1
|
Dokic I, Moustafa M, Tessonnier T, Meister S, Ciamarone F, Akbarpour M, Krunic D, Haberer T, Debus J, Mairani A, Abdollahi A. Ultrahigh Dose Rate Helium Ion Beams: Minimizing Brain Tissue Damage while Preserving Tumor Control. Mol Cancer Ther 2025; 24:763-771. [PMID: 39739545 PMCID: PMC12046314 DOI: 10.1158/1535-7163.mct-24-0536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 11/16/2024] [Accepted: 12/19/2024] [Indexed: 01/02/2025]
Abstract
Ultrahigh dose rate radiotherapy (RT) with electrons and protons has shown potential for cancer treatment by effectively targeting tumors while sparing healthy tissues (FLASH effect). This study aimed to investigate the potential FLASH sparing effect of ultrahigh dose rate helium ion irradiation, focusing on acute brain injury and subcutaneous tumor response in a preclinical in vivo setting. Raster-scanned helium ion beams were used to compare the effects of standard dose rate (SDR; at 0.2 Gy/second) and FLASH (at 141 Gy/second) RT on healthy brain tissue. Irradiation-induced brain injury was studied in C57BL/6 mice via DNA damage response, using nuclear γH2AX as a marker for double-strand breaks. The integrity of neurovascular and immune compartments was assessed through CD31+ microvascular density and activation of microglia/macrophages. IBA1+ ramified and CD68+ phagocytic microglia/macrophages were quantified, along with the expression of inducible nitric oxide synthetase. Tumor response to SDR (0.2 Gy/second) and FLASH (250 Gy/second) RT was evaluated in an A549 carcinoma model, using tumor volume and Kaplan-Meier survival as endpoints. The results showed that helium FLASH RT significantly reduced acute brain tissue injury compared with SDR, evidenced by lower levels of double-strand breaks and preserved the neurovascular endothelium. Additionally, FLASH RT reduced neuroinflammatory signals compared with SDR, as indicated by fewer CD68+ inducible nitric oxide synthetase-positive microglia/macrophages. FLASH RT achieved tumor control comparable with that of SDR RT. To the best of our knowledge, this is the first study to report the FLASH sparing effect of raster scanning helium ion RT in vivo, highlighting its potential for neuroprotection and effective tumor control.
Collapse
Affiliation(s)
- Ivana Dokic
- Clinical Cooperation Unit Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Molecular and Translational Radiation Oncology, Department of Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
- German Cancer Consortium (DKTK) Core-Center Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Oncology (NCRO), Heidelberg University and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
| | - Mahmoud Moustafa
- Clinical Cooperation Unit Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Molecular and Translational Radiation Oncology, Department of Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
- German Cancer Consortium (DKTK) Core-Center Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Oncology (NCRO), Heidelberg University and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
- Department of Clinical Pathology, Suez Canal University, Ismailia, Egypt
| | - Thomas Tessonnier
- Clinical Cooperation Unit Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
| | - Sarah Meister
- Clinical Cooperation Unit Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Molecular and Translational Radiation Oncology, Department of Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
- German Cancer Consortium (DKTK) Core-Center Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Oncology (NCRO), Heidelberg University and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
| | - Federica Ciamarone
- Clinical Cooperation Unit Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Molecular and Translational Radiation Oncology, Department of Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
- German Cancer Consortium (DKTK) Core-Center Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Oncology (NCRO), Heidelberg University and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
| | - Mahdi Akbarpour
- Clinical Cooperation Unit Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Molecular and Translational Radiation Oncology, Department of Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
- German Cancer Consortium (DKTK) Core-Center Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Oncology (NCRO), Heidelberg University and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
| | - Damir Krunic
- Light Microscopy Facility, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Thomas Haberer
- Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
| | - Jürgen Debus
- Clinical Cooperation Unit Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Molecular and Translational Radiation Oncology, Department of Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
- German Cancer Consortium (DKTK) Core-Center Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Oncology (NCRO), Heidelberg University and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
| | - Andrea Mairani
- Clinical Cooperation Unit Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
- National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Amir Abdollahi
- Clinical Cooperation Unit Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Molecular and Translational Radiation Oncology, Department of Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
- German Cancer Consortium (DKTK) Core-Center Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Oncology (NCRO), Heidelberg University and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
| |
Collapse
|
2
|
Shen Y, Lin P. The Role of Cytokines in Postherpetic Neuralgia. J Integr Neurosci 2025; 24:25829. [PMID: 40302252 DOI: 10.31083/jin25829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/02/2024] [Accepted: 10/23/2024] [Indexed: 05/02/2025] Open
Abstract
Nerve injury is a significant cause of postherpetic neuralgia (PHN). It is marked by upregulated expression of cytokines secreted by immune cells such as tumor necrosis factor alpha, interleukin 1 beta (IL-1β), IL-6, IL-18, and IL-10. In neuropathic pain (NP) due to nerve injury, cytokines are important for the induction of neuroinflammation, activation of glial cells, and expression of cation channels. The release of chemokines due to nerve injury promotes immune cell infiltration, recruiting inflammatory cytokines and further amplifying the inflammatory response. The resulting disequilibrium in neuroimmune response and neuroinflammation leads to a reduction of nerve fibers, altered nerve excitability, and neuralgia. PHN is a typical NP and cytokines may induce PHN by promoting central and peripheral sensitization. Currently, treating PHN is challenging and research on the role of cytokine signaling pathways in PHN is lacking. This review summarizes the potential mechanisms of cytokine-mediated PHN and discusses the cytokine signaling pathways associated with the central and peripheral sensitization of PHN. By elucidating the mechanisms of cytokines, the cells and molecules that regulate cytokines, and their signaling systems in PHN, this review reveals important research developments regarding cytokines and their signaling pathways mediating PHN, highlighting new targets of action for the development of analgesic drugs.
Collapse
Affiliation(s)
- Yunyan Shen
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, 310053 Hangzhou, Zhejiang, China
| | - Ping Lin
- Department of Geriatrics, Hangzhou Third People's Hospital, 310009 Hangzhou, Zhejiang, China
| |
Collapse
|
3
|
Dokic I, Moustafa M, Tessonnier T, Meister S, Ciamarone F, Akbarpour M, Krunic D, Haberer T, Debus J, Mairani A, Abdollahi A. Ultra-High Dose Rate Helium Ion Beams: Minimizing Brain Tissue Damage while Preserving Tumor Control. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.13.598785. [PMID: 38915610 PMCID: PMC11195254 DOI: 10.1101/2024.06.13.598785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Ultra-high dose rate radiotherapy with electrons and protons has shown potential for cancer treatment by effectively targeting tumors while sparing healthy tissues (FLASH effect). This study aimed to investigate the potential FLASH sparing effect of ultra-high-dose rate helium ion irradiation, focusing on acute brain injury and subcutaneous tumor response in a preclinical in vivo setting. Raster-scanned helium ion beams were used to compare the effects of standard dose rate (SDR at 0.2 Gy/s) and FLASH (at 141 Gy/s) radiotherapy on healthy brain tissue. Irradiation-induced brain injury was studied in C57BL/6 mice via DNA damage response, using nuclear γH2AX as a marker for double-strand breaks (DSB). The integrity of neurovascular and immune compartments was assessed through CD31 + microvascular density and activation of microglia/macrophages. Iba1+ ramified and CD68 + phagocytic microglia/macrophages were quantified, along with the expression of inducible nitric oxide synthetase (iNOS). Tumor response to SDR (0.2 Gy/s) and FLASH (250 Gy/s) radiotherapy was evaluated in A549 carcinoma model, using tumor volume and Kaplan-Meier survival as endpoints. The results showed that helium FLASH radiotherapy significantly reduced acute brain tissue injury compared to SDR, evidenced by lower levels of DSB and preserved neurovascular endothelium. Additionally, FLASH radiotherapy reduced neuroinflammatory signals compared to SDR, as indicated by fewer CD68+ iNOS+ microglia/macrophages. FLASH radiotherapy achieved tumor control comparable to that of SDR radiotherapy. This study is the first to report the FLASH sparing effect of raster scanning helium ion radiotherapy in vivo, highlighting its potential for neuroprotection and effective tumor control.
Collapse
|
4
|
Shweta, Sharma K, Shakarad M, Agrawal N, Maurya SK. Drosophila glial system: an approach towards understanding molecular complexity of neurodegenerative diseases. Mol Biol Rep 2024; 51:1146. [PMID: 39532789 DOI: 10.1007/s11033-024-10075-w] [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: 09/07/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
Glia is pivotal in regulating neuronal stem cell proliferation, functioning, and nervous system homeostasis, significantly influencing neuronal health and disorders. Dysfunction in glial activity is a key factor in the development and progression of brain pathology. However, a deeper understanding of the intricate nature of glial cells and their diverse role in neurological disorders is still required. To this end, we conducted data mining to retrieve literature from PubMed and Google Scholar using the keywords: glia, Drosophila, neurodegeneration, and mammals. The retrieved literature was manually screened and used to comprehensively understand and present the different glial types in Drosophila, i.e., perineurial, subperineurial, cortex, astrocyte-like and ensheathing glia, their relevance with mammalian counterparts, mainly microglia and astrocytes, and their potential to reveal complex neuron-glial molecular networks in managing neurodegenerative processes.
Collapse
Affiliation(s)
- Shweta
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, 110007, India
| | - Khushboo Sharma
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Mallikarjun Shakarad
- Evolutionary Biology Laboratory, Department of Zoology, University of Delhi, Delhi, 110007, India
| | - Namita Agrawal
- Fly Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, 110007, India
| | - Shashank Kumar Maurya
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, 110007, India.
| |
Collapse
|
5
|
Lana JF, Navani A, Jeyaraman M, Santos N, Pires L, Santos GS, Rodrigues IJ, Santos D, Mosaner T, Azzini G, da Fonseca LF, de Macedo AP, Huber SC, de Moraes Ferreira Jorge D, Purita J. Sacral Bioneuromodulation: The Role of Bone Marrow Aspirate in Spinal Cord Injuries. Bioengineering (Basel) 2024; 11:461. [PMID: 38790327 PMCID: PMC11118755 DOI: 10.3390/bioengineering11050461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/26/2024] [Accepted: 04/26/2024] [Indexed: 05/26/2024] Open
Abstract
Spinal cord injury (SCI) represents a severe trauma to the nervous system, leading to significant neurological damage, chronic inflammation, and persistent neuropathic pain. Current treatments, including pharmacotherapy, immobilization, physical therapy, and surgical interventions, often fall short in fully addressing the underlying pathophysiology and resultant disabilities. Emerging research in the field of regenerative medicine has introduced innovative approaches such as autologous orthobiologic therapies, with bone marrow aspirate (BMA) being particularly notable for its regenerative and anti-inflammatory properties. This review focuses on the potential of BMA to modulate inflammatory pathways, enhance tissue regeneration, and restore neurological function disrupted by SCI. We hypothesize that BMA's bioactive components may stimulate reparative processes at the cellular level, particularly when applied at strategic sites like the sacral hiatus to influence lumbar centers and higher neurological structures. By exploring the mechanisms through which BMA influences spinal repair, this review aims to establish a foundation for its application in clinical settings, potentially offering a transformative approach to SCI management that extends beyond symptomatic relief to promoting functional recovery.
Collapse
Affiliation(s)
- José Fábio Lana
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (N.S.); (L.P.); (I.J.R.); (D.S.); (T.M.); (G.A.); (L.F.d.F.); (A.P.d.M.); (S.C.H.); (D.d.M.F.J.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
- Clinical Research, Anna Vitória Lana Institute (IAVL), Indaiatuba 13334-170, SP, Brazil
- Medical School, Max Planck University Center (UniMAX), Indaiatuba 13343-060, SP, Brazil
| | - Annu Navani
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
- Medical School, Max Planck University Center (UniMAX), Indaiatuba 13343-060, SP, Brazil
- Comprehensive Spine & Sports Center, Campbell, CA 95008, USA
| | - Madhan Jeyaraman
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
- Department of Orthopaedics, ACS Medical College and Hospital, Chennai 600077, Tamil Nadu, India
| | - Napoliane Santos
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (N.S.); (L.P.); (I.J.R.); (D.S.); (T.M.); (G.A.); (L.F.d.F.); (A.P.d.M.); (S.C.H.); (D.d.M.F.J.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
| | - Luyddy Pires
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (N.S.); (L.P.); (I.J.R.); (D.S.); (T.M.); (G.A.); (L.F.d.F.); (A.P.d.M.); (S.C.H.); (D.d.M.F.J.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
| | - Gabriel Silva Santos
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (N.S.); (L.P.); (I.J.R.); (D.S.); (T.M.); (G.A.); (L.F.d.F.); (A.P.d.M.); (S.C.H.); (D.d.M.F.J.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
| | - Izair Jefthé Rodrigues
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (N.S.); (L.P.); (I.J.R.); (D.S.); (T.M.); (G.A.); (L.F.d.F.); (A.P.d.M.); (S.C.H.); (D.d.M.F.J.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
| | - Douglas Santos
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (N.S.); (L.P.); (I.J.R.); (D.S.); (T.M.); (G.A.); (L.F.d.F.); (A.P.d.M.); (S.C.H.); (D.d.M.F.J.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
| | - Tomas Mosaner
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (N.S.); (L.P.); (I.J.R.); (D.S.); (T.M.); (G.A.); (L.F.d.F.); (A.P.d.M.); (S.C.H.); (D.d.M.F.J.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
| | - Gabriel Azzini
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (N.S.); (L.P.); (I.J.R.); (D.S.); (T.M.); (G.A.); (L.F.d.F.); (A.P.d.M.); (S.C.H.); (D.d.M.F.J.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
| | - Lucas Furtado da Fonseca
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (N.S.); (L.P.); (I.J.R.); (D.S.); (T.M.); (G.A.); (L.F.d.F.); (A.P.d.M.); (S.C.H.); (D.d.M.F.J.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
- Medical School, Federal University of São Paulo (UNIFESP), São Paulo 04024-002, SP, Brazil
| | - Alex Pontes de Macedo
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (N.S.); (L.P.); (I.J.R.); (D.S.); (T.M.); (G.A.); (L.F.d.F.); (A.P.d.M.); (S.C.H.); (D.d.M.F.J.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
| | - Stephany Cares Huber
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (N.S.); (L.P.); (I.J.R.); (D.S.); (T.M.); (G.A.); (L.F.d.F.); (A.P.d.M.); (S.C.H.); (D.d.M.F.J.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
| | - Daniel de Moraes Ferreira Jorge
- Department of Orthopedics, Brazilian Institute of Regenerative Medicine (BIRM), Indaiatuba 13334-170, SP, Brazil; (J.F.L.); (N.S.); (L.P.); (I.J.R.); (D.S.); (T.M.); (G.A.); (L.F.d.F.); (A.P.d.M.); (S.C.H.); (D.d.M.F.J.)
