1
|
Paumier A, Verre J, Runel G, Chlasta J, Tribolo S, Chanut S. Anas barbariae 200K Modulates Cell Stiffness and Oxidative Stress in Microglial Cells In Vitro. Int J Mol Sci 2025; 26:1451. [PMID: 40003917 PMCID: PMC11855513 DOI: 10.3390/ijms26041451] [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: 09/27/2024] [Revised: 01/24/2025] [Accepted: 02/01/2025] [Indexed: 02/27/2025] Open
Abstract
Anas barbariae 200K, a homeopathic medicine, is traditionally used for influenza-like illnesses. We investigated the effects of Anas barbariae 200K on microglial cells, a subpopulation of macrophages specific to the central nervous system often used to study the inflammatory processes and oxidative stress generated during influenza-like episodes. The study demonstrates the effect of Anas barbariae 200K on cell stiffness and the reactive oxygen species production using atomic force microscopy and fluorescence microscopy techniques, respectively. Our results showed that Anas barbariae 200K rapidly increased cell stiffness in resting cells by 41% compared with the vehicle. In inflamed cells, cell stiffness was decreased by 21% when treated with Anas barbariae 200K compared with the vehicle. Finally, Anas barbariae 200K caused a reorganisation of filamentous actin, with marked relocation of actin at the cell extremities. Moreover, Anas barbariae 200K significantly decreased the reactive oxygen species (ROS) production in inflamed microglial cells by 40% (total intracellular ROS) and by 67% (mitochondrial ROS) compared with the vehicle. These results strongly suggest an effect of Anas barbariae 200K at a cellular level on cell stiffness and actin cytoskeleton. This sheds light on the biological mechanism of action of this homeopathic preparation.
Collapse
Affiliation(s)
- Anne Paumier
- Laboratoires BOIRON, Research Department, 2 Avenue de l’Ouest Lyonnais, 69510 Messimy, France; (A.P.); (J.V.); (S.C.)
| | - Justine Verre
- Laboratoires BOIRON, Research Department, 2 Avenue de l’Ouest Lyonnais, 69510 Messimy, France; (A.P.); (J.V.); (S.C.)
| | - Gaël Runel
- BioMeca, 60C Avenue Rockfeller, 69008 Lyon, France;
| | | | - Sandra Tribolo
- Laboratoires BOIRON, Research Department, 2 Avenue de l’Ouest Lyonnais, 69510 Messimy, France; (A.P.); (J.V.); (S.C.)
| | - Stéphanie Chanut
- Laboratoires BOIRON, Research Department, 2 Avenue de l’Ouest Lyonnais, 69510 Messimy, France; (A.P.); (J.V.); (S.C.)
| |
Collapse
|
2
|
Salikhova DI, Shedenkova MO, Sudina AK, Belousova EV, Krasilnikova IA, Nekrasova AA, Nefedova ZA, Frolov DA, Fatkhudinov TK, Makarov AV, Surin AM, Savostyanov KV, Goldshtein DV, Bakaeva ZV. Neuroprotective and anti-inflammatory properties of proteins secreted by glial progenitor cells derived from human iPSCs. Front Cell Neurosci 2024; 18:1449063. [PMID: 39165834 PMCID: PMC11333358 DOI: 10.3389/fncel.2024.1449063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 07/19/2024] [Indexed: 08/22/2024] Open
Abstract
Currently, stem cells technology is an effective tool in regenerative medicine. Cell therapy is based on the use of stem/progenitor cells to repair or replace damaged tissues or organs. This approach can be used to treat various diseases, such as cardiovascular, neurological diseases, and injuries of various origins. The mechanisms of cell therapy therapeutic action are based on the integration of the graft into the damaged tissue (replacement effect) and the ability of cells to secrete biologically active molecules such as cytokines, growth factors and other signaling molecules that promote regeneration (paracrine effect). However, cell transplantation has a number of limitations due to cell transportation complexity and immune rejection. A potentially more effective therapy is using only paracrine factors released by stem cells. Secreted factors can positively affect the damaged tissue: promote forming new blood vessels, stimulate cell proliferation, and reduce inflammation and apoptosis. In this work, we have studied the anti-inflammatory and neuroprotective effects of proteins with a molecular weight below 100 kDa secreted by glial progenitor cells obtained from human induced pluripotent stem cells. Proteins secreted by glial progenitor cells exerted anti-inflammatory effects in a primary glial culture model of LPS-induced inflammation by reducing nitric oxide (NO) production through inhibition of inducible NO synthase (iNOS). At the same time, added secreted proteins neutralized the effect of glutamate, increasing the number of viable neurons to control values. This effect is a result of decreased level of intracellular calcium, which, at elevated concentrations, triggers apoptotic death of neurons. In addition, secreted proteins reduce mitochondrial depolarization caused by glutamate excitotoxicity and help maintain higher NADH levels. This therapy can be successfully introduced into clinical practice after additional preclinical studies, increasing the effectiveness of rehabilitation of patients with neurological diseases.
