1
|
Yurkina DM, Shcherbakov KA, Romanova EA, Tvorogova AV, Feoktistov AM, Georgiev GP, Yashin DV, Sashchenko LP. Shortened PGLYRP1 Peptides Regulate Antitumor Activity of Cytotoxic Lymphocytes via TREM-1 Receptor: From Biology to Bioinformatics. Int J Mol Sci 2025; 26:4069. [PMID: 40362307 PMCID: PMC12071940 DOI: 10.3390/ijms26094069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/14/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
The pro-inflammatory immune response plays an important role in protecting the body from pathogens and tumors. In this study, we were able to identify three peptides of the innate immunity protein PGLYRP1 (Tag7) that could regulate the activity of the TREM-1 receptor. TREM-1 receptor activation on monocytes triggers the appearance of antitumor lymphocytes. All three peptides studied (17.0, N9, and N15) bind with the TREM-1 receptor with the Kds 1.32 ± 0.2 nM, 9.66 ± 0.5 nM, and 7.43 ± 0.4 nM, respectively. An N9 peptide inhibiting the activity of the receptor was identified in addition to two peptides (N9 and N15) that jointly trigger the activation of the receptor. The conducted molecular docking study revealed amino acid residues (Ile57, Ile58, Glu106, Ser108, Leu110, Tyr116, Pro118, Pro119, Arg130, and Val 132), necessary for various functions of peptides, providing important knowledge for understanding the mechanism of activation of this receptor that can also serve as a basis for the development of therapeutic drugs to regulate its activity in the treatment of autoimmune diseases and tumors.
Collapse
Affiliation(s)
- Daria M. Yurkina
- Institute of Gene Biology (RAS), 119334 Moscow, Russia; (D.M.Y.); (K.A.S.); (E.A.R.); (A.V.T.); (A.M.F.); (G.P.G.); (L.P.S.)
| | - Kirill A. Shcherbakov
- Institute of Gene Biology (RAS), 119334 Moscow, Russia; (D.M.Y.); (K.A.S.); (E.A.R.); (A.V.T.); (A.M.F.); (G.P.G.); (L.P.S.)
| | - Elena A. Romanova
- Institute of Gene Biology (RAS), 119334 Moscow, Russia; (D.M.Y.); (K.A.S.); (E.A.R.); (A.V.T.); (A.M.F.); (G.P.G.); (L.P.S.)
| | - Anna V. Tvorogova
- Institute of Gene Biology (RAS), 119334 Moscow, Russia; (D.M.Y.); (K.A.S.); (E.A.R.); (A.V.T.); (A.M.F.); (G.P.G.); (L.P.S.)
| | - Alexey M. Feoktistov
- Institute of Gene Biology (RAS), 119334 Moscow, Russia; (D.M.Y.); (K.A.S.); (E.A.R.); (A.V.T.); (A.M.F.); (G.P.G.); (L.P.S.)
- Institute of Molecular Biology (RAS), 119334 Moscow, Russia
| | - Georgii P. Georgiev
- Institute of Gene Biology (RAS), 119334 Moscow, Russia; (D.M.Y.); (K.A.S.); (E.A.R.); (A.V.T.); (A.M.F.); (G.P.G.); (L.P.S.)
| | - Denis V. Yashin
- Institute of Gene Biology (RAS), 119334 Moscow, Russia; (D.M.Y.); (K.A.S.); (E.A.R.); (A.V.T.); (A.M.F.); (G.P.G.); (L.P.S.)
| | - Lidia P. Sashchenko
- Institute of Gene Biology (RAS), 119334 Moscow, Russia; (D.M.Y.); (K.A.S.); (E.A.R.); (A.V.T.); (A.M.F.); (G.P.G.); (L.P.S.)
| |
Collapse
|
2
|
Temur BZ, Timucin AC, Atik AE, Kocagoz T, Can O. Peptide-Based Regulation of TNF-α-Mediated Cytotoxicity. Biomolecules 2025; 15:559. [PMID: 40338229 PMCID: PMC12024540 DOI: 10.3390/biom15040559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 05/09/2025] Open
Abstract
Tumor necrosis factor alpha (TNF-α) is a pro-inflammatory cytokine associated with TNF receptor 1 (TNFR1) and TNF receptor 2 (TNFR2), which play important roles in several inflammatory diseases. There is a growing interest in developing alternative molecules that can be used as TNF blockers. In this study, we focused on TNF-α-, TNFR1-, and TNFR2-mimicking peptides to inhibit TNF-α receptor binding in various ways. Six peptides (OB1, OB2, OB5, OB6, OB7, and OB8) were developed to bind TNFR1, TNFR2, and TNF-α. OB1 and OB2 bound to TNF-α with lower Kd values of 300 and 46.7 nM, respectively, compared to previously published sequences. These synthetic peptides directly and indirectly inhibited TNF-α in vitro without cytotoxicity to L929 cells, and OB1 significantly inhibited apoptosis in the presence of hTNF-α. Peptides developed in this study may prove to be useful for therapeutic inhibition of TNF-α.
Collapse
MESH Headings
- Tumor Necrosis Factor-alpha/metabolism
- Tumor Necrosis Factor-alpha/pharmacology
- Peptides/pharmacology
- Peptides/chemistry
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Receptors, Tumor Necrosis Factor, Type I/antagonists & inhibitors
- Receptors, Tumor Necrosis Factor, Type II/metabolism
- Receptors, Tumor Necrosis Factor, Type II/antagonists & inhibitors
- Receptors, Tumor Necrosis Factor, Type II/chemistry
- Apoptosis/drug effects
- Animals
- Mice
- Humans
- Cell Line
- Protein Binding
Collapse
Affiliation(s)
- Betul Zehra Temur
- Department of Medical Biotechnology, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Atasehir, 34752 Istanbul, Turkey; (B.Z.T.); (T.K.)
| | - Ahmet Can Timucin
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Acibadem Mehmet Ali Aydinlar University, Atasehir, 34752 İstanbul, Turkey;
- Graduate School of Natural and Applied Sciences, Acibadem Mehmet Ali Aydinlar University, Atasehir, 34752 İstanbul, Turkey
| | - Ahmet Emin Atik
- Turgut Ilaclari A.S., 41400 Kocaeli, Turkey;
- Department of Natural Sciences, Faculty of Engineering and Natural Sciences, Acibadem Mehmet Ali Aydinlar University, Atasehir, 34752 Istanbul, Turkey
| | - Tanil Kocagoz
- Department of Medical Biotechnology, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Atasehir, 34752 Istanbul, Turkey; (B.Z.T.); (T.K.)
- Departmen of Medical Microbiology, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Atasehir, 34752 Istanbul, Turkey
| | - Ozge Can
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Acibadem Mehmet Ali Aydinlar University, Atasehir, 34752 Istanbul, Turkey
| |
Collapse
|
3
|
Félix Vélez NE, Tu K, Guo P, Reeves RR, Aguado BA. Secreted Cytokines From Inflammatory Macrophages Modulate Sex Differences in Valvular Interstitial Cells on Hydrogel Biomaterials. J Biomed Mater Res A 2025; 113:e37885. [PMID: 39995146 PMCID: PMC11875511 DOI: 10.1002/jbm.a.37885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/18/2024] [Accepted: 02/04/2025] [Indexed: 02/26/2025]
Abstract
Patients with aortic valve stenosis (AVS) experience fibrosis and/or calcification in valve tissue, which leads to heart failure if left untreated. Inflammation is a hallmark of AVS, and secreted cytokines from pro-inflammatory macrophages are thought to contribute to valve fibro-calcification by driving the activation of valvular interstitial cells (VICs) to myofibroblasts. However, the molecular mechanisms by which inflammatory cytokines differentially regulate myofibroblast activation as a function of biological sex are not fully defined. Here, we developed an in vitro hydrogel culture platform to culture male and female valvular interstitial cells (VICs) and characterize the sex-specific effects of inflammatory cytokines on VIC activation to myofibroblasts and osteoblast-like cells. Our data reveal that tumor necrosis factor alpha (TNF-α) modulates female-specific myofibroblast activation via MAPK/ERK signaling, nuclear chromatin availability, and osteoblast-like differentiation via RUNX2 nuclear localization. In parallel, our data also suggest that male-specific myofibroblast deactivation in response to TNF-α occurs via alternative pathways outside of MAPK/ERK signaling. Collectively, hydrogel biomaterials as cell culture platforms are critical for distinguishing sex differences in cellular phenotypes.
Collapse
Affiliation(s)
- Nicole E. Félix Vélez
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92037, USA
| | - Kristi Tu
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92037, USA
| | - Peng Guo
- Nikon Imaging Center, Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Ryan R. Reeves
- Sulpizio Cardiovascular Center, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Brian A. Aguado
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92037, USA
- Program in Materials Science and Engineering, University of California, San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
4
|
Relouw FJA, Kox M, Taal HR, Koch BCP, Prins MWJ, van Riel NAW. Mathematical model of the inflammatory response to acute and prolonged lipopolysaccharide exposure in humans. NPJ Syst Biol Appl 2024; 10:146. [PMID: 39638779 PMCID: PMC11621538 DOI: 10.1038/s41540-024-00473-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 11/18/2024] [Indexed: 12/07/2024] Open
Abstract
One in five deaths worldwide is associated with sepsis, which is defined as organ dysfunction caused by a dysregulated host response to infection. An increased understanding of the pathophysiology of sepsis could provide improved approaches for early detection and treatment. Here we describe the development and validation of a mechanistic mathematical model of the inflammatory response, making use of a combination of in vitro and human in vivo data obtained from experiments where bacterial lipopolysaccharide (LPS) was used to induce an inflammatory response. The new model can simulate the responses to both acute and prolonged inflammatory stimuli in an experimental setting, as well as the response to infection in the clinical setting. This model serves as a foundation for a sepsis simulation model with a potentially wide range of applications in different disciplines involved with sepsis research.