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
| | - Joseph Purita
- Regenerative Medicine, Orthoregen International Course, Indaiatuba 13334-170, SP, Brazil; (A.N.); (J.P.)
- Medical School, Max Planck University Center (UniMAX), Indaiatuba 13343-060, SP, Brazil
| |
Collapse
|
6
|
Floare ML, Wharton SB, Simpson JE, Aeschlimann D, Hoggard N, Hadjivassiliou M. Cerebellar degeneration in gluten ataxia is linked to microglial activation. Brain Commun 2024; 6:fcae078. [PMID: 38510211 PMCID: PMC10953628 DOI: 10.1093/braincomms/fcae078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/16/2024] [Accepted: 03/05/2024] [Indexed: 03/22/2024] Open
Abstract
Gluten sensitivity has long been recognized exclusively for its gastrointestinal involvement; however, more recent research provides evidence for the existence of neurological manifestations that can appear in combination with or independent of the small bowel manifestations. Amongst all neurological manifestations of gluten sensitivity, gluten ataxia is the most commonly occurring one, accounting for up to 40% of cases of idiopathic sporadic ataxia. However, despite its prevalence, its neuropathological basis is still poorly defined. Here, we provide a neuropathological characterization of gluten ataxia and compare the presence of neuroinflammatory markers glial fibrillary acidic protein, ionized calcium-binding adaptor molecule 1, major histocompatibility complex II and cluster of differentiation 68 in the central nervous system of four gluten ataxia cases to five ataxia controls and seven neurologically healthy controls. Our results demonstrate that severe cerebellar atrophy, cluster of differentiation 20+ and cluster of differentiation 8+ lymphocytic infiltration in the cerebellar grey and white matter and a significant upregulation of microglial immune activation in the cerebellar granular layer, molecular layer and cerebellar white matter are features of gluten ataxia, providing evidence for the involvement of both cellular and humoral immune-mediated processes in gluten ataxia pathogenesis.
Collapse
Affiliation(s)
- Mara-Luciana Floare
- Sheffield Institute for Translational Neuroscience, The University of Sheffield, Sheffield S10 2HQ, UK
| | - Stephen B Wharton
- Sheffield Institute for Translational Neuroscience, The University of Sheffield, Sheffield S10 2HQ, UK
| | - Julie E Simpson
- Sheffield Institute for Translational Neuroscience, The University of Sheffield, Sheffield S10 2HQ, UK
| | - Daniel Aeschlimann
- Matrix Biology and Tissue Repair Research Unit, College of Biomedical and Life Sciences, School of Dentistry, Cardiff University, Cardiff CF14 4XY, UK
| | - Nigel Hoggard
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield S10 2JF, UK
| | - Marios Hadjivassiliou
- Academic Department of Neuroscience, Sheffield Teaching Hospitals NHS Trust, Royal Hallamshire Hospital, Sheffield S10 2JF, UK
| |
Collapse
|
7
|
El-Helaly A, Abou-El-Naga AM, Alshehri KM, El-Dein MA. Miracle Tree ( Moringa oleifera) Attuned GFAP and Synaptophysin Levels, Oxidative Stress and Biomarkers in Cerebellar Fluorosis of Pregnant Rats. Pak J Biol Sci 2023; 26:628-650. [PMID: 38334155 DOI: 10.3923/pjbs.2023.628.650] [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] [Indexed: 02/10/2024]
Abstract
<b>Background and Objective:</b> Cerebellar fluorosis is a health issue associated with excessive exposure to fluoride (F) either in direct or indirect ways as pesticides, drinking water and caries preventing prescriptions. It is characterized by elevation in oxidative stress, inflammation, demyelination and Purkinje cell loss. <i>Moringa oleifera</i> (M), is a widely cultivated plant used as a health-booster agent in modulating various disorders because of its high content of vitamins and minerals. The beneficial effect of moringa against fluoride-induced cerebellar toxicity in pregnant rats was investigated in this study. <b>Materials and Methods:</b> Twenty pregnant rats were administered daily 300 mg kg<sup></sup><sup>1</sup> <i>M. oleifera</i> aqueous extract incorporated with 10 mg kg<sup></sup><sup>1</sup> of F intoxication from the 1st day of gestation until the 20th day. Following the termination of the trial, sera were collected and cerebellar tissue was removed for further examinations, along with the assessment of maternity. <b>Results:</b> The <i>M. oleifera</i> significantly normalized serum FSH, LH, progesterone, dopamine and serotonin levels of F-intoxicated mothers. Additionally, <i>M. oleifera</i> markedly prevented the lipid peroxidation and DNA fragmentation indicated by the tail length and moment in comet assay (-34.4 and -75.3%, respectively, when compared to the fluoride intoxicated group), while sustaining the levels of SOD and CAT revealing its antioxidant activity. The <i>M. oleifera</i> regressed the cerebellar α-amylase (-25.4%) and acetylcholinesterase activity (-40.6%), also attenuated GFAP (-73.4%, p<0.0001), synaptophysin level (216.6%, p<0.0001) and IL-6 expression (-91.2%) comparing to fluoride only treated mothers. <b>Conclusion:</b> Histological and ultrastructural examinations confirmed the recuperating effects of <i>M. oleifera</i> on mothers' cerebellar tissue intoxicated with fluoride indicated by intact folia and restored Purkinje cells number and architecture. The maternal study emphasized the anti-abortifacient activity of moringa against fluoride induced-fetotoxicity.
Collapse
|
8
|
Khan D, Bedner P, Müller J, Lülsberg F, Henning L, Prinz M, Steinhäuser C, Muhammad S. TGF-β Activated Kinase 1 (TAK1) Is Activated in Microglia After Experimental Epilepsy and Contributes to Epileptogenesis. Mol Neurobiol 2023; 60:3413-3422. [PMID: 36862288 PMCID: PMC10122619 DOI: 10.1007/s12035-023-03290-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 02/19/2023] [Indexed: 03/03/2023]
Abstract
Increasing evidence suggests that inflammation promotes epileptogenesis. TAK1 is a central enzyme in the upstream pathway of NF-κB and is known to play a central role in promoting neuroinflammation in neurodegenerative diseases. Here, we investigated the cellular role of TAK1 in experimental epilepsy. C57Bl6 and transgenic mice with inducible and microglia-specific deletion of Tak1 (Cx3cr1CreER:Tak1fl/fl) were subjected to the unilateral intracortical kainate mouse model of temporal lobe epilepsy (TLE). Immunohistochemical staining was performed to quantify different cell populations. The epileptic activity was monitored by continuous telemetric electroencephalogram (EEG) recordings over a period of 4 weeks. The results show that TAK1 was activated predominantly in microglia at an early stage of kainate-induced epileptogenesis. Tak1 deletion in microglia resulted in reduced hippocampal reactive microgliosis and a significant decrease in chronic epileptic activity. Overall, our data suggest that TAK1-dependent microglial activation contributes to the pathogenesis of chronic epilepsy.
Collapse
Affiliation(s)
- Dilaware Khan
- Department of Neurosurgery, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany.,Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Peter Bedner
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Julia Müller
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Fabienne Lülsberg
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Lukas Henning
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Christian Steinhäuser
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - Sajjad Muhammad
- Department of Neurosurgery, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany. .,Department of Neurosurgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
| |
Collapse
|
9
|
Ferreyra S, González S. Therapeutic potential of progesterone in spinal cord injury-induced neuropathic pain: At the crossroads between neuroinflammation and N-methyl-D-aspartate receptor. J Neuroendocrinol 2023; 35:e13181. [PMID: 35924434 DOI: 10.1111/jne.13181] [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: 04/27/2022] [Revised: 06/13/2022] [Accepted: 06/19/2022] [Indexed: 10/17/2022]
Abstract
In recent decades, an area of active research has supported the notion that progesterone promotes a wide range of remarkable protective actions in experimental models of nervous system trauma or disease, and has also provided a strong basis for considering this steroid as a promising molecule for modulating the complex maladaptive changes that lead to neuropathic pain, especially after spinal cord injury. In this review, we intend to give the readers a brief appraisal of the main mechanisms underlying the increased excitability of the spinal circuit in the pain pathway after trauma, with particular emphasis on those mediated by the activation of resident glial cells, the subsequent release of proinflammatory cytokines and their impact on N-methyl-D-aspartate receptor function. We then summarize the available preclinical data pointing to progesterone as a valuable repurposing molecule for blocking critical cellular and molecular events that occur in the dorsal horn of the injured spinal cord and are related to the development of chronic pain. Since the treatment and management of neuropathic pain after spinal injury remains challenging, the potential therapeutic value of progesterone opens new traslational perspectives to prevent central pain.
Collapse
Affiliation(s)
- Sol Ferreyra
- Instituto de Biología y Medicina Experimental, Laboratorio de Nocicepción y Dolor Neuropático, CONICET, Buenos Aires, Argentina
| | - Susana González
- Instituto de Biología y Medicina Experimental, Laboratorio de Nocicepción y Dolor Neuropático, CONICET, Buenos Aires, Argentina
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Buenos Aires, Argentina
| |
Collapse
|
10
|
Lampiasi N, Bonaventura R, Deidda I, Zito F, Russo R. Inflammation and the Potential Implication of Macrophage-Microglia Polarization in Human ASD: An Overview. Int J Mol Sci 2023; 24:2703. [PMID: 36769026 PMCID: PMC9916462 DOI: 10.3390/ijms24032703] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/23/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
Autism spectrum disorder (ASD) is a heterogeneous collection of neurodevelopmental disorders, difficult to diagnose and currently lacking treatment options. The possibility of finding reliable biomarkers useful for early identification would offer the opportunity to intervene with treatment strategies to improve the life quality of ASD patients. To date, there are many recognized risk factors for the development of ASD, both genetic and non-genetic. Although genetic and epigenetic factors may play a critical role, the extent of their contribution to ASD risk is still under study. On the other hand, non-genetic risk factors include pollution, nutrition, infection, psychological states, and lifestyle, all together known as the exposome, which impacts the mother's and fetus's life, especially during pregnancy. Pathogenic and non-pathogenic maternal immune activation (MIA) and autoimmune diseases can cause various alterations in the fetal environment, also contributing to the etiology of ASD in offspring. Activation of monocytes, macrophages, mast cells and microglia and high production of pro-inflammatory cytokines are indeed the cause of neuroinflammation, and the latter is involved in ASD's onset and development. In this review, we focused on non-genetic risk factors, especially on the connection between inflammation, macrophage polarization and ASD syndrome, MIA, and the involvement of microglia.
Collapse
Affiliation(s)
- Nadia Lampiasi
- Istituto per la Ricerca e l’Innovazione Biomedica IRIB, Consiglio Nazionale delle Ricerche, Via Ugo La Malfa 153, 90146 Palermo, Italy
| | | | | | | | | |
Collapse
|
11
|
Resveratrol Inhibits Oxidative Stress and Regulates M1/M2-Type Polarization of Microglia via Mediation of the Nrf2/Shh Signaling Cascade after OGD/R Injury In Vitro. J Pers Med 2022; 12:jpm12122087. [PMID: 36556306 PMCID: PMC9782981 DOI: 10.3390/jpm12122087] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 12/09/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
AIMS Microglia are closely related to the occurrence and development of oxidative stress. Cerebral ischemia leads to abnormal activation of microglia. Resveratrol can regulate M1/M2-type microglia polarization, but the underlying mechanism is not well understood, although the Nrf2 and Shh signaling pathways may be involved. Given that resveratrol activates Shh, the present study examined whether this is mediated by Nrf2 signaling. METHODS N9 microglia were pretreated with drugs before oxygen-glucose deprivation/reoxygenation (OGD/R). HT22 neurons were also used for conditional co-culture with microglia. Cell viability was measured by CCK-8 assay. MDA levels and SOD activity in the supernatant were detected by TBA and WST-1, respectively. Immunofluorescence detected Nrf2 and Gli1 nuclear translocation. The levels of CD206, Arg1, iNOS, TNF-α, Nrf2, HO-1, NQO1, Shh, Ptc, Smo, Gli1 protein and mRNA were measured by Western blotting or RT-qPCR. Annexin V-FITC Flow Cytometric Analysis detected apoptosis. RESULTS Resveratrol and Nrf2 activator RTA-408 enhanced the viability of microglia, reduced oxidative stress, promoted M2-type microglia polarization and activated Nrf2 and Shh signaling. ML385, a selective inhibitor of Nrf2, decreased the viability of microglia, aggravated oxidative stress, promoted M1-type microglia polarization and inhibited Nrf2 and Shh signaling. Moreover, resveratrol and RTA-408-treated microglia can reduce the apoptosis and increase the viability of HT22 neurons, while ML385-treated microglia aggravated the apoptosis and weakened the viability of HT22 neurons. CONCLUSIONS These results demonstrated that resveratrol may inhibit oxidative stress, regulate M1/M2-type polarization of microglia and decrease neuronal injury in conditional co-culture of neurons and microglia via the mediation of the Nrf2/Shh signaling cascade after OGD/R injury in vitro.
