victor Grijalva
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine
| | - Arnab Chattopadhyay
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine
| | - Jeremy Papesh
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine
| | - David Meriwether
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine
| | | | - Alan M Fogelman
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine
| | - Srinivasa T Reddy
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine
- Department of Molecular and Medical Pharmacology
- Molecular Toxicology Interdepartmental Degree Program, University of California Los Angeles
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | | |
Collapse
|
15
|
Guo Y, Li W, Qian M, Jiang T, Guo P, Du Q, Lin N, Xie X, Wu Z, Lin D, Liu D. D-4F Ameliorates Contrast Media-Induced Oxidative Injuries in Endothelial Cells via the AMPK/PKC Pathway. Front Pharmacol 2021; 11:556074. [PMID: 33658920 PMCID: PMC7917283 DOI: 10.3389/fphar.2020.556074] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 11/30/2020] [Indexed: 01/23/2023] Open
Abstract
Endothelial dysfunction is involved in the pathophysiological processes of contrast media (CM)–induced acute kidney injury (CI-AKI) after vascular angiography or intervention. Previous study found that apolipoprotein A-I (apoA-I) mimetic peptide, D-4F, alleviates endothelial impairments via upregulating heme oxygenase-1 (HO-1) expression and scavenging excessively generated reactive oxygen species (ROS). However, whether D-4F could ameliorate oxidative injuries in endothelial cells through suppressing ROS production remains unclear. In this study, a representative nonionic iodinated CM, iodixanol, was chosen for the in vitro and in vivo studies. Endothelial cell viability was assayed using micrographs, lactate dehydrogenase (LDH) activity, and cell counting kit-8 (CCK-8). Apoptosis was detected using flow cytometry analysis and caspase-3 activation. Endothelial inflammation was tested using monocyte adhesion assay and adhesion molecule expression. ROS production was detected by measuring the formation of lipid peroxidation malondialdehyde (MDA) through the thiobarbituric acid reactive substance (TBARS) assay. Peroxynitrite (ONOO⁻) formation was tested using the 3-nitrotyrosine ELISA kit. Iodixanol impaired cell viability, promoted vascular cell adhesion molecule-1 (VCAM-1) and intercellular cell adhesion molecule-1 (ICAM-1) expression, and induced cell apoptosis in human umbilical vein endothelial cells (HUVECs). However, D-4F mitigated these injuries. Furthermore, iodixanol induced the phosphorylation of protein kinase C (PKC) beta II, p47, Rac1, and endothelial nitric oxide synthase (eNOS) at Thr495, which elicited ROS release and ONOO⁻ generation. D-4F inhibited NADPH oxidase (NOX) activation, ROS production, and ONOO⁻ formation via the AMP-activated protein kinase (AMPK)/PKC pathway. Additionally, after an intravascular injection of iodixanol in Sprague Dawley rats, iodixanol induced a remarkable inflammatory response in arterial endothelial cells, although significant apoptosis and morphological changes were not observed. D-4F alleviated the vessel inflammation resulting from iodixanol in vivo. Collectively, besides scavenging ROS, D-4F could also suppress ROS production and ONOO⁻ formation through the AMPK/PKC pathway, which ameliorated oxidative injuries in endothelial cells. Hence, D-4F might serve as a potential agent in preventing CI-AKI.
Collapse
Affiliation(s)
- Yansong Guo
- Department of Cardiology, Fujian Provincial Hospital, Fujian Provincial Key Laboratory of Cardiovascular Disease, Fujian Cardiovascular Institute, Fujian Provincial Center for Geriatrics, Provincial Clinical Medicine College of Fujian Medical University, Fuzhou, China
| | - Wei Li
- Department of Cardiology, the Affiliated Xiamen Cardiovascular Hospital of Xiamen University, Medical College of Xiamen University, Xiamen, China
| | - Mingming Qian
- Department of Cardiology, the Affiliated Xiamen Cardiovascular Hospital of Xiamen University, Medical College of Xiamen University, Xiamen, China
| | - Ting Jiang
- MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, High-field NMR Research Center, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, China
| | - Ping Guo
- Department of Cardiology, the Affiliated Xiamen Cardiovascular Hospital of Xiamen University, Medical College of Xiamen University, Xiamen, China
| | - Qian Du
- Department of Cardiology, the Affiliated Xiamen Cardiovascular Hospital of Xiamen University, Medical College of Xiamen University, Xiamen, China
| | - Na Lin
- Department of Cardiology, Fujian Provincial Hospital, Fujian Provincial Key Laboratory of Cardiovascular Disease, Fujian Cardiovascular Institute, Fujian Provincial Center for Geriatrics, Provincial Clinical Medicine College of Fujian Medical University, Fuzhou, China
| | - Xianwei Xie
- Department of Cardiology, Fujian Provincial Hospital, Fujian Provincial Key Laboratory of Cardiovascular Disease, Fujian Cardiovascular Institute, Fujian Provincial Center for Geriatrics, Provincial Clinical Medicine College of Fujian Medical University, Fuzhou, China
| | - Zhiyong Wu
- Department of Cardiology, Fujian Provincial Hospital, Fujian Provincial Key Laboratory of Cardiovascular Disease, Fujian Cardiovascular Institute, Fujian Provincial Center for Geriatrics, Provincial Clinical Medicine College of Fujian Medical University, Fuzhou, China
| | - Donghai Lin
- MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, High-field NMR Research Center, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, China
| | - Donghui Liu
- Department of Cardiology, Fujian Provincial Hospital, Fujian Provincial Key Laboratory of Cardiovascular Disease, Fujian Cardiovascular Institute, Fujian Provincial Center for Geriatrics, Provincial Clinical Medicine College of Fujian Medical University, Fuzhou, China.,Department of Cardiology, the Affiliated Xiamen Cardiovascular Hospital of Xiamen University, Medical College of Xiamen University, Xiamen, China
| |
Collapse
|
16
|
Rao N, Rho JG, Um W, EK PK, Nguyen VQ, Oh BH, Kim W, Park JH. Hyaluronic Acid Nanoparticles as Nanomedicine for Treatment of Inflammatory Diseases. Pharmaceutics 2020; 12:E931. [PMID: 33003609 PMCID: PMC7600604 DOI: 10.3390/pharmaceutics12100931] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 09/24/2020] [Accepted: 09/28/2020] [Indexed: 02/06/2023] Open
Abstract
Owing to their unique biological functions, hyaluronic acid (HA) and its derivatives have been explored extensively for biomedical applications such as tissue engineering, drug delivery, and molecular imaging. In particular, self-assembled HA nanoparticles (HA-NPs) have been used widely as target-specific and long-acting nanocarriers for the delivery of a wide range of therapeutic or diagnostic agents. Recently, it has been demonstrated that empty HA-NPs without bearing any therapeutic agent can be used therapeutically for the treatment of inflammatory diseases via modulating inflammatory responses. In this review, we aim to provide an overview of the significant achievements in this field and highlight the potential of HA-NPs for the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- N.Vijayakameswara Rao
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Korea; (N.V.R.); (W.U.); (P.K.E.); (V.Q.N.); (B.H.O.)
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei 10617, Taiwan
| | - Jun Gi Rho
- Department of Molecular Science & Technology, Ajou University, Suwon 16499, Korea;
| | - Wooram Um
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Korea; (N.V.R.); (W.U.); (P.K.E.); (V.Q.N.); (B.H.O.)
| | - Pramod Kumar EK
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Korea; (N.V.R.); (W.U.); (P.K.E.); (V.Q.N.); (B.H.O.)
| | - Van Quy Nguyen
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Korea; (N.V.R.); (W.U.); (P.K.E.); (V.Q.N.); (B.H.O.)
| | - Byeong Hoon Oh
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Korea; (N.V.R.); (W.U.); (P.K.E.); (V.Q.N.); (B.H.O.)
| | - Wook Kim
- Department of Molecular Science & Technology, Ajou University, Suwon 16499, Korea;
| | - Jae Hyung Park
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Korea; (N.V.R.); (W.U.); (P.K.E.); (V.Q.N.); (B.H.O.)
- Department Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea
| |
Collapse
|
17
|
Swaminathan SK, Zhou AL, Ahlschwede KM, Curran GL, Lowe VJ, Li L, Kandimalla KK. High-Density Lipoprotein Mimetic Peptide 4F Efficiently Crosses the Blood-Brain Barrier and Modulates Amyloid- β Distribution between Brain and Plasma. J Pharmacol Exp Ther 2020; 375:308-316. [PMID: 32778535 DOI: 10.1124/jpet.120.265876] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 07/24/2020] [Indexed: 12/24/2022] Open
Abstract
Treatments to elevate high-density lipoprotein (HDL) levels in plasma have decreased cerebrovascular amyloid -β (Aβ) deposition and mitigated cognitive decline in Alzheimer disease (AD) transgenic mice. Since the major protein component of HDL particles, apolipoprotein A-I (ApoA-I), has very low permeability at the blood-brain barrier (BBB), we investigated 4F, an 18-amino-acid ApoA-I/HDL mimetic peptide, as a therapeutic alternative. Specifically, we examined the BBB permeability of 4F and its effects on [125I]Aβ trafficking from brain to blood and from blood to brain. After systemic injection in mice, the BBB permeability of [125I]4F, estimated as the permeability-surface area (PS) product, ranged between 2 and 5 × 10-6 ml/g per second in various brain regions. The PS products of [125I]4F were ∼1000-fold higher compared with those determined for [125I]ApoA-I. Moreover, systemic infusion with 4F increased the brain efflux of intracerebrally injected [125I]Aβ42. Conversely, 4F infusion decreased the brain influx of systemically injected [125I]Aβ42. Interestingly, 4F did not significantly alter the brain influx of [125I]Aβ40. To corroborate the in vivo findings, we evaluated the effects of 4F on [125I]Aβ42 transcytosis across polarized human BBB endothelial cell (hCMEC/D3) monolayers. Treatment with 4F increased the abluminal-to-luminal flux and decreased the luminal-to-abluminal flux of [125I]Aβ42 across the hCMEC/D3 monolayers. Additionally, 4F decreased the endothelial accumulation of fluorescein-labeled Aβ42 in the hCMEC/D3 monolayers. These findings provide a mechanistic interpretation for the reductions in brain Aβ burden reported in AD mice after oral 4F administration, which represents a novel strategy for treating AD and cerebral amyloid angiopathy. SIGNIFICANCE STATEMENT: The brain permeability of the ApoA-I mimetic peptide 4F was estimated to be ∼1000-fold greater than ApoA-I after systemic injection of radiolabeled peptide/protein in mice. Further, 4F treatment increased the brain efflux of amyloid -β and also decreased its brain influx, as evaluated in mice and in blood-brain barrier cell monolayers. Thus, 4F represents a potential therapeutic strategy to mitigate brain amyloid accumulation in cerebral amyloid angiopathy and Alzheimer disease.