Collapse
Affiliation(s)
- Freek J A Relouw
- Department of Intensive Care Medicine, Radboud university medical center, Nijmegen, The Netherlands.
- Department of Neonatal and Paediatric Intensive Care, Division of Neonatology, Erasmus University Medical Center, Rotterdam, The Netherlands.
- Department of Hospital Pharmacy, Erasmus University Medical Center, Rotterdam, The Netherlands.
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.
- Institute of Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands.
| | - Matthijs Kox
- Department of Intensive Care Medicine, Radboud university medical center, Nijmegen, The Netherlands
| | - H Rob Taal
- Department of Neonatal and Paediatric Intensive Care, Division of Neonatology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Birgit C P Koch
- Department of Hospital Pharmacy, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Menno W J Prins
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
- Institute of Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
- Department of Applied Physics, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Natal A W van Riel
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.
- Institute of Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands.
| |
Collapse
|
5
|
Nolan RP, Printz MA. Modeling the subcutaneous pharmacokinetics of antibodies co-administered with rHuPH20. Clin Transl Sci 2024; 17:e13788. [PMID: 38561908 PMCID: PMC10985223 DOI: 10.1111/cts.13788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/15/2024] [Accepted: 03/20/2024] [Indexed: 04/04/2024] Open
Abstract
Predicting the subcutaneous (SC) pharmacokinetics (PK) of antibodies in humans is challenging, with clinical data currently being the only reliable data source for modeling SC absorption and bioavailability. Recombinant human hyaluronidase PH20 (rHuPH20) is an enzyme that facilitates SC delivery of high-dose, high-volume therapeutics. Numerous monoclonal antibodies have been co-administered SC with rHuPH20 in a clinical setting, establishing an extensive PK database. The goal of this work is to demonstrate how aggregated clinical data can be leveraged in a universal modeling framework for characterizing SC antibody PK, resulting in parameterization that can be used in predictive simulations of new antibodies. Data for 10 individual antibodies co-administered SC with rHuPH20 were obtained from publicly available sources. PK modeling of each antibody was conducted using the same model structure, but uniquely parameterized. The model structure consisted of a two-compartment model to capture linear kinetics, plus a target-binding mechanism to accommodate nonlinear kinetics driven by antibody-target complex formation and elimination. The clinical PK profiles for all antibodies were accurately described using the universal modeling framework. The SC PK parameters of absorption and bioavailability were consistent across the range of antibody and target properties evaluated. SC administration with rHuPH20 yielded a 30% increase in absorption rate on average and similar or better bioavailability. These parameter values can serve as initial conditions for model-based PK predictions for new antibodies co-administered SC with rHuPH20 to enable evaluation of optimal SC dose and schedule regimens prior to and during clinical development.
Collapse
|
6
|
Yurkina DM, Romanova EA, Tvorogova AV, Naydenysheva ZK, Feoktistov AV, Yashin DV, Sashchenko LP. The 12-Membered TNFR1 Peptide, as Well as the 16-Membered and 6-Membered TNF Peptides, Regulate TNFR1-Dependent Cytotoxic Activity of TNF. Int J Mol Sci 2024; 25:3900. [PMID: 38612709 PMCID: PMC11011327 DOI: 10.3390/ijms25073900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/29/2024] [Accepted: 03/30/2024] [Indexed: 04/14/2024] Open
Abstract
Understanding the exact mechanisms of the activation of proinflammatory immune response receptors is very important for the targeted regulation of their functioning. In this work, we were able to identify the sites of the molecules in the proinflammatory cytokine TNF (tumor necrosis factor) and its TNFR1 (tumor necrosis factor receptor 1), which are necessary for the two-stage cytotoxic signal transduction required for tumor cell killing. A 12-membered TNFR1 peptide was identified and synthesized, interacting with the ligands of this receptor protein's TNF and Tag7 and blocking their binding to the receptor. Two TNF cytokine peptides interacting with different sites of TNFR1 receptors were identified and synthesized. It has been demonstrated that the long 16-membered TNF peptide interferes with the binding of TNFR1 ligands to this receptor, and the short 6-membered peptide interacts with the receptor site necessary for the transmission of a cytotoxic signal into the cell after the ligands' interaction with the binding site. This study may help in the development of therapeutic approaches to regulate the activity of the cytokine TNF.
Collapse
Affiliation(s)
- Daria M. Yurkina
- Institute of Gene Biology (RAS), Moscow 119334, Russia; (D.M.Y.); (E.A.R.); (Z.K.N.); (L.P.S.)
| | - Elena A. Romanova
- Institute of Gene Biology (RAS), Moscow 119334, Russia; (D.M.Y.); (E.A.R.); (Z.K.N.); (L.P.S.)
| | - Anna V. Tvorogova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, Moscow 119334, Russia;
| | - Zlata K. Naydenysheva
- Institute of Gene Biology (RAS), Moscow 119334, Russia; (D.M.Y.); (E.A.R.); (Z.K.N.); (L.P.S.)
| | - Alexey V. Feoktistov
- Institute of Gene Biology (RAS), Moscow 119334, Russia; (D.M.Y.); (E.A.R.); (Z.K.N.); (L.P.S.)
- Engelhardt Institute of Molecular Biology (RAS), Moscow 119334, Russia
| | - Denis V. Yashin
- Institute of Gene Biology (RAS), Moscow 119334, Russia; (D.M.Y.); (E.A.R.); (Z.K.N.); (L.P.S.)
| | - Lidia P. Sashchenko
- Institute of Gene Biology (RAS), Moscow 119334, Russia; (D.M.Y.); (E.A.R.); (Z.K.N.); (L.P.S.)
| |
Collapse
|
7
|
Kaur S, Saini AK, Tuli HS, Garg N, Joshi H, Varol M, Kaur J, Chhillar AK, Saini RV. Polymer-mediated nanoformulations: a promising strategy for cancer immunotherapy. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:1311-1326. [PMID: 37695334 DOI: 10.1007/s00210-023-02699-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 08/29/2023] [Indexed: 09/12/2023]
Abstract
Engineering polymer-based nano-systems have attracted many researchers owing to their unique qualities like shape, size, porosity, mechanical strength, biocompatibility, and biodegradability. Both natural and synthetic polymers can be tuned to get desired surface chemistry and functionalization to improve the efficacy of cancer therapy by promoting targeted delivery to the tumor site. Recent advancements in cancer immunoediting have been able to manage both primary tumor and metastatic lesions via activation of the immune system. The combinations of nano-biotechnology and immunotherapeutic agents have provided positive outcomes by enhancing the host immune response in cancer therapy. The nanoparticles have been functionalized using antibodies, targeted antigens, small molecule ligands, and other novel agents that can interact with biological systems at nanoscale levels. Several polymers, such as polyethylene glycol (PEG), poly(lactic-co-glycolic acid) (PLGA), poly(ε-caprolactone) (PCL), and chitosan, have been approved by the Food and Drug Administration for clinical use in biomedicine. The polymeric nanoformulations such as polymers-antibody/antigen conjugates and polymeric drug conjugates are currently being explored as nanomedicines that can target cancer cells directly or target immune cells to promote anti-cancer immunotherapy. In this review, we focus on scientific developments and advancements on engineered polymeric nano-systems in conjugation with immunotherapeutic agents targeting the tumor microenvironment to improve their efficacy and the safety for better clinical outcomes.
Collapse
Affiliation(s)
- Simranjit Kaur
- Department of Bioscience and Technology, MMEC, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana, 133207, India
| | - Adesh K Saini
- Department of Bioscience and Technology, MMEC, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana, 133207, India
- Central Research Cell, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana, 133207, India
| | - Hardeep Singh Tuli
- Department of Bioscience and Technology, MMEC, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana, 133207, India
| | - Nancy Garg
- Department of Bioscience and Technology, MMEC, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana, 133207, India
| | - Hemant Joshi
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Mehmet Varol
- Department of Molecular Biology and Genetics, Faculty of Science, Mugla Sitki Kocman University, Mugla, Turkey
| | - Jagjit Kaur
- Graduate School of Biomedical Engineering, Faculty of Engineering, The University of New South Wales, Sydney, 2052, Australia
| | - Anil K Chhillar
- Centre for Biotechnology, M.D. University, Rohtak, Haryana, 124 001, India
| | - Reena V Saini
- Department of Bioscience and Technology, MMEC, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana, 133207, India.
- Central Research Cell, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana, 133207, India.
| |
Collapse
|
8
|
Stanilov N, Velikova T, Stanilova S. Navigating the Cytokine Seas: Targeting Cytokine Signaling Pathways in Cancer Therapy. Int J Mol Sci 2024; 25:1009. [PMID: 38256080 PMCID: PMC10815616 DOI: 10.3390/ijms25021009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/09/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Cancer remains one of the leading causes of morbidity and mortality worldwide, necessitating continuous efforts to develop effective therapeutic strategies. Over the years, advancements in our understanding of the complex interplay between the immune system and cancer cells have led to the development of immunotherapies that revolutionize cancer treatment. Cytokines, as key regulators of the immune response, are involved in both the initiation and progression of cancer by affecting inflammation and manipulating multiple intracellular signaling pathways that regulate cell growth, proliferation, and migration. Cytokines, as key regulators of inflammation, have emerged as promising candidates for cancer therapy. This review article aims to provide an overview of the significance of cytokines in cancer development and therapy by highlighting the importance of targeting cytokine signaling pathways as a potential therapeutic approach.