Collapse
|
12
|
MiR-200c-3p inhibits LPS-induced M1 polarization of BV2 cells by targeting RIP2. Genes Genomics 2022; 44:477-486. [PMID: 35013887 PMCID: PMC8921044 DOI: 10.1007/s13258-021-01210-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 12/17/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Microglia are important immune cells, which can be induced by lipopolysaccharide (LPS) into M1 phenotype that express pro-inflammatory cytokines. Some studies have shown that microRNAs play critical roles in microglial activation. OBJECTIVE This study was designed to investigate the role of miR-200c-3p in regulating inflammatory responses of LPS-treated BV2 cells. METHODS The expression of miR-200c-3p in BV2 cells was detected by real-time PCR. Receptor-interacting protein 2 (RIP2) was predicted as a target gene of miR-200c-3p. Their relationship was verified by dual-luciferase reporter assay. The function of miR-200c-3p and RIP2 in microglial polarization and NF-κB signaling was further evaluated. RESULTS LPS treatment reduced miR-200c-3p expression in a dose-dependent and time-dependent manner in BV2 cells. LPS treatment increased the expression of M1 phenotype markers inducible nitric oxide synthase (iNOS) and major histocompatibility complex class (MHC)-II, promoted the release of pro-inflammatory cytokines interleukin (IL)-1β, IL-6 and tumor necrosis factor (TNF)-α, and enhanced the nuclear translocation and phosphorylation of nuclear factor-kappaB (NF-κB) p65. Reversely, miR-200c-3p mimics down-regulated the levels of these inflammatory factors. Furthermore, RIP2 was identified to be a direct target of miR-200c-3p. RIP2 knockdown had a similar effect to miR-200c-3p mimics. Overexpression of RIP2 eliminated the inhibitory effect of miR-200c-3p on LPS-induced M1 polarization and NF-κB activation in BV2 cells. CONCLUSIONS MiR-200c-3p mimics suppressed LPS-induced microglial M1 polarization and NF-κB activation by targeting RIP2. MiR-200c-3p/RIP2 might be a potential therapeutic target for the treatment of neuroinflammation-associated diseases.
Collapse
|
13
|
Jaffa AA, Jaffa MA, Moussa M, Ahmed IA, Karam M, Aldeen KS, Al Sayegh R, El-Achkar GA, Nasrallah L, Yehya Y, Habib A, Ziyadeh FN, Eid AH, Kobeissy FH, Jaffa AA. Modulation of Neuro-Inflammatory Signals in Microglia by Plasma Prekallikrein and Neuronal Cell Debris. Front Pharmacol 2021; 12:743059. [PMID: 34867349 PMCID: PMC8636058 DOI: 10.3389/fphar.2021.743059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 10/28/2021] [Indexed: 11/13/2022] Open
Abstract
Microglia, the resident phagocytes of the central nervous system and one of the key modulators of the innate immune system, have been shown to play a major role in brain insults. Upon activation in response to neuroinflammation, microglia promote the release of inflammatory mediators as well as promote phagocytosis. Plasma prekallikrein (PKall) has been recently implicated as a mediator of neuroinflammation; nevertheless, its role in mediating microglial activation has not been investigated yet. In the current study, we evaluate the mechanisms through which PKall contributes to microglial activation and release of inflammatory cytokines assessing PKall-related receptors and their dynamics. Murine N9-microglial cells were exposed to PKall (2.5 ng/ml), lipopolysaccharide (100 ng/ml), bradykinin (BK, 0.1 μM), and neuronal cell debris (16.5 μg protein/ml). Gene expression of bradykinin 2 receptor (B2KR), protease-activated receptor 2 (PAR-2), along with cytokines and fibrotic mediators were studied. Bioinformatic analysis was conducted to correlate altered protein changes with microglial activation. To assess receptor dynamics, HOE-140 (1 μM) and GB-83 (2 μM) were used to antagonize the B2KR and PAR-2 receptors, respectively. Also, the role of autophagy in modulating microglial response was evaluated. Data from our work indicate that PKall, LPS, BK, and neuronal cell debris resulted in the activation of microglia and enhanced expression/secretion of inflammatory mediators. Elevated increase in inflammatory mediators was attenuated in the presence of HOE-140 and GB-83, implicating the engagement of these receptors in the activation process coupled with an increase in the expression of B2KR and PAR-2. Finally, the inhibition of autophagy significantly enhanced the release of the cytokine IL-6 which were validated via bioinformatics analysis demonstrating the role of PKall in systematic and brain inflammatory processes. Taken together, we demonstrated that PKall can modulate microglial activation via the engagement of PAR-2 and B2KR where PKall acts as a neuromodulator of inflammatory processes.
Collapse
Affiliation(s)
- Aneese A Jaffa
- Department of Biology, Faculty of Arts and Sciences, American University of Beirut, Beirut, Lebanon
| | - Miran A Jaffa
- Epidemiology and Population Health Department, Faculty of Health Sciences, American University of Beirut, Beirut, Lebanon
| | - Mayssam Moussa
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ibrahim A Ahmed
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Mia Karam
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Kawthar Sharaf Aldeen
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Rola Al Sayegh
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,INSERM-UMR1149, Centre de Recherche sur l'Inflammation, and Sorbonne Paris Cité, Laboratoire d'Excellence Inflamex, Faculté de Médecine, Universite de Paris, Paris, France
| | - Ghewa A El-Achkar
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Leila Nasrallah
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Yara Yehya
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Aida Habib
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,INSERM-UMR1149, Centre de Recherche sur l'Inflammation, and Sorbonne Paris Cité, Laboratoire d'Excellence Inflamex, Faculté de Médecine, Universite de Paris, Paris, France
| | - Fuad N Ziyadeh
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,Department of Internal Medicine, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar.,Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Firas H Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ayad A Jaffa
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
14
|
Strath LJ, Totsch SK, Quinn TL, Menard M, Philip George A, Lukens SL, Simmons J, Zhang Y, Sorge RE. The effect of the Standard American Diet on Iba-1 immunoreactivity in the spinal cord before and after peripheral inflammatory injury in rats. PHARMANUTRITION 2021. [DOI: 10.1016/j.phanu.2021.100278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
15
|
Chhetri G. Emerging roles of IL-34 in neurodegenerative and neurological infectious disease. Int J Neurosci 2021; 133:660-671. [PMID: 34347576 DOI: 10.1080/00207454.2021.1963962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Neurological infections are often devastating in their clinical presentation. Although significant advances have made in neuroimaging techniques and molecular tools for diagnosis, as well as in anti-infective therapy, these diseases always difficult to diagnose and treat. Neuroparasitic infections and virus infections lead to neurological infections. In the nervous system, various cytokines and chemokines act as neuroinflammatory agents, neuromodulators, regulate neurodevelopment, and synaptic transmission. Among the most important cytokines, interleukins (ILs) are a large group of immunomodulatory proteins that elicit a wide variety of responses in cells and tissues. These ILs are involved in pro and anti-inflammatory effects, systemic inflammation, immune system modulation and play crucial roles in fighting cancer, infectious disease, and neurological disorders. Interleukin-34 (IL-34) identified by screening a comprehensive human protein library containing ∼3400 secreted and extracellular domain proteins in a human monocyte viability assay. Recent evidence has disclosed the crucial roles of IL-34 in the proliferation and differentiation of mononuclear phagocyte lineage cells, osteoclastogenesis, and inflammation. Additionally, IL-34 plays an important role in development, homeostasis, and disease. Dysregulation in IL-34 function can lead to various inflammatory and infectious diseases (e.g. Inflammatory bowel disease, liver fibrosis, Systemic Lupus erythematosus, rheumatoid arthritis), neurological disorders (e.g. Alzheimer disease) and neurological infectious disease (e.g. West Nile virus disease). In this review, we explore the biological role of IL-34 in addition to various impairments caused by dysregulation in IL-34 and discuss their potential links that may lead to important therapeutic and/or preventive strategies for these disorders.
Collapse
Affiliation(s)
- Gaurav Chhetri
- School of Pharmacy, Shanghai Jiao Tong University, Minhang, Shanghai, P.R. China
| |
Collapse
|
16
|
Pautrat A, Vassal F, Casteillo F, Magné N, Péoc'h M, Forest F. Glioma Associated Microglia/Macrophages Distribution is Conserved in Glioblastoma Regrowth: A Morphometric Study. Cancer Invest 2021; 39:621-626. [PMID: 34151673 DOI: 10.1080/07357907.2021.1944179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
We compared the morphological aspect of glioblastoma-associated microglia/macrophages cells in 15 paired recurrent glioblastomas to check the ability of glioblastoma to recreate its microenvironment. The absolute number of GAMs is lower in normal tissue (21/mm2) than in the isolated tumor cells area (100-112/mm2) than in the solid tumor area (212-220/mm2) (p < 0.01). The morphology of GAMs remained the same in each tumor area with a reduced covered area by cell processes (196 to 216/mm2) than in normal tissue (708/mm2) (p < 0.01). In paired tumors, GAMs morphology remained the same in successive resections and was not modified by the treatments.
Collapse
Affiliation(s)
- Arnaud Pautrat
- Department of Pathology, University Jean Monnet, Saint Etienne, France
| | - François Vassal
- Department of Neurosurgery, University Jean Monnet, Saint Etienne, France
| | | | - Nicolas Magné
- Department of Radiotherapy, University Jean Monnet, Saint Etienne, France
| | - Michel Péoc'h
- Department of Pathology, University Jean Monnet, Saint Etienne, France
| | - Fabien Forest
- Department of Pathology, University Jean Monnet, Saint Etienne, France
| |
Collapse
|
17
|
IKBKB siRNA-Encapsulated Poly (Lactic- co-Glycolic Acid) Nanoparticles Diminish Neuropathic Pain by Inhibiting Microglial Activation. Int J Mol Sci 2021; 22:ijms22115657. [PMID: 34073390 PMCID: PMC8203094 DOI: 10.3390/ijms22115657] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 05/19/2021] [Accepted: 05/21/2021] [Indexed: 01/08/2023] Open
Abstract
Activation of nuclear factor-kappa B (NF-κB) in microglia plays a decisive role in the progress of neuropathic pain, and the inhibitor of kappa B (IκB) is a protein that blocks the activation of NF-κB and is degraded by the inhibitor of NF-κB kinase subunit beta (IKBKB). The role of IKBKB is to break down IκB, which blocks the activity of NF-kB. Therefore, it prevents the activity of NK-kB. This study investigated whether neuropathic pain can be reduced in spinal nerve ligation (SNL) rats by reducing the activity of microglia by delivering IKBKB small interfering RNA (siRNA)-encapsulated poly (lactic-co-glycolic acid) (PLGA) nanoparticles. PLGA nanoparticles, as a carrier for the delivery of IKBKB genes silencer, were used because they have shown potential to enhance microglial targeting. SNL rats were injected with IKBKB siRNA-encapsulated PLGA nanoparticles intrathecally for behavioral tests on pain response. IKBKB siRNA was delivered for suppressing the expression of IKBKB. In rats injected with IKBKB siRNA-encapsulated PLGA nanoparticles, allodynia caused by mechanical stimulation was reduced, and the secretion of pro-inflammatory mediators due to NF-κB was reduced. Delivering IKBKB siRNA through PLGA nanoparticles can effectively control the inflammatory response and is worth studying as a treatment for neuropathic pain.
Collapse
|
18
|
Abstract
Tissue-resident macrophages are present in most tissues with developmental, self-renewal, or functional attributes that do not easily fit into a textbook picture of a plastic and multifunctional macrophage originating from hematopoietic stem cells; nor does it fit a pro- versus anti-inflammatory paradigm. This review presents and discusses current knowledge on the developmental biology of macrophages from an evolutionary perspective focused on the function of macrophages, which may aid in study of developmental, inflammatory, tumoral, and degenerative diseases. We also propose a framework to investigate the functions of macrophages in vivo and discuss how inherited germline and somatic mutations may contribute to the roles of macrophages in diseases.