Collapse
Affiliation(s)
- Suresh K Swaminathan
- Department of Pharmaceutics and Brain Barriers Research Center (S.K.S., A.L.Z., K.M.A., K.K.K.) and Department of Experimental and Clinical Pharmacology (L.L.), University of Minnesota, College of Pharmacy, Minneapolis, Minnesota; Department of Pharmaceutical Sciences, College of Pharmacy, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois (K.M.A.); and Departments of Radiology (G.L.C., V.J.L.) and Neurology (G.L.C.), Mayo Clinic, College of Medicine, Rochester, Minnesota
| | - Andrew L Zhou
- Department of Pharmaceutics and Brain Barriers Research Center (S.K.S., A.L.Z., K.M.A., K.K.K.) and Department of Experimental and Clinical Pharmacology (L.L.), University of Minnesota, College of Pharmacy, Minneapolis, Minnesota; Department of Pharmaceutical Sciences, College of Pharmacy, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois (K.M.A.); and Departments of Radiology (G.L.C., V.J.L.) and Neurology (G.L.C.), Mayo Clinic, College of Medicine, Rochester, Minnesota
| | - Kristen M Ahlschwede
- Department of Pharmaceutics and Brain Barriers Research Center (S.K.S., A.L.Z., K.M.A., K.K.K.) and Department of Experimental and Clinical Pharmacology (L.L.), University of Minnesota, College of Pharmacy, Minneapolis, Minnesota; Department of Pharmaceutical Sciences, College of Pharmacy, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois (K.M.A.); and Departments of Radiology (G.L.C., V.J.L.) and Neurology (G.L.C.), Mayo Clinic, College of Medicine, Rochester, Minnesota
| | - Geoffry L Curran
- Department of Pharmaceutics and Brain Barriers Research Center (S.K.S., A.L.Z., K.M.A., K.K.K.) and Department of Experimental and Clinical Pharmacology (L.L.), University of Minnesota, College of Pharmacy, Minneapolis, Minnesota; Department of Pharmaceutical Sciences, College of Pharmacy, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois (K.M.A.); and Departments of Radiology (G.L.C., V.J.L.) and Neurology (G.L.C.), Mayo Clinic, College of Medicine, Rochester, Minnesota
| | - Val J Lowe
- Department of Pharmaceutics and Brain Barriers Research Center (S.K.S., A.L.Z., K.M.A., K.K.K.) and Department of Experimental and Clinical Pharmacology (L.L.), University of Minnesota, College of Pharmacy, Minneapolis, Minnesota; Department of Pharmaceutical Sciences, College of Pharmacy, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois (K.M.A.); and Departments of Radiology (G.L.C., V.J.L.) and Neurology (G.L.C.), Mayo Clinic, College of Medicine, Rochester, Minnesota
| | - Ling Li
- Department of Pharmaceutics and Brain Barriers Research Center (S.K.S., A.L.Z., K.M.A., K.K.K.) and Department of Experimental and Clinical Pharmacology (L.L.), University of Minnesota, College of Pharmacy, Minneapolis, Minnesota; Department of Pharmaceutical Sciences, College of Pharmacy, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois (K.M.A.); and Departments of Radiology (G.L.C., V.J.L.) and Neurology (G.L.C.), Mayo Clinic, College of Medicine, Rochester, Minnesota
| | - Karunya K Kandimalla
- Department of Pharmaceutics and Brain Barriers Research Center (S.K.S., A.L.Z., K.M.A., K.K.K.) and Department of Experimental and Clinical Pharmacology (L.L.), University of Minnesota, College of Pharmacy, Minneapolis, Minnesota; Department of Pharmaceutical Sciences, College of Pharmacy, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois (K.M.A.); and Departments of Radiology (G.L.C., V.J.L.) and Neurology (G.L.C.), Mayo Clinic, College of Medicine, Rochester, Minnesota
| |
Collapse
|
18
|
Zou J, Wang G, Li H, Yu X, Tang C. IgM natural antibody T15/E06 in atherosclerosis. Clin Chim Acta 2020; 504:15-22. [DOI: 10.1016/j.cca.2020.01.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 01/23/2020] [Accepted: 01/23/2020] [Indexed: 11/28/2022]
|
19
|
Apolipoprotein-AI mimetic peptides D-4F and L-5F decrease hepatic inflammation and increase insulin sensitivity in C57BL/6 mice. PLoS One 2020; 15:e0226931. [PMID: 31914125 PMCID: PMC6948736 DOI: 10.1371/journal.pone.0226931] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 12/06/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Apolipoprotein-AI (apo-AI) is the major apolipoprotein found in high density lipoprotein particles (HDLs). We previously demonstrated that apo-AI injected directly into high-fat diet fed mice improved insulin sensitivity associated with decreased hepatic inflammation. While our data provides compelling proof of concept, apoA-I mimetic peptides are more clinically feasible. The aim of this study was to test whether apo-AI mimetic peptide (D-4F and L-5F) treatment will emulate the effects of full-length apo-AI to improve insulin sensitivity. METHODS Male C57BL/6 mice were fed a high-fat diet for 16 weeks before receiving D4F mimetic peptide administered via drinking water or L5F mimetic peptide administered by intraperitoneal injection bi-weekly for a total of five weeks. Glucose tolerance and insulin tolerance tests were conducted to assess the effects of the peptides on insulin resistance. Effects of the peptides on inflammation, gluconeogenic enzymes and lipid synthesis were assessed by real-time PCR of key markers involved in the respective pathways. RESULTS Treatment with apo-AI mimetic peptides D-4F and L-5F showed: (i) improved blood glucose clearance (D-4F 1.40-fold AUC decrease compared to HFD, P<0.05; L-4F 1.17-fold AUC decrease compared to HFD, ns) in the glucose tolerance test; (ii) improved insulin tolerance (D-4F 1.63-fold AUC decrease compared to HFD, P<0.05; L-5F 1.39-fold AUC compared to HFD, P<0.05) in the insulin tolerance test. The metabolic test results were associated with (i) decreased hepatic inflammation of SAA1, IL-1β IFN-γ and TNFα (2.61-5.97-fold decrease compared to HFD, P<0.05) for both mimetics; (ii) suppression of hepatic mRNA expression of gluconeogenesis-associated genes (PEPCK and G6Pase; 1.66-3.01-fold decrease compared to HFD, P<0.001) for both mimetics; (iii) lipogenic-associated genes, (SREBP1c and ChREBP; 2.15-3.31-fold decrease compared to HFD, P<0.001) for both mimetics and; (iv) reduced hepatic macrophage infiltration (F4/80 and CD68; 1.77-2.15-fold compared to HFD, P<0.001) for both mimetics. CONCLUSION Apo-AI mimetic peptides treatment led to improved glucose homeostasis. This effect is associated with reduced expression of inflammatory markers in the liver and reduced infiltration of macrophages, suggesting an overall suppression of hepatic inflammation. We also showed altered expression of genes associated with gluconeogenesis and lipid synthesis, suggesting that glucose and lipid synthesis is suppressed. These findings suggest that apoA-I mimetic peptides could be a new therapeutic option to reduce hepatic inflammation that contributes to the development of overnutrition-induced insulin resistance.
Collapse
|
20
|
Kornmueller K, Vidakovic I, Prassl R. Artificial High Density Lipoprotein Nanoparticles in Cardiovascular Research. Molecules 2019; 24:E2829. [PMID: 31382521 PMCID: PMC6695986 DOI: 10.3390/molecules24152829] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/31/2019] [Accepted: 08/01/2019] [Indexed: 02/07/2023] Open
Abstract
Lipoproteins are endogenous nanoparticles which are the major transporter of fats and cholesterol in the human body. They play a key role in the regulatory mechanisms of cardiovascular events. Lipoproteins can be modified and manipulated to act as drug delivery systems or nanocarriers for contrast agents. In particular, high density lipoproteins (HDL), which are the smallest class of lipoproteins, can be synthetically engineered either as nascent HDL nanodiscs or spherical HDL nanoparticles. Reconstituted HDL (rHDL) particles are formed by self-assembly of various lipids and apolipoprotein AI (apo-AI). A variety of substances including drugs, nucleic acids, signal emitting molecules, or dyes can be loaded, making them efficient nanocarriers for therapeutic applications or medical diagnostics. This review provides an overview about synthesis techniques, physicochemical properties of rHDL nanoparticles, and structural determinants for rHDL function. We discuss recent developments utilizing either apo-AI or apo-AI mimetic peptides for the design of pharmaceutical rHDL formulations. Advantages, limitations, challenges, and prospects for clinical translation are evaluated with a special focus on promising strategies for the treatment and diagnosis of atherosclerosis and cardiovascular diseases.