Collapse
Affiliation(s)
- Diana I. Salikhova
- Laboratory of Cellular Biotechnology, Research Institute of Molecular and Cellular Medicine, Medical Institute of RUDN University, Moscow, Russia
- Laboratory of Stem Cell Genetics, Research Centre for Medical Genetics, Moscow, Russia
| | - Margarita O. Shedenkova
- Laboratory of Cellular Biotechnology, Research Institute of Molecular and Cellular Medicine, Medical Institute of RUDN University, Moscow, Russia
- Laboratory of Stem Cell Genetics, Research Centre for Medical Genetics, Moscow, Russia
| | - Anastasya K. Sudina
- Laboratory of Cellular Biotechnology, Research Institute of Molecular and Cellular Medicine, Medical Institute of RUDN University, Moscow, Russia
- Laboratory of Stem Cell Genetics, Research Centre for Medical Genetics, Moscow, Russia
| | - Ekaterina V. Belousova
- Laboratory of Cellular Biotechnology, Research Institute of Molecular and Cellular Medicine, Medical Institute of RUDN University, Moscow, Russia
- Laboratory of Stem Cell Genetics, Research Centre for Medical Genetics, Moscow, Russia
| | - Irina A. Krasilnikova
- Medical Genetic Center, National Medical Research Center for Children’s Health, Moscow, Russia
| | - Anastasya A. Nekrasova
- Medical Genetic Center, National Medical Research Center for Children’s Health, Moscow, Russia
| | - Zlata A. Nefedova
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Daniil A. Frolov
- Institute of Information Technologies, MIREA-Russian Technological University, Moscow, Russia
| | - Timur Kh. Fatkhudinov
- Laboratory of Cellular Biotechnology, Research Institute of Molecular and Cellular Medicine, Medical Institute of RUDN University, Moscow, Russia
| | - Andrey V. Makarov
- Laboratory of Cellular Biotechnology, Research Institute of Molecular and Cellular Medicine, Medical Institute of RUDN University, Moscow, Russia
| | - Alexander M. Surin
- Laboratory of Fundamental and Applied Problems of Pain, Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Kirill V. Savostyanov
- Medical Genetic Center, National Medical Research Center for Children’s Health, Moscow, Russia
| | - Dmitry V. Goldshtein
- Laboratory of Cellular Biotechnology, Research Institute of Molecular and Cellular Medicine, Medical Institute of RUDN University, Moscow, Russia
- Laboratory of Stem Cell Genetics, Research Centre for Medical Genetics, Moscow, Russia
| | - Zanda V. Bakaeva
- Medical Genetic Center, National Medical Research Center for Children’s Health, Moscow, Russia
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| |
Collapse
|
3
|
Deepika BK, Apoorva NH, Joel PR, B B, Sudheer SP. Enhanced osteogenic differentiation potential of Arnica montana and Bellis perennis in C3H10T1/2 multipotent mesenchymal stem cells. Mol Biol Rep 2024; 51:596. [PMID: 38683461 DOI: 10.1007/s11033-024-09509-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 04/02/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Arnica montana and Bellis perennis are two medicinal plants that are thought to accelerate bone repair in homoeopathic literature. Mesenchymal stem cells (MSCs) are multipotent stem cells with the ability to differentiate and regenerate bone or osteogenesis. Hence, we aimed to determine the role of Arnica montana and Bellis perennis on the osteogenic differentiation of the C3H10T1/2 stem cell line. METHODS AND RESULTS The cell proliferation of Arnica montana and Bellis perennis was evaluated by MTT assay. Osteogenic differentiation of C3H10T1/2 was induced by the addition of β-glycerophosphate, ascorbic acid and dexamethasone in the differentiation medium over 3 weeks. Cells were treated with Arnica montana and Bellis perennis individually as well as in combination. The osteogenic differentiation potential of Arnica montana and Bellis perennis to differentiate C3H10T1/2 into osteoblasts was measured by alkaline phosphatase activity, alizarin red staining and the expression of Osteocalcin using immunostaining and qRT-PCR. Arnica montana and Bellis perennis could enhance C3H10T1/2 cell proliferation at 1600 µg. Further, the compound showed the ability to augment osteogenesis as confirmed by increased expression of alkaline phosphatase and enhanced calcium accumulation as seen by the Alizarin Red staining and quantification. Enhanced osteogenesis was further supported by the increased expression of osteocalcin in the treated cells with individual and combined doses of Arnica montana and Bellis perennis. Therefore, the findings provide additional support for the positive impact of Arnica montana and Bellis perennis on bone formation. CONCLUSIONS Our findings suggest that homoeopathic compounds Arnica montana and Bellis perennis can augment osteogenesis individually as well as in combination.