Collapse
Affiliation(s)
- Nehemiah Cox
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Maria Pokrovskii
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Rocio Vicario
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Frederic Geissmann
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| |
Collapse
|
19
|
Li H, Wang Y, Wang B, Li M, Liu J, Yang H, Shi Y. Baicalin and Geniposide Inhibit Polarization and Inflammatory Injury of OGD/R-Treated Microglia by Suppressing the 5-LOX/LTB4 Pathway. Neurochem Res 2021; 46:1844-1858. [PMID: 33891262 PMCID: PMC8187209 DOI: 10.1007/s11064-021-03305-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 02/25/2021] [Accepted: 03/17/2021] [Indexed: 11/27/2022]
Abstract
Cerebral ischemia causes severe neurological disorders and neuronal dysfunction. Baicalin (BC), geniposide (GP), and their combination (BC/GP) have been shown to inhibit post-ischemic inflammatory injury by inhibiting the 5-LOX/CysLTs pathway. The aims of this study were to observe the inhibitory effects of BC/GP on the activation of microglial cells induced by oxygen glucose deprivation and reoxygenation (OGD/R) and to investigate whether the 5-LOX/LTB4 pathway was involved in these effects. Molecular docking showed that BC and GP exhibited considerable binding activity with LTB4 synthase LTA4H. BV-2 microglia were transfected with a 5-LOX overexpression lentiviral vector, and then OGD/R was performed. The effects of different concentrations of BC, GP, and BC/GP (6.25 μM, 12.5 μM, and 25 μM) on cell viability and apoptosis of microglia were evaluated by MTT and flow cytometry. The expression of TNF-α, IL-1β, NF-κB, and pNF-κB also was measured by ELISA, Western blots and immunofluorescence. Western blots and qRT-PCR analysis were used to determine the levels of CD11b, CD206, and 5-LOX pathway proteins. Results showed that BC, GP, and BC/GP reduced the apoptosis caused by OGD/R in a dose-dependent manner, and cell viability was significantly increased at a concentration of 12.5 μM. OGD/R significantly increased the release of TNF-α, IL-1β, NF-κB, pNF-κB, and CD11b. These effects were suppressed by BC, GP, and BC/GP, and the OGD/R-induced transfer of NF-κB p65 from the ctytoplasm to the nucleus was inhibited in microglia. Interestingly, the LTB4 inhibitor, U75302, exhibited the same effect. Also, BC, GP, and BC/GP significantly reduced the expression of 5-LOX pathway proteins. These results demonstrated that BC/GP inhibited OGD/R-induced polarization in BV2 microglia by regulating the 5-LOX/LTB4 signaling pathways and attenuating the inflammatory response. Our results supported the theoretical basis for additional in-depth study of the function of BC/GP and the value of determining its unique target, which might provide a new therapeutic strategy for ischemic cerebrovascular disease.
Collapse
Affiliation(s)
- HuiMin Li
- Key Laboratory of Pharmacodynamic Mechanism and Material Basis of Traditional Chinese Medicine, Shaanxi Provincial Administration of Traditional Chinese Medicine, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Yan Wang
- Key Laboratory of Pharmacodynamic Mechanism and Material Basis of Traditional Chinese Medicine, Shaanxi Provincial Administration of Traditional Chinese Medicine, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Bin Wang
- Key Laboratory of Pharmacodynamic Mechanism and Material Basis of Traditional Chinese Medicine, Shaanxi Provincial Administration of Traditional Chinese Medicine, Shaanxi University of Chinese Medicine, Xianyang, 712046, China.
| | - Min Li
- Key Laboratory of Pharmacodynamic Mechanism and Material Basis of Traditional Chinese Medicine, Shaanxi Provincial Administration of Traditional Chinese Medicine, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - JiPing Liu
- Key Laboratory of Pharmacodynamic Mechanism and Material Basis of Traditional Chinese Medicine, Shaanxi Provincial Administration of Traditional Chinese Medicine, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - HongLian Yang
- Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - YongHeng Shi
- Key Laboratory of Pharmacodynamic Mechanism and Material Basis of Traditional Chinese Medicine, Shaanxi Provincial Administration of Traditional Chinese Medicine, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| |
Collapse
|
20
|
Erisgin Z. Melamine exposure from the weaning period causes apoptosis, inflammation, and damage to the blood-brain barrier. J Chem Neuroanat 2021; 113:101939. [PMID: 33639231 DOI: 10.1016/j.jchemneu.2021.101939] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/20/2021] [Accepted: 02/20/2021] [Indexed: 11/28/2022]
Abstract
The aim of this study to investigate the effects of melamine exposure from the weaning period on the developing brain in rats. Female Wistar albino rats (21 days old, n = 18) were divided into 3 groups, all animals were weighed daily and dose-adjusted. For 3 weeks, 0.1 mL of saline was administered by oral gavage to the control group, 50 mg/kg of melamine to the second group, and 75 mg/kg of melamine to the third group were administered by oral gavage by dissolving in 0.1 mL of saline. On the postnatal 45th day, the rats were sacrificed under anesthesia and brain tissues placed in neutral formalin. After routine tissue processing, brain sections were stained with hematoxylin&eosin(H&E) and Terminal deoxynucleotidyl transferase(TdT) dUTP Nick-End Labeling(TUNEL), IBA-1, Glial Fibrillar Acidic Protein(GFAP), Tumor necrosis factor-α (TNF-α), and SMI-70 antibodies as immunohistochemically. In the results, according to apoptotic index(AI) results, there was a significant increase in the 75 mg/kg and 50 mg/kg melamine groups compared to the control groups (p < 005). There was a significant increase in the number of anti- TNF-α positive neurons and the number of anti-GFAP positive astrocytes in both melamine groups compared to the control group (p < 001). In terms of SMI-71, an increase was found in the 75 mg/kg group compared to other groups (p < 001), while no significant difference was found between the groups in terms of IBA-1 (p > 0.05). It has been observed an increase in dilatation of blood vessels, inflammatory cell infiltration, and endothelial cell degeneration in the 50 mg/kg and 75 mg/kg melamine groups compared to the control group (p < 0.01). there was no statistically significant difference in the body and brain weight between both melamine treatment groups (75 mg/kg and 50 mg/kg) and the control group (p > 005). Melamine exposure (50 mg/kg and 75 mg/kg) from the weaning period causes apoptosis and inflammation in the developing brain, and the disruptions in the blood-brain barrier (BBB) significantly increase exposure to 75 mg/kg.
Collapse
Affiliation(s)
- Zuleyha Erisgin
- Giresun University Faculty of Medicine Department of Histology and Embryology, Giresun, Turkey.
| |
Collapse
|
21
|
Retinal Inflammation, Cell Death and Inherited Retinal Dystrophies. Int J Mol Sci 2021; 22:ijms22042096. [PMID: 33672611 PMCID: PMC7924201 DOI: 10.3390/ijms22042096] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/12/2021] [Accepted: 02/18/2021] [Indexed: 12/15/2022] Open
Abstract
Inherited retinal dystrophies (IRDs) are a group of retinal disorders that cause progressive and severe loss of vision because of retinal cell death, mainly photoreceptor cells. IRDs include retinitis pigmentosa (RP), the most common IRD. IRDs present a genetic and clinical heterogeneity that makes it difficult to achieve proper treatment. The progression of IRDs is influenced, among other factors, by the activation of the immune cells (microglia, macrophages, etc.) and the release of inflammatory molecules such as chemokines and cytokines. Upregulation of tumor necrosis factor alpha (TNFα), a pro-inflammatory cytokine, is found in IRDs. This cytokine may influence photoreceptor cell death. Different cell death mechanisms are proposed, including apoptosis, necroptosis, pyroptosis, autophagy, excessive activation of calpains, or parthanatos for photoreceptor cell death. Some of these cell death mechanisms are linked to TNFα upregulation and inflammation. Therapeutic approaches that reduce retinal inflammation have emerged as useful therapies for slowing down the progression of IRDs. We focused this review on the relationship between retinal inflammation and the different cell death mechanisms involved in RP. We also reviewed the main anti-inflammatory therapies for the treatment of IRDs.
Collapse
|
22
|
Muniz Santana Bastos E, Bispo da Silva A, Cerqueira Coelho PL, Pereira Borges JM, Amaral da Silva VD, Moreau da Cunha VH, Costa SL. Anti-inflammatory activity of Jatropha curcas L. in brain glial cells primary cultures. JOURNAL OF ETHNOPHARMACOLOGY 2021; 264:113201. [PMID: 32814081 DOI: 10.1016/j.jep.2020.113201] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 07/09/2020] [Accepted: 07/17/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Jatropha curcas L. (Euphorbiaceae), a medicinal plant known in Brazil as "Pinhão Manso", is highly adaptable, being cultivated in different tropical and subtropical regions of the world. Antimicrobial, antioxidant and antiinflammatory activities have been attributed to different parts of the plant. In the central nervous sytem (CNS), neuroinflammation is mediated by glial cells, mainly by astrocytes and microglia, a process that plays an important role in neurodegenerative diseases and other CNS disorders. In this study, we investigated the anti-inflammatory activity of the methanolic extract obtained from the leaves of J. curcas L. (MEJc) in primary cultures of glial cells submited to inflammatory stimulus. MATERIALS AND METHODS Primary cultures of glial cells obtained from the cerebral cortex of neonate Wistar rats were treated with MEJc (0.1-50,000 μg mL-1) and its fractions (FnJc) (0.1 μg mL-1) with or without lipopolysaccharide of Escherichia coli (LPS) (1 μg mL-1). Cell viability was determined with MTT test. Modifications in glial cell morphology were investigated by means of phase contrast microscopy and May-Grünwald staining. The reactivity of astrocytes and microglia were investigated with immunocytochemistry for GFAP, Iba1 and transcription factor NF-kB, as well as with Greiss reaction to determine the nitric oxide (NO) production. RESULTS MEJc at 0.1-1000 μg mL-1 was non-toxic to glial cells and the DE50 was 10.794 μg mL-1. The treatment with LPS induced the activation of astrocytes and microglia marked by morphological modifications and changes in the expression of GFAP and Iba1, as well as the increase in NF-kB expression and NO production. Treatment with MEJc inhibited the morphological modifications, changes in GFAP and Iba1 expression, and the increase in NF-kB and NO production induced by LPS. CONCLUSION This study demonstrates that the MEJc and its fractions modulate inflammatory response of astrocytes and microglia to LPS and may be considered as a potential therapeutic strategy for neuroinflammation-related diseases.
Collapse
Affiliation(s)
- Eduardo Muniz Santana Bastos
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Federal University of Bahia - Institute of Health Sciences, 40100-902, Salvador, BA, Brazil; Department of Biotechnology, Institute of Health Sciences, Federal University of Bahia, 40100-902, Salvador, BA, Brazil
| | - Alessandra Bispo da Silva
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Federal University of Bahia - Institute of Health Sciences, 40100-902, Salvador, BA, Brazil
| | - Paulo Lucas Cerqueira Coelho
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Federal University of Bahia - Institute of Health Sciences, 40100-902, Salvador, BA, Brazil
| | - Julita Maria Pereira Borges
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Federal University of Bahia - Institute of Health Sciences, 40100-902, Salvador, BA, Brazil
| | - Victor Diogenes Amaral da Silva
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Federal University of Bahia - Institute of Health Sciences, 40100-902, Salvador, BA, Brazil
| | - Vitor Hugo Moreau da Cunha
- Department of Biotechnology, Institute of Health Sciences, Federal University of Bahia, 40100-902, Salvador, BA, Brazil.
| | - Silvia Lima Costa
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Federal University of Bahia - Institute of Health Sciences, 40100-902, Salvador, BA, Brazil.
| |
Collapse
|
23
|
Rawlings S, Takechi R, Lavender AP. Effects of sub-concussion on neuropsychological performance and its potential mechanisms: A narrative review. Brain Res Bull 2020; 165:56-62. [PMID: 33011196 DOI: 10.1016/j.brainresbull.2020.09.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/12/2020] [Accepted: 09/25/2020] [Indexed: 12/12/2022]
Abstract
Concussion and mild traumatic brain injury (mTBI) are recognised as serious medical events that are relatively common in contact sports. Recently, the seemingly non-injurious phenomenon of sub-concussion has gained interest among neuroscience researchers and early studies are showing that there may be some acute and chronic effects on brain health and function with repeated sub-concussive events of the type seen in soccer, where players strike the ball with the head, and collision sports like the rugby codes. The aim of this narrative review is to describe sub-concussion and the current understanding of short and long term effects of repeated minor impacts that have been found to occur in human and animal models. Here, potential mechanisms for cognitive dysfunction following sub-concussion and recommend directions for future research are discussed. The Potential mechanisms of injuries resulting from sub-concussion such as changes in blood brain barrier integrity, neuroinflammation, cognitive impairment, and oxidative stress damage, among other changes in central nervous system function vary considerably making understanding of the underlying causative mechanism challenging for researchers. Some evidence suggests a link between impaired cerebrovascular function and cognitive impairment which poses a potential mechanism linking the two. It is hoped that this review helps guide researchers toward a potential direction of investigations.
Collapse
Affiliation(s)
- Samuel Rawlings
- School of Physiotherapy and Exercise Science, Faculty of Health Sciences, Curtin University, Perth, Australia
| | - Ryusuke Takechi
- Curtin Health Innovation Research Institute, Curtin University, Perth, Australia; School of Public Health, Faculty of Health Sciences, Curtin University, Perth, Australia
| | - Andrew P Lavender
- School of Physiotherapy and Exercise Science, Faculty of Health Sciences, Curtin University, Perth, Australia; School of Science, Psychology and Sport, Federation University Australia, Ballarat, Australia.
| |
Collapse
|
24
|
Gavegnano C, Haile W, Koneru R, Hurwitz SJ, Kohler JJ, Tyor WR, Schinazi RF. Novel method to quantify phenotypic markers of HIV-associated neurocognitive disorder in a murine SCID model. J Neurovirol 2020; 26:838-845. [PMID: 32901392 DOI: 10.1007/s13365-020-00842-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 03/18/2020] [Accepted: 04/13/2020] [Indexed: 10/23/2022]
Abstract
Despite combined antiretroviral therapy (cART), HIV infection in the CNS persists with reported increases in activation of macrophages (MΦ), microglia, and surrounding astrocytes/neurons, conferring HIV-induced inflammation. Chronic inflammation results in HIV-associated neurocognitive disorders (HAND) with reported occurrence of up to half of individuals with HIV infection. The existing HAND mouse model used by laboratories including ours, and the effect of novel agents on its pathology present with labor-intensive and time-consuming limitations since brain sections and immunohistochemistry assays have to be performed and analyzed. A novel flow cytometry-based system to objectively quantify phenotypic effects of HIV using a SCID mouse HAND model was developed which demonstrated that the HIV-infected mice had significant increases in astrogliosis, loss of neuronal dendritic marker, activation of murine microglia, and human macrophage explants compared to uninfected control mice. HIV p24 could also be quantified in the brains of the infected mice. Correlation of these impairments with HIV-induced brain inflammation and previous behavioral abnormalities studies in mice suggests that this model can be used as a fast and relevant throughput methodology to quantify preclinical testing of novel treatments for HAND.