Collapse
Affiliation(s)
- Karin Kornmueller
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Biophysics, Medical University of Graz, Neue Stiftingtalstraße 6/IV, 8010 Graz, Austria
| | - Ivan Vidakovic
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Biophysics, Medical University of Graz, Neue Stiftingtalstraße 6/IV, 8010 Graz, Austria
| | - Ruth Prassl
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Biophysics, Medical University of Graz, Neue Stiftingtalstraße 6/IV, 8010 Graz, Austria.
| |
Collapse
|
21
|
Macpherson ME, Halvorsen B, Yndestad A, Ueland T, Mollnes TE, Berge RK, Rashidi A, Otterdal K, Gregersen I, Kong XY, Holven KB, Aukrust P, Fevang B, Jørgensen SF. Impaired HDL Function Amplifies Systemic Inflammation in Common Variable Immunodeficiency. Sci Rep 2019; 9:9427. [PMID: 31263122 PMCID: PMC6603020 DOI: 10.1038/s41598-019-45861-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 06/13/2019] [Indexed: 12/16/2022] Open
Abstract
Common variable immunodeficiency (CVID) is the most common symptomatic primary immunodeficiency, characterized by inadequate antibody responses and recurrent bacterial infections. Paradoxically, a majority of CVID patients have non-infectious inflammatory and autoimmune complications, associated with systemic immune activation. Our aim was to explore if HDL, known to have anti-inflammatory properties, had impaired function in CVID patients and thereby contributed to their inflammatory phenotype. We found reduced HDL cholesterol levels in plasma of CVID patients compared to healthy controls, particularly in patients with inflammatory and autoimmune complications, correlating negatively with inflammatory markers CRP and sCD25. Reverse cholesterol transport capacity testing showed reduced serum acceptance capacity for cholesterol in CVID patients with inflammatory and autoimmune complications. They also had reduced cholesterol efflux capacity from macrophages to serum and decreased expression of ATP-binding cassette transporter ABCA1. Human HDL suppressed TLR2-induced TNF release less in blood mononuclear cells from CVID patients, associated with decreased expression of transcriptional factor ATF3. Our data suggest a link between impaired HDL function and systemic inflammation in CVID patients, particularly in those with autoimmune and inflammatory complications. This identifies HDL as a novel therapeutic target in CVID as well as other more common conditions characterized by sterile inflammation or autoimmunity.
Collapse
Affiliation(s)
- Magnhild E Macpherson
- Research Institute of Internal Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway.
- Section of Clinical Immunology and Infectious Diseases, Department of Rheumatology, Dermatology and Infectious Diseases, Oslo University Hospital, Rikshospitalet, Oslo, Norway.
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Arne Yndestad
- Research Institute of Internal Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Faculty of Health Sciences and K.G. Jebsen TREC, University of Tromsø, Tromsø, Norway
| | - Tom E Mollnes
- Department of Immunology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Research Laboratory, Nordland Hospital, Bodø, Norway
- Faculty of Health Sciences and K.G. Jebsen TREC, University of Tromsø, Tromsø, Norway
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway
| | - Rolf K Berge
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Azita Rashidi
- Research Institute of Internal Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Kari Otterdal
- Research Institute of Internal Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Ida Gregersen
- Research Institute of Internal Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Xiang Y Kong
- Research Institute of Internal Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Kirsten B Holven
- Department of Nutrition, Institute for Basic Medical Sciences, University of Oslo, Oslo, Norway
- Norwegian National Advisory Unit on Familial Hypercholesterolemia, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Department of Rheumatology, Dermatology and Infectious Diseases, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Faculty of Health Sciences and K.G. Jebsen TREC, University of Tromsø, Tromsø, Norway
| | - Børre Fevang
- Research Institute of Internal Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Department of Rheumatology, Dermatology and Infectious Diseases, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Silje F Jørgensen
- Research Institute of Internal Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Department of Rheumatology, Dermatology and Infectious Diseases, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
22
|
White CR, Datta G, Wilson L, Palgunachari MN, Anantharamaiah GM. The apoA-I mimetic peptide 4F protects apolipoprotein A-I from oxidative damage. Chem Phys Lipids 2019; 219:28-35. [PMID: 30707910 DOI: 10.1016/j.chemphyslip.2019.01.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 01/10/2019] [Accepted: 01/28/2019] [Indexed: 02/07/2023]
Abstract
High density lipoprotein (HDL) is prone to modification by the oxidizing and chlorinating agent hypochlorite anion (OCl-). Oxidation of apolipoprotein (apo) A-I, the major protein in HDL, reduces ABCA-1 mediated cholesterol efflux and other protective responses to HDL. The apoA-I mimetic peptide 4F has been shown to undergo oxidation; however, the ability of the peptide to mediate cholesterol efflux remains intact. Here, we show that 4F protects apoA-I from hypochlorite-mediated oxidation. Mass spectral analysis of apoA-I shows that tyrosine residues that are prone to hypochlorite-mediated chlorination are protected in the presence of 4F. Furthermore, 4F enhances the cholesterol efflux ability of apoA-I to a greater extent than either 4F or apoA-I alone, even after hypochlorite oxidation. These observations suggest that apoA-I in lipid complexes may be protected by the presence of 4F, resulting in the preservation of its anti-inflammatory and anti-atherogenic properties. These studies also form the basis for the future studies of nanoparticles possessing both apoA-I and 4F.
Collapse
Affiliation(s)
- C Roger White
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, United States
| | - Geeta Datta
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, United States
| | - Landon Wilson
- Targeted Metabolomics and Proteomics Laboratory, University of Alabama at Birmingham, Birmingham, AL, 35294, United States
| | - Mayakonda N Palgunachari
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, United States
| | - G M Anantharamaiah
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, United States; Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, 35294, United States.
| |
Collapse
|
23
|
Oxidized low density lipoproteins: The bridge between atherosclerosis and autoimmunity. Possible implications in accelerated atherosclerosis and for immune intervention in autoimmune rheumatic disorders. Autoimmun Rev 2018; 17:366-375. [DOI: 10.1016/j.autrev.2017.11.028] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 11/13/2017] [Indexed: 02/07/2023]
|
24
|
Casteleijn MG, Parkkila P, Viitala T, Koivuniemi A. Interaction of lecithin:cholesterol acyltransferase with lipid surfaces and apolipoprotein A-I-derived peptides. J Lipid Res 2018; 59:670-683. [PMID: 29438987 PMCID: PMC5880497 DOI: 10.1194/jlr.m082685] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 01/22/2018] [Indexed: 12/12/2022] Open
Abstract
LCAT is an enzyme responsible for the formation of cholesteryl esters from unesterified cholesterol (UC) and phospholipid (PL) molecules in HDL particles. However, it is poorly understood how LCAT interacts with lipoproteins and how apoA-I activates it. Here we have studied the interactions between LCAT and lipids through molecular simulations. In addition, we studied the binding of LCAT to apoA-I-derived peptides, and their effect on LCAT lipid association-utilizing experiments. Results show that LCAT anchors itself to lipoprotein surfaces by utilizing nonpolar amino acids located in the membrane-binding domain and the active site tunnel opening. Meanwhile, the membrane-anchoring hydrophobic amino acids attract cholesterol molecules next to them. The results also highlight the role of the lid-loop in the lipid binding and conformation of LCAT with respect to the lipid surface. The apoA-I-derived peptides from the LCAT-activating region bind to LCAT and promote its lipid surface interactions, although some of these peptides do not bind lipids individually. The transfer free-energy of PL from the lipid bilayer into the active site is consistent with the activation energy of LCAT. Furthermore, the entry of UC molecules into the active site becomes highly favorable by the acylation of SER181.
Collapse
Affiliation(s)
- Marco G Casteleijn
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Petteri Parkkila
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Tapani Viitala
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Artturi Koivuniemi
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
25
|
Haas MJ, Plazarte M, Chamseddin A, Onstead-Haas L, Wong NCW, Plazarte G, Mooradian AD. Inhibition of hepatic apolipoprotein A-I gene expression by histamine. Eur J Pharmacol 2018; 823:49-57. [PMID: 29378195 DOI: 10.1016/j.ejphar.2018.01.035] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 01/17/2018] [Accepted: 01/23/2018] [Indexed: 01/12/2023]
Abstract
In a recent high throughput analysis to identify drugs that alter hepatic apolipoprotein A-I (apo A-I) expression, histamine receptor one (H1) antagonists emerged as potential apo A-1 inducing drugs. Thus the present study was undertaken to identify some of the underlying molecular mechanisms of the effect of antihistaminic drugs on apo AI production. Apo A-I levels were measured by enzyme immunoassay and Western blots. Apo A-I mRNA levels were measured by reverse transcription real-time PCR using glyceraldehyde-3-phosphate dehydrogenase (GAPDH) mRNA as the internal control. The effects of histamine and antihistamines on apo A-I gene were determined by transient transfection of plasmids containing the apo A-I gene promoter. Histamine repressed while (H1) receptor antagonist azelastine increased apo A-I protein and mRNA levels within 48 h in a dose-dependent manner. Azelastine and histamine increased and suppressed, respectively, apo A-I gene promoter activity through a peroxisome proliferator activated receptor α response element. Treatment of HepG2 cells with other H1 receptor antagonists including fexofenadine, cetirizine, and diphenhydramine increased apo A-I levels in a dose-dependent manner while treatment with H2 receptor antagonists including cimetidine, famotidine, and ranitidine had no effect. We conclude that H1 receptor signaling is a novel pathway of apo A1 gene expression and therefore could be an important therapeutic target for enhancing de-novo apo A-1 synthesis.