Collapse
Affiliation(s)
- Noyko Stanilov
- Medical Faculty, Trakia University, 6000 Stara Zagora, Bulgaria;
| | - Tsvetelina Velikova
- Medical Faculty, Sofia University St. Kliment Ohridski, 1 Kozyak Str., 1407 Sofia, Bulgaria
| | - Spaska Stanilova
- Department of Molecular Biology, Immunology and Medical Genetics, Medical Faculty, Trakia University, 6000 Stara Zagora, Bulgaria;
| |
Collapse
|
9
|
Szwed-Georgiou A, Płociński P, Kupikowska-Stobba B, Urbaniak MM, Rusek-Wala P, Szustakiewicz K, Piszko P, Krupa A, Biernat M, Gazińska M, Kasprzak M, Nawrotek K, Mira NP, Rudnicka K. Bioactive Materials for Bone Regeneration: Biomolecules and Delivery Systems. ACS Biomater Sci Eng 2023; 9:5222-5254. [PMID: 37585562 PMCID: PMC10498424 DOI: 10.1021/acsbiomaterials.3c00609] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 07/31/2023] [Indexed: 08/18/2023]
Abstract
Novel tissue regeneration strategies are constantly being developed worldwide. Research on bone regeneration is noteworthy, as many promising new approaches have been documented with novel strategies currently under investigation. Innovative biomaterials that allow the coordinated and well-controlled repair of bone fractures and bone loss are being designed to reduce the need for autologous or allogeneic bone grafts eventually. The current engineering technologies permit the construction of synthetic, complex, biomimetic biomaterials with properties nearly as good as those of natural bone with good biocompatibility. To ensure that all these requirements meet, bioactive molecules are coupled to structural scaffolding constituents to form a final product with the desired physical, chemical, and biological properties. Bioactive molecules that have been used to promote bone regeneration include protein growth factors, peptides, amino acids, hormones, lipids, and flavonoids. Various strategies have been adapted to investigate the coupling of bioactive molecules with scaffolding materials to sustain activity and allow controlled release. The current manuscript is a thorough survey of the strategies that have been exploited for the delivery of biomolecules for bone regeneration purposes, from choosing the bioactive molecule to selecting the optimal strategy to synthesize the scaffold and assessing the advantages and disadvantages of various delivery strategies.
Collapse
Affiliation(s)
- Aleksandra Szwed-Georgiou
- Department
of Immunology and Infectious Biology, Faculty of Biology and Environmental
Protection, University of Lodz, Lodz 90-136, Poland
| | - Przemysław Płociński
- Department
of Immunology and Infectious Biology, Faculty of Biology and Environmental
Protection, University of Lodz, Lodz 90-136, Poland
| | - Barbara Kupikowska-Stobba
- Biomaterials
Research Group, Lukasiewicz Research Network
- Institute of Ceramics and Building Materials, Krakow 31-983, Poland
| | - Mateusz M. Urbaniak
- Department
of Immunology and Infectious Biology, Faculty of Biology and Environmental
Protection, University of Lodz, Lodz 90-136, Poland
- The
Bio-Med-Chem Doctoral School, University of Lodz and Lodz Institutes
of the Polish Academy of Sciences, University
of Lodz, Lodz 90-237, Poland
| | - Paulina Rusek-Wala
- Department
of Immunology and Infectious Biology, Faculty of Biology and Environmental
Protection, University of Lodz, Lodz 90-136, Poland
- The
Bio-Med-Chem Doctoral School, University of Lodz and Lodz Institutes
of the Polish Academy of Sciences, University
of Lodz, Lodz 90-237, Poland
| | - Konrad Szustakiewicz
- Department
of Polymer Engineering and Technology, Faculty of Chemistry, Wroclaw University of Technology, Wroclaw 50-370, Poland
| | - Paweł Piszko
- Department
of Polymer Engineering and Technology, Faculty of Chemistry, Wroclaw University of Technology, Wroclaw 50-370, Poland
| | - Agnieszka Krupa
- Department
of Immunology and Infectious Biology, Faculty of Biology and Environmental
Protection, University of Lodz, Lodz 90-136, Poland
| | - Monika Biernat
- Biomaterials
Research Group, Lukasiewicz Research Network
- Institute of Ceramics and Building Materials, Krakow 31-983, Poland
| | - Małgorzata Gazińska
- Department
of Polymer Engineering and Technology, Faculty of Chemistry, Wroclaw University of Technology, Wroclaw 50-370, Poland
| | - Mirosław Kasprzak
- Biomaterials
Research Group, Lukasiewicz Research Network
- Institute of Ceramics and Building Materials, Krakow 31-983, Poland
| | - Katarzyna Nawrotek
- Faculty
of Process and Environmental Engineering, Lodz University of Technology, Lodz 90-924, Poland
| | - Nuno Pereira Mira
- iBB-Institute
for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de
Lisboa, Lisboa 1049-001, Portugal
- Associate
Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior
Técnico, Universidade de Lisboa, Lisboa 1049-001, Portugal
- Instituto
Superior Técnico, Universidade de Lisboa, Lisboa 1049-001, Portugal
| | - Karolina Rudnicka
- Department
of Immunology and Infectious Biology, Faculty of Biology and Environmental
Protection, University of Lodz, Lodz 90-136, Poland
| |
Collapse
|
10
|
Zhang F, Yang Q, Tang S, Jiang S, Zhao Q, Li J, Xu C, Liu J, Fu Y. CD38-targeted and erythrocyte membrane camouflaged nanodrug delivery system for photothermal and chemotherapy in multiple myeloma. Int J Pharm 2023; 643:123241. [PMID: 37479101 DOI: 10.1016/j.ijpharm.2023.123241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 06/19/2023] [Accepted: 07/15/2023] [Indexed: 07/23/2023]
Abstract
Multiple myeloma (MM) is a malignant and incurable disease. Chemotherapy is currently the primary treatment option for MM. However, chemotherapeutic drugs can interrupt treatment because of serious side effects. Therefore, development of novel therapeutics for MM is essential. In this study, we designed and constructed an innovative nanoparticle-based drug delivery system, P-R@Ni3P-BTZ, and investigated its feasibility, effectiveness, and safety both in vitro and in vivo. P-R@Ni3P-BTZ is a nanocomposite that consists of two parts: (1) the drug carrier (Ni3P), which integrates photothermal therapy (PTT) with chemotherapy by loading bortezomib (BTZ); and (2) the shell (P-R), a CD38 targeting peptide P-modified red blood cell membrane nanovesicles. In vitro and in vivo, it was proven that P-R@Ni3P-BTZ exhibits remarkable antitumor effects by actively targeting CD38 + MM cells. P-R@Ni3P-BTZ significantly induces the accumulation of intracellular reactive oxygen species (ROS) and increases the apoptosis of MM cells, which underlies the primary mechanism of its antitumor effects. In addition, P-R@Ni3P exhibits good biocompatibility and biosafety, both in vitro and in vivo. Overall, P-R@Ni3P-BTZ is a specific and efficient MM therapeutic method.
Collapse
Affiliation(s)
- Fangrong Zhang
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha 410013, People's Republic of China
| | - Qin Yang
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha 410013, People's Republic of China
| | - Sishi Tang
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha 410013, People's Republic of China
| | - Siyi Jiang
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha 410013, People's Republic of China
| | - Qiangqiang Zhao
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha 410013, People's Republic of China
| | - Jian Li
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha 410013, People's Republic of China
| | - Cong Xu
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha 410013, People's Republic of China
| | - Jing Liu
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha 410013, People's Republic of China.
| | - Yunfeng Fu
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha 410013, People's Republic of China.
| |
Collapse
|
11
|
Pang Q, You L, Meng X, Li Y, Deng T, Li D, Zhu B. Regulation of the JAK/STAT signaling pathway: The promising targets for cardiovascular disease. Biochem Pharmacol 2023; 213:115587. [PMID: 37187275 DOI: 10.1016/j.bcp.2023.115587] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 05/05/2023] [Accepted: 05/08/2023] [Indexed: 05/17/2023]
Abstract
Individuals have known that Janus kinase (JAK) signal transducer and activator of transcription (STAT) signaling pathway was involved in the growth of the cell, cell differentiation courses advancement, immune cellular survival, as well as hematopoietic system advancement. Researches in the animal models have already uncovered a JAK/STAT regulatory function in myocardial ischemia-reperfusion injury (MIRI), acute myocardial infarction (MI), hypertension, myocarditis, heart failure, angiogenesis and fibrosis. Evidences originating in these studies indicate a therapeutic JAK/STAT function in cardiovascular diseases (CVDs). In this retrospection, various JAK/STAT functions in the normal and ill hearts were described. Moreover, the latest figures about JAK/STAT were summarized under the background of CVDs. Finally, we discussed the clinical transformation prospects and technical limitations of JAK/STAT as the potential therapeutic targets for CVDs. This collection of evidences has essential meanings for the clinical application of JAK/STAT as medicinal agents for CVDs. In this retrospection, various JAK/STAT functions in the normal and ill hearts were described. Moreover, the latest figures about JAK/STAT were summarized under the background of CVDs. Finally, we discussed the clinical transformation prospects and toxicity of JAK/STAT inhibitors as potential therapeutic targets for CVDs. This collection of evidences has essential meanings for the clinical application of JAK/STAT as medicinal agents for CVDs.
