1
|
Ganguly K, Adhikary K, Acharjee A, Acharjee P, Trigun SK, Mutlaq AS, Ashique S, Yasmin S, Alshahrani AM, Ansari MY. Biological significance and pathophysiological role of Matrix Metalloproteinases in the Central Nervous System. Int J Biol Macromol 2024; 280:135967. [PMID: 39322129 DOI: 10.1016/j.ijbiomac.2024.135967] [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: 06/22/2024] [Revised: 09/21/2024] [Accepted: 09/21/2024] [Indexed: 09/27/2024]
Abstract
Matrix Metalloproteinases (MMPs), which are endopeptidase reliant on zinc, are low in embryonic tissues but increases in response to a variety of physiological stimulus and pathological stresses. Neuro-glial cells, endothelial cells, fibroblasts, and leucocytes secrete MMPs, which cleave extracellular matrix proteins in a time-dependent manner. MMPs affect synaptic plasticity and the development of short-term memory by controlling the size, shape, and excitatory synapses' function through the lateral diffusion of receptors. In addition, MMPs influence the Extracellular Matrix proteins in the Peri-Neuronal Net at the Neuro-glial interface, which aids in the establishment of long-term memory. Through modulating neuronal, and glial cells migration, differentiation, Neurogenesis, and survival, MMPs impact brain development in mammals. In adult brains, MMPs play a beneficial role in physiological plasticity, which includes learning, memory consolidation, social interaction, and complex behaviors, by proteolytically altering a wide variety of factors, including growth factors, cytokines, receptors, DNA repair enzymes, and matrix proteins. Additionally, stress, depression, addiction, hepatic encephalopathy, and stroke may all have negative effects on MMPs. In addition to their role in glioblastoma development, MMPs influence neurological diseases such as epilepsy, schizophrenia, autism spectrum disorder, brain damage, pain, neurodegeneration, and Alzheimer's and Parkinson's. To help shed light on the potential of MMPs as a therapeutic target for neurodegenerative diseases, this review summarizes their regulation, mode of action, and participation in brain physiological plasticity and pathological damage. Finally, by employing different MMP-based nanotools and inhibitors, MMPs may also be utilized to map the anatomical and functional connectome of the brain, analyze its secretome, and treat neurodegenerative illnesses.
Collapse
Affiliation(s)
- Krishnendu Ganguly
- Department of Medical Lab Technology, Paramedical College Durgapur, Helen Keller Sarani, Durgapur 713212, West Bengal, India.
| | - Krishnendu Adhikary
- Department of Medical Lab Technology, Paramedical College Durgapur, Helen Keller Sarani, Durgapur 713212, West Bengal, India.
| | - Arup Acharjee
- Molecular Omics Laboratory, Department of Zoology, University of Allahabad, Allahabad, Uttar Pradesh, India.
| | - Papia Acharjee
- Biochemistry Section, Department of Zoology, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India.
| | - Surendra Kumar Trigun
- Biochemistry Section, Department of Zoology, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India.
| | | | - Sumel Ashique
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Department of Pharmaceutics, Bengal College of Pharmaceutical Sciences & Research, Durgapur 713212, West Bengal, India.
| | - Sabina Yasmin
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia.
| | - Asma M Alshahrani
- Department of Clinical Pharmacy, Faculty of Pharmacy, King Khalid University, Abha, Saudi Arabia; Department of Clinical Pharmacy, Shaqra University, Saudi Arabia.
| | - Mohammad Yousuf Ansari
- MM college of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana 133207, India.