Collapse
Affiliation(s)
- Michael J Haas
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Florida Jacksonville College of Medicine, 653-1 West 8th Street L14, Jacksonville, FL 32209, United States.
| | - Monica Plazarte
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Florida Jacksonville College of Medicine, 653-1 West 8th Street L14, Jacksonville, FL 32209, United States
| | - Ayham Chamseddin
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Florida Jacksonville College of Medicine, 653-1 West 8th Street L14, Jacksonville, FL 32209, United States
| | - Luisa Onstead-Haas
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Florida Jacksonville College of Medicine, 653-1 West 8th Street L14, Jacksonville, FL 32209, United States
| | - Norman C W Wong
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta, Canada
| | - Gabriela Plazarte
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Florida Jacksonville College of Medicine, 653-1 West 8th Street L14, Jacksonville, FL 32209, United States
| | - Arshag D Mooradian
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Florida Jacksonville College of Medicine, 653-1 West 8th Street L14, Jacksonville, FL 32209, United States
| |
Collapse
|
26
|
Li D, Fawaz MV, Morin EE, Sviridov D, Ackerman R, Olsen K, Remaley AT, Schwendeman A. Effect of Synthetic High Density Lipoproteins Modification with Polyethylene Glycol on Pharmacokinetics and Pharmacodynamics. Mol Pharm 2018; 15:83-96. [PMID: 29141139 PMCID: PMC6435036 DOI: 10.1021/acs.molpharmaceut.7b00734] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Synthetic high density lipoprotein nanoparticles (sHDLs) capable of mobilizing excess cholesterol from atherosclerotic arteries and delivering it to the liver for elimination have been shown to reduce plaque burden in patients. Unfortunately, sHDLs have a narrow therapeutic index and relative to the endogenous HDL shorter circulation half-life. Surface modification with polyethylene glycol (PEG) was investigated for its potential to extend sHDL circulation in vivo. Various amounts (2.5, 5, and 10%) and different chain lengths (2 and 5 kDa) of PEG-modified lipids were incorporated in sHDL's lipid membrane. Incorporating PEG did not reduce the ability of sHDL to facilitate cholesterol efflux, nor did it inhibit cholesterol uptake by the liver cells. By either adding more PEG or using PEG of longer chain lengths, the circulation half-life was extended. Addition of PEG also increased the area under the curve for the phospholipid component of sHDL (p < 0.05), but not for the apolipoprotein A-I peptide component of sHDL, suggesting sHDL is remodeled by endogenous lipoproteins in vivo. The extended phospholipid circulation led to a higher mobilization of plasma free cholesterol, a biomarker for facilitation of reverse cholesterol transport. The area under the cholesterol mobilization increased about 2-4-fold (p < 0.05), with greater increases observed for longer PEG chains and higher molar percentages of incorporated PEGylated lipids. Mobilized cholesterol was associated primarily with the HDL fraction, led to a transient increase in VLDL cholesterol, and returned to baseline 24 h postdose. Overall, PEGylation of sHDL led to beneficial changes in sHDL particle pharmacokinetic and pharmacodynamic behaviors.
Collapse
Affiliation(s)
- Dan Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, MI 48109
| | - Maria V. Fawaz
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, MI 48109
| | - Emily E. Morin
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, MI 48109
| | - Denis Sviridov
- National Heart, Lung and Blood Institute, National Institutes of Health, Building 10 – 2C433, 10 Center Drive, MSC 1666, Bethesda, MD 20892
| | - Rose Ackerman
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, MI 48109
| | - Karl Olsen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, MI 48109
| | - Alan T. Remaley
- National Heart, Lung and Blood Institute, National Institutes of Health, Building 10 – 2C433, 10 Center Drive, MSC 1666, Bethesda, MD 20892
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church Street, Ann Arbor, MI 48109
- Biointerfaces Institute, University of Michigan, NCRC, 2800 Plymouth Road, Ann Arbor, MI 48109
| |
Collapse
|
27
|
Kudinov VA, Zakharova TS, Torkhovskaya TI, Ipatova OM, Archakov AI. [Pharmacological targets for dislipidemies correction. Opportunities and prospects of therapeutic usage]. BIOMEDITSINSKAIA KHIMIIA 2018; 64:66-83. [PMID: 29460837 DOI: 10.18097/pbmc20186401066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Literature data on influence of existing and new groups of drug preparations for dyslipidemias correction are systemized, and molecular mechanisms of their effects are reviewed. The results of experimental and clinical investigations aimed at revealing of new pharmacological targets of dyslipidemias correction were analyzed. The approaches for activation of high density lipoproteins functionality are described. The implementation of alternative preparations with new alternative mechanisms of action may be suggested to improve the effectiveness of traditional treatment in the future.
Collapse
Affiliation(s)
- V A Kudinov
- Institute of Biomedical Chemistry, Moscow, Russia
| | | | | | - O M Ipatova
- Institute of Biomedical Chemistry, Moscow, Russia
| | - A I Archakov
- Institute of Biomedical Chemistry, Moscow, Russia
| |
Collapse
|
28
|
Antioxidative activity of high-density lipoprotein (HDL): Mechanistic insights into potential clinical benefit. BBA CLINICAL 2017; 8:66-77. [PMID: 28936395 PMCID: PMC5597817 DOI: 10.1016/j.bbacli.2017.07.002] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 07/21/2017] [Accepted: 07/24/2017] [Indexed: 12/29/2022]
Abstract
Uptake of low-density lipoprotein (LDL) particles by macrophages represents a key step in the development of atherosclerotic plaques, leading to the foam cell formation. Chemical modification of LDL is however necessary to induce this process. Proatherogenic LDL modifications include aggregation, enzymatic digestion and oxidation. LDL oxidation by one-electron (free radicals) and two-electron oxidants dramatically increases LDL affinity to macrophage scavenger receptors, leading to rapid LDL uptake and fatty streak formation. Circulating high-density lipoprotein (HDL) particles, primarily small, dense, protein-rich HDL3, provide potent protection of LDL from oxidative damage by free radicals, resulting in the inhibition of the generation of pro-inflammatory oxidized lipids. HDL-mediated inactivation of lipid hydroperoxides involves their initial transfer from LDL to HDL and subsequent reduction to inactive hydroxides by redox-active Met residues of apolipoprotein A-I. Several HDL-associated enzymes are present at elevated concentrations in HDL3 relative to large, light HDL2 and can be involved in the inactivation of short-chain oxidized phospholipids. Therefore, HDL represents a multimolecular complex capable of acquiring and inactivating proatherogenic lipids. Antioxidative function of HDL can be impaired in several metabolic and inflammatory diseases. Structural and compositional anomalies in the HDL proteome and lipidome underlie such functional deficiency. Concomitant normalization of the metabolism, circulating levels, composition and biological activities of HDL particles, primarily those of small, dense HDL3, can constitute future therapeutic target.
Collapse
|
29
|
Liu D, Wu M, Du Q, Ding Z, Qian M, Tong Z, Xu W, Zhang L, Chang H, Wang Y, Huang C, Lin D. The apolipoprotein A-I mimetic peptide, D-4F, restrains neointimal formation through heme oxygenase-1 up-regulation. J Cell Mol Med 2017; 21:3810-3820. [PMID: 28767201 PMCID: PMC5706511 DOI: 10.1111/jcmm.13290] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Accepted: 05/27/2017] [Indexed: 12/21/2022] Open
Abstract
D‐4F, an apolipoprotein A‐I (apoA‐I) mimetic peptide, possesses distinctly anti‐atherogenic effects. However, the biological functions and mechanisms of D‐4F on the hyperplasia of vascular smooth muscle cells (VSMCs) remain unclear. This study aimed to determine its roles in the proliferation and migration of VSMCs. In vitro, D‐4F inhibited VSMC proliferation and migration induced by ox‐LDL in a dose‐dependent manner. D‐4F up‐regulated heme oxygenase‐1 (HO‐1) expression in VSMCs, and the PI3K/Akt/AMP‐activated protein kinase (AMPK) pathway was involved in these processes. HO‐1 down‐regulation with siRNA or inhibition with zinc protoporphyrin (Znpp) impaired the protective effects of D‐4F on the oxidative stress and the proliferation and migration of VSMCs. Moreover, down‐regulation of ATP‐binding cassette transporter A1 (ABCA1) abolished the activation of Akt and AMPK, the up‐regulation of HO‐1 and the anti‐oxidative effects of D‐4F. In vivo, D‐4F restrained neointimal formation and oxidative stress of carotid arteries in balloon‐injured Sprague Dawley rats. And inhibition of HO‐1 with Znpp decreased the inhibitory effects of D‐4F on neointimal formation and ROS production in arteries. In conclusion, D‐4F inhibited VSMC proliferation and migration in vitro and neointimal formation in vivo through HO‐1 up‐regulation, which provided a novel prophylactic and therapeutic strategy for anti‐restenosis of arteries.