Collapse
Affiliation(s)
- Bhat K Deepika
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya Deemed to Be University, University Road, Deralakatte, Mangalore, Karnataka, 575018, India
| | - Nagendra H Apoorva
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya Deemed to Be University, University Road, Deralakatte, Mangalore, Karnataka, 575018, India
| | - Pinto R Joel
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya Deemed to Be University, University Road, Deralakatte, Mangalore, Karnataka, 575018, India
| | - Bipasha B
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya Deemed to Be University, University Road, Deralakatte, Mangalore, Karnataka, 575018, India
| | - Shenoy P Sudheer
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya Deemed to Be University, University Road, Deralakatte, Mangalore, Karnataka, 575018, India.
| |
Collapse
|
4
|
Verre J, Boisson M, Paumier A, Tribolo S, Boujedaini N. Anti-inflammatory effects of Arnica montana (mother tincture and homeopathic dilutions) in various cell models. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:117064. [PMID: 37598770 DOI: 10.1016/j.jep.2023.117064] [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: 06/28/2023] [Revised: 08/07/2023] [Accepted: 08/17/2023] [Indexed: 08/22/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The plant Arnica montana L. has been shown to alleviate inflammation, pain and swelling associated with trauma, and post-operative clinical conditions, yet the mechanism of action is not well understood. AIM OF THE STUDY The study was designed to investigate the effect of Arnica montana (A. montana) mother tincture and homeopathic dilutions on inflammation markers, oxidative stress and cell migration in diverse cell culture models. MATERIALS AND METHODS We tested A. montana mother tincture and a range of homeopathic dilutions in different human and murine cell culture models to demonstrate their anti-inflammatory properties by measuring the inflammatory markers: tumor necrosis factor alpha (TNFα), interleukin-6 (IL-6), cyclooxygenase-2 (COX-2), monocyte chemoattractant protein-1 (MCP-1), intercellular adhesion molecule (ICAM-1), reactive oxygen species (ROS) and cell migration. The inflammatory markers were measured by ELISA assays. The intracellular oxidative stress (ROS) in microglial cells was measured using Deep Red CellROX probe. The cell migration was examined by wound healing using the Oris Cell migration assay. RESULTS These data showed the ability of A. montana (mother tincture and mainly 1C dilution) to significantly reduce TNFα production in inflamed macrophages compared with vehicle (control). They significantly reduced both IL-6 and MCP-1 in inflamed human microglial cells and significantly decreased COX-2 expression in inflamed murine fibroblasts. Moreover, A. montana mother tincture reduced the cell migration whereas 9C dilution significantly enhanced the migration of fibroblast cells compared with vehicle. The expression of ICAM-1 was significantly reduced with A. montana mother tincture and 1C, 3C, 5C, and 9C dilutions in inflamed human endothelial cells compared with vehicle. A. montana mother tincture and 1C, 3C, 5C and 9C dilutions induced a significant and consistent effect on ROS production in inflamed murine microglial cells. A. montana 1C had the largest impact on ROS production. CONCLUSIONS Mother tincture and 1C dilution of A. montana showed anti-inflammatory properties assessed by measurement of several markers (pro-inflammatory cytokines, adhesion molecule, ROS) in various human and murine cell models. In addition, A. montana 3C, 5C, 9C dilutions have anti-inflammatory and antioxidant effects as highlighted on both primary endothelial cells and murine microglial cells.