Collapse
Affiliation(s)
- Qiuyu Pang
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Lu You
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xiangmin Meng
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yumeng Li
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Tian Deng
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Deyong Li
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Bingmei Zhu
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
12
|
Tanito K, Nii T, Yokoyama Y, Oishi H, Shibata M, Hijii S, Kaneko R, Tateishi C, Ito S, Kishimura A, Mori T, Katayama Y. Engineered macrophages acting as a trigger to induce inflammation only in tumor tissues based on arginase 1-responsive TNF-α accelerated release. J Control Release 2023:S0168-3659(23)00260-2. [PMID: 37080897 DOI: 10.1016/j.jconrel.2023.04.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 03/23/2023] [Accepted: 04/05/2023] [Indexed: 04/22/2023]
Abstract
Herein, we report engineered macrophages, termed "MacTrigger," acting as a trigger to induce an inflammatory environment only in tumor tissues. This led to intensive anti-tumor effects based on the removal potential of foreign substances. The strength of this study is the utilization of two unique functions of macrophages: (1) their ability to migrate to tumor tissues and (2) polarization into the anti-inflammatory M2 phenotype in the presence of tumor tissues. The MacTrigger accelerated the release of inflammatory cytokines, tumor necrosis factor-alpha (TNF-α), when it was polarized to the M2 phenotype. When the MacTrigger was administered to tumor-bearing mice, tumor growth was significantly inhibited compared with the non-treatment group, the un-transfected macrophages group, and the group with engineered macrophages capable of randomly releasing TNF-α. Additionally, the ratio of the M1 phenotype to the M2 phenotype in tumor tissues was >1 only in the MacTrigger group. Moreover, the ratios of natural killer cells and CD8+T cells in tumor tissues were increased compared with other groups. These results indicate that MacTrigger can induce inflammation in tumor tissues, leading to effective anti-tumor effects. In normal tissues, especially the liver, notable side effects were not observed. This is because, in the liver, the MacTrigger was not polarized to the M2 phenotype and could not induce inflammation. These results suggest that the MacTrigger is a "trigger" that can induce inflammation only in tumor tissues, then allowing the body to attack tumor tissues through the innate immunity system.
Collapse
Affiliation(s)
- Kenta Tanito
- Graduate School of Systems Life Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Teruki Nii
- Graduate School of Systems Life Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan; Department of Applied Chemistry, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan.
| | - Yuta Yokoyama
- Graduate School of Systems Life Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Haruka Oishi
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Mayuka Shibata
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Shoichi Hijii
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Ryosuke Kaneko
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Chuya Tateishi
- Graduate School of Systems Life Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Shoko Ito
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Akihiro Kishimura
- Graduate School of Systems Life Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan; Department of Applied Chemistry, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan; Center for Future Chemistry, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan; International Research Center for Molecular Systems, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Takeshi Mori
- Graduate School of Systems Life Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan; Department of Applied Chemistry, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan; Center for Future Chemistry, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Yoshiki Katayama
- Graduate School of Systems Life Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan; Department of Applied Chemistry, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan; Center for Future Chemistry, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan; International Research Center for Molecular Systems, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan; Center for Advanced Medical Innovation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; Department of Biomedical Engineering, Chung Yuan Christian University, 200 Chung Pei Road, Chung Li 32023, Taiwan, ROC.
| |
Collapse
|
13
|
Nanavati K, Rutherfurd-Markwick K, Lee SJ, Bishop NC, Ali A. Effect of curcumin supplementation on exercise-induced muscle damage: a narrative review. Eur J Nutr 2022; 61:3835-3855. [PMID: 35831667 PMCID: PMC9596560 DOI: 10.1007/s00394-022-02943-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 06/15/2022] [Indexed: 11/26/2022]
Abstract
Curcumin, a natural polyphenol extracted from turmeric, is a potent antioxidant and anti-inflammatory agent. In the past few decades, curcumin's ability to impact chronic inflammatory conditions such as metabolic syndrome, arthritis, and cancer has been widely researched, along with growing interest in understanding its role in exercise-induced muscle damage (EIMD). EIMD impacts individuals differently depending on the type (resistance exercise, high-intensity interval training, and running), intensity, and duration of the exercise. Exercise disrupts the muscles' ultrastructure, raises inflammatory cytokine levels, and can cause swelling in the affected limb, a reduction in range of motion (ROM), and a reduction in muscular force-producing capacity. This review focuses on the metabolism, pharmacokinetics of various brands of curcumin supplements, and the effect of curcumin supplementation on EIMD regarding muscle soreness, activity of creatine kinase (CK), and production of inflammatory markers. Curcumin supplementation in the dose range of 90-5000 mg/day can decrease the subjective perception of muscle pain intensity, increase antioxidant capacity, and reduce CK activity, which reduces muscle damage when consumed close to exercise. Consumption of curcumin also improves muscle performance and has an anti-inflammatory effect, downregulating the production of pro-inflammatory cytokines, including TNF-α, IL-6, and IL-8. Curcumin may also improve oxidative capacity without hampering training adaptations in untrained and recreationally active individuals. The optimal curcumin dose to ameliorate EIMD is challenging to assess as its effect depends on the curcumin concentration in the supplement and its bioavailability.
Collapse
Affiliation(s)
- K. Nanavati
- School of Sport, Exercise, and Nutrition, Massey University, Auckland, New Zealand
| | | | - S. J. Lee
- School of Food and Advanced Technology, Massey University, Auckland, New Zealand
| | - N. C. Bishop
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - A. Ali
- School of Sport, Exercise, and Nutrition, Massey University, Auckland, New Zealand
| |
Collapse
|
14
|
Ross A, Sauce-Guevara MA, Alarcon EI, Mendez-Rojas MA. Peptide Biomaterials for Tissue Regeneration. Front Bioeng Biotechnol 2022; 10:893936. [PMID: 35992354 PMCID: PMC9388858 DOI: 10.3389/fbioe.2022.893936] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 06/06/2022] [Indexed: 11/21/2022] Open
Abstract
Expanding the toolbox of therapeutic materials for soft tissue and organ repair has become a critical component of tissue engineering. While animal- and plant-derived proteins are the foundation for developing biomimetic tissue constructs, using peptides as either constituents or frameworks for the materials has gained increasing momentum in recent years. This mini review discusses recent advances in peptide-based biomaterials' design and application. We also discuss some of the future challenges posed and opportunities opened by peptide-based structures in the field of tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Alex Ross
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, Canada
- Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Mildred A. Sauce-Guevara
- Department of Chemical and Biological Sciences, Universidad de Las Américas Puebla, Puebla, Mexico
| | - Emilio I. Alarcon
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, Canada
- Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Miguel A. Mendez-Rojas
- Department of Chemical and Biological Sciences, Universidad de Las Américas Puebla, Puebla, Mexico
| |
Collapse
|
15
|
Jiao L, Dong Q, Zhai W, Zhao W, Shi P, Wu Y, Zhou X, Gao Y. A PD-L1 and VEGFR2 dual targeted peptide and its combination with irradiation for cancer immunotherapy. Pharmacol Res 2022; 182:106343. [DOI: 10.1016/j.phrs.2022.106343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 06/03/2022] [Accepted: 07/01/2022] [Indexed: 10/17/2022]
|
16
|
Noack L, Bundkirchen K, Xu B, Gylstorff S, Zhou Y, Köhler K, Jantaree P, Neunaber C, Nowak AJ, Relja B. Acute Intoxication With Alcohol Reduces Trauma-Induced Proinflammatory Response and Barrier Breakdown in the Lung via the Wnt/β-Catenin Signaling Pathway. Front Immunol 2022; 13:866925. [PMID: 35663960 PMCID: PMC9159919 DOI: 10.3389/fimmu.2022.866925] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/07/2022] [Indexed: 11/13/2022] Open
Abstract