Collapse
Affiliation(s)
- Donghui Liu
- Department of Cardiology, The Affiliated Cardiovascular Hospital of Xiamen University, Medical College of Xiamen University, Xiamen, China
| | - Mengzhang Wu
- Department of Cardiology, The Affiliated Cardiovascular Hospital of Xiamen University, Medical College of Xiamen University, Xiamen, China.,Union Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Qian Du
- Department of Cardiology, The Affiliated Cardiovascular Hospital of Xiamen University, Medical College of Xiamen University, Xiamen, China
| | - Zhenzhen Ding
- Department of Cardiology, The Affiliated Cardiovascular Hospital of Xiamen University, Medical College of Xiamen University, Xiamen, China.,Union Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Mingming Qian
- Department of Cardiology, The Affiliated Cardiovascular Hospital of Xiamen University, Medical College of Xiamen University, Xiamen, China
| | - Zijia Tong
- Department of Cardiology, The Affiliated Cardiovascular Hospital of Xiamen University, Medical College of Xiamen University, Xiamen, China.,Union Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Wenqi Xu
- High-field NMR Research Center, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, China
| | - Le Zhang
- Department of Cardiology, The Affiliated Cardiovascular Hospital of Xiamen University, Medical College of Xiamen University, Xiamen, China
| | - He Chang
- Department of Cardiology, The Affiliated Cardiovascular Hospital of Xiamen University, Medical College of Xiamen University, Xiamen, China
| | - Yan Wang
- Department of Cardiology, The Affiliated Cardiovascular Hospital of Xiamen University, Medical College of Xiamen University, Xiamen, China
| | - Caihua Huang
- Department of Physical Education, Xiamen University of Technology, Xiamen, China
| | - Donghai Lin
- High-field NMR Research Center, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, China
| |
Collapse
|
30
|
Zhao Y, Leman LJ, Search DJ, Garcia RA, Gordon DA, Maryanoff BE, Ghadiri MR. Self-Assembling Cyclic d,l-α-Peptides as Modulators of Plasma HDL Function. A Supramolecular Approach toward Antiatherosclerotic Agents. ACS CENTRAL SCIENCE 2017; 3:639-646. [PMID: 28691076 PMCID: PMC5492419 DOI: 10.1021/acscentsci.7b00154] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Indexed: 05/26/2023]
Abstract
There is great interest in developing new modes of therapy for atherosclerosis to treat coronary heart disease and stroke, particularly ones that involve modulation of high-density lipoproteins (HDLs). Here, we describe a new supramolecular chemotype for altering HDL morphology and function. Guided by rational design and SAR-driven peptide sequence enumerations, we have synthesized and determined the HDL remodeling activities of over 80 cyclic d,l-α-peptides. We have identified a few distinct sequence motifs that are effective in vitro in remodeling human and mouse plasma HDLs to increase the concentration of lipid-poor pre-beta HDLs, which are key initial acceptors of cholesterol in the reverse cholesterol transport (RCT) process, and concomitantly promote cholesterol efflux from macrophage cells. Functional assays with various control peptides, such as scrambled sequences, linear and enantiomeric cyclic peptide variants, and backbone-modified structures that limit peptide self-assembly, provide strong support for the supramolecular mode of action. Importantly, when the lead cyclic peptide c[wLwReQeR] was administered to mice (ip), it also promoted the formation of small, lipid-poor HDLs in vivo, displayed good plasma half-life (∼6 h), did not appear to have adverse side effects, and exerted potent anti-inflammatory effects in an acute in vivo inflammation assay. Given that previously reported HDL remodeling peptides have been based on α-helical apoA-I mimetic architectures, the present study, involving a new structural class, represents a promising step toward new potential therapeutics to combat atherosclerosis.
Collapse
Affiliation(s)
- Yannan Zhao
- Department
of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Luke J. Leman
- Department
of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Debra J. Search
- Cardiovascular
Drug Discovery, Bristol-Myers Squibb Company, Pennington, New Jersey 08534, United States
| | - Ricardo A. Garcia
- Cardiovascular
Drug Discovery, Bristol-Myers Squibb Company, Pennington, New Jersey 08534, United States
| | - David A. Gordon
- Cardiovascular
Drug Discovery, Bristol-Myers Squibb Company, Pennington, New Jersey 08534, United States
| | - Bruce E. Maryanoff
- Department
of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - M. Reza Ghadiri
- Department
of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| |
Collapse
|
31
|
Liu D, Ding Z, Wu M, Xu W, Qian M, Du Q, Zhang L, Cui Y, Zheng J, Chang H, Huang C, Lin D, Wang Y. The apolipoprotein A-I mimetic peptide, D-4F, alleviates ox-LDL-induced oxidative stress and promotes endothelial repair through the eNOS/HO-1 pathway. J Mol Cell Cardiol 2017; 105:77-88. [PMID: 28274624 DOI: 10.1016/j.yjmcc.2017.01.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Revised: 12/30/2016] [Accepted: 01/27/2017] [Indexed: 02/08/2023]
Abstract
Apolipoprotein A-I (apoA-I) mimetic peptide exerts many anti-atherogenic properties. However, the underlying mechanisms related to the endothelial protective effects remain elusive. In this study, the apoA-I mimetic peptide, D-4F, was used. Proliferation assay, wound healing, and transwell migration experiments showed that D-4F improved the impaired endothelial proliferation and migration resulting from ox-LDL. Endothelial adhesion molecules expression and monocyte adhesion assay demonstrated that D-4F inhibited endothelial inflammation. Caspase-3 activation and TUNEL stain indicated that D-4F reduced endothelial cell apoptosis. A pivotal anti-oxidant enzyme, heme oxygenase-1 (HO-1) was upregulated by D-4F. The Akt/AMPK/eNOS pathways were involved in the expression of HO-1 induced by D-4F. Moreover, the anti-oxidation, pro-proliferation, and pro-migration capacities of D-4F were diminished by the inhibitors of both eNOS (L-NAME) and HO-1 (Znpp). Additionally, downregulation of ATP-binding cassette transporter A1 (ABCA1) by siRNA abolished the activation of Akt, AMPK and eNOS, and reduced the upregulation of HO-1 triggered by D-4F. Furthermore, D-4F promoted the reendothelialization of injured intima in carotid artery injury model of C57BL/6J mice in vivo. In summary, these findings suggested that D-4F might be a powerful candidate in the protection of endothelial cells and the prevention of cardiovascular disease (CVD).
Collapse
Affiliation(s)
- Donghui Liu
- Department of Cardiology, the Affiliated Xiamen Cardiovascular Hospital of Xiamen University, Medical College of Xiamen University, Xiamen 361004, China.
| | - Zhenzhen Ding
- Department of Cardiology, the Affiliated Xiamen Cardiovascular Hospital of Xiamen University, Medical College of Xiamen University, Xiamen 361004, China; Union Clinical Medical College of Fujian Medical University, Fuzhou 350001, China
| | - Mengzhang Wu
- Department of Cardiology, the Affiliated Xiamen Cardiovascular Hospital of Xiamen University, Medical College of Xiamen University, Xiamen 361004, China; Union Clinical Medical College of Fujian Medical University, Fuzhou 350001, China
| | - Wenqi Xu
- High-field NMR Research Center, MOE Key Laboratory of Spectrochemical Analysis& Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Mingming Qian
- Department of Cardiology, the Affiliated Xiamen Cardiovascular Hospital of Xiamen University, Medical College of Xiamen University, Xiamen 361004, China
| | - Qian Du
- Department of Cardiology, the Affiliated Xiamen Cardiovascular Hospital of Xiamen University, Medical College of Xiamen University, Xiamen 361004, China
| | - Le Zhang
- Department of Cardiology, the Affiliated Xiamen Cardiovascular Hospital of Xiamen University, Medical College of Xiamen University, Xiamen 361004, China
| | - Ye Cui
- Department of Ob/Gyn and Neonatal and Reproductive Medicine, the People's Liberation Army 174th Hospital and the Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Jianlan Zheng
- Department of Ob/Gyn and Neonatal and Reproductive Medicine, the People's Liberation Army 174th Hospital and the Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - He Chang
- Department of Cardiology, the Affiliated Xiamen Cardiovascular Hospital of Xiamen University, Medical College of Xiamen University, Xiamen 361004, China
| | - Caihua Huang
- Department of Physical Education, Xiamen University of Technology, Xiamen 361021, China
| | - Donghai Lin
- High-field NMR Research Center, MOE Key Laboratory of Spectrochemical Analysis& Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Yan Wang
- Department of Cardiology, the Affiliated Xiamen Cardiovascular Hospital of Xiamen University, Medical College of Xiamen University, Xiamen 361004, China.
| |
Collapse
|
32
|
Meriwether D, Sulaiman D, Wagner A, Grijalva V, Kaji I, Williams KJ, Yu L, Fogelman S, Volpe C, Bensinger SJ, Anantharamaiah GM, Shechter I, Fogelman AM, Reddy ST. Transintestinal transport of the anti-inflammatory drug 4F and the modulation of transintestinal cholesterol efflux. J Lipid Res 2016; 57:1175-93. [PMID: 27199144 DOI: 10.1194/jlr.m067025] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Indexed: 01/28/2023] Open
Abstract
The site and mechanism of action of the apoA-I mimetic peptide 4F are incompletely understood. Transintestinal cholesterol efflux (TICE) is a process involved in the clearance of excess cholesterol from the body. While TICE is responsible for at least 30% of the clearance of neutral sterols from the circulation into the intestinal lumen, few pharmacological agents have been identified that modulate this pathway. We show first that circulating 4F selectively targets the small intestine (SI) and that it is predominantly transported into the intestinal lumen. This transport of 4F into the SI lumen is transintestinal in nature, and it is modulated by TICE. We also show that circulating 4F increases reverse cholesterol transport from macrophages and cholesterol efflux from lipoproteins via the TICE pathway. We identify the cause of this modulation of TICE either as 4F being a cholesterol acceptor with respect to enterocytes, from which 4F enhances cholesterol efflux, or as 4F being an intestinal chaperone with respect to TICE. Our results assign a novel role for 4F as a modulator of the TICE pathway and suggest that the anti-inflammatory functions of 4F may be a partial consequence of the codependent intestinal transport of both 4F and cholesterol.