Collapse
Affiliation(s)
- Christina Gavegnano
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University, Atlanta, GA, 30322, USA.,Emory Center for AIDS Research (CFAR), Emory University, Atlanta, GA, 30322, USA
| | - Woldeab Haile
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30209, USA.,Atlanta Veterans Affairs Medical Center, Decatur, GA, 30033, USA
| | - Raj Koneru
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30209, USA.,Atlanta Veterans Affairs Medical Center, Decatur, GA, 30033, USA
| | - Selwyn J Hurwitz
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University, Atlanta, GA, 30322, USA.,Emory Center for AIDS Research (CFAR), Emory University, Atlanta, GA, 30322, USA
| | - James J Kohler
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University, Atlanta, GA, 30322, USA.,Emory Center for AIDS Research (CFAR), Emory University, Atlanta, GA, 30322, USA
| | - William R Tyor
- Emory Center for AIDS Research (CFAR), Emory University, Atlanta, GA, 30322, USA. .,Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30209, USA. .,Atlanta Veterans Affairs Medical Center, Decatur, GA, 30033, USA.
| | - Raymond F Schinazi
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University, Atlanta, GA, 30322, USA. .,Emory Center for AIDS Research (CFAR), Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
25
|
Worthen RJ, Garzon Zighelboim SS, Torres Jaramillo CS, Beurel E. Anti-inflammatory IL-10 administration rescues depression-associated learning and memory deficits in mice. J Neuroinflammation 2020; 17:246. [PMID: 32828124 PMCID: PMC7443292 DOI: 10.1186/s12974-020-01922-1] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 08/10/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Major depressive disorder is a widespread mood disorder. One of the most debilitating symptoms patients often experience is cognitive impairment. Recent findings suggest that inflammation is associated with depression and impaired cognition. Pro-inflammatory cytokines are elevated in the blood of depressed patients and impair learning and memory processes, suggesting that an anti-inflammatory approach might be beneficial for both depression and cognition. METHODS We subjected mice to the learned helplessness paradigm and evaluated novel object recognition and spatial memory. Mice were treated with IL-10 intranasally or/and microglia cells were depleted using PLX5622. Statistical differences were tested using ANOVA or t tests. RESULTS We first established a mouse model of depression in which learning and memory are impaired. We found that learned helplessness (LH) impairs novel object recognition (NOR) and spatial working memory. LH mice also exhibit reduced hippocampal dendritic spine density and increased microglial activation compared to non-shocked (NS) mice or mice that were subjected to the learned helpless paradigm but did not exhibit learned helplessness (non-learned helpless or NLH). These effects are mediated by microglia, as treatment with PLX5622, which depletes microglia, restores learning and memory and hippocampal dendritic spine density in LH mice. However, PLX5622 also impairs learning and memory and reduces hippocampal dendritic spine density in NLH mice, suggesting that microglia in NLH mice produce molecules that promote learning and memory. We found that microglial interleukin (IL)-10 levels are reduced in LH mice, and IL-10 administration is sufficient to restore NOR, spatial working memory, and hippocampal dendritic spine density in LH mice, and in NLH mice treated with PLX5622 consistent with a pro-cognitive role for IL-10. CONCLUSIONS Altogether these data demonstrate the critical role of IL-10 in promoting learning and memory after learned helplessness.
Collapse
Affiliation(s)
- Ryan J Worthen
- Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Gautier Building room 415, 1011 NW 15th Street, Miami, FL, 33136, USA
| | - Susan S Garzon Zighelboim
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Gautier Building room, 4151011 NW 15th Street, Miami, FL, 33136, USA
| | - Camila S Torres Jaramillo
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Gautier Building room, 4151011 NW 15th Street, Miami, FL, 33136, USA
| | - Eleonore Beurel
- Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Gautier Building room 415, 1011 NW 15th Street, Miami, FL, 33136, USA.
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Gautier Building room, 4151011 NW 15th Street, Miami, FL, 33136, USA.
| |
Collapse
|
26
|
Ahmed ME, Selvakumar GP, Kempuraj D, Raikwar SP, Thangavel R, Bazley K, Wu K, Khan O, Khan A, Zaheer S, Iyer S, Burton C, James D, Zaheer A. Glia Maturation Factor (GMF) Regulates Microglial Expression Phenotypes and the Associated Neurological Deficits in a Mouse Model of Traumatic Brain Injury. Mol Neurobiol 2020; 57:4438-4450. [PMID: 32737763 DOI: 10.1007/s12035-020-02040-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 07/24/2020] [Indexed: 12/20/2022]
Abstract
Traumatic brain injury (TBI) induces inflammatory responses through microglial activation and polarization towards a more inflammatory state that contributes to the deleterious secondary brain injury. Glia maturation factor (GMF) is a pro-inflammatory protein that is responsible for neuroinflammation following insult to the brain, such as in TBI. We hypothesized that the absence of GMF in GMF-knockout (GMF-KO) mice would regulate microglial activation state and the M1/M2 phenotypes following TBI. We used the weight drop model of TBI in C57BL/6 mice wild-type (WT) and GMF-KO mice. Immunofluorescence staining, Western blot, and ELISA assays were performed to confirm TBI-induced histopathological and neuroinflammatory changes. Behavioral analysis was done to check motor coordination ability and cognitive function. We demonstrated that the deletion of GMF in GMF-KO mice significantly limited lesion volume, attenuated neuronal loss, inhibited gliosis, and activated microglia adopted predominantly anti-inflammatory (M2) phenotypes. Using an ELISA method, we found a gradual decrease in pro-inflammatory cytokines (TNF-α and IL-6) and upregulation of anti-inflammatory cytokines (IL-4 and IL-10) in GMF-KO mice compared with WT mice, thus, promoting the transition of microglia towards a more predominantly anti-inflammatory (M2) phenotype. GMF-KO mice showed significant improvement in motor ability, memory, and cognition. Overall, our results demonstrate that GMF deficiency regulates microglial polarization, which ameliorates neuronal injury and behavioral impairments following TBI in mice and concludes that GMF is a regulator of neuroinflammation and an ideal therapeutic target for the treatment of TBI.
Collapse
Affiliation(s)
- Mohammad Ejaz Ahmed
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA
| | - Govindhasamy Pushpavathi Selvakumar
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA
| | - Duraisamy Kempuraj
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA
| | - Sudhanshu P Raikwar
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA
| | - Ramasamy Thangavel
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA
| | - Kieran Bazley
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Kristopher Wu
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Osaid Khan
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Asher Khan
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Smita Zaheer
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Shankar Iyer
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA
| | | | | | - Asgar Zaheer
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.
| |
Collapse
|
27
|
Chambel SS, Tavares I, Cruz CD. Chronic Pain After Spinal Cord Injury: Is There a Role for Neuron-Immune Dysregulation? Front Physiol 2020; 11:748. [PMID: 32733271 PMCID: PMC7359877 DOI: 10.3389/fphys.2020.00748] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 06/08/2020] [Indexed: 12/14/2022] Open
Abstract
Spinal cord injury (SCI) is a devastating event with a tremendous impact in the life of the affected individual and family. Traumatic injuries related to motor vehicle accidents, falls, sports, and violence are the most common causes. The majority of spinal lesions is incomplete and occurs at cervical levels of the cord, causing a disruption of several ascending and descending neuronal pathways. Additionally, many patients develop chronic pain and describe it as burning, stabbing, shooting, or shocking and often arising with no stimulus. Less frequently, people with SCI also experience pain out of context with the stimulus (e.g., light touch). While abolishment of the endogenous descending inhibitory circuits is a recognized cause for chronic pain, an increasing number of studies suggest that uncontrolled release of pro- and anti-inflammatory mediators by neurons, glial, and immune cells is also important in the emergence and maintenance of SCI-induced chronic pain. This constitutes the topic of the present mini-review, which will focus on the importance of neuro-immune dysregulation for pain after SCI.
Collapse
Affiliation(s)
- Sílvia S Chambel
- Department of Biomedicine, Experimental Biology Unit, Faculty of Medicine, University of Porto, Porto, Portugal.,Translational NeuroUrology Group, Instituto de Investigação e Inovação em Saúde - i3S, Universidade do Porto, Porto, Portugal
| | - Isaura Tavares
- Department of Biomedicine, Experimental Biology Unit, Faculty of Medicine, University of Porto, Porto, Portugal.,Pain Research Group, Instituto de Investigação e Inovação em Saúde - i3S, Universidade do Porto, Porto, Portugal
| | - Célia D Cruz
- Department of Biomedicine, Experimental Biology Unit, Faculty of Medicine, University of Porto, Porto, Portugal.,Translational NeuroUrology Group, Instituto de Investigação e Inovação em Saúde - i3S, Universidade do Porto, Porto, Portugal
| |
Collapse
|
28
|
Jørgensen LØ, Hyrlov KH, Elkjaer ML, Weber AB, Pedersen AE, Svenningsen ÅF, Illes Z. Cladribine modifies functional properties of microglia. Clin Exp Immunol 2020; 201:328-340. [PMID: 32492189 PMCID: PMC7419928 DOI: 10.1111/cei.13473] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 05/04/2020] [Accepted: 05/18/2020] [Indexed: 01/13/2023] Open
Abstract
Cladribine (CdA), an oral prodrug approved for the treatment of relapsing multiple sclerosis, selectively depletes lymphocytes. CdA passes the blood–brain barrier, suggesting a potential effect on central nervous system (CNS) resident cells. We examined if CdA modifies the phenotype and function of naive and activated primary mouse microglia, when applied in the concentrations 0·1–1 μM that putatively overlap human cerebrospinal fluid (CSF) concentrations. Primary microglia cultures without stimulation or in the presence of proinflammatory lipopolysaccharide (LPS) or anti‐inflammatory interleukin (IL)‐4 were treated with different concentrations of CdA for 24 h. Viability was assessed by MTT [3‐(4,5‐dimethylthiazol‐2‐yl)‐2,5‐diphenyltetrazolium bromide] assay. Phagocytotic ability and morphology were examined by flow cytometry and random migration using IncuCyte Zoom and TrackMate. Change in gene expression was examined by quantitative polymerase chain reaction (qPCR) and protein secretion by Meso Scale Discovery. We found that LPS and IL‐4 up‐regulated deoxycytidine kinase (DCK) expression. Only activated microglia were affected by CdA, and this was unrelated to viability. CdA 0·1–1 μM significantly reduced granularity, phagocytotic ability and random migration of activated microglia. CdA 10 μM increased the IL‐4‐induced gene expression of arginase 1 (Arg1) and LPS‐induced expression of IL‐1β, tumor necrosis factor (TNF), inducible nitric oxide synthase (iNOS) and Arg1, but protein secretion remained unaffected. CdA 10 μM potentiated the increased expression of anti‐inflammatory TNF receptor 2 (TNF‐R2) but not TNF‐R1 induced by LPS. This suggests that microglia acquire a less activated phenotype when treated with 0·1–1 μM CdA that putatively overlaps human CSF concentrations. This may be related to the up‐regulated gene expression of DCK upon activation, and suggests a potential alternative mechanism of CdA with direct effect on CNS resident cells.
Collapse
Affiliation(s)
- L Ø Jørgensen
- Department of Neurology, Odense University Hospital, Odense, Denmark.,Neurology Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Neurobiology Research Unit, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - K H Hyrlov
- Department of Neurology, Odense University Hospital, Odense, Denmark.,Neurology Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Neurobiology Research Unit, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - M L Elkjaer
- Department of Neurology, Odense University Hospital, Odense, Denmark.,Neurology Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Neurobiology Research Unit, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - A B Weber
- Department of Neurology, Odense University Hospital, Odense, Denmark.,Neurology Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Neurobiology Research Unit, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - A E Pedersen
- Merck A/S, Søborg, Denmark.,Department of Odontology, University of Copenhagen, Copenhagen, Denmark
| | - Å Fex Svenningsen
- Neurology Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Neurobiology Research Unit, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Z Illes
- Department of Neurology, Odense University Hospital, Odense, Denmark.,Neurology Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Neurobiology Research Unit, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
29
|
Jiang M, Li Z, Zhu G. Immunological regulatory effect of flavonoid baicalin on innate immune toll-like receptors. Pharmacol Res 2020; 158:104890. [PMID: 32389860 DOI: 10.1016/j.phrs.2020.104890] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 04/01/2020] [Accepted: 04/30/2020] [Indexed: 02/07/2023]
Abstract
As an essential component of the innate immune system, Toll-like receptors (TLRs) are a family of well-recognized ligand-binding receptors found in various organisms and initiate host immune responses. Activation of TLRs signaling pathways lead to the induction of numerous genes that function in host defense. Baicalin is a natural compound from the dry raw root of Scutellaria baicalensis (S. baicalensis) and it has been found to exhibit several pharmaceutical actions, such as anti-inflammation, anti-tumor and antivirus. These biological activities are mainly related to the regulatory effect of baicalin on the host immune response. In this review, we provide an overview of the regulation of baicalin on TLRs signaling pathways in various pathological conditions, and highlight potential targets for the development of the regulatory effect of natural compound from traditional Chinese medicine on innate immune system.