Background Trauma is the third leading cause of mortality worldwide. Upon admission, up to 50% of traumatized patients are acutely intoxicated with alcohol, which might lead to aberrant immune responses. An excessive and uncontrolled inflammatory response to injury is associated with damage to trauma-distant organs. We hypothesize that, along with inflammation-induced apoptosis, the activation of the Wnt/β-catenin signaling pathway would cause breakdown of the lung barrier and the development of lung injury after trauma. It remains unclear whether ethanol intoxication (EI) prior to trauma and hemorrhagic shock will attenuate inflammation and organ injury. Methods In this study, 14 male C57BL/6J mice were randomly assigned to two groups and exposed either to EtOH or to NaCl as a control by an oral gavage before receiving a femur fracture (Fx) and hemorrhagic shock, followed by resuscitation (THFx). Fourteen sham animals received either EtOH or NaCl and underwent surgical procedures without THFx induction. After 24 h, oil red O staining of fatty vacuoles in the liver was performed. Histological lung injury score (LIS) was assessed to analyze the trauma-induced RLI. Gene expression of Cxcl1, Il-1β, Muc5ac, Tnf, and Tnfrsf10b as well as CXCL1, IL-1β, and TNF protein levels in the lung tissue and bronchoalveolar lavage fluid were determined by RT-qPCR, ELISA, and immunohistological analyses. Infiltrating polymorphonuclear leukocytes (PMNLs) were examined via immunostaining. Apoptosis was detected by activated caspase-3 expression in the lung tissue. To confirm active Wnt signaling after trauma, gene expression of Wnt3a and its inhibitor sclerostin (Sost) was determined. Protein expression of A20 and RIPK4 as possible modulators of the Wnt signaling pathway was analyzed via immunofluorescence. Results Significant fatty changes in the liver confirmed the acute EI. Histopathology and decreased Muc5ac expression revealed an increased lung barrier breakdown and concomitant lung injury after THFx versus sham. EI prior trauma decreased lung injury. THFx increased not only the gene expression of pro-inflammatory markers but also the pulmonary infiltration with PMNL and apoptosis versus sham, while EI prior to THFx reduced those changes significantly. EI increased the THFx-reduced gene expression of Sost and reduced the THFx-induced expression of Wnt3a. While A20, RIPK4, and membranous β-catenin were significantly reduced after trauma, they were enhanced upon EI. Conclusion These findings suggest that acute EI alleviates the uncontrolled inflammatory response and lung barrier breakdown after trauma by suppressing the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Laurens Noack
- Department of Radiology and Nuclear Medicine, Experimental Radiology, Otto-von-Guericke University, Magdeburg, Germany
| | | | - Baolin Xu
- Department of Radiology and Nuclear Medicine, Experimental Radiology, Otto-von-Guericke University, Magdeburg, Germany.,Trauma Department, Hannover Medical School, Hannover, Germany
| | - Severin Gylstorff
- Department of Radiology and Nuclear Medicine, Experimental Radiology, Otto-von-Guericke University, Magdeburg, Germany
| | - Yuzhuo Zhou
- Department of Radiology and Nuclear Medicine, Experimental Radiology, Otto-von-Guericke University, Magdeburg, Germany.,Trauma Department, Hannover Medical School, Hannover, Germany
| | - Kernt Köhler
- Institute of Veterinary Pathology, Justus Liebig University Giessen, Giessen, Germany
| | - Phatcharida Jantaree
- Institute of Experimental Internal Medicine, Otto-von-Guericke University, Magdeburg, Germany
| | | | - Aleksander J Nowak
- Department of Radiology and Nuclear Medicine, Experimental Radiology, Otto-von-Guericke University, Magdeburg, Germany
| | - Borna Relja
- Department of Radiology and Nuclear Medicine, Experimental Radiology, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
17
|
Salvoza N, Bedin C, Saccani A, Tiribelli C, Rosso N. The Beneficial Effects of Triterpenic Acid and Acteoside in an In Vitro Model of Nonalcoholic Steatohepatitis (NASH). Int J Mol Sci 2022; 23:3562. [PMID: 35408923 PMCID: PMC8998673 DOI: 10.3390/ijms23073562] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/18/2022] [Accepted: 03/21/2022] [Indexed: 02/04/2023] Open
Abstract
Triterpenic acid (TA) and acteoside (ACT), the major components of APPLIVER and ACTEOS, respectively, have been reported to exert hepatoprotective effects, but the molecular mechanisms remain elusive, particularly in the NAFLD/NASH context. We assessed their effects in our well-established in vitro model resembling the pathophysiological mechanisms involved in NASH. Human hepatocytes and hepatic stellate cells were exposed to free fatty acids (FFA) alone or in combination with APPLIVER and ACTEOS as a mono- or co-culture. Steatosis, inflammation, generation of reactive oxygen species (ROS), and collagen deposition were determined. ACTEOS reduced both the TNF-α and ROS production, and, most importantly, attenuated collagen deposition elicited by the excess of FFA in the co-culture model. APPLIVER also showed inhibition of both TNF-α production and collagen deposition caused by FFA accumulation. The compounds alone did not induce any cellular effects. The present study showed the efficacy of APPLIVER and ACTEOS on pathophysiological mechanisms related to NASH. These in vitro data suggest that these compounds deserve further investigation for possible use in NASH treatment.
Collapse
Affiliation(s)
- Noel Salvoza
- Fondazione Italiana Fegato—ONLUS, Area Science Park, Basovizza SS14 km 163.5, 34149 Trieste, Italy;
- Philippine Council for Health Research and Development, DOST-Bicutan, Taguig City 1631, Philippines
| | - Chiara Bedin
- ABResearch S.R.L., Via dell’Impresa 1, 36040 Brendola, Italy; (C.B.); (A.S.)
| | - Andrea Saccani
- ABResearch S.R.L., Via dell’Impresa 1, 36040 Brendola, Italy; (C.B.); (A.S.)
| | - Claudio Tiribelli
- Fondazione Italiana Fegato—ONLUS, Area Science Park, Basovizza SS14 km 163.5, 34149 Trieste, Italy;
| | - Natalia Rosso
- Fondazione Italiana Fegato—ONLUS, Area Science Park, Basovizza SS14 km 163.5, 34149 Trieste, Italy;
| |
Collapse
|
18
|
Zhu X, Hu Z, Yu T, Hu H, Zhao Y, Li C, Zhu Q, Wang M, Zhai P, He L, Riaz Rajoka MS, Song X, He Z. The Antiviral Effects of Jasminin via Endogenous TNF-α and the Underlying TNF-α-Inducing Action. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27051598. [PMID: 35268699 PMCID: PMC8911969 DOI: 10.3390/molecules27051598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/25/2022] [Accepted: 02/26/2022] [Indexed: 12/03/2022]
Abstract
Previous studies have reported that recombinant tumor necrosis factor (TNF)-α has powerful antiviral activity but severe systematic side effects. Jasminin is a common bioactive component found in Chinese herbal medicine beverage “Jasmine Tea”. Here, we report that jasminin-induced endogenous TNF-α showed antiviral activity in vitro. The underlying TNF-α-inducing action of jasminin was also investigated in RAW264.7 cells. The level of endogenous TNF-α stimulated by jasminin was first analyzed by an enzyme-linked immunosorbent assay (ELISA) from the cell culture supernatant of RAW264.7 cells. The supernatants were then collected to investigate the potential antiviral effect against herpes simplex virus 1 (HSV-1). The antiviral effects of jasminin alone or its supernatants were evaluated by a plaque reduction assay. The potential activation of the PI3K–Akt pathway, three main mitogen-activated protein kinases (MAPKs), and nuclear factor (NF)–κB signaling pathways that induce TNF-α production were also investigated. Jasminin induces TNF-α protein expression in RAW264.7 cells without additional stimuli 10-fold more than the control. No significant up-expression of type I, II, and III interferons; interleukins 2 and 10; nor TNF-β were observed by the jasminin stimuli. The supernatants, containing jasminin-induced-TNF-α, showed antiviral activity against HSV-1. The jasminin-stimulated cells caused the simultaneous activation of the Akt, MAPKs, and NF–κB signal pathways. Furthermore, the pretreatment of the cells with the Akt, MAPKs, and NF–κB inhibitors effectively suppressed jasminin-induced TNF-α production. Our research provides evidence that endogenous TNF-α can be used as a strategy to encounter viral infections. Additionally, the Akt, MAPKs, and NF–κB signaling pathways are involved in the TNF-α synthesis that induced by jasminin.
Collapse
Affiliation(s)
- Xiaohong Zhu
- Affiliated Longhua People’s Hospital, Southern Medical University, Shenzhen 518172, China;
- School of Pharmaceutical Sciences, School of Basic Medicine, Health Science Center, Shenzhen University, Shenzhen 518000, China; (Z.H.); (T.Y.); (H.H.); (Y.Z.); (C.L.)
| | - Ziwei Hu
- School of Pharmaceutical Sciences, School of Basic Medicine, Health Science Center, Shenzhen University, Shenzhen 518000, China; (Z.H.); (T.Y.); (H.H.); (Y.Z.); (C.L.)
| | - Tian Yu
- School of Pharmaceutical Sciences, School of Basic Medicine, Health Science Center, Shenzhen University, Shenzhen 518000, China; (Z.H.); (T.Y.); (H.H.); (Y.Z.); (C.L.)
| | - Hao Hu
- School of Pharmaceutical Sciences, School of Basic Medicine, Health Science Center, Shenzhen University, Shenzhen 518000, China; (Z.H.); (T.Y.); (H.H.); (Y.Z.); (C.L.)
| | - Yunshi Zhao
- School of Pharmaceutical Sciences, School of Basic Medicine, Health Science Center, Shenzhen University, Shenzhen 518000, China; (Z.H.); (T.Y.); (H.H.); (Y.Z.); (C.L.)
| | - Chenyang Li
- School of Pharmaceutical Sciences, School of Basic Medicine, Health Science Center, Shenzhen University, Shenzhen 518000, China; (Z.H.); (T.Y.); (H.H.); (Y.Z.); (C.L.)
| | - Qinchang Zhu
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China; (Q.Z.); (M.W.)
| | - Mingzhong Wang
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China; (Q.Z.); (M.W.)
| | - Peng Zhai
- Faculty of Information Technology, Macau University of Science and Technology, Macau 999078, China;
| | - Longxia He
- Department of Otorhinolaryngology-Head and Neck Surgery, Chengdu Integrated TCM&Western Medicine Hospital, Chengdu 610017, China;
| | - Muhammad Shahid Riaz Rajoka
- Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan;
| | - Xun Song
- School of Pharmaceutical Sciences, School of Basic Medicine, Health Science Center, Shenzhen University, Shenzhen 518000, China; (Z.H.); (T.Y.); (H.H.); (Y.Z.); (C.L.)
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China; (Q.Z.); (M.W.)
- Correspondence: (X.S.); (Z.H.)
| | - Zhendan He
- School of Pharmaceutical Sciences, School of Basic Medicine, Health Science Center, Shenzhen University, Shenzhen 518000, China; (Z.H.); (T.Y.); (H.H.); (Y.Z.); (C.L.)
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China; (Q.Z.); (M.W.)