| |
Collapse
|
2
|
Yang F, Yang L, Kuroda Y, Lai S, Takahashi Y, Sayo T, Namiki T, Nakajima K, Sano S, Inoue S, Tsuruta D, Katayama I. Disorganisation of basement membrane zone architecture impairs melanocyte residence in vitiligo. J Pathol 2024; 264:30-41. [PMID: 38989633 DOI: 10.1002/path.6321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 04/22/2024] [Accepted: 05/22/2024] [Indexed: 07/12/2024]
Abstract
The basement membrane zone is the interface between the epidermis and dermis, and it is disrupted in several skin conditions. Here, we report the results of a comprehensive investigation into the structural and molecular factors of the basement membrane zone in vitiligo, a dermatological disorder characterised by depigmented patches on the skin. Using electron microscopy and immunofluorescence staining, we confirmed abnormal basement membrane zone morphology and disrupted basement membrane zone architecture in human vitiliginous skin. Furthermore, we identified elevated expression of matrix metalloproteinase 2 (MMP2) in human dermal fibroblasts as a key factor responsible for basement membrane zone matrix degradation. In our in vitro and ex vivo models, overexpression of MMP2 in fibroblasts led to basement membrane zone disruption and melanocyte disappearance. Importantly, we reveal that the loss of melanocytes in vitiligo is primarily linked to their weakened adhesion to the basement membrane, mediated by binding between integrin β1 and laminin and discoidin domain receptor 1 and collagen IV. Finally, inhibition of matrix metalloproteinase 2 expression reversed depigmentation in a mouse model of vitiligo. In conclusion, our research shows the importance of basement membrane zone integrity in melanocyte residence and offers new avenues for therapeutic interventions to address this challenging skin condition. © 2024 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Fei Yang
- Department of Pigmentation Research and Therapeutics, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
- Biological Science Research Laboratories, Kao Corporation, Odawara, Japan
| | - Lingli Yang
- Department of Pigmentation Research and Therapeutics, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Yasutaka Kuroda
- Department of Pigmentation Research and Therapeutics, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
- Biological Science Research Laboratories, Kao Corporation, Odawara, Japan
| | - Sylvia Lai
- Department of Pigmentation Research and Therapeutics, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Yoshito Takahashi
- Biological Science Research Laboratories, Kao Corporation, Odawara, Japan
| | - Tetsuya Sayo
- Biological Science Research Laboratories, Kao Corporation, Odawara, Japan
| | - Takeshi Namiki
- Department of Dermatology, Graduate School and Faculty of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kimiko Nakajima
- Department of Dermatology, Kochi Medical School, Kochi University, Kochi, Japan
| | - Shigetoshi Sano
- Department of Dermatology, Kochi Medical School, Kochi University, Kochi, Japan
| | - Shintaro Inoue
- Department of Cosmetic Health Science, Gifu Pharmaceutical University, Gifu, Japan
| | - Daisuke Tsuruta
- Department of Dermatology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Ichiro Katayama
- Department of Pigmentation Research and Therapeutics, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| |
Collapse
|
3
|
Padarath K, Deroubaix A, Naicker P, Stoychev S, Kramvis A. Comparative Proteomic Analysis of Huh7 Cells Transfected with Sub-Saharan African Hepatitis B Virus (Sub)genotypes Reveals Potential Oncogenic Factors. Viruses 2024; 16:1052. [PMID: 39066215 PMCID: PMC11281506 DOI: 10.3390/v16071052] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024] Open
Abstract
In sub-Saharan Africa (SSA), the (sub)genotypes A1, D3, and E of the hepatitis B virus (HBV) prevail. Individuals infected with subgenotype A1 have a 4.5-fold increased risk of HCC compared to those infected with other (sub)genotypes. The effect of (sub)genotypes on protein expression and host signalling has not been studied. Mass spectrometry was used to analyse the proteome of Huh7 cells transfected with replication-competent clones. Proteomic analysis revealed significantly differentially expressed proteins between SSA (sub)genotypes. Different (sub)genotypes have the propensity to dysregulate specific host signalling pathways. Subgenotype A1 resulted in dysregulation within the Ras pathway. Ras-associated protein, RhoC, was significantly upregulated in cells transfected with subgenotype A1 compared to those transfected with other (sub)genotypes, on both a proteomic (>1.5-fold) and mRNA level (p < 0.05). Two of the main cellular signalling pathways involving RHOC, MAPK and PI3K/Akt/mTOR, regulate cell growth, motility, and survival. Downstream signalling products of these pathways have been shown to increase MMP2 and MMP9 expression. An extracellular MMP2 and MMP9 ELISA revealed a non-significant increase in MMP2 and MMP9 in the cells transfected with A1 compared to the other (sub)genotypes (p < 0.05). The upregulated Ras-associated proteins have been implicated as oncoproteins in various cancers and could contribute to the increased hepatocarcinogenic potential of A1.
Collapse
Affiliation(s)
- Kiyasha Padarath
- Hepatitis Virus Diversity Unit, Department of Internal Medicine, School of Clinical Medicine, Faculty of Health Science, University of Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa (A.D.)
| | - Aurélie Deroubaix
- Hepatitis Virus Diversity Unit, Department of Internal Medicine, School of Clinical Medicine, Faculty of Health Science, University of Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa (A.D.)
- Life Sciences Imaging Facility, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa
| | - Previn Naicker
- Future Production Chemicals, Council for Scientific and Industrial Research, Pretoria 0184, South Africa;
| | - Stoyan Stoychev
- ReSyn Biosciences, Johannesburg 2000, South Africa;
- Evosep Biosystems, 5230 Odense, Denmark
| | - Anna Kramvis
- Hepatitis Virus Diversity Unit, Department of Internal Medicine, School of Clinical Medicine, Faculty of Health Science, University of Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa (A.D.)