Collapse
Affiliation(s)
- Justine Verre
- Laboratoires BOIRON, Research Department, 2 Avenue de l'Ouest Lyonnais, 69510, Messimy, France.
| | - Marie Boisson
- Laboratoires BOIRON, Research Department, 2 Avenue de l'Ouest Lyonnais, 69510, Messimy, France.
| | - Anne Paumier
- Laboratoires BOIRON, Research Department, 2 Avenue de l'Ouest Lyonnais, 69510, Messimy, France.
| | - Sandra Tribolo
- Laboratoires BOIRON, Research Department, 2 Avenue de l'Ouest Lyonnais, 69510, Messimy, France.
| | - Naoual Boujedaini
- Laboratoires BOIRON, Research Department, 2 Avenue de l'Ouest Lyonnais, 69510, Messimy, France.
| |
Collapse
|
5
|
Rath S, Jema JP, Kesavan K, Mallick S, Pradhan J, Chainy GBN, Nayak D, Kaushik S, Dandapat J. Arsenic album 30C exhibits crystalline nano structure of arsenic trioxide and modulates innate immune markers in murine macrophage cell lines. Sci Rep 2024; 14:745. [PMID: 38185726 PMCID: PMC10772077 DOI: 10.1038/s41598-024-51319-w] [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: 06/01/2023] [Accepted: 01/03/2024] [Indexed: 01/09/2024] Open
Abstract
Macrophages are associated with innate immune response and M1-polarized macrophages exhibit pro-inflammatory functions. Nanoparticles of natural or synthetic compounds are potential triggers of innate immunity. As2O3 is the major component of the homeopathic drug, Arsenic album 30C.This has been claimed to have immune-boosting activities, however, has not been validated experimentally. Here we elucidated the underlying mechanism of Ars. alb 30C-mediated immune priming in murine macrophage cell line. Transmission Electron Microscopy (TEM) and X-ray diffraction (XRD) used for the structural analysis of the drug reveals the presence of crystalline As2O3 nanoparticles of cubic structure. Similarly, signatures of M1-macrophage polarization were observed by surface enhanced Raman scattering (SERS) in RAW 264.7 cells with concomitant over expression of M1 cell surface marker, CD80 and transcription factor, NF-κB, respectively. We also observed a significant increase in pro-inflammatory cytokines like iNOS, TNF-α, IL-6, and COX-2 expression with unaltered ROS and apoptosis in drug-treated cells. Enhanced expression of Toll-like receptors 3 and 7 were observed both in transcriptional and translational levels after the drug treatment. In sum, our findings for the first time indicated the presence of crystalline As2O3 cubic nanostructure in Ars. alb 30C which facilitates modulation of innate immunity by activating macrophage polarization.
Collapse
Affiliation(s)
- Suvasmita Rath
- Centre of Environment, Climate Change and Public Health, Utkal University, Vani Vihar, Bhubaneswar, Odisha, India
- Post Graduate Department of Biotechnology, Utkal University, Bhubaneswar, Odisha, India
| | - Jyoti Prava Jema
- Post Graduate Department of Biotechnology, Utkal University, Bhubaneswar, Odisha, India
| | - Kamali Kesavan
- CSIR-Institute of Minerals and Materials Technology, Bhubaneswar, Odisha, India
| | - Sagar Mallick
- CSIR-Institute of Minerals and Materials Technology, Bhubaneswar, Odisha, India
| | - Jyotsnarani Pradhan
- Post Graduate Department of Biotechnology, Utkal University, Bhubaneswar, Odisha, India
| | | | - Debadatta Nayak
- Central Council for Research in Homeopathy, New Delhi, India
| | - Subhash Kaushik
- Central Council for Research in Homeopathy, New Delhi, India
| | - Jagneshwar Dandapat
- Post Graduate Department of Biotechnology, Utkal University, Bhubaneswar, Odisha, India.