- Correspondence: (X.S.); (Z.H.)
| |
Collapse
|
19
|
Piri H, Sharifi S, Nigjeh S, Haghdoost-Yazdi H. Dopaminergic neuronal death in the substantia nigra associates with change in serum levels of TNF-α and IL-1β; evidence from early experimental model of Parkinson's disease. Neurol Res 2022; 44:544-553. [PMID: 34986749 DOI: 10.1080/01616412.2021.2024726] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
PURPOSE Studies have shown that inflammation plays a key role in etiology of Parkinson's disease (PD). However, human studies which have evaluated association between PD and serum levels of tumor necrosis factor-alpha (TNF-α) and interleukin-1 beta (IL-1β) have reported conflicting results. In this study, serum and striatum levels of these cytokines were evaluated in 6-hydroxydopamine (6-OHDA) animal model of PD. METHOD The neurotoxin of 6-OHDA was injected into medial forebrain bundle of right hemisphere and behavioral tests were carried out to eight weeks thereafter to evaluate severity of PD and its progress. Blood was collected before the toxin and in second and eight weeks after that. Survival of dopaminergic (DAergic) neurons in substantia nigra was assessed by immunohistochemistry. TNF-α and IL-1β levels were determined using ELISA kits. RESULT Severity of behavioral symptoms was gradually increased in 6-OHDA-treated rats. They showed a decrease in serum TNF-α level in the eight week and increase in IL-1β both in the second and eight weeks. They were divided into two subgroups, symptomatic and asymptomatic with severe and moderate degrees in DAergic neuronal death. Significant decrease in serum TNF-α was only observed in the symptomatic subgroup but IL-1β increased in both subgroups. Also, striatal levels of both cytokines were higher in the lesioned hemisphere. CONCLUSION Increase in serum IL-1β level can reflect moderate degree of lesion in substantia nigra and thereby is used for prognosis of PD before its clinical symptoms are appeared. On the other hand, an increase in serum TNF-α is appeared in advanced stage of PD.
Collapse
Affiliation(s)
- Hossein Piri
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Sahar Sharifi
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Sepideh Nigjeh
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Hashem Haghdoost-Yazdi
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran
| |
Collapse
|
20
|
Abstract
The Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling pathway was discovered more than a quarter-century ago. As a fulcrum of many vital cellular processes, the JAK/STAT pathway constitutes a rapid membrane-to-nucleus signaling module and induces the expression of various critical mediators of cancer and inflammation. Growing evidence suggests that dysregulation of the JAK/STAT pathway is associated with various cancers and autoimmune diseases. In this review, we discuss the current knowledge about the composition, activation, and regulation of the JAK/STAT pathway. Moreover, we highlight the role of the JAK/STAT pathway and its inhibitors in various diseases.
Collapse
Affiliation(s)
- Xiaoyi Hu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu, 610041, Sichuan, P. R. China
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, 610041, Chengdu, P. R. China
| | - Jing Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu, 610041, Sichuan, P. R. China
| | - Maorong Fu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu, 610041, Sichuan, P. R. China
| | - Xia Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu, 610041, Sichuan, P. R. China.
- Department of Gynecology and Obstetrics, Development and Related Disease of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, 610041, Chengdu, P. R. China.
| | - Wei Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu, 610041, Sichuan, P. R. China.
| |
Collapse
|
21
|
Zeng Y, Xiang Y, Sheng R, Tomás H, Rodrigues J, Gu Z, Zhang H, Gong Q, Luo K. Polysaccharide-based nanomedicines for cancer immunotherapy: A review. Bioact Mater 2021; 6:3358-3382. [PMID: 33817416 PMCID: PMC8005658 DOI: 10.1016/j.bioactmat.2021.03.008] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/19/2021] [Accepted: 03/02/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer immunotherapy is an effective antitumor approach through activating immune systems to eradicate tumors by immunotherapeutics. However, direct administration of "naked" immunotherapeutic agents (such as nucleic acids, cytokines, adjuvants or antigens without delivery vehicles) often results in: (1) an unsatisfactory efficacy due to suboptimal pharmacokinetics; (2) strong toxic and side effects due to low targeting (or off-target) efficiency. To overcome these shortcomings, a series of polysaccharide-based nanoparticles have been developed to carry immunotherapeutics to enhance antitumor immune responses with reduced toxicity and side effects. Polysaccharides are a family of natural polymers that hold unique physicochemical and biological properties, as they could interact with immune system to stimulate an enhanced immune response. Their structures offer versatility in synthesizing multifunctional nanocomposites, which could be chemically modified to achieve high stability and bioavailability for delivering therapeutics into tumor tissues. This review aims to highlight recent advances in polysaccharide-based nanomedicines for cancer immunotherapy and propose new perspectives on the use of polysaccharide-based immunotherapeutics.
Collapse
Affiliation(s)
- Yujun Zeng
- Huaxi MR Research Center (HMRRC), Department of Radiology, Department of Neurosurgery, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yufan Xiang
- Huaxi MR Research Center (HMRRC), Department of Radiology, Department of Neurosurgery, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ruilong Sheng
- CQM-Centro de Quimica da Madeira, MMRG, Universidade da Madeira, Campus da Penteada, 9000-390, Funchal, Madeira, Portugal
| | - Helena Tomás
- CQM-Centro de Quimica da Madeira, MMRG, Universidade da Madeira, Campus da Penteada, 9000-390, Funchal, Madeira, Portugal
| | - João Rodrigues
- CQM-Centro de Quimica da Madeira, MMRG, Universidade da Madeira, Campus da Penteada, 9000-390, Funchal, Madeira, Portugal
| | - Zhongwei Gu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Department of Neurosurgery, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| | - Hu Zhang
- Amgen Bioprocessing Centre, Keck Graduate Institute, Claremont, CA, 91711, USA
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC), Department of Radiology, Department of Neurosurgery, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, Department of Neurosurgery, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| |
Collapse
|
22
|
Larsson J, Hoppe E, Gautrois M, Cvijovic M, Jirstrand M. Second-generation TNFα turnover model for improved analysis of test compound interventions in LPS challenge studies. Eur J Pharm Sci 2021; 165:105937. [PMID: 34260892 DOI: 10.1016/j.ejps.2021.105937] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/31/2021] [Accepted: 07/04/2021] [Indexed: 11/30/2022]
Abstract
This study presents a non-linear mixed effects model describing tumour necrosis factor alpha (TNFα) release after lipopolysaccharide (LPS) provocations in absence or presence of anti-inflammatory test compounds. Inter-occasion variability and the pharmacokinetics of two test compounds have been added to this second-generation model, and the goal is to produce a framework of how to model TNFα response in LPS challenge studies in vivo and demonstrate its general applicability regardless of occasion or type of test compound. Model improvements based on experimental data were successfully implemented and provided a robust model for TNFα response after LPS provocation, as well as reliable estimates of the median pharmacodynamic parameters. The two test compounds, Test Compound A and roflumilast, showed 81.1% and 74.9% partial reduction of TNFα response, respectively, and the potency of Test Compound A was estimated to 0.166 µmol/L. Comparing this study with previously published work reveals that our model leads to biologically reasonable output, handles complex data pooled from different studies, and highlights the importance of accurately distinguishing the stimulatory effect of LPS from the inhibitory effect of the test compound.
Collapse
Affiliation(s)
- Julia Larsson
- Fraunhofer-Chalmers Centre, Chalmers Science Park, 412 88 Gothenburg, Sweden.; Department of Mathematical Sciences, Chalmers University of Technology and University of Gothenburg, 412 96 Gothenburg, Sweden..
| | | | | | - Marija Cvijovic
- Department of Mathematical Sciences, Chalmers University of Technology and University of Gothenburg, 412 96 Gothenburg, Sweden
| | - Mats Jirstrand
- Fraunhofer-Chalmers Centre, Chalmers Science Park, 412 88 Gothenburg, Sweden
| |
Collapse
|
23
|
PK/PD Modeling of the PDE7 Inhibitor-GRMS-55 in a Mouse Model of Autoimmune Hepatitis. Pharmaceutics 2021; 13:pharmaceutics13050597. [PMID: 33919375 PMCID: PMC8143339 DOI: 10.3390/pharmaceutics13050597] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/12/2021] [Accepted: 04/19/2021] [Indexed: 01/03/2023] Open
Abstract
This study aimed to assess the efficacy and explore the mechanisms of action of a potent phosphodiesterase (PDE)7A and a moderate PDE4B inhibitor GRMS-55 in a mouse model of autoimmune hepatitis (AIH). The concentrations of GRMS-55 and relevant biomarkers were measured in the serum of BALB/c mice with concanavalin A (ConA)-induced hepatitis administered with GRMS-55 at two dose levels. A semi-mechanistic PK/PD/disease progression model describing the time courses of measured biomarkers was developed. The emetogenicity as a potential side effect of the studied compound was evaluated in the α2-adrenoceptor agonist-induced anesthesia model. The results indicate that liver damage observed in mice challenged with ConA was mainly mediated by TNF-α and IFN-γ. GRMS-55 decreased the levels of pro-inflammatory mediators and the transaminase activities in the serum of mice with AIH. The anti-inflammatory properties of GRMS-55, resulting mainly from PDE7A inhibition, led to a high hepatoprotective activity in mice with AIH, which was mediated by an inhibition of pro-inflammatory signaling. GRMS-55 did not induce the emetic-like behavior. The developed PK/PD/disease progression model may be used in future studies to assess the potency and explore the mechanisms of action of new investigational compounds for the treatment of AIH.