Collapse
Affiliation(s)
- Ming Jiang
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan, Hubei, 430030, China
| | - Zhuoneng Li
- Centers for Disease Control and Prevention of Wuhan, China
| | - Guangxun Zhu
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan, Hubei, 430030, China.
| |
Collapse
|
30
|
Chagas LDS, Sandre PC, Ribeiro e Ribeiro NCA, Marcondes H, Oliveira Silva P, Savino W, Serfaty CA. Environmental Signals on Microglial Function during Brain Development, Neuroplasticity, and Disease. Int J Mol Sci 2020; 21:ijms21062111. [PMID: 32204421 PMCID: PMC7139373 DOI: 10.3390/ijms21062111] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 12/12/2019] [Accepted: 12/13/2019] [Indexed: 12/15/2022] Open
Abstract
Recent discoveries on the neurobiology of the immunocompetent cells of the central nervous system (CNS), microglia, have been recognized as a growing field of investigation on the interactions between the brain and the immune system. Several environmental contexts such as stress, lesions, infectious diseases, and nutritional and hormonal disorders can interfere with CNS homeostasis, directly impacting microglial physiology. Despite many encouraging discoveries in this field, there are still some controversies that raise issues to be discussed, especially regarding the relationship between the microglial phenotype assumed in distinct contexts and respective consequences in different neurobiological processes, such as disorders of brain development and neuroplasticity. Also, there is an increasing interest in discussing microglial–immune system cross-talk in health and in pathological conditions. In this review, we discuss recent literature concerning microglial function during development and homeostasis. In addition, we explore the contribution of microglia to synaptic disorders mediated by different neuroinflammatory outcomes during pre- and postnatal development, with long-term consequences impacting on the risk and vulnerability to the emergence of neurodevelopmental, neurodegenerative, and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Luana da Silva Chagas
- Laboratory of Neural Plasticity Neurobiology Department, Biology Institute, Federal Fluminense University, Niteroi 24020-141, Brazil; (L.d.S.C.); (P.C.S.); (N.C.A.R.eR.); (H.M.); (P.O.S.)
| | - Poliana Capucho Sandre
- Laboratory of Neural Plasticity Neurobiology Department, Biology Institute, Federal Fluminense University, Niteroi 24020-141, Brazil; (L.d.S.C.); (P.C.S.); (N.C.A.R.eR.); (H.M.); (P.O.S.)
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil
| | - Natalia Cristina Aparecida Ribeiro e Ribeiro
- Laboratory of Neural Plasticity Neurobiology Department, Biology Institute, Federal Fluminense University, Niteroi 24020-141, Brazil; (L.d.S.C.); (P.C.S.); (N.C.A.R.eR.); (H.M.); (P.O.S.)
| | - Henrique Marcondes
- Laboratory of Neural Plasticity Neurobiology Department, Biology Institute, Federal Fluminense University, Niteroi 24020-141, Brazil; (L.d.S.C.); (P.C.S.); (N.C.A.R.eR.); (H.M.); (P.O.S.)
| | - Priscilla Oliveira Silva
- Laboratory of Neural Plasticity Neurobiology Department, Biology Institute, Federal Fluminense University, Niteroi 24020-141, Brazil; (L.d.S.C.); (P.C.S.); (N.C.A.R.eR.); (H.M.); (P.O.S.)
- National Institute of Science and Technology on Neuroimmunomodulation –INCT-NIM, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil
| | - Wilson Savino
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation –INCT-NIM, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil
- Correspondence: (W.S.); (C.A.S.)
| | - Claudio A. Serfaty
- Laboratory of Neural Plasticity Neurobiology Department, Biology Institute, Federal Fluminense University, Niteroi 24020-141, Brazil; (L.d.S.C.); (P.C.S.); (N.C.A.R.eR.); (H.M.); (P.O.S.)
- National Institute of Science and Technology on Neuroimmunomodulation –INCT-NIM, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil
- Correspondence: (W.S.); (C.A.S.)
| |
Collapse
|
31
|
Benzimidazole Containing Acetamide Derivatives Attenuate Neuroinflammation and Oxidative Stress in Ethanol-Induced Neurodegeneration. Biomolecules 2020; 10:biom10010108. [PMID: 31936383 PMCID: PMC7023260 DOI: 10.3390/biom10010108] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/25/2019] [Accepted: 01/04/2020] [Indexed: 12/13/2022] Open
Abstract
Oxidative stress-induced neuroinflammation is the prominent feature of neurodegenerative disorders, and is characterized by a gradual decline of structure and function of neurons. Many biochemical events emerge thanks to the result of this neurodegeneration, and ultimately provoke neuroinflammation, activation of microglia, and oxidative stress, leading to neuronal death. This cascade not only explains the complexity of events taking place across different stages, but also depicts the need for more effective therapeutic agents. The present study was designed to investigate the neuroprotective effects of newly synthesized benzimidazole containing acetamide derivatives, 3a (2-(4-methoxyanilino)-N-[1-(4-methylbenzene-1-sulfonyl)-1H-benzimidazol-2-yl] acetamide) and 3b (2-(Dodecylamino)-N-[1-(4-methylbenzene-1-sulfonyl)-1H-benzimidazol-2-yl] acetamide) against ethanol-induced neurodegeneration in the rat model. Both derivatives were characterized spectroscopically by proton NMR (1H-NMR) and carbon-13 NMR (13C-NMR) and evaluated for neuroprotective potential using different pharmacological approaches. In vivo experiments demonstrated that ethanol triggered neurodegeneration characterized by impaired antioxidant enzymes and elevated oxidative stress. Furthermore, ethanol administration induced neuroinflammation, as demonstrated by elevated expression of tumor necrotic factor (TNF-α), nuclear factor κB (NF-κB), cyclooxygenase-2 (COX2), and ionized calcium-binding adapter molecule-1 (Iba-1), which was further validated by enzyme-linked immunosorbent assay (ELISA). Treatment with 3a and 3b ameliorated the ethanol-induced oxidative stress, neuroinflammation, and memory impairment. The affinity of synthesized derivatives towards various receptors involved in neurodegeneration was assessed through docking analysis. The versatile nature of benzimidazole nucleus and its affinity toward several receptors suggested that it could be a multistep targeting neuroprotectant. As repetitive clinical trials of neuroprotectants targeting a single step of the pathological process have failed previously, our results suggested that a neuroprotective strategy of acting at different stages may be more advantageous to intervene in the vicious cycles of neuroinflammation.
Collapse
|
32
|
Dubey T, Chinnathambi S. Brahmi (Bacopa monnieri): An ayurvedic herb against the Alzheimer's disease. Arch Biochem Biophys 2019; 676:108153. [PMID: 31622587 DOI: 10.1016/j.abb.2019.108153] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 10/08/2019] [Accepted: 10/11/2019] [Indexed: 01/16/2023]
Abstract
Ayurveda is the medicinal science, dealing with utilization of naturally available plant products for treatment. A wide variety of neuroprotective herbs have been reported in Ayurveda. Brahmi, Bacopa monnieri is a nootropic ayurvedic herb known to be effective in neurological disorders from ancient times. Numerous approaches including natural and synthetic compounds have been applied against Alzheimer's disease. Amyloid-β and Tau are the hallmarks proteins of several neuronal dysfunctions resulting in Alzheimer's disease. Tau is a microtubule-associated protein known to be involved in progression of Alzheimer's disease. The generation of reaction oxygen species, increased neuroinflammation and neurotoxicity are the major physiological dysfunctions associated with Tau aggregates, which leads to dementia and behavioural deficits. Bacoside A, Bacoside B, Bacosaponins, Betulinic acid, etc; are the bioactive component of Brahmi belonging to various chemical families. Each chemical component known have its significant role in neuroprotection. The neuroprotective properties of Brahmi and its bioactive components including reduction of ROS, neuroinflammation, aggregation inhibition of Amyloid-β and improvement of cognitive and learning behaviour. Here on basis of earlier studies we hypothesize the inhibitory role of Brahmi against Tau-mediated toxicity. The overall studies have concluded that Brahmi can be used as a lead formulation for treatment of Alzheimer's disease and other neurological disorders.
Collapse
Affiliation(s)
- Tushar Dubey
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, 411008, Pune, India; Academy of Scientific and Innovative Research (AcSIR), 411008, Pune, India
| | - Subashchandrabose Chinnathambi
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, 411008, Pune, India; Academy of Scientific and Innovative Research (AcSIR), 411008, Pune, India.
| |
Collapse
|
33
|
Vanden-Hehir S, Cairns SA, Lee M, Zoupi L, Shaver MP, Brunton VG, Williams A, Hulme AN. Alkyne-Tagged PLGA Allows Direct Visualization of Nanoparticles In Vitro and Ex Vivo by Stimulated Raman Scattering Microscopy. Biomacromolecules 2019; 20:4008-4014. [PMID: 31408325 PMCID: PMC6794644 DOI: 10.1021/acs.biomac.9b01092] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Indexed: 12/19/2022]
Abstract
Polymeric nanoparticles (NPs) are attractive candidates for the controlled and targeted delivery of therapeutics in vitro and in vivo. However, detailed understanding of the uptake, location, and ultimate cellular fate of the NPs is necessary to satisfy safety concerns, which is difficult because of the nanoscale size of these carriers. In this work, we show how small chemical labels can be appended to poly(lactic acid-co-glycolic acid) (PLGA) to synthesize NPs that can then be imaged by stimulated Raman scattering microscopy, a vibrational imaging technique that can elucidate bond-specific information in biological environments, such as the identification of alkyne signatures in modified PLGA terpolymers. We show that both deuterium and alkyne labeled NPs can be imaged within primary rat microglia, and the alkyne NPs can also be imaged in ex vivo cortical mouse brain tissue. Immunohistochemical analysis confirms that the NPs localize in microglia in the mouse brain tissue, demonstrating that these NPs have the potential to deliver therapeutics selectively to microglia.
Collapse
Affiliation(s)
- Sally Vanden-Hehir
- EaStCHEM
School of Chemistry, University of Edinburgh, David Brewster Road, Edinburgh, EH9 3FJ, United Kingdom
| | - Stefan A. Cairns
- EaStCHEM
School of Chemistry, University of Edinburgh, David Brewster Road, Edinburgh, EH9 3FJ, United Kingdom
| | - Martin Lee
- Edinburgh
Cancer Research UK Centre, University of
Edinburgh, Western General Hospital, Crewe Road South, Edinburgh, EH4 2XR, United Kingdom
| | - Lida Zoupi
- MRC
Centre for Regenerative Medicine, The University
of Edinburgh, Edinburgh
BioQuarter, 5, Little France Drive, Edinburgh, EH16 4UU, United Kingdom
| | - Michael P. Shaver
- EaStCHEM
School of Chemistry, University of Edinburgh, David Brewster Road, Edinburgh, EH9 3FJ, United Kingdom
| | - Valerie G. Brunton
- Edinburgh
Cancer Research UK Centre, University of
Edinburgh, Western General Hospital, Crewe Road South, Edinburgh, EH4 2XR, United Kingdom
| | - Anna Williams
- MRC
Centre for Regenerative Medicine, The University
of Edinburgh, Edinburgh
BioQuarter, 5, Little France Drive, Edinburgh, EH16 4UU, United Kingdom
| | - Alison N. Hulme
- EaStCHEM
School of Chemistry, University of Edinburgh, David Brewster Road, Edinburgh, EH9 3FJ, United Kingdom
| |
Collapse
|
34
|
Koga M, Nakagawa S, Kato A, Kusumi I. Caffeic acid reduces oxidative stress and microglial activation in the mouse hippocampus. Tissue Cell 2019; 60:14-20. [DOI: 10.1016/j.tice.2019.07.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 07/19/2019] [Accepted: 07/21/2019] [Indexed: 12/19/2022]
|
35
|
Phipps JA, Dixon MA, Jobling AI, Wang AY, Greferath U, Vessey KA, Fletcher EL. The renin-angiotensin system and the retinal neurovascular unit: A role in vascular regulation and disease. Exp Eye Res 2019; 187:107753. [PMID: 31408629 DOI: 10.1016/j.exer.2019.107753] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/31/2019] [Accepted: 08/05/2019] [Indexed: 01/16/2023]
Abstract
The retina is known to have a local renin-angiotensin system (RAS) and dysfunction in the RAS is often associated with diseases of the retinal vasculature that cause irreversible vision loss. Regulation of the retinal vasculature to meet the metabolic needs of the tissues occurs through a mechanism called neurovascular coupling, which is critical for maintaining homeostatic function and support for neurons. Neurovascular coupling is the process by which support cells, including glia, regulate blood vessel calibre and blood flow in response to neural activity. In retinal vascular diseases, this coupling mechanism is often disrupted. However, the role that angiotensin II (Ang II), the main effector peptide of the RAS, has in regulating both the retinal vasculature and neurovascular coupling is not fully understood. As components of the RAS are located on the principal neurons, glia and blood vessels of the retina, it is possible that Ang II has a role in regulating communication and function between these three cell types, and therefore the capacity to regulate neurovascular coupling. This review focuses on components of the RAS located on the retinal neurovascular unit, and the potential of this system to contribute to blood flow modulation in the healthy and compromised retina.