Collapse
Affiliation(s)
- David Meriwether
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA Department of Medical and Molecular Pharmacology, University of California Los Angeles, Los Angeles, CA
| | - Dawoud Sulaiman
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA Molecular Toxicology Interdepartmental Degree Program, University of California Los Angeles, Los Angeles, CA
| | - Alan Wagner
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Victor Grijalva
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Izumi Kaji
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Kevin J Williams
- Department of Medical and Molecular Pharmacology, University of California Los Angeles, Los Angeles, CA
| | - Liqing Yu
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD
| | - Spencer Fogelman
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Carmen Volpe
- Division of Laboratory Animal Medicine, University of California Los Angeles, Los Angeles, CA
| | - Steven J Bensinger
- Department of Medical and Molecular Pharmacology, University of California Los Angeles, Los Angeles, CA Department of Microbiology, Immunology and Molecular Genetics, University of California Los Angeles, Los Angeles, CA
| | - G M Anantharamaiah
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Ishaiahu Shechter
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Alan M Fogelman
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Srinivasa T Reddy
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA Department of Medical and Molecular Pharmacology, University of California Los Angeles, Los Angeles, CA Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| |
Collapse
|
33
|
Lee-Rueckert M, Escola-Gil JC, Kovanen PT. HDL functionality in reverse cholesterol transport--Challenges in translating data emerging from mouse models to human disease. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:566-83. [PMID: 26968096 DOI: 10.1016/j.bbalip.2016.03.004] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Revised: 02/26/2016] [Accepted: 03/04/2016] [Indexed: 12/18/2022]
Abstract
Whereas LDL-derived cholesterol accumulates in atherosclerotic lesions, HDL particles are thought to facilitate removal of cholesterol from the lesions back to the liver thereby promoting its fecal excretion from the body. Because generation of cholesterol-loaded macrophages is inherent to atherogenesis, studies on the mechanisms stimulating the release of cholesterol from these cells and its ultimate excretion into feces are crucial to learn how to prevent lesion development or even induce lesion regression. Modulation of this key anti-atherogenic pathway, known as the macrophage-specific reverse cholesterol transport, has been extensively studied in several mouse models with the ultimate aim of applying the emerging knowledge to humans. The present review provides a detailed comparison and critical analysis of the various steps of reverse cholesterol transport in mouse and man. We attempt to translate this in vivo complex scenario into practical concepts, which could serve as valuable tools when developing novel HDL-targeted therapies.
Collapse
|
34
|
Mooradian AD, Haas MJ. Targeting high-density lipoproteins: increasing de novo production versus decreasing clearance. Drugs 2016; 75:713-22. [PMID: 25895465 DOI: 10.1007/s40265-015-0390-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Although cardiovascular mortality has been decreasing in industrialized countries, there continues to be a substantial residual risk; thus, novel therapeutic agents and new targets of therapy have been sought. One highly plausible therapeutic target is high-density lipoprotein (HDL). HDL is a key player in reverse cholesterol transport and possesses a slew of other cardioprotective properties; however, recent trials with agents known to increase HDL levels have generally not shown any reduction in cardiovascular events. Further analysis of these trials suggest that fibrates have consistently reduced some cardiovascular outcomes, at least in the subgroup of patients with high serum triglycerides and low HDL cholesterol (HDLc) levels. Since fibrates, unlike niacin or cholesterol ester transfer protein inhibitors, increase HDLc level mostly through the stimulation of apolipoprotein A-I production, it is suggested that the quality and functionality of HDL are enhanced when de novo synthesis rather than inhibition of turnover is the mechanism of increasing HDL level. In this communication, the evidence for and against the cardioprotective properties of HDL is reviewed and the contemporary clinical trials are discussed.
Collapse
Affiliation(s)
- Arshag D Mooradian
- Department of Medicine, University of Florida College of Medicine, 655 West 11th Street, Jacksonville, FL, 32209, USA,
| | | |
Collapse
|
35
|
Zamanian-Daryoush M, DiDonato JA. Apolipoprotein A-I and Cancer. Front Pharmacol 2015; 6:265. [PMID: 26617517 PMCID: PMC4642354 DOI: 10.3389/fphar.2015.00265] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 10/23/2015] [Indexed: 12/22/2022] Open
Abstract
High-density lipoprotein (HDL) and apolipoprotein A-I (apoA-I), the predominant protein in plasma HDL, have long been the focus of intense studies in the field of atherosclerosis and cardiovascular disease. ApoA-I, in large part, is responsible for HDL assembly and its main atheroprotective function, that of shuttling excess cholesterol from peripheral tissues to the liver for excretion (reverse cholesterol transport). Recently, a protective role for HDL in cancer was suggested from several large clinical studies where an inverse relationship between plasma HDL-cholesterol (HDL-C) levels and risk of developing cancer was noted. This notion has now been tested and found to be supported in mouse tumor studies, where increasing levels of apoA-I/HDL were discovered to protect against tumor development and provision of human apoA-I was therapeutic against established tumors. This mini-review discusses the emerging role of apoA-I in tumor biology and its potential as cancer therapeutic.
Collapse
Affiliation(s)
- Maryam Zamanian-Daryoush
- Department of Cellular and Molecular Medicine, and Center for Cardiovascular Diagnostics and Prevention, Cleveland Clinic, Cleveland OH, USA
| | - Joseph A DiDonato
- Department of Cellular and Molecular Medicine, and Center for Cardiovascular Diagnostics and Prevention, Cleveland Clinic, Cleveland OH, USA
| |
Collapse
|
36
|
Lee GY, Kim JH, Choi KY, Yoon HY, Kim K, Kwon IC, Choi K, Lee BH, Park JH, Kim IS. Hyaluronic acid nanoparticles for active targeting atherosclerosis. Biomaterials 2015; 53:341-8. [DOI: 10.1016/j.biomaterials.2015.02.089] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 02/13/2015] [Accepted: 02/19/2015] [Indexed: 02/02/2023]
|
37
|
Navab M, Chattopadhyay A, Hough G, Meriwether D, Fogelman SI, Wagner AC, Grijalva V, Su F, Anantharamaiah GM, Hwang LH, Faull KF, Reddy ST, Fogelman AM. Source and role of intestinally derived lysophosphatidic acid in dyslipidemia and atherosclerosis. J Lipid Res 2015; 56:871-87. [PMID: 25646365 DOI: 10.1194/jlr.m056614] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
We previously reported that i) a Western diet increased levels of unsaturated lysophosphatidic acid (LPA) in small intestine and plasma of LDL receptor null (LDLR(-/-)) mice, and ii) supplementing standard mouse chow with unsaturated (but not saturated) LPA produced dyslipidemia and inflammation. Here we report that supplementing chow with unsaturated (but not saturated) LPA resulted in aortic atherosclerosis, which was ameliorated by adding transgenic 6F tomatoes. Supplementing chow with lysophosphatidylcholine (LysoPC) 18:1 (but not LysoPC 18:0) resulted in dyslipidemia similar to that seen on adding LPA 18:1 to chow. PF8380 (a specific inhibitor of autotaxin) significantly ameliorated the LysoPC 18:1-induced dyslipidemia. Supplementing chow with LysoPC 18:1 dramatically increased the levels of unsaturated LPA species in small intestine, liver, and plasma, and the increase was significantly ameliorated by PF8380 indicating that the conversion of LysoPC 18:1 to LPA 18:1 was autotaxin dependent. Adding LysoPC 18:0 to chow increased levels of LPA 18:0 in small intestine, liver, and plasma but was not altered by PF8380 indicating that conversion of LysoPC 18:0 to LPA 18:0 was autotaxin independent. We conclude that i) intestinally derived unsaturated (but not saturated) LPA can cause atherosclerosis in LDLR(-/-) mice, and ii) autotaxin mediates the conversion of unsaturated (but not saturated) LysoPC to LPA.
Collapse
Affiliation(s)
- Mohamad Navab
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - Arnab Chattopadhyay
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - Greg Hough
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - David Meriwether
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736 Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - Spencer I Fogelman
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - Alan C Wagner
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - Victor Grijalva
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - Feng Su
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - G M Anantharamaiah
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Lin H Hwang
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - Kym F Faull
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - Srinivasa T Reddy
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736 Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736 Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| | - Alan M Fogelman
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095-1736
| |
Collapse
|
38
|
Abstract
Besides their well-documented function of reverse transport of cholesterol, high-density lipoproteins (HDLs) display pleiotropic effects due to their antioxidant, antithrombotic, anti-inflammatory and antiapoptotic properties that may play a major protective role in acute stroke, in particular by limiting the deleterious effects of ischaemia on the blood-brain barrier (BBB) and on the parenchymal cerebral compartment. HDLs may also modulate leukocyte and platelet activation, which may also represent an important target that would justify the use of HDL-based therapy in acute stroke. In this review, we will present an update of all the recent findings in HDL biology that could support a potential clinical use of HDL therapy in ischaemic stroke.
Collapse
|
39
|
Amar MJA, Sakurai T, Sakurai-Ikuta A, Sviridov D, Freeman L, Ahsan L, Remaley AT. A novel apolipoprotein C-II mimetic peptide that activates lipoprotein lipase and decreases serum triglycerides in apolipoprotein E-knockout mice. J Pharmacol Exp Ther 2014; 352:227-35. [PMID: 25395590 DOI: 10.1124/jpet.114.220418] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Apolipoprotein A-I (apoA-I) mimetic peptides are currently being developed as possible new agents for the treatment of cardiovascular disease based on their ability to promote cholesterol efflux and their other beneficial antiatherogenic properties. Many of these peptides, however, have been reported to cause transient hypertriglyceridemia due to inhibition of lipolysis by lipoprotein lipase (LPL). We describe a novel bihelical amphipathic peptide (C-II-a) that contains an amphipathic helix (18A) for binding to lipoproteins and stimulating cholesterol efflux as well as a motif based on the last helix of apolipoprotein C-II (apoC-II) that activates lipolysis by LPL. The C-II-a peptide promoted cholesterol efflux from ATP-binding cassette transporter ABCA1-transfected BHK cells similar to apoA-I mimetic peptides. Furthermore, it was shown in vitro to be comparable to the full-length apoC-II protein in activating lipolysis by LPL. When added to serum from a patient with apoC-II deficiency, it restored normal levels of LPL-induced lipolysis and also enhanced lipolysis in serum from patients with type IV and V hypertriglyceridemia. Intravenous injection of C-II-a (30 mg/kg) in apolipoprotein E-knockout mice resulted in a significant reduction of plasma cholesterol and triglycerides of 38 ± 6% and 85 ± 7%, respectively, at 4 hours. When coinjected with the 5A peptide (60 mg/kg), the C-II-a (30 mg/kg) peptide was found to completely block the hypertriglyceridemic effect of the 5A peptide in C57Bl/6 mice. In summary, C-II-a is a novel peptide based on apoC-II, which promotes cholesterol efflux and lipolysis and may therefore be useful for the treatment of apoC-II deficiency and other forms of hypertriglyceridemia.