Collapse
|
24
|
Geng DH, Ju Z, Xiao T, Zhou S, Huang L, Liu L, Zhou X, Wang L, Tong LT. Peptides YYGGEGSSSEQG and SESEM Inhibit TNF-α-Induced Smooth Muscle Cells Proliferation and Migration Through Their Bindings to TNF-α Receptor. Int J Pept Res Ther 2021. [DOI: 10.1007/s10989-020-10097-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
25
|
Pan H, Xie Y, Lu W, Chen Y, Lu Z, Zhen J, Wang W, Shang A. Engineering an enhanced thrombin-based GLP-1 analog with long-lasting glucose-lowering and efficient weight reduction. RSC Adv 2019; 9:30707-30714. [PMID: 35529389 PMCID: PMC9072222 DOI: 10.1039/c9ra06771j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 09/20/2019] [Indexed: 11/21/2022] Open
Abstract
Peptides are considered as potent therapeutic drugs primarily due to the exquisite potency and selectivity to targets. However, the development and clinical application of peptide drugs were severely limited by the poor in vivo lifespans. Here, we designed an improved small albumin-binding polypeptide that can associate with human serum albumin (HSA) and liberate the bioactive peptide. Using glucagon-like peptide-1 (GLP-1) as a model, two new long-lasting GLP-1 analogs (termed XTS1 and XTS2) containing an albumin-binding domain, a protease-cleavable linker and a mutated GLP-1(A8Aib) were designed to demonstrate the sustained release of GLP-1 due to the plasma thrombin (TBN) digestion. Two XTS peptides were prepared of high purity (>99%) and accurate molecular weight determined by reversed high-performance liquid chromatography and mass spectrometry, respectively. In vitro measurements of surface plasmon resonance indicated that XTS1 associate with serum albumins of all species with higher affinity compared with XTS2. Metabolic stability of XTS1 in vitro in human plasma was also better than that of XTS2. Protease cleavage assay results of XTS peptides demonstrated the controlled-release of transient GLP-1 from the XTS1 and XTS2 mixture after thrombin-catalyzed hydrolysis. Then the intraperitoneal glucose tolerance test (IPGTT) showed that the glucose-lowering efficacies of XTS1 were in a dosage-dependent manner within the range of 0.1–0.9 mg kg−1. In addition, XTS1 showed similar hypoglycemic intensity and significantly longer action duration compared to Liraglutide in both multiple IPGTTs and hypoglycemic duration test. Apparently extended plasma half-lives of ∼2.3 and ∼3.5 days were observed after a single subcutaneous administration of XTS1 (0.9 mg kg−1) in rats and cynomolgus monkeys, respectively. Furthermore, twice-weekly subcutaneously dosed XTS1 in db/db mice achieved long-term beneficial effects on body weight, hemoglobin A1C (HbA1C) lowering and the function of pancreatic beta cells. These studies support that XTS1 exerts potential as a therapeutic drug for the treatment of T2DM. Peptides are considered as potent therapeutic drugs primarily due to the exquisite potency and selectivity to targets.![]()
Collapse
Affiliation(s)
- Hongchao Pan
- Department of Laboratory Medicine, Shanghai Simple Gene Medical Laboratory Shanghai 200025 P.R. China
| | - Yini Xie
- Department of Laboratory Medicine, The People's Hospital of Jiedong Jieyang 515500 P. R. China
| | - Wenying Lu
- Department of Experimental Medicine Center, The Sixth People's Hospital of Yancheng City Yancheng 224001 P. R. China
| | - Yin Chen
- Key Laboratory of Biological Medicine, Department of Life Science and Technology, Jinan University 51000 P. R. China
| | - Zhao Lu
- Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University Nanjing 210000 P. R. China
| | - Jun Zhen
- Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University Nanjing 210000 P. R. China
| | - Weiwei Wang
- Department of Experimental Medicine Center, The Sixth People's Hospital of Yancheng City Yancheng 224001 P. R. China
| | - Anquan Shang
- Department of Laboratory Medicine, Tongji Hospital of Tongji University Shanghai 200065 P. R. China
| |
Collapse
|
26
|
Yan Q, Chen X, Gong H, Qiu P, Xiao X, Dang S, Hong A, Ma Y. Delivery of a TNF-α-derived peptide by nanoparticles enhances its antitumor activity by inducing cell-cycle arrest and caspase-dependent apoptosis. FASEB J 2018; 32:fj201800377R. [PMID: 30161002 DOI: 10.1096/fj.201800377r] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Prostate cancer is the second-most common malignancy of the male genitourinary system. TNF-α has attracted intense attention as a potential therapeutic agent against various cancers. However, its therapeutic application is restricted by short half life and severe toxic side-effects. In this study, we constructed a stable nanodrug, called TNF-α-derived polypeptide (P16)-conjugated, chitosan (CTS)-modified selenium nanoparticle (SC; SCP), which is composed of SC as a slow-release carrier conjugated to P16. SCP had significant inhibitory effects on multiple types of tumor cells, especially DU145 prostate cancer cells, but not on RWPE-1 normal human prostate epithelial cells. SCP could induce G0/G1 cell-cycle arrest and apoptosis in DU145 cells more effectively than could P16 and TNF-α. In DU145 xenograft tumor models, SCP exerted much stronger antitumor effects than P16 or estramustine (the clinical drug for prostate cancer) but caused fewer toxic side-effects. In addition, SCP significantly inhibited proliferation and accelerated apoptosis in DU145 xenograft tumors. Further mechanistic studies revealed that SCP exerted antitumor effects via activation of the p38 MAPK/JNK pathway, thus inducing G0/G1 cell-cycle arrest and caspase-dependent apoptosis. These findings suggest that SCP may represent a potential long-lasting therapeutic agent for human prostate cancer with fewer side effects.-Yan, Q., Chen, X., Gong, H., Qiu, P., Xiao, X., Dang, S., Hong, A., Ma, Y. Delivery of a TNF-α-derived peptide by nanoparticles enhances its antitumor activity by inducing cell-cycle arrest and caspase-dependent apoptosis.
Collapse
Affiliation(s)
- Qiuxia Yan
- Department of Cellular Biology, National Engineering Research Center of Genetic Medicine, Key Laboratory of Bioengineering Medicine of Guangdong Province, Institute of Biomedicine, Jinan University, Guangzhou, China
- Center for Reproductive Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| | - Xueming Chen
- Department of Cellular Biology, National Engineering Research Center of Genetic Medicine, Key Laboratory of Bioengineering Medicine of Guangdong Province, Institute of Biomedicine, Jinan University, Guangzhou, China
| | - Huizhen Gong
- Department of Cellular Biology, National Engineering Research Center of Genetic Medicine, Key Laboratory of Bioengineering Medicine of Guangdong Province, Institute of Biomedicine, Jinan University, Guangzhou, China
| | - Pei Qiu
- Department of Cellular Biology, National Engineering Research Center of Genetic Medicine, Key Laboratory of Bioengineering Medicine of Guangdong Province, Institute of Biomedicine, Jinan University, Guangzhou, China
| | - Xing Xiao
- Department of Cellular Biology, National Engineering Research Center of Genetic Medicine, Key Laboratory of Bioengineering Medicine of Guangdong Province, Institute of Biomedicine, Jinan University, Guangzhou, China
| | - Shiying Dang
- Department of Cellular Biology, National Engineering Research Center of Genetic Medicine, Key Laboratory of Bioengineering Medicine of Guangdong Province, Institute of Biomedicine, Jinan University, Guangzhou, China
| | - An Hong
- Department of Cellular Biology, National Engineering Research Center of Genetic Medicine, Key Laboratory of Bioengineering Medicine of Guangdong Province, Institute of Biomedicine, Jinan University, Guangzhou, China
| | - Yi Ma
- Department of Cellular Biology, National Engineering Research Center of Genetic Medicine, Key Laboratory of Bioengineering Medicine of Guangdong Province, Institute of Biomedicine, Jinan University, Guangzhou, China
| |
Collapse
|
27
|
Advances in structural design of lipid-based nanoparticle carriers for delivery of macromolecular drugs, phytochemicals and anti-tumor agents. Adv Colloid Interface Sci 2017; 249:331-345. [PMID: 28477868 DOI: 10.1016/j.cis.2017.04.006] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 03/13/2017] [Accepted: 04/17/2017] [Indexed: 12/19/2022]
Abstract
The present work highlights recent achievements in development of nanostructured dispersions and biocolloids for drug delivery applications. We emphasize the key role of biological small-angle X-ray scattering (BioSAXS) investigations for the nanomedicine design. A focus is given on controlled encapsulation of small molecular weight phytochemical drugs in lipid-based nanocarriers as well as on encapsulation of macromolecular siRNA, plasmid DNA, peptide and protein pharmaceuticals in nanostructured nanoparticles that may provide efficient intracellular delivery and triggered drug release. Selected examples of utilisation of the BioSAXS method for characterization of various types of liquid crystalline nanoorganizations (liposome, spongosome, cubosome, hexosome, and nanostructured lipid carriers) are discussed in view of the successful encapsulation and protection of phytochemicals and therapeutic biomolecules in the hydrophobic or the hydrophilic compartments of the nanocarriers. We conclude that the structural design of the nanoparticulate carriers is of crucial importance for the therapeutic outcome and the triggered drug release from biocolloids.