Collapse
Affiliation(s)
- Joanna A Phipps
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Michael A Dixon
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Andrew I Jobling
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Anna Y Wang
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Ursula Greferath
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Kirstan A Vessey
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Erica L Fletcher
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, 3010, Victoria, Australia.
| |
Collapse
|
36
|
Jiang Y, Peng T, Gaur U, Silva M, Little P, Chen Z, Qiu W, Zhang Y, Zheng W. Role of Corticotropin Releasing Factor in the Neuroimmune Mechanisms of Depression: Examination of Current Pharmaceutical and Herbal Therapies. Front Cell Neurosci 2019; 13:290. [PMID: 31312123 PMCID: PMC6614517 DOI: 10.3389/fncel.2019.00290] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 06/14/2019] [Indexed: 12/17/2022] Open
Abstract
Approximately 3% of the world population suffers from depression, which is one of the most common form of mental disorder. Recent findings suggest that an interaction between the nervous system and immune system might be behind the pathophysiology of various neurological and psychiatric disorders, including depression. Neuropeptides have been shown to play a major role in mediating response to stress and inducing immune activation or suppression. Corticotropin releasing factor (CRF) is a major regulator of the hypothalamic pituitary adrenal (HPA) axis response. CRF is a stress-related neuropeptide whose dysregulation has been associated with depression. In this review, we summarized the role of CRF in the neuroimmune mechanisms of depression, and the potential therapeutic effects of Chinese herbal medicines (CHM) as well as other agents. Studying the network of CRF and immune responses will help to enhance our understanding of the pathogenesis of depression. Additionally, targeting this important network may aid in developing novel treatments for this debilitating psychiatric disorder.
Collapse
Affiliation(s)
- Yizhou Jiang
- Center of Reproduction, Development and Aging and Institute of Translation Medicine, Faculty of Health Sciences, University of Macau, Macau, China.,Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Tangming Peng
- Center of Reproduction, Development and Aging and Institute of Translation Medicine, Faculty of Health Sciences, University of Macau, Macau, China.,Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Neurosurgical Clinical Research Center of Sichuan Province, Luzhou, China
| | - Uma Gaur
- Center of Reproduction, Development and Aging and Institute of Translation Medicine, Faculty of Health Sciences, University of Macau, Macau, China
| | - Marta Silva
- Center of Reproduction, Development and Aging and Institute of Translation Medicine, Faculty of Health Sciences, University of Macau, Macau, China
| | - Peter Little
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD, Australia
| | - Zhong Chen
- Key Laboratory of Medical Neurobiology of the Ministry of Health of China, College of Pharmaceutical Sciences, Institute of Pharmacology and Toxicology, Zhejiang University, Hangzhou, China
| | - Wei Qiu
- The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yandong Zhang
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Wenhua Zheng
- Center of Reproduction, Development and Aging and Institute of Translation Medicine, Faculty of Health Sciences, University of Macau, Macau, China
| |
Collapse
|
37
|
Matta SM, Hill-Yardin EL, Crack PJ. The influence of neuroinflammation in Autism Spectrum Disorder. Brain Behav Immun 2019; 79:75-90. [PMID: 31029798 DOI: 10.1016/j.bbi.2019.04.037] [Citation(s) in RCA: 247] [Impact Index Per Article: 41.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 04/15/2019] [Accepted: 04/24/2019] [Indexed: 12/12/2022] Open
Abstract
Autism Spectrum Disorder (ASD) is a neurodevelopmental disorder characterised by deficits in social communication and restricted or repetitive behaviours. The clinical presentation of ASD is highly variable and diagnosis is based on the presence of impaired social communication and repetitive and/or restricted behaviours. Although the precise pathophysiologies underlying ASD are unclear, growing evidence supports a role for dysregulated neuroinflammation. The potential involvement of microglia and astrocytes reactive to inflammatory stimuli in ASD has generated much interest due to their varied roles including in mounting an immune response and regulating synaptic function. Increased numbers of reactive microglial and astrocytes in both ASD postmortem tissue and animal models have been reported. Whether dysregulation of glial subtypes exacerbates alterations in neural connectivity in the brain of autistic patients is not well explored. A role for the gut-brain axis involving microbial-immune-neuronal cross talk is also a growing area of neuroinflammation research. Greater understanding of these interactions under patho/physiological conditions and the identification of consistent immune profile abnormalities can potentially lead to more reliable diagnostic measures and treatments in ASD.
Collapse
Affiliation(s)
- Samantha M Matta
- Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Elisa L Hill-Yardin
- School of Health & Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia; Department of Physiology, The University of Melbourne, Parkville, VIC 3010, Australia.
| | - Peter J Crack
- Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
38
|
Jian M, Kwan JSC, Bunting M, Ng RCL, Chan KH. Adiponectin suppresses amyloid-β oligomer (AβO)-induced inflammatory response of microglia via AdipoR1-AMPK-NF-κB signaling pathway. J Neuroinflammation 2019; 16:110. [PMID: 31128596 PMCID: PMC6535190 DOI: 10.1186/s12974-019-1492-6] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 04/30/2019] [Indexed: 01/07/2023] Open
Abstract
Background Microglia-mediated neuroinflammation is important in Alzheimer’s disease (AD) pathogenesis. Extracellular deposition of β-amyloid (Aβ), a major pathological hallmark of AD, can induce microglia activation. Adiponectin (APN), an adipocyte-derived adipokine, exerts anti-inflammatory effects in the periphery and brain. Chronic APN deficiency leads to cognitive impairment and AD-like pathologies in aged mice. Here, we aim to study the role of APN in regulating microglia-mediated neuroinflammation in AD. Methods Inflammatory response of cultured microglia (BV2 cells) to AβO and effects of APN were studied by measuring levels of proinflammatory cytokines (tumor necrosis factor α [TNFα] and interleukin-1β [IL-1β]) in cultured medium before and after exposure to AβO, with and without APN pretreatment. Adiponectin receptor 1 (AdipoR1) and receptor 2 (AdipoR2) were targeted by small interference RNA. To study the neuroprotective effect of APN, cultured HT-22 hippocampal cells were treated with conditioned medium of AβO-exposed BV2 cells or were co-cultured with BV2 cells in transwells. The cytotoxicity of HT-22 hippocampal cells was assessed by MTT reduction. We generated APN-deficient AD mice (APN−/−5xFAD) by crossing APN-knockout mice with 5xFAD mice to determine the effects of APN deficiency on microglia-mediated neuroinflammation in AD. Results AdipoR1 and AdipoR2 were expressed in BV2 cells and microglia of mice. Pretreatment with APN for 2 h suppressed TNFα and IL-1β release induced by AβO in BV2 cells. Additionally, APN rescued the decrease of AMPK phosphorylation and suppressed nuclear translocation of nuclear factor kappa B (NF-κB) induced by AβO. Compound C, an inhibitor of AMPK, abolished these effects of APN. Knockdown of AdipoR1, but not AdipoR2 in BV2 cells, inhibited the ability of APN to suppress proinflammatory cytokine release induced by AβO. Moreover, pretreatment with APN inhibited the cytotoxicity of HT-22 cells co-cultured with AβO-exposed BV2 cells. Lastly, APN deficiency exacerbated microglia activation in 9-month-old APN−/−5xFAD mice associated with upregulation of TNFα and IL-1β in the cortex and hippocampus. Conclusions Our findings demonstrate that APN inhibits inflammatory response of microglia to AβO via AdipoR1-AMPK-NF-κB signaling, and APN deficiency aggravates microglia activation and neuroinflammation in AD mice. APN may be a novel therapeutic agent for inhibiting neuroinflammation in AD. Electronic supplementary material The online version of this article (10.1186/s12974-019-1492-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Min Jian
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, 8/F, 21 Sassoon Road, Pokfulam, Hong Kong, Special Administrative Region of China
| | - Jason Shing-Cheong Kwan
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, 8/F, 21 Sassoon Road, Pokfulam, Hong Kong, Special Administrative Region of China.,Neuroimmunology and Neuroinflammation Research Laboratory, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, Special Administrative Region of China
| | - Myriam Bunting
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, 8/F, 21 Sassoon Road, Pokfulam, Hong Kong, Special Administrative Region of China
| | - Roy Chun-Laam Ng
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, 8/F, 21 Sassoon Road, Pokfulam, Hong Kong, Special Administrative Region of China. .,Neuroimmunology and Neuroinflammation Research Laboratory, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, Special Administrative Region of China.
| | - Koon Ho Chan
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, 8/F, 21 Sassoon Road, Pokfulam, Hong Kong, Special Administrative Region of China. .,Neuroimmunology and Neuroinflammation Research Laboratory, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, Special Administrative Region of China. .,Research Center of Heart, Brain, Hormone and Healthy Aging, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, Special Administrative Region of China. .,Hong Kong University Alzheimer's Disease Research Network, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, Special Administrative Region of China. .,Department of Medicine, The University of Hong Kong, 4/F, Professorial Block, Queen Mary Hospital, 102 Pokfulam Road, Pokfulam, Hong Kong, Special Administrative Region of China.
| |
Collapse
|
39
|
Non-cell-autonomous actions of α-synuclein: Implications in glial synucleinopathies. Prog Neurobiol 2018; 169:158-171. [DOI: 10.1016/j.pneurobio.2018.06.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 07/24/2017] [Accepted: 06/30/2018] [Indexed: 01/11/2023]
|
40
|
Chen SX, Wang SK, Yao PW, Liao GJ, Na XD, Li YY, Zeng WA, Liu XG, Zang Y. Early CALP2 expression and microglial activation are potential inducers of spinal IL-6 up-regulation and bilateral pain following motor nerve injury. J Neurochem 2018; 145:154-169. [DOI: 10.1111/jnc.14317] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 01/28/2018] [Accepted: 01/30/2018] [Indexed: 12/21/2022]
Affiliation(s)
- Shao-Xia Chen
- Pain Research Center and Department of Physiology; Zhongshan Medical School of Sun Yat-Sen University; Guangzhou China
- Department of Anesthesiology; Cancer Center, Sun Yat-Sen University; State Key Laboratory of Oncology in South China; Collaborative, Innovation Center for Cancer Medicine; Guangzhou China
| | - Shao-Kun Wang
- Pain Research Center and Department of Physiology; Zhongshan Medical School of Sun Yat-Sen University; Guangzhou China
| | - Pei-Wen Yao
- Pain Research Center and Department of Physiology; Zhongshan Medical School of Sun Yat-Sen University; Guangzhou China
| | - Guang-Jie Liao
- Pain Research Center and Department of Physiology; Zhongshan Medical School of Sun Yat-Sen University; Guangzhou China
| | - Xiao-Dong Na
- Pain Research Center and Department of Physiology; Zhongshan Medical School of Sun Yat-Sen University; Guangzhou China
- Department of Pathophysiology; Zhongshan Medical School of Sun Yat-Sen University; Guangzhou China
| | - Yong-Yong Li
- Pain Research Center and Department of Physiology; Zhongshan Medical School of Sun Yat-Sen University; Guangzhou China
| | - Wei-an Zeng
- Department of Anesthesiology; Cancer Center, Sun Yat-Sen University; State Key Laboratory of Oncology in South China; Collaborative, Innovation Center for Cancer Medicine; Guangzhou China
| | - Xian-Guo Liu
- Pain Research Center and Department of Physiology; Zhongshan Medical School of Sun Yat-Sen University; Guangzhou China
| | - Ying Zang
- Pain Research Center and Department of Physiology; Zhongshan Medical School of Sun Yat-Sen University; Guangzhou China
| |
Collapse
|
41
|
TNF-α-sensitive brain pericytes activate microglia by releasing IL-6 through cooperation between IκB-NFκB and JAK-STAT3 pathways. Brain Res 2018; 1692:34-44. [PMID: 29702085 DOI: 10.1016/j.brainres.2018.04.023] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 04/17/2018] [Accepted: 04/20/2018] [Indexed: 01/18/2023]
Abstract
Interleukin (IL)-6 is an important mediator of neurovascular dysfunction, neurodegeneration and/or neuroinflammation. We previously reported that brain pericytes released higher levels of IL-6 than did glial cells (astrocytes and microglia) in response to tumor necrosis factor (TNF)-α. Moreover, pericytes stimulated with TNF-α enhanced activation of BV-2 microglia. In this study, we investigated the mechanisms of TNF-α mediated induction of IL-6 release from brain pericytes and astrocytes and whether pericyte-derived IL-6 would facilitate activation of BV-2 microglia. Using rat brain pericyte and astrocyte primary cultures and pharmacological inhibitors, we found that, TNF-α induced the highest levels of IL-6 release from pericytes by activating the inhibitor kappa B (IκB)-nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) and Janus family of tyrosine kinase (JAK)-signal transducer and activator of transcription (STAT)3 pathways. STAT3 contributed to TNF-α induced nuclear translocation of phospho-NFκB in pericytes. TNF-α-induced IL-6 release in astrocytes was mediated by NFκB but not by STAT3. The presence of pericytes amplified TNF-α-induced iNOS mRNA expression in BV-2 microglia. This effect was blocked by a neutralizing antibody for IL-6. These findings indicated that crosstalk between the IκB-NFκB and JAK-STAT3 pathways is a pericyte specific mechanism, not occurring in astrocytes, for TNF-α-induced IL-6 release. IL-6 derived from pericytes enhanced microglial activation. Our findings increase understanding of the role of pericyte-microglia crosstalk in the brain under neuroinflammatory conditions and suggest a potentially attractive therapeutic target for brain inflammation.