| |
Collapse
|
4
|
Chen X, Ran X, Wei X, Zhu L, Chen S, Liao Z, Xu K, Xia W. Bioactive glass 1393 promotes angiogenesis and accelerates wound healing through ROS/P53/MMP9 signaling pathway. Regen Ther 2024; 26:132-144. [PMID: 38872979 PMCID: PMC11169082 DOI: 10.1016/j.reth.2024.05.016] [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: 03/29/2024] [Revised: 05/20/2024] [Accepted: 05/26/2024] [Indexed: 06/15/2024] Open
Abstract
Compared to bioactive glass 45S5, bioactive glass 1393 has shown greater potential in activating tissue cells and promoting angiogenesis for bone repair. Nevertheless, the effect of bioactive glass 1393 in the context of wound healing remains extensively unexplored, and its mechanism in wound healing remains unclear. Considering that angiogenesis is a critical stage in wound healing, we hypothesize that bioactive glass 1393 may facilitate wound healing through the stimulation of angiogenesis. To validate this hypothesis and further explore the mechanisms underlying its pro-angiogenic effects, we investigated the impact of bioactive glass 1393 on wound healing angiogenesis through both in vivo and in vitro studies. The research demonstrated that bioactive glass 1393 accelerated wound healing by promoting the formation of granulation, deposition of collagen, and angiogenesis. The results of Western blot analysis and immunofluorescence staining revealed that bioactive glass 1393 up-regulated the expression of angiogenesis-related factors. Additionally, bioactive glass 1393 inhibited the expression of ROS and P53 to promote angiogenesis. Furthermore, bioactive glass 1393 stimulated angiogenesis through the P53 signaling pathway, as evidenced by P53 activation assays. Collectively, these findings indicate that bioactive glass 1393 accelerates wound healing by promoting angiogenesis via the ROS/P53/MMP9 signaling pathway.
Collapse
Affiliation(s)
- Xuenan Chen
- National Key Clinical Specialty(Wound Healing), Burn and Wound Healing Center, The First Affliated Hospital of Wenzhou Medical University, Wenzhou, China
- College of Life and Environmental Sciences, Wenzhou University, Zhejiang, China
| | - Xinyu Ran
- National Key Clinical Specialty(Wound Healing), Burn and Wound Healing Center, The First Affliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xuebo Wei
- National Key Clinical Specialty(Wound Healing), Burn and Wound Healing Center, The First Affliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lifei Zhu
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China
| | - Shaodong Chen
- Department of Orthopaedics, Lishui People's Hospital, Zhejiang, China
| | - Zhiyong Liao
- College of Life and Environmental Sciences, Wenzhou University, Zhejiang, China
| | - Ke Xu
- National Key Clinical Specialty(Wound Healing), Burn and Wound Healing Center, The First Affliated Hospital of Wenzhou Medical University, Wenzhou, China
- College of Life and Environmental Sciences, Wenzhou University, Zhejiang, China
| | - Weidong Xia
- National Key Clinical Specialty(Wound Healing), Burn and Wound Healing Center, The First Affliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
5
|
Popa MA, Mihai CM, Șuică VI, Antohe F, Dubey RK, Leeners B, Simionescu M. Dihydrotestosterone Augments the Angiogenic and Migratory Potential of Human Endothelial Progenitor Cells by an Androgen Receptor-Dependent Mechanism. Int J Mol Sci 2024; 25:4862. [PMID: 38732080 PMCID: PMC11084206 DOI: 10.3390/ijms25094862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Endothelial progenitor cells (EPCs) play a critical role in cardiovascular regeneration. Enhancement of their native properties would be highly beneficial to ensuring the proper functioning of the cardiovascular system. As androgens have a positive effect on the cardiovascular system, we hypothesized that dihydrotestosterone (DHT) could also influence EPC-mediated repair processes. To evaluate this hypothesis, we investigated the effects of DHT on cultured human EPCs' proliferation, viability, morphology, migration, angiogenesis, gene and protein expression, and ability to integrate into cardiac tissue. The results showed that DHT at different concentrations had no cytotoxic effect on EPCs, significantly enhanced the cell proliferation and viability and induces fast, androgen-receptor-dependent formation of capillary-like structures. DHT treatment of EPCs regulated gene expression of androgen receptors and the genes and proteins involved in cell migration and angiogenesis. Importantly, DHT stimulation promoted EPC migration and the cells' ability to adhere and integrate into murine cardiac slices, suggesting it has a role in promoting tissue regeneration. Mass spectrometry analysis further highlighted the impact of DHT on EPCs' functioning. In conclusion, DHT increases the proliferation, migration, and androgen-receptor-dependent angiogenesis of EPCs; enhances the cells' secretion of key factors involved in angiogenesis; and significantly potentiates cellular integration into heart tissue. The data offer support for potential therapeutic applications of DHT in cardiovascular regeneration and repair processes.