Collapse
|
28
|
Ma M, Ma Y, Zhang GJ, Liao R, Jiang XF, Yan XX, Bie FJ, Li XB, Lv YH. Eugenol alleviated breast precancerous lesions through HER2/PI3K-AKT pathway-induced cell apoptosis and S-phase arrest. Oncotarget 2017; 8:56296-56310. [PMID: 28915591 PMCID: PMC5593562 DOI: 10.18632/oncotarget.17626] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Accepted: 04/19/2017] [Indexed: 01/19/2023] Open
Abstract
Eugenol can be separated from the oil extract of clove bud, and has many pharmacological functions such as anticancer and transdermal absorption. HER2/PI3K-AKT is a key signaling pathway in the development of breast cancer. In this study, 80 μM eugenol could significantly inhibit the proliferation of HER-2 positive MCF-10AT cells and the inhibition rate was up to 32.8%, but had no obvious inhibitory effect on MCF-7 and MCF-10A cells with HER2 weak expression. Eugenol also significantly induced human breast precancerous lesion MCF-10AT cell apoptosis and cell cycle S-phase arrest, but the biological effects nearly disappeared after HER2 over-expression through transfecting pcDNA3.1-HER2. In MCF-10AT cells treated by 180 μM eugenol, the protein expressions of HER2, AKT, PDK1, p85, Bcl2, NF-κB, Bad and Cyclin D1 were decreased and the decreased rates were respectively 63.0%, 60.0%, 52.9%, 62.9%, 37.1%, 47.2%, 61.7%, 59.1%, while the p21, p27 and Bax expression were increased by 4.48-, 4.76- and 2.57-fold respectively. In the rat models of breast precancerous lesion, 1 mg eugenol for external use significantly inhibited the progress of breast precancerous lesion and the occurrence rate of breast precancerous lesions and invasive carcinomas was decreased by about 30.5%. Furthermore eugenol for external (1 mg) markedly decreased the protein expressions of HER2 (62.9%), AKT (58.6%), PDK1 (56.4%), p85 (54.3%), Bcl2 (59.3%), NF-κB (65.7%), Bad (64.0%), Cyclin D1 (43.0%), while p21, p27 and Bax protein expressions were respectively increased 1.83-, 2.52- and 2.51-fold. The results showed eugenol could significantly inhibit the development of breast precancerous lesions by blocking HER2/PI3K-AKT signaling network. So eugenol may be a promising external drug for breast precancerous lesions.
Collapse
Affiliation(s)
- Min Ma
- College of Traditional Chinese Medicine, Institute of Integrated Traditional Chinese and Western Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Yi Ma
- Institute of Biomedicine, Department of Cellular Biology, Key Laboratory of Bioengineering Medicine of Guangdong Province, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Gui-Juan Zhang
- The First Affiliated Hospital of Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Rui Liao
- College of Traditional Chinese Medicine, Institute of Integrated Traditional Chinese and Western Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Xue-Feng Jiang
- College of Traditional Chinese Medicine, Institute of Integrated Traditional Chinese and Western Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Xian-Xin Yan
- College of Traditional Chinese Medicine, Institute of Integrated Traditional Chinese and Western Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Feng-Jie Bie
- College of Traditional Chinese Medicine, Institute of Integrated Traditional Chinese and Western Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Xiao-Bo Li
- College of Traditional Chinese Medicine, Institute of Integrated Traditional Chinese and Western Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Yan-Hong Lv
- College of Traditional Chinese Medicine, Institute of Integrated Traditional Chinese and Western Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China
| |
Collapse
|
29
|
Xu G, Gu H, Hu B, Tong F, Liu D, Yu X, Zheng Y, Gu J. PEG- b-(PELG- g-PLL) nanoparticles as TNF-α nanocarriers: potential cerebral ischemia/reperfusion injury therapeutic applications. Int J Nanomedicine 2017; 12:2243-2254. [PMID: 28356740 PMCID: PMC5367577 DOI: 10.2147/ijn.s130842] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Brain ischemia/reperfusion (I/R) injury (BI/RI) is a leading cause of death and disability worldwide. However, the outcome of pharmacotherapy for BI/RI remains unsatisfactory. Innovative approaches for enhancing drug sensitivity and recovering neuronal activity in BI/RI treatment are urgently needed. The purpose of this study was to evaluate the protective effects of tumor necrosis factor (TNF)-α-loaded poly(ethylene glycol)-b-(poly(ethylenediamine L-glutamate)-g-poly(L-lysine)) (TNF-α/PEG-b-(PELG-g-PLL)) nanoparticles on BI/RI. The particle size of PEG-b-(PELG-g-PLL) and the loading and release rates of TNF-α were determined. The nanoparticle cytotoxicity was evaluated in vitro using rat cortical neurons. Sprague Dawley rats were preconditioned with free TNF-α or TNF-α/PEG-b-(PELG-g-PLL) polyplexes and then subjected to 2 hours ischemia and 22 hours reperfusion. Brain edema was assessed using the brain edema ratio, and the antioxidative activity was assessed by measuring the superoxide dismutase (SOD) activity and the malondialdehyde (MDA) content in the brain tissue. We further estimated the inflammatory activity and apoptosis level by determining the levels of interleukin-4 (IL-4), IL-6, IL-8, IL-10, and nitric oxide (NO), as well as the expression of glial fibrillary acidic protein (GFAP), intercellular adhesion molecule-1 (ICAM-1), and cysteine aspartase-3 (caspase-3), in the brain tissue. We provide evidence that TNF-α preconditioning attenuated the oxidative stress injury, the inflammatory activity, and the apoptosis level in I/R-induced cerebral injury, while the application of block copolymer PEG-b-(PELG-g-PLL) as a potential TNF-α nanocarrier with sustained release significantly enhanced the bioavailability of TNF-α. We propose that the block copolymer PEG-b-(PELG-g-PLL) may function as a potent nanocarrier for augmenting BI/RI pharmacotherapy, with unprecedented clinical benefits. Further studies are needed to better clarify the underlying mechanisms.
Collapse
Affiliation(s)
- Guangtao Xu
- Department of Pathology and Chemistry, Provincial Key Laboratory of Infectious Diseases and Immunopathology, Collaborative and Creative Center, Molecular Diagnosis and Personalized Medicine, Shantou University Medical College, Shantou, Guangdong
- Department of Pathology, Provincial Key Discipline of Pharmacology, Jiaxing University Medical College, Jiaxing, Zhejiang, People’s Republic of China
| | - Huan Gu
- Department of Pathology and Chemistry, Provincial Key Laboratory of Infectious Diseases and Immunopathology, Collaborative and Creative Center, Molecular Diagnosis and Personalized Medicine, Shantou University Medical College, Shantou, Guangdong
- Department of Physics, University of Maryland, College Park, Annapolis, MD, USA
| | - Bo Hu
- Department of Chemical Pathology, Jiaxing Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Jiaxing, Zhejiang, People’s Republic of China
| | - Fei Tong
- Department of Pathology, Provincial Key Discipline of Pharmacology, Jiaxing University Medical College, Jiaxing, Zhejiang, People’s Republic of China
| | - Daojun Liu
- Department of Pathology and Chemistry, Provincial Key Laboratory of Infectious Diseases and Immunopathology, Collaborative and Creative Center, Molecular Diagnosis and Personalized Medicine, Shantou University Medical College, Shantou, Guangdong
| | - Xiaojun Yu
- Department of Pathology and Chemistry, Provincial Key Laboratory of Infectious Diseases and Immunopathology, Collaborative and Creative Center, Molecular Diagnosis and Personalized Medicine, Shantou University Medical College, Shantou, Guangdong
| | - Yongxia Zheng
- Department of Pathology and Chemistry, Provincial Key Laboratory of Infectious Diseases and Immunopathology, Collaborative and Creative Center, Molecular Diagnosis and Personalized Medicine, Shantou University Medical College, Shantou, Guangdong
- Department of Pathology, Provincial Key Discipline of Pharmacology, Jiaxing University Medical College, Jiaxing, Zhejiang, People’s Republic of China
| | - Jiang Gu
- Department of Pathology and Chemistry, Provincial Key Laboratory of Infectious Diseases and Immunopathology, Collaborative and Creative Center, Molecular Diagnosis and Personalized Medicine, Shantou University Medical College, Shantou, Guangdong
- Correspondence: Jiang Gu, Department of Pathology, Provincial Key Laboratory of Infectious Diseases and Immunopathology, Shantou University Medical College, 22 Xinling Road, Shantou, Guangdong 515041, People’s Republic of China, Tel +86 754 8895 0207, Email
| |
Collapse
|
30
|
Zheng M, Wu Z, Wu A, Huang Z, He N, Xie X. MiR-145 promotes TNF-α-induced apoptosis by facilitating the formation of RIP1-FADDcaspase-8 complex in triple-negative breast cancer. Tumour Biol 2016; 37:8599-607. [PMID: 26733177 DOI: 10.1007/s13277-015-4631-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 12/10/2015] [Indexed: 12/14/2022] Open
Abstract
Researches indicate that the dysregulation of microRNA (miRNA) is involved in tumorigenesis. Among such dysregulated miRNAs in cancer, miR-145 is reported to be downregulated in multiple cancers. In this study, we demonstrated the downregulation of miR-145 in triple-negative breast cancer (TNBC) tissues and TNBC cell lines by quantitative reverse-transcription polymerase chain reaction (RT-PCR) analysis. Furthermore, we found that the tumor necrosis factor-alpha (TNF-α)-induced apoptosis was expanded by the transfection of miR-145 in MDA-MB-231 which belongs to the TNBC cell lines. We then indicated that the mechanism by which miR-145 promotes the TNF-α-induced apoptosis is dependent on the formation of RIP1-FADD-caspase-8 complex. The cellular inhibitor of apoptosis (cIAP1), which is the inhibitor of apoptosis protein, was found to be a target of miR-145 in MDA-MB-231 cells. As a result of cIAP1 overexpression, the promotion of miR-145 on TNF-α-induced apoptosis was inhibited in MDA-MB-231 cells. Therefore, our results indicate that miR-145 acts as a tumor suppressor in TNBC, suggesting that the miR-145-cIAP1 axis might be a potential therapeutic target for TNBC.
Collapse
Affiliation(s)
- Min Zheng
- Department of Breast Surgery, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Zhihao Wu
- Department of Breast Surgery, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Anqi Wu
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, 325035, China
| | - Zhenyu Huang
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, 325035, China
| | - Na He
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, 325035, China
| | - Xiaohong Xie
- Department of Breast Surgery, Zhejiang Provincial Hospital of Traditional Chinese Medicine, No. 54 Youdian Road, Hangzhou City, Zhejiang Province, 310006, China.
| |
Collapse
|