- Centre of Excellence in Integrated Omics and Computational Biology, Utkal University, Bhubaneswar, Odisha, 751004, India.
| |
Collapse
|
6
|
Behera S, Sharma M, Lal R, Regar RK, Tripathi D, Gupta P, Kumar GVN, Verma D, Kaushik S, Khurana A. Investigation of The Effect of Metal-based Medicine Arsenicum album on Humoral Immune Response in SRBC-immunized Mice. Endocr Metab Immune Disord Drug Targets 2024; 24:1622-1627. [PMID: 38192149 DOI: 10.2174/0118715303273277231125150713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/05/2023] [Accepted: 10/16/2023] [Indexed: 01/10/2024]
Abstract
BACKGROUND In complementary and alternative medicinal systems, the Arsenicum album in ultra-high dilution was used in various therapeutic conditions, considering its effects on the body's immune system, including the COVID-19 pandemic. However, scientific evidence regarding its immunomodulatory effects is insufficient. OBJECTIVES The current study aimed to investigate the immunomodulatory effects of Arsenicum album in an experimental mouse model. MATERIALS AND METHODS Immunomodulatory activity of potentized dilutions of Arsenicum album i.e., 6C, 30C, 200C in BALB/c mice was evaluated by humoral antibody titer and delayedtype hypersensitivity assays wherein a fixed concentration (0.5 ml of 1× 109 cells/ml) of freshly prepared sheep RBC was administered as a foreign antigen to generate primary and secondary antibodies. RESULTS Arsenicum album showed significant immunomodulatory activity by increasing primary antibody titer evaluated on day 21 of the treatment in all the dilutions as compared to SRBC and vehicle control group in humoral immune response assay without showing any effect on delayed-type hypersensitivity. CONCLUSION The results of this preliminary study indicate that oral administration of Arsenicum album has the potential to augment primary humoral response at all dilutions. Hence, the possibility of using the Arsenicum album could be explored to treat immunological conditions, infections, etc., as an alternative therapy alongwith modern medicines.
Collapse
Affiliation(s)
- Sangita Behera
- Department of Pharmacology, Dr. D.P. Rastogi, Central Council for Research in Homoeopathy (DDPR-CRIH), Sector 24, Noida, Uttar Pradesh, 201301, India
| | - Mahima Sharma
- Department of Pharmacology, Dr. D.P. Rastogi, Central Council for Research in Homoeopathy (DDPR-CRIH), Sector 24, Noida, Uttar Pradesh, 201301, India
| | - Roshan Lal
- Department of Pharmacology, Dr. D.P. Rastogi, Central Council for Research in Homoeopathy (DDPR-CRIH), Sector 24, Noida, Uttar Pradesh, 201301, India
| | - Raj Kumar Regar
- Department of Pharmacology, Dr. D.P. Rastogi, Central Council for Research in Homoeopathy (DDPR-CRIH), Sector 24, Noida, Uttar Pradesh, 201301, India
| | - Deepika Tripathi
- Department of Pharmacology, Dr. D.P. Rastogi, Central Council for Research in Homoeopathy (DDPR-CRIH), Sector 24, Noida, Uttar Pradesh, 201301, India
| | - Pankaj Gupta
- Department of Pharmacology, Dr. D.P. Rastogi, Central Council for Research in Homoeopathy (DDPR-CRIH), Sector 24, Noida, Uttar Pradesh, 201301, India
| | | | - Digvijay Verma
- Drug Standarsise Unit, Central Council for Research in Homoeopathy, New Delhi, India
| | - Subhash Kaushik
- Drug Standarsise Unit, Central Council for Research in Homoeopathy, New Delhi, India
| | - Anil Khurana
- Drug Standarsise Unit, Central Council for Research in Homoeopathy, New Delhi, India
| |
Collapse
|