Collapse
Affiliation(s)
- Marcelo J A Amar
- Lipoprotein Metabolism Section, Cardio-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Toshihiro Sakurai
- Lipoprotein Metabolism Section, Cardio-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Akiko Sakurai-Ikuta
- Lipoprotein Metabolism Section, Cardio-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Denis Sviridov
- Lipoprotein Metabolism Section, Cardio-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Lita Freeman
- Lipoprotein Metabolism Section, Cardio-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Lusana Ahsan
- Lipoprotein Metabolism Section, Cardio-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Alan T Remaley
- Lipoprotein Metabolism Section, Cardio-Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
40
|
Barylski M, Toth PP, Nikolic D, Banach M, Rizzo M, Montalto G. Emerging therapies for raising high-density lipoprotein cholesterol (HDL-C) and augmenting HDL particle functionality. Best Pract Res Clin Endocrinol Metab 2014; 28:453-61. [PMID: 24840270 DOI: 10.1016/j.beem.2013.11.001] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
High-density lipoprotein (HDL) particles are highly complex polymolecular aggregates capable of performing a remarkable range of atheroprotective functions. Considerable research is being performed throughout the world to develop novel pharmacologic approaches to: (1) promote apoprotein A-I and HDL particle biosynthesis; (2) augment capacity for reverse cholesterol transport so as to reduce risk for the development and progression of atherosclerotic disease; and (3) modulate the functionality of HDL particles in order to increase their capacity to antagonize oxidation, inflammation, thrombosis, endothelial dysfunction, insulin resistance, and other processes that participate in arterial wall injury. HDL metabolism and the molecular constitution of HDL particles are highly complex and can change in response to both acute and chronic alterations in the metabolic milieu. To date, some of these interventions have been shown to positively impact rates of coronary artery disease progression. However, none of them have as yet been shown to significantly reduce risk for cardiovascular events. In the next 3-5 years a variety of pharmacologic interventions for modulating HDL metabolism and functionality will be tested in large, randomized, prospective outcomes trials. It is hoped that one or more of these therapeutic approaches will result in the ability to further reduce risk for cardiovascular events once low-density lipoprotein cholesterol and non-HDL-cholesterol targets have been attained.
Collapse
Affiliation(s)
- Marcin Barylski
- Department of Internal Medicine and Cardiological Rehabilitation, Medical University of Lodz, Lodz, Poland.
| | - Peter P Toth
- CGH Medical Center, Sterling, IL 61081, USA; University of Illinois School of Medicine, Peoria, IL, USA.
| | - Dragana Nikolic
- Biomedical Department of Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy.
| | - Maciej Banach
- Nephrology and Hypertension, Medical University of Lodz, Zeromskiego 113, 90-549 Lodz, Poland.
| | - Manfredi Rizzo
- Biomedical Department of Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy; Euro-Mediterranean Institute of Science and Technology, Palermo, Italy.
| | - Giuseppe Montalto
- Biomedical Department of Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy.
| |
Collapse
|
41
|
Abstract
High-density lipoprotein (HDL) is a complex mixture of lipoproteins that is associated with many minor proteins and lipids that influence the function of HDL. Although HDL is a promising marker and potential therapeutic target based on its epidemiological data and the effects of healthy HDL in vitro in endothelial cells and macrophages, as well as based on infusion studies of reconstituted HDL in patients with hypercholesterolemia, it remains still uncertain whether or not HDL cholesterol–raising drugs will improve outcomes. Recent studies suggest that HDL becomes modified in patients with coronary artery disease or acute coronary syndrome because of oxidative processes that result in alterations in its proteome composition (proteome remodelling) leading to HDL dysfunction.
Collapse
Affiliation(s)
- Thomas F. Lüscher
- From Department of Cardiology, University Heart Center (T.F.L., U.L.), and Department of Clinical Chemistry (A.v.E.), University Hospital Zurich, Zurich, Switzerland; Division of Cardiovascular Research, Institute of Physiology, University of Zurich, Zurich, Switzerland (T.F.L., U.L.); and Department of Medicine, University of California, Los Angeles, CA (A.M.F.)
| | - Ulf Landmesser
- From Department of Cardiology, University Heart Center (T.F.L., U.L.), and Department of Clinical Chemistry (A.v.E.), University Hospital Zurich, Zurich, Switzerland; Division of Cardiovascular Research, Institute of Physiology, University of Zurich, Zurich, Switzerland (T.F.L., U.L.); and Department of Medicine, University of California, Los Angeles, CA (A.M.F.)
| | - Arnold von Eckardstein
- From Department of Cardiology, University Heart Center (T.F.L., U.L.), and Department of Clinical Chemistry (A.v.E.), University Hospital Zurich, Zurich, Switzerland; Division of Cardiovascular Research, Institute of Physiology, University of Zurich, Zurich, Switzerland (T.F.L., U.L.); and Department of Medicine, University of California, Los Angeles, CA (A.M.F.)
| | - Alan M. Fogelman
- From Department of Cardiology, University Heart Center (T.F.L., U.L.), and Department of Clinical Chemistry (A.v.E.), University Hospital Zurich, Zurich, Switzerland; Division of Cardiovascular Research, Institute of Physiology, University of Zurich, Zurich, Switzerland (T.F.L., U.L.); and Department of Medicine, University of California, Los Angeles, CA (A.M.F.)
| |
Collapse
|
42
|
|
43
|
Valanti E, Tsompanidis A, Sanoudou D. Pharmacogenomics in the development and characterization of atheroprotective drugs. Methods Mol Biol 2014; 1175:259-300. [PMID: 25150873 DOI: 10.1007/978-1-4939-0956-8_11] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Atherosclerosis is the main cause of cardiovascular disease (CVD) and can lead to stroke, myocardial infarction, and death. The clinically available atheroprotective drugs aim mainly at reducing the levels of circulating low-density lipoprotein (LDL), increasing high-density lipoprotein (HDL), and attenuating inflammation. However, the cardiovascular risk remains high, along with morbidity, mortality, and incidence of adverse drug events. Pharmacogenomics is increasingly contributing towards the characterization of existing atheroprotective drugs, the evaluation of novel ones, and the identification of promising, unexplored therapeutic targets, at the global molecular pathway level. This chapter presents highlights of pharmacogenomics investigations and discoveries that have contributed towards the elucidation of pharmacological atheroprotection, while opening the way to new therapeutic approaches.
Collapse
Affiliation(s)
- Efi Valanti
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Athens, 115 27, Greece
| | | | | |
Collapse
|
44
|
Sharifov OF, Nayyar G, Ternovoy VV, Palgunachari MN, Garber DW, Anantharamaiah G, Gupta H. Comparison of anti-endotoxin activity of apoE and apoA mimetic derivatives of a model amphipathic peptide 18A. Innate Immun 2013; 20:867-80. [PMID: 24323453 DOI: 10.1177/1753425913514621] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Endotoxemia is a major cause of chronic inflammation, and is an important pathogenic factor in the development of metabolic syndrome and atherosclerosis. Human apolipoprotein E (apoE) and apoA-I are protein components of high-density lipoprotein, which have strong anti-endotoxin activity. Here, we compared anti-endotoxin activity of Ac-hE18A-NH2 and 4F peptides, modified from model amphipathic helical 18A peptide, to mimic, respectively, apoE and apoA-I properties. Ac-hE18A-NH2, stronger than 4F, inhibited endotoxin activity and disaggregated Escherichia coli 055:B5 (wild smooth serotype). Ac-hE18A-NH2 and 4F inhibited endotoxin activity of E. coli 026:B6 (rough-like serotype) to a similar degree. This suggests that Ac-hE18A-NH2 as a dual-domain molecule might interact with both the lipid A and headgroup of smooth LPS, whereas 4F binds lipid A. In C57BL/6 mice, Ac-hE18A-NH2 was superior to 4F in inhibiting the inflammatory responses mediated by E. coli 055:B5, but not E. coli 026:B6. However, in THP-1 cells, isolated human primary leukocytes, and whole human blood, Ac-hE18A-NH2 reduced responses more strongly than 4F to both E. coli serotypes either when peptides were pre-incubated or co-incubated with LPS, indicating that Ac-hE18A-NH2 also has strong anti-inflammatory effects independent of endotoxin-neutralizing properties. In conclusion, Ac-hE18A-NH2 is more effective than 4F in inhibiting LPS-mediated inflammation, which opens prospective clinical applications for Ac-hE18A-NH2.