Collapse
|
42
|
Lee JM, Kim CJ, Park JM, Song MK, Kim YJ. Effect of treadmill exercise on spatial navigation impairment associated with cerebellar Purkinje cell loss following chronic cerebral hypoperfusion. Mol Med Rep 2018; 17:8121-8128. [PMID: 29693705 PMCID: PMC5983984 DOI: 10.3892/mmr.2018.8893] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 03/20/2018] [Indexed: 12/16/2022] Open
Abstract
In addition to roles in motor coordination, the cerebellum is also associated with cognitive function. The aim of the present study was to investigate the effect of treadmill exercise on spatial navigation deficit induced by chronic cerebral hypoperfusion (CCH). Furthermore, whether decreased loss of Purkinje cells, which contain the calcium-binding protein in the posterior lobe of the cerebellum, attenuates the spatial navigation deficit induced by CCH was also investigated. Wistar rats were randomly divided into three groups: Sham group, bilateral common carotid arteries occlusion (BCCAO) group and a BCCAO + exercise (Ex) group. The rats in the BCCAO + Ex group ran on a treadmill for 30 min once a day for 8 weeks, starting at 4 weeks post-birth. CCH was induced by performing BCCAO at 12 weeks post-birth. The Morris water maze test was performed to determine the spatial navigation function of the rats. To investigate the histological features of the cerebellum in all of the experimental groups post-treatment, terminal deoxynucleotidyl transferase dUTP nick end labeling staining, as well as immunohistochemical analysis revealing the expression of calbindin, parvalbumin, glial fibrillary acidic protein, ionized calcium-binding adaptor molecule 1 and caspase-3, was performed. The results of the present study revealed that treadmill exercise improved spatial navigation, decreased the expression of reactive astrocytes and microglial cells, and decreased apoptotic rates in the cerebellar vermis post-CCH. Treadmill exercise also attenuated the loss of Purkinje cells following CCH. The number of Purkinje cells was revealed to be negatively correlated with spatial navigation performance. These results indicate that treadmill exercise may attenuate spatial navigation impairment via inhibition of Purkinje cell loss in the posterior lobe of the cerebellum following CCH. Therefore, treadmill exercise may represent a therapeutic strategy for the treatment of patients with spatial navigation impairment following CCH.
Collapse
Affiliation(s)
- Jae-Min Lee
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Chang-Ju Kim
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jong-Min Park
- Department of Nursing, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Min Kyung Song
- Department of Nursing, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Youn-Jung Kim
- Department of Basic Nursing Science, College of Nursing Science, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
43
|
Wu F, Luo T, Mei Y, Liu H, Dong J, Fang Y, Peng J, Guo Y. Simvastatin alters M1/M2 polarization of murine BV2 microglia via Notch signaling. J Neuroimmunol 2018; 316:56-64. [DOI: 10.1016/j.jneuroim.2017.12.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 11/12/2017] [Accepted: 12/17/2017] [Indexed: 01/10/2023]
|
44
|
Santoro A, Spinelli CC, Martucciello S, Nori SL, Capunzo M, Puca AA, Ciaglia E. Innate immunity and cellular senescence: The good and the bad in the developmental and aged brain. J Leukoc Biol 2018; 103:509-524. [PMID: 29389023 DOI: 10.1002/jlb.3mr0118-003r] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 01/12/2018] [Accepted: 01/12/2018] [Indexed: 12/21/2022] Open
Abstract
Ongoing studies evidence cellular senescence in undifferentiated and specialized cells from tissues of all ages. Although it is believed that senescence plays a wider role in several stress responses in the mature age, its participation in certain physiological and pathological processes throughout life is coming to light. The "senescence machinery" has been observed in all brain cell populations, including components of innate immunity (e.g., microglia and astrocytes). As the beneficial versus detrimental implications of senescence is an open question, we aimed to analyze the contribution of immune responses in regulatory mechanisms governing its distinct functions in healthy (development, organogenesis, danger patrolling events) and diseased brain (glioma, neuroinflammation, neurodeneration), and the putative connection between cellular and molecular events governing the 2 states. Particularly this review offers new insights into the complex roles of senescence both as a chronological event as age advances, and as a molecular mechanism of brain homeostasis through the important contribution of innate immune responses and their crosstalk with neighboring cells in brain parenchyma. We also highlight the impact of the recently described glymphatic system and brain lymphatic vasculature in the interplay between peripheral and central immune surveillance and its potential implication during aging. This will open new ways to understand brain development, its deterioration during aging, and the occurrence of several oncological and neurodegenerative diseases.
Collapse
Affiliation(s)
- Antonietta Santoro
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana," University of Salerno, Via Salvatore Allende, Baronissi, Italy
| | | | | | - Stefania Lucia Nori
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana," University of Salerno, Via Salvatore Allende, Baronissi, Italy
| | - Mario Capunzo
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana," University of Salerno, Via Salvatore Allende, Baronissi, Italy
| | - Annibale Alessandro Puca
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana," University of Salerno, Via Salvatore Allende, Baronissi, Italy.,Cardiovascular Research Unit, IRCCS MultiMedica, Milan, Italy
| | - Elena Ciaglia
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana," University of Salerno, Via Salvatore Allende, Baronissi, Italy
| |
Collapse
|
45
|
Lim S, Chun Y, Lee JS, Lee SJ. Neuroinflammation in Synucleinopathies. Brain Pathol 2018; 26:404-9. [PMID: 26940152 DOI: 10.1111/bpa.12371] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 01/08/2016] [Accepted: 02/24/2016] [Indexed: 12/25/2022] Open
Abstract
The causes of most neurodegenerative diseases are attributed to multiple genetic and environmental factors interacting with one another. Above all, inflammation in the nervous system has been implicated in many neurodegenerative diseases. Still, the roles of neuroinflammation in disease mechanisms and the triggers of inflammatory responses in disease-inflicted brain tissues seem to remain unclear. This review will examine previous studies that had been done from genetic, pathological and epidemiological perspectives. These studies assess the involvement of neuroinflammation in synucleinopathies, a group of neurodegenerative diseases that are characterized by deposition of α-synuclein aggregates such as Parkinson's disease, dementia with Lewy bodies and multiple system atrophy. The review will also discuss the role of α-synuclein aggregates in triggering inflammatory responses from glial cells. It is expected that a precise assessment of the roles and mechanisms of neuroinflammation in neurodegenerative diseases will pave the way for the development of disease-modifying drugs.
Collapse
Affiliation(s)
- Somin Lim
- Department of Biomedical Sciences, Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Yewon Chun
- Department of Biomedical Sciences, Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Jun Sung Lee
- Department of Biomedical Sciences, Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Seung-Jae Lee
- Department of Biomedical Sciences, Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, 03080, Korea
| |
Collapse
|
46
|
Manickavasagam D, Novak K, Oyewumi MO. Therapeutic Delivery of Simvastatin Loaded in PLA-PEG Polymersomes Resulted in Amplification of Anti-inflammatory Effects in Activated Microglia. AAPS JOURNAL 2017; 20:18. [DOI: 10.1208/s12248-017-0176-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 11/21/2017] [Indexed: 01/18/2023]
|
47
|
Wang J, Li D, Hou J, Lei H. Protective effects of geniposide and ginsenoside Rg1 combination treatment on rats following cerebral ischemia are mediated via microglial microRNA‑155‑5p inhibition. Mol Med Rep 2017; 17:3186-3193. [PMID: 29257264 DOI: 10.3892/mmr.2017.8221] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 11/02/2017] [Indexed: 11/06/2022] Open
Abstract
Geniposide, an active component of Gardenia, has been reported to protect against cerebral ischemia in animals. Ginsenoside Rg1, a component of Panax notoginseng, is usually administered in combination with Gardenia for the treatment of acute ischemic stroke; however, there are unknown effects of ginsenoside Rg1 that require further investigation. In the present study, the effects of geniposide and ginsensoide Rg1 combination treatment on focal cerebral ischemic stroke were investigated. For in vivo analysis, male rats were separated into three groups, including the (control), model and geniposide + ginsenoside Rg1 groups (n=8 per group). A middle cerebral artery occlusion model was established as the model group. The treatment group was treated with geniposide (30 mg/kg, tail vein injection) + ginsenoside Rg1 (6 mg/kg, tail vein injection), and the model group received saline instead. Neurobehavioral deficits, infarct volume, brain edema, and the expression of microRNA (miR)‑155‑5p and CD11b by reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) and immunohistochemistry, were assessed following 24 h of ischemia. For in vitro analysis, BV2 mouse microglial cells were cultured and exposed to geniposide (40 µg/ml) + ginsenoside Rg1 (8 µg/ml) during various durations of oxygen‑glucose deprivation (OGD). The expression levels of miR‑155‑5p, pri‑miR‑155 and pre‑miR‑155 were detected by RT‑qPCR. The results demonstrated that increases in brain infarct volume, edema volume, CD11b‑positive cells and miR‑155‑5p levels were alleviated following geniposide + ginsenoside administration in rats exposed to ischemia. Furthermore, geniposide + ginsenoside Rg1 treatment suppressed the miR‑155‑5p, pri‑miR‑155 and pre‑miR‑155 expression levels in OGD‑injured BV2 microglial cells. The results of the present study demonstrated that tail vein administration of geniposide in combination with ginsenoside Rg1 protected against focal cerebral ischemia in rats through inhibition of microglial miR‑155‑5p following ischemic injury, which may serve as a novel therapeutic agent for the treatment of strokes.
Collapse
Affiliation(s)
- Jun Wang
- Institute of Basic Theory, China Academy of Chinese Medical Sciences, Beijing 100700, P.R. China
| | - Dan Li
- Jingjinji United Innovation Pharmaceutical Research Company, Beijing 100083, P.R. China
| | - Jincai Hou
- Jingjinji United Innovation Pharmaceutical Research Company, Beijing 100083, P.R. China
| | - Hongtao Lei
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, P.R. China
| |
Collapse
|
48
|
da Fonseca ACC, Amaral R, Garcia C, Geraldo LH, Matias D, Lima FRS. Microglia in Cancer: For Good or for Bad? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 949:245-261. [PMID: 27714693 DOI: 10.1007/978-3-319-40764-7_12] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Glioblastoma is a malignant tumor of astrocytic origin that is highly invasive, proliferative and angiogenic. Despite current advances in multimodal therapies, such as surgery, radio- and chemotherapy, the outcome for patients with glioblastoma is nearly always fatal. The glioblastoma microenvironment has a tremendous influence over the tumor growth and spread. Microglia and macrophages are abundant cells in the tumor mass. Increasing evidence indicates that glioblastoma recruits these cell populations and signals in a way that microglia and macrophages are subverted to promote tumor progression. In this chapter, we discuss some aspects of the interaction between microglia and glioblastoma, consequences of this interaction for tumor progression and the possibility of microglial cells being used as therapeutic vectors, which opens up new alternatives for the development of GBM therapies targeting microglia.
Collapse
Affiliation(s)
- Anna Carolina Carvalho da Fonseca
- Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Rackele Amaral
- Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Celina Garcia
- Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Luiz Henrique Geraldo
- Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Diana Matias
- Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Rio de Janeiro, RJ, 21949-590, Brazil
| | - Flavia Regina Souza Lima
- Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Rio de Janeiro, RJ, 21949-590, Brazil.
| |
Collapse
|
49
|
Wang J, Yang C, Zhao Q, Zhu Z, Li Y, Yang P. Microglia activation induced by serum of SLE patients. J Neuroimmunol 2017; 310:135-142. [PMID: 28778438 DOI: 10.1016/j.jneuroim.2017.07.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 06/30/2017] [Accepted: 07/17/2017] [Indexed: 01/10/2023]
Abstract
To investigate the potential involvement of microglia in the neuropathology of systemic lupus erythematosus (SLE), we examined whether SLE patient sera could activate BV2 microglia in vitro. Exposure to SLE patient sera resulted in morphological changes in the microglia, an increase in MHC II and CD86 protein expression, and an obvious release of nitric oxide and proinflammatory cytokines. However, the SLE sera did not induce a specific change in the production of immunoregulatory cytokines. Inactivating complements or neutralizing proinflammatory cytokines in the SLE sera did not suppress microglial activation. Our results highlight the potential role of microglia in neuroinflammation in SLE patients.
Collapse
Affiliation(s)
- Jianing Wang
- Department of Rheumatology and Immunology, First Affiliated Hospital, China Medical University, Shenyang 110001, People's Republic of China
| | - Chunshu Yang
- Department of 1st Cancer Institute, First Affiliated Hospital, China Medical University, Shenyang 110001, People's Republic of China
| | - Qi Zhao
- Department of Rheumatology and Immunology, First Affiliated Hospital, China Medical University, Shenyang 110001, People's Republic of China
| | - Ziwei Zhu
- Department of Rheumatology and Immunology, First Affiliated Hospital, China Medical University, Shenyang 110001, People's Republic of China
| | - Yujia Li
- Department of Rheumatology and Immunology, First Affiliated Hospital, China Medical University, Shenyang 110001, People's Republic of China
| | - Pingting Yang
- Department of Rheumatology and Immunology, First Affiliated Hospital, China Medical University, Shenyang 110001, People's Republic of China.
| |
Collapse
|
50
|
Lai JCY, Rocha-Ferreira E, Ek CJ, Wang X, Hagberg H, Mallard C. Immune responses in perinatal brain injury. Brain Behav Immun 2017; 63:210-223. [PMID: 27865947 DOI: 10.1016/j.bbi.2016.10.022] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 10/28/2016] [Accepted: 10/30/2016] [Indexed: 12/13/2022] Open
Abstract
The perinatal period has often been described as immune deficient. However, it has become clear that immune responses in the neonate following exposure to microbes or as a result of tissue injury may be substantial and play a role in perinatal brain injury. In this article we will review the immune cell composition under normal physiological conditions in the perinatal period, both in the human and rodent. We will summarize evidence of the inflammatory responses to stimuli and discuss how neonatal immune activation, both in the central nervous system and in the periphery, may contribute to perinatal hypoxic-ischemic brain injury.
Collapse
Affiliation(s)
- Jacqueline C Y Lai
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30 Gothenburg, Sweden
| | - Eridan Rocha-Ferreira
- Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30 Gothenburg, Sweden
| | - C Joakim Ek
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30 Gothenburg, Sweden
| | - Xiaoyang Wang
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30 Gothenburg, Sweden
| | - Henrik Hagberg
- Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30 Gothenburg, Sweden
| | - Carina Mallard
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30 Gothenburg, Sweden.
| |
Collapse
|