Collapse
Affiliation(s)
- Mirel Adrian Popa
- Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (M.A.P.); (C.M.M.); (V.I.Ș.); (F.A.)
| | - Cristina Maria Mihai
- Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (M.A.P.); (C.M.M.); (V.I.Ș.); (F.A.)
| | - Viorel Iulian Șuică
- Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (M.A.P.); (C.M.M.); (V.I.Ș.); (F.A.)
| | - Felicia Antohe
- Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (M.A.P.); (C.M.M.); (V.I.Ș.); (F.A.)
| | - Raghvendra K. Dubey
- Department for Reproductive Endocrinology, University Zurich, 8006 Zürich, Switzerland; (R.K.D.); (B.L.)
| | - Brigitte Leeners
- Department for Reproductive Endocrinology, University Zurich, 8006 Zürich, Switzerland; (R.K.D.); (B.L.)
| | - Maya Simionescu
- Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (M.A.P.); (C.M.M.); (V.I.Ș.); (F.A.)
| |
Collapse
|
6
|
Han PP, Han Y, Shen XY, Gao ZK, Bi X. Enriched environment-induced neuroplasticity in ischemic stroke and its underlying mechanisms. Front Cell Neurosci 2023; 17:1210361. [PMID: 37484824 PMCID: PMC10360187 DOI: 10.3389/fncel.2023.1210361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 06/26/2023] [Indexed: 07/25/2023] Open
Abstract
Stroke is a common cerebrovascular disease that can interrupt local blood flow in the brain, causing neuronal damage or even death, resulting in varying degrees of neurological dysfunction. Neuroplasticity is an important neurological function that helps neurons reorganize and regain function after injury. After cerebral ischemia, neuroplasticity changes are critical factors for restoring brain function. An enriched environment promotes increased neuroplasticity, thereby aiding stroke recovery. In this review, we discuss the positive effects of the enriched environment on neuroplasticity after cerebral ischemia, including synaptic plasticity, neurogenesis, and angiogenesis. In addition, we also introduce some studies on the clinical application of enriched environments in the rehabilitation of post-stroke patients, hoping that they can provide some inspiration for doctors and therapists looking for new approaches to stroke rehabilitation.
Collapse
Affiliation(s)
- Ping-Ping Han
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Yu Han
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China
| | - Xin-Ya Shen
- Graduate School of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhen-Kun Gao
- Graduate School of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xia Bi
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| |
Collapse
|
7
|
Frommer ML, Langridge BJ, Awad L, Jasionowska S, Denton CP, Abraham DJ, Abu-Hanna J, Butler PEM. Single-Cell Analysis of ADSC Interactions with Fibroblasts and Endothelial Cells in Scleroderma Skin. Cells 2023; 12:1784. [PMID: 37443817 PMCID: PMC10341100 DOI: 10.3390/cells12131784] [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: 06/02/2023] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
Adipose-derived stem cells (ADSCs) as part of autologous fat grafting have anti-fibrotic and anti-inflammatory effects, but the exact mechanisms of action remain unknown. By simulating the interaction of ADSCs with fibroblasts and endothelial cells (EC) from scleroderma (SSc) skin in silico, we aim to unravel these mechanisms. Publicly available single-cell RNA sequencing data from the stromal vascular fraction of 3 lean patients and biopsies from the skin of 10 control and 12 patients with SSc were obtained from the GEO and analysed using R and Seurat. Differentially expressed genes were used to compare the fibroblast and EC transcriptome between controls and SSc. GO and KEGG functional enrichment was performed. Ligand-receptor interactions of ADSCs with fibroblasts and ECs were explored with LIANA. Pro-inflammatory and extracellular matrix (ECM) interacting fibroblasts were identified in SSc. Arterial, capillary, venous and lymphatic ECs showed a pro-fibrotic and pro-inflammatory transcriptome. Most interactions with both cell types were based on ECM proteins. Differential interactions identified included NTN1, VEGFD, MMP2, FGF2, and FNDC5. The ADSC secretome may disrupt vascular and perivascular inflammation hubs in scleroderma by promoting angiogenesis and especially lymphangiogenesis. Key phenomena observed after fat grafting remain unexplained, including modulation of fibroblast behaviour.
Collapse
Affiliation(s)
- Marvin L. Frommer
- Charles Wolfson Centre for Reconstructive Surgery, Royal Free Hospital, London NW3 2QG, UK; (B.J.L.); (J.A.-H.); (P.E.M.B.)
- Department of Surgical Biotechnology, Division of Surgery & Interventional Science, University College London, London NW3 2QG, UK
- Department of Plastic Surgery, Royal Free Hospital, London NW3 2QG, UK
| | - Benjamin J. Langridge
- Charles Wolfson Centre for Reconstructive Surgery, Royal Free Hospital, London NW3 2QG, UK; (B.J.L.); (J.A.-H.); (P.E.M.B.)
- Department of Surgical Biotechnology, Division of Surgery & Interventional Science, University College London, London NW3 2QG, UK
- Department of Plastic Surgery, Royal Free Hospital, London NW3 2QG, UK
| | - Laura Awad
- Charles Wolfson Centre for Reconstructive Surgery, Royal Free Hospital, London NW3 2QG, UK; (B.J.L.); (J.A.-H.); (P.E.M.B.)
- Department of Plastic Surgery, Royal Free Hospital, London NW3 2QG, UK
| | - Sara Jasionowska
- Charles Wolfson Centre for Reconstructive Surgery, Royal Free Hospital, London NW3 2QG, UK; (B.J.L.); (J.A.-H.); (P.E.M.B.)
- Department of Plastic Surgery, Royal Free Hospital, London NW3 2QG, UK
| | - Christopher P. Denton
- Centre for Rheumatology, Department of Inflammation, Division of Medicine, University College London, London NW3 2QG, UK
| | - David J. Abraham
- Centre for Rheumatology, Department of Inflammation, Division of Medicine, University College London, London NW3 2QG, UK
| | - Jeries Abu-Hanna
- Charles Wolfson Centre for Reconstructive Surgery, Royal Free Hospital, London NW3 2QG, UK; (B.J.L.); (J.A.-H.); (P.E.M.B.)
- Division of Medical Sciences, University of Oxford, Oxford OX3 9DU, UK
| | - Peter E. M. Butler
- Charles Wolfson Centre for Reconstructive Surgery, Royal Free Hospital, London NW3 2QG, UK; (B.J.L.); (J.A.-H.); (P.E.M.B.)
- Department of Surgical Biotechnology, Division of Surgery & Interventional Science, University College London, London NW3 2QG, UK
- Department of Plastic Surgery, Royal Free Hospital, London NW3 2QG, UK
| |
Collapse
|
8
|
Winkelman MA, Dai G. Bioengineered perfused human brain microvascular networks enhance neural progenitor cell survival, neurogenesis, and maturation. SCIENCE ADVANCES 2023; 9:eaaz9499. [PMID: 37163593 PMCID: PMC10171804 DOI: 10.1126/sciadv.aaz9499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 04/10/2023] [Indexed: 05/12/2023]
Abstract
Neural progenitor cells (NPCs) have the capability to self-renew and differentiate into neurons and glial cells. In the adult brain, NPCs are found near brain microvascular networks (BMVNs) in specialized microenvironments called the neurovascular niche (NVN). Although several in vitro NVN models have been previously reported, most do not properly recapitulate the intimate cellular interactions between NPCs and perfused brain microvessels. Here, we developed perfused BMVNs composed of primary human brain endothelial cells, pericytes, and astrocytes within microfluidic devices. When induced pluripotent stem cell-derived NPCs were introduced into BMVNs, we found that NPC survival, neurogenesis, and maturation were enhanced. The application of flow during BMVN coculture was also beneficial for neuron differentiation. Collectively, our work highlighted the important role of BMVNs and flow in NPC self-renewal and neurogenesis, as well as demonstrated our model's potential to study the biological and physical interactions of human NVN in vitro.
Collapse
Affiliation(s)
- Max A. Winkelman
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | | |
Collapse
|
9
|
Li L, Li X, Han R, Wu M, Ma Y, Chen Y, Zhang H, Li Y. Therapeutic Potential of Chinese Medicine for Endogenous Neurogenesis: A Promising Candidate for Stroke Treatment. Pharmaceuticals (Basel) 2023; 16:ph16050706. [PMID: 37242489 DOI: 10.3390/ph16050706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 04/26/2023] [Accepted: 05/03/2023] [Indexed: 05/28/2023] Open
Abstract
Strokes are a leading cause of morbidity and mortality in adults worldwide. Extensive preclinical studies have shown that neural-stem-cell-based treatments have great therapeutic potential for stroke. Several studies have confirmed that the effective components of traditional Chinese medicine can protect and maintain the survival, proliferation, and differentiation of endogenous neural stem cells through different targets and mechanisms. Therefore, the use of Chinese medicines to activate and promote endogenous nerve regeneration and repair is a potential treatment option for stroke patients. Here, we summarize the current knowledge regarding neural stem cell strategies for ischemic strokes and the potential effects of these Chinese medicines on neuronal regeneration.
Collapse
Affiliation(s)
- Lin Li
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiao Li
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Rui Han
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Meirong Wu
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yaolei Ma
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yuzhao Chen
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Han Zhang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yue Li
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Ministry of Education, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| |
Collapse
|
10
|
Tanabe H, Suzuki T, Ohishi T, Isemura M, Nakamura Y, Unno K. Effects of Epigallocatechin-3-Gallate on Matrix Metalloproteinases in Terms of Its Anticancer Activity. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28020525. [PMID: 36677584 PMCID: PMC9862901 DOI: 10.3390/molecules28020525] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/29/2022] [Accepted: 12/31/2022] [Indexed: 01/06/2023]
Abstract
Epidemiological studies have shown that the consumption of green tea has beneficial effects against cancer. Basic studies have provided evidence that epigallocatechin gallate (EGCG) is a major contributor to these effects. Matrix metalloproteinases (MMPs) are zinc-dependent metalloproteinases with the ability to degrade the extracellular matrix proteins and are involved in various diseases including cancer in which MMPs have a critical role in invasion and metastasis. In this review, we discuss the effects of EGCG on several types of MMPs in the context of its anticancer activity. In the promoter region, MMPs have binding sites for at least one transcription factor of AP-1, Sp1, and NF-κB, and EGCG can downregulate these transcription factors through signaling pathways mediated by reactive oxygen species. EGCG can also decrease nuclear ERK, p38, heat shock protein-27 (Hsp27), and β-catenin levels, leading to suppression of MMPs' expression. Other mechanisms by which EGCG inhibits MMPs include direct binding to MMPs to prevent their activation and downregulation of NF-κB to suppress the production of inflammatory cytokines such as TNFα and IL-1β. Findings from studies on EGCG presented here may be useful in the development of more effective anti-MMP agents, which would give beneficial effects on cancer and other diseases.
Collapse
Affiliation(s)
- Hiroki Tanabe
- Faculty of Health and Welfare Science, Nayoro City University, Nayoro 096-8641, Hokkaido, Japan
- Correspondence: (H.T.); (T.O.)
| | - Takuji Suzuki
- Department of Food Science and Nutrition, Faculty of Human Life and Science, Doshisha Women’s College of Liberal Arts, Kyoto 602-0893, Japan
| | - Tomokazu Ohishi
- Institute of Microbial Chemistry (BIKAKEN), Numazu, Microbial Chemistry Research Foundation, Numazu 410-0301, Shizuoka, Japan
- Institute of Microbial Chemistry (BIKAKEN), Laboratory of Oncology, Microbial Chemistry Research Foundation, Shinagawa, Tokyo 141-0021, Japan
- Correspondence: (H.T.); (T.O.)
| | - Mamoru Isemura
- Tea Science Center, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan
| | - Yoriyuki Nakamura
- Tea Science Center, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan
| | - Keiko Unno
- Tea Science Center, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan
| |
Collapse
|
11
|
Denaro S, D’Aprile S, Alberghina C, Pavone AM, Torrisi F, Giallongo S, Longhitano L, Mannino G, Lo Furno D, Zappalà A, Giuffrida R, Tibullo D, Li Volti G, Vicario N, Parenti R. Neurotrophic and immunomodulatory effects of olfactory ensheathing cells as a strategy for neuroprotection and regeneration. Front Immunol 2022; 13:1098212. [PMID: 36601122 PMCID: PMC9806219 DOI: 10.3389/fimmu.2022.1098212] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Abstract
Accumulating evidence sustains glial cells as critical players during central nervous system (CNS) development, homeostasis and disease. Olfactory ensheathing cells (OECs), a type of specialized glia cells sharing properties with both Schwann cells and astrocytes, are of critical importance in physiological condition during olfactory system development, supporting its regenerative potential throughout the adult life. These characteristics prompted research in the field of cell-based therapy to test OEC grafts in damaged CNS. Neuroprotective mechanisms exerted by OEC grafts are not limited to axonal regeneration and cell differentiation. Indeed, OEC immunomodulatory properties and their phagocytic potential encourage OEC-based approaches for tissue regeneration in case of CNS injury. Herein we reviewed recent advances on the immune role of OECs, their ability to modulate CNS microenvironment via bystander effects and the potential of OECs as a cell-based strategy for tissue regeneration.
Collapse
Affiliation(s)
- Simona Denaro
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Simona D’Aprile
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Cristiana Alberghina
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Anna Maria Pavone
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Filippo Torrisi
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Sebastiano Giallongo
- Section of Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Lucia Longhitano
- Section of Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Giuliana Mannino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Debora Lo Furno
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Agata Zappalà
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Rosario Giuffrida
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Daniele Tibullo
- Section of Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Giovanni Li Volti
- Section of Biochemistry, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Nunzio Vicario
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy,*Correspondence: Nunzio Vicario, ; Rosalba Parenti,
| | - Rosalba Parenti
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy,*Correspondence: Nunzio Vicario, ; Rosalba Parenti,
| |
Collapse
|
12
|
Bang SJ, Lee J, Jeon GW, Jun YH. Erythropoietin Reduces Death and Neurodevelopmental Impairment in Neonatal Hypoxic-Ischemic Encephalopathy. NEONATAL MEDICINE 2022. [DOI: 10.5385/nm.2022.29.4.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Purpose: Erythropoietin (EPO) is a promising neuroprotective drug. We investigated whether EPO has beneficial effects on neurodevelopmental outcomes in infants with hypoxic-ischemic encephalopathy (HIE).Methods: We retrospectively reviewed the data of 56 infants with HIE born at or after 35 weeks of gestation who were admitted to Inha University Hospital between 2012 and 2021. Patients were divided into two groups based on EPO use and compared. In the EPO group, patients were administered 1,000 U/kg of EPO on days 1, 2, 3, 5, and 7, starting within 24 hours after birth. The primary outcome was death or neurodevelopmental impairment (NDI) at the age of 12 months.Results: EPO was administered to 38 infants, and 18 did not receive EPO. Only 37.5% of patients with HIE (21/56) and 60% of patients with moderate-to-severe HIE (21/35) received therapeutic hypothermia. Among all patients with HIE, death or NDI (21.1 % vs. 50.0%; odds ratio [OR], 0.09; 95% confidence interval [CI], 0.01 to 0.78; P=0.029) and brain injury on imaging (42.1% vs. 83.3%; OR, 0.16; 95% CI, 0.03 to 0.92; P=0.040) were significantly lower in the EPO group than in the control group. Among patients with moderate-to-severe HIE, brain injury on imaging (54.2% vs. 90.9%; OR, 0.04; 95% CI, 0.002 to 0.700; P=0.027) was significantly lower in the EPO group than in the control group.Conclusion: EPO administration significantly reduced mortality and NDI in infants with HIE. EPO can be considered an adjunctive therapeutic agent for neonatal HIE.
Collapse
|
13
|
RGS16 regulated by let-7c-5p promotes glioma progression by activating PI3K-AKT pathway. Front Med 2022; 17:143-155. [PMID: 36414916 DOI: 10.1007/s11684-022-0929-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 04/21/2022] [Indexed: 11/24/2022]
Abstract
Gliomas are the most common central nervous system tumours; they are highly aggressive and have a poor prognosis. RGS16 belongs to the regulator of G-protein signalling (RGS) protein family, which plays an important role in promoting various cancers, such as breast cancer, pancreatic cancer, and colorectal cancer. Moreover, previous studies confirmed that let-7c-5p, a well-known microRNA, can act as a tumour suppressor to regulate the progression of various tumours by inhibiting the expression of its target genes. However, whether RGS16 can promote the progression of glioma and whether it is regulated by miR let-7c-5p are still unknown. Here, we confirmed that RGS16 is upregulated in glioma tissues and that high expression of RGS16 is associated with poor survival. Ectopic deletion of RGS16 significantly suppressed glioma cell proliferation and migration both in vitro and in vivo. Moreover, RGS16 was validated as a direct target gene of miR let-7c-5p. The overexpression of miR let-7c-5p obviously downregulated the expression of RGS16, and knocking down miR let-7c-5p had the opposite effect. Thus, we suggest that the suppression of RGS16 by miR let-7c-5p can promote glioma progression and may serve as a potential prognostic biomarker and therapeutic target in glioma.
Collapse
|
14
|
Deng G, Fu TJ, Liu CP. Increased expression of Myosin X contributes to the metastasis in patients with laryngeal squamous cell carcinoma. Mol Genet Genomics 2022; 297:1529-1536. [PMID: 35951144 PMCID: PMC9596522 DOI: 10.1007/s00438-022-01934-x] [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: 11/01/2021] [Accepted: 07/23/2022] [Indexed: 11/06/2022]
Abstract
Laryngeal Squamous Cell Carcinoma (LSCC) is one of the most common malignancy in Head and neck cancer for which the mechanism underlying its metastasis is poorly understood. Myosin X, a molecular motor in cells has been demonstrated to play an important role in cell migration. However, whether Myosin X is involved in the metastasis of LSCC remains unclear. To investigate the expression of Myosin X and its implication in the metastasis of LSCC, we recruited 30 patients with LSCC and 6 patients with vocal cord polyp range from October 2016 to October 2018. Tissue samples were obtained during surgery and the expression of Myosin X, Cortactin, MMP2, MMP9, E-cadherin, and β-catenin in tissue samples were evaluated by RT-PCR, Western blot, immunohistochemistry or ELISA. Patients with LSCC were further followed-up 2 year after surgery for metastasis analysis. We found that the level of Myosin X, Cortactin, MMP2, and MMP9 was much higher in poorly differentiated LSCC compared to that in moderately and highly LSCC, as well as the control tissues. In contrast, the expression of epithelial-mesenchymal transition related marker, E-cadherin, and β-catenin, were much lower in poorly differentiated LSCC tissues compared to that in moderately and highly differentiated LSCC tissues, as well as the control tissues. Moreover, the expression of Myosin X was positively correlated with Cortactin, MMP2, and MMP9 levels. Increased expression of Myosin X in LSCC tissues was related to higher risk of metastasis. In conclusion, our findings showed that. Myosin X augments the expression of Cortactin, MMP2 and MMP9, which could upregulate the cell migration and the matrix degradation, and consequently reduce the expression of E-cadherin and β-catenin, thereby activating epithelial-mesenchymal transformation and promoting the metastasis of LSCC. Targeting Myosin X may have potential therapeutic effect in the metastasis of LSCC.
Collapse
Affiliation(s)
- Gang Deng
- Department of Otolaryngology-Head and Neck Surgery, Wuhan No.1 Hospital, Wuhan, People's Republic of China
| | - Tie-Jun Fu
- Department of Otolaryngology-Head and Neck Surgery, Shiyan Hospital of Integrated Traditional and Western Medicine, Shiyan, People's Republic of China
| | - Cui-Ping Liu
- Department of Otolaryngology-Head and Neck Surgery, Second People's Hospital of Gansu Province, No. 1 He Zheng West Street, Chengguan District, Lanzhou, 730000, Gansu, People's Republic of China.
| |
Collapse
|
15
|
Victor S, Rocha-Ferreira E, Rahim A, Hagberg H, Edwards D. New possibilities for neuroprotection in neonatal hypoxic-ischemic encephalopathy. Eur J Pediatr 2022; 181:875-887. [PMID: 34820702 PMCID: PMC8897336 DOI: 10.1007/s00431-021-04320-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/11/2021] [Accepted: 11/12/2021] [Indexed: 12/26/2022]
Abstract
Around 0.75 million babies worldwide suffer from moderate or severe hypoxic-ischemic encephalopathy (HIE) each year resulting in around 400,000 babies with neurodevelopmental impairment. In 2010, neonatal HIE was associated with 2.4% of the total Global Burden of Disease. Therapeutic hypothermia (TH), a treatment that is now standard of care in high-income countries, provides proof of concept that strategies that aim to improve neurodevelopment are not only possible but can also be implemented to clinical practice. While TH is beneficial, neonates with moderate or severe HIE treated with TH still experience devastating complications: 48% (range: 44-53) combined death or moderate/severe disability. There is a concern that TH may not be effective in low- and middle-income countries. Therapies that further improve outcomes are desperately needed, and in high-income countries, they must be tested in conjunction with TH. We have in this review focussed on pharmacological treatment options (e.g. erythropoietin, allopurinol, melatonin, cannabidiol, exendin-4/exenatide). Erythropoietin and allopurinol show promise and are progressing towards the clinic with ongoing definitive phase 3 randomised placebo-controlled trials. However, there remain global challenges for the next decade. Conclusion: There is a need for more optimal animal models, greater industry support/sponsorship, increased use of juvenile toxicology, dose-ranging studies with pharmacokinetic-pharmacodynamic modelling, and well-designed clinical trials to avoid exposure to harmful medications or abandoning putative treatments. What is Known: • Therapeutic hypothermia is beneficial in neonatal hypoxic-ischemic encephalopathy. • Neonates with moderate or severe hypoxic-ischemic encephalopathy treated with therapeutic hypothermia still experience severe sequelae. What is New: • Erythropoietin, allopurinol, melatonin, cannabidiol, and exendin-4/exenatide show promise in conjunction with therapeutic hypothermia. • There is a need for more optimal animal models, greater industry support/sponsorship, increased use of juvenile toxicology, dose-ranging studies with pharmacokinetic-pharmacodynamic modelling, and well-designed clinical trials.
Collapse
Affiliation(s)
- Suresh Victor
- Centre for the Developing Brain, Department of Perinatal Imaging and Health, School of Biomedical Engineering and Imaging Sciences, King’s College London, 1st Floor, South Wing, St Thomas’ Hospital, Westmister Bridge Road, London, UK
| | - Eridan Rocha-Ferreira
- Centre for Perinatal Medicine and Health, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ahad Rahim
- UCL School of Pharmacy, University College London, London, UK
| | - Henrik Hagberg
- Centre for Perinatal Medicine and Health, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - David Edwards
- Centre for the Developing Brain, Department of Perinatal Imaging and Health, School of Biomedical Engineering and Imaging Sciences, King’s College London, 1st Floor, South Wing, St Thomas’ Hospital, Westmister Bridge Road, London, UK
| |
Collapse
|
16
|
Zheng Z, Chen J, Chopp M. Mechanisms of Plasticity Remodeling and Recovery. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00011-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
17
|
Kaur D, Behl T, Sehgal A, Singh S, Sharma N, Badavath VN, Ul Hassan SS, Hasan MM, Bhatia S, Al-Harassi A, Khan H, Bungau S. Unravelling the potential neuroprotective facets of erythropoietin for the treatment of Alzheimer's disease. Metab Brain Dis 2022; 37:1-16. [PMID: 34436747 DOI: 10.1007/s11011-021-00820-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 08/09/2021] [Indexed: 02/06/2023]
Abstract
During the last three decades, recombinant DNA technology has produced a wide range of hematopoietic and neurotrophic growth factors, including erythropoietin (EPO), which has emerged as a promising protein drug in the treatment of several diseases. Cumulative studies have recently indicated the neuroprotective role of EPO in preclinical models of acute and chronic neurodegenerative disorders, including Alzheimer's disease (AD). AD is one of the most prevalent neurodegenerative illnesses in the elderly, characterized by the accumulation of extracellular amyloid-