Collapse
Affiliation(s)
- Oleg F Sharifov
- Department of Medicine, University of Alabama at Birmingham, AL, USA
| | - Gaurav Nayyar
- Department of Medicine, University of Alabama at Birmingham, AL, USA
| | | | | | - David W Garber
- Department of Medicine, University of Alabama at Birmingham, AL, USA
| | - Gm Anantharamaiah
- Department of Medicine, University of Alabama at Birmingham, AL, USA Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, AL, USA
| | - Himanshu Gupta
- Department of Medicine, University of Alabama at Birmingham, AL, USA VA Medical Center, Birmingham, AL, USA
| |
Collapse
|
45
|
Leman LJ, Maryanoff BE, Ghadiri MR. Molecules that mimic apolipoprotein A-I: potential agents for treating atherosclerosis. J Med Chem 2013; 57:2169-96. [PMID: 24168751 DOI: 10.1021/jm4005847] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Certain amphipathic α-helical peptides can functionally mimic many of the properties of full-length apolipoproteins, thereby offering an approach to modulate high-density lipoprotein (HDL) for combating atherosclerosis. In this Perspective, we summarize the key findings and advances over the past 25 years in the development of peptides that mimic apolipoproteins, especially apolipoprotein A-I (apoA-I). This assemblage of information provides a reasonably clear picture of the state of the art in the apolipoprotein mimetic field, an appreciation of the potential for such agents in pharmacotherapy, and a sense of the opportunities for optimizing the functional properties of HDL.
Collapse
Affiliation(s)
- Luke J Leman
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute , 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | | | | |
Collapse
|
46
|
Hafiane A, Genest J. HDL, Atherosclerosis, and Emerging Therapies. CHOLESTEROL 2013; 2013:891403. [PMID: 23781332 PMCID: PMC3678415 DOI: 10.1155/2013/891403] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 04/22/2013] [Accepted: 04/30/2013] [Indexed: 12/21/2022]
Abstract
This review aims to provide an overview on the properties of high-density lipoproteins (HDLs) and their cardioprotective effects. Emergent HDL therapies will be presented in the context of the current understanding of HDL function, metabolism, and protective antiatherosclerotic properties. The epidemiological association between levels of HDL-C or its major apolipoprotein (apoA-I) is strong, graded, and coherent across populations. HDL particles mediate cellular cholesterol efflux, have antioxidant properties, and modulate vascular inflammation and vasomotor function and thrombosis. A link of causality has been cast into doubt with Mendelian randomization data suggesting that genes causing HDL-C deficiency are not associated with increased cardiovascular risk, nor are genes associated with increased HDL-C, with a protective effect. Despite encouraging data from small studies, drugs that increase HDL-C levels have not shown an effect on major cardiovascular end-points in large-scale clinical trials. It is likely that the cholesterol mass within HDL particles is a poor biomarker of therapeutic efficacy. In the present review, we will focus on novel therapeutic avenues and potential biomarkers of HDL function. A better understanding of HDL antiatherogenic functions including reverse cholesterol transport, vascular protective and antioxidation effects will allow novel insight on novel, emergent therapies for cardiovascular prevention.
Collapse
Affiliation(s)
| | - Jacques Genest
- Faculty of Medicine, Center for Innovative Medicine, McGill University Health Center, Royal Victoria Hospital, McGill University, 687 Pine Avenue West, Montreal, QC, Canada H3A 1A1
| |
Collapse
|
47
|
Damiano MG, Mutharasan RK, Tripathy S, McMahon KM, Thaxton CS. Templated high density lipoprotein nanoparticles as potential therapies and for molecular delivery. Adv Drug Deliv Rev 2013; 65:649-62. [PMID: 22921597 DOI: 10.1016/j.addr.2012.07.013] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Revised: 07/13/2012] [Accepted: 07/23/2012] [Indexed: 01/04/2023]
Abstract
High density lipoproteins (HDLs) are dynamic natural nanoparticles best known for their role in cholesterol transport and the inverse correlation that exists between blood HDL levels and the risk of developing coronary heart disease. In addition, enhanced HDL-cholesterol uptake has been demonstrated in several human cancers. As such, the use of HDL as a therapeutic and as a vehicle for systemic delivery of drugs and as imaging agents is increasingly important. HDLs exist on a continuum from the secreted HDL-scaffolding protein, apolipoprotein A-1 (Apo A1), to complex, spherical "mature" HDLs. Aspects of HDL particles including their size, shape, and surface chemical composition are being recognized as critical to their diverse biological functions. Here we review HDL biology; strategies for synthesizing HDLs; data supporting the clinical use and benefit of directly administered HDL; a rationale for developing synthetic methods for spherical, mature HDLs; and, the potential to employ HDLs as therapies, imaging agents, and drug delivery vehicles. Importantly, methods that utilize nanoparticle templates to control synthetic HDL size, shape, and surface chemistry are highlighted.
Collapse
Affiliation(s)
- Marina G Damiano
- Department of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208, USA
| | | | | | | | | |
Collapse
|
48
|
Mahdy Ali K, Wonnerth A, Huber K, Wojta J. Cardiovascular disease risk reduction by raising HDL cholesterol--current therapies and future opportunities. Br J Pharmacol 2013; 167:1177-94. [PMID: 22725625 DOI: 10.1111/j.1476-5381.2012.02081.x] [Citation(s) in RCA: 164] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Since the first discovery of an inverse correlation between high-density lipoprotein-cholesterol (HDL-C) levels and coronary heart disease in the 1950s the life cycle of HDL, its role in atherosclerosis and the therapeutic modification of HDL-C levels have been major research topics. The Framingham study and others that followed could show that HDL-C is an independent cardiovascular risk factor and that the increase of HDL-C of only 10 mg·L(-1) leads to a risk reduction of 2-3%. While statin therapy and therefore low-density lipoprotein-cholesterol (LDL-C) reduction could lower coronary heart disease considerably; cardiovascular morbidity and mortality still occur in a significant portion of subjects already receiving therapy. Therefore, new strategies and therapies are needed to further reduce the risk. Raising HDL-C was thought to achieve this goal. However, established drug therapies resulting in substantial HDL-C increase are scarce and their effect is controversial. Furthermore, it is becoming increasingly evident that HDL particle functionality is at least as important as HDL-C levels since HDL particles not only promote reverse cholesterol transport from the periphery (mainly macrophages) to the liver but also exert pleiotropic effects on inflammation, haemostasis and apoptosis. This review deals with the biology of HDL particles, the established and future therapeutic options to increase HDL-C and discusses the results and conclusions of the most important studies published in the last years. Finally, an outlook on future diagnostic tools and therapeutic opportunities regarding coronary artery disease is given.
Collapse
Affiliation(s)
- K Mahdy Ali
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | | | | | | |
Collapse
|
49
|
High-density lipoprotein in uremic patients: metabolism, impairment, and therapy. Int Urol Nephrol 2013; 46:27-39. [PMID: 23443874 DOI: 10.1007/s11255-012-0366-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Accepted: 12/12/2012] [Indexed: 01/21/2023]
Abstract
Several studies have shown that HDL has altered antioxidant and anti-inflammatory effects in chronic uremia, either by the reduction in its antioxidant enzymes or by the impairment of their activity. Systemic oxidative stress, which is highly prevalent in chronic kidney disease (CKD) patients, has been shown to decrease antioxidant and anti-inflammatory effects of HDL and even transform it into a pro-oxidant and pro-inflammatory agent. For this reason, we believe that the propensity for accelerated cardiovascular disease in CKD is facilitated by a few key features of this disease, namely, oxidative stress, inflammation, hypertension, and disorders of lipid metabolism. In a nutshell, oxidative stress and inflammation enhance atherosclerosis leading to increased cardiovascular mortality and morbidity in this population. In this detailed review, we highlight the current knowledge on HDL dysfunction and impairment in chronic kidney disease as well as the available therapy.
Collapse
|
50
|
Chattopadhyay A, Navab M, Hough G, Gao F, Meriwether D, Grijalva V, Springstead JR, Palgnachari MN, Namiri-Kalantari R, Su F, Van Lenten BJ, Wagner AC, Anantharamaiah GM, Farias-Eisner R, Farias-Eisener R, Reddy ST, Fogelman AM. A novel approach to oral apoA-I mimetic therapy. J Lipid Res 2013; 54:995-1010. [PMID: 23378594 DOI: 10.1194/jlr.m033555] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Transgenic tomato plants were constructed with an empty vector (EV) or a vector expressing an apoA-I mimetic peptide, 6F. EV or 6F tomatoes were harvested, lyophilized, ground into powder, added to Western diet (WD) at 2.2% by weight, and fed to LDL receptor-null (LDLR(-/-)) mice at 45 mg/kg/day 6F. After 13 weeks, the percent of the aorta with lesions was 4.1 ± 4%, 3.3 ± 2.4%, and 1.9 ± 1.4% for WD, WD + EV, and WD + 6F, respectively (WD + 6F vs. WD, P = 0.0134; WD + 6F vs. WD + EV, P = 0.0386; WD + EV vs. WD, not significant). While body weight did not differ, plasma serum amyloid A (SAA), total cholesterol, triglycerides, and lysophosphatidic acid (LPA) levels were less in WD + 6F mice; P < 0.0295. HDL cholesterol and paroxonase-1 activity (PON) were higher in WD + 6F mice (P = 0.0055 and P = 0.0254, respectively), but not in WD + EV mice. Plasma SAA, total cholesterol, triglycerides, LPA, and 15-hydroxyeicosatetraenoic acid (HETE) levels positively correlated with lesions (P < 0.0001); HDL cholesterol and PON were inversely correlated (P < 0.0001). After feeding WD + 6F: i) intact 6F was detected in small intestine (but not in plasma); ii) small intestine LPA was decreased compared with WD + EV (P < 0.0469); and iii) small intestine LPA 18:2 positively correlated with the percent of the aorta with lesions (P < 0.0179). These data suggest that 6F acts in the small intestine and provides a novel approach to oral apoA-I mimetic therapy.
Collapse
Affiliation(s)
- Arnab Chattopadhyay
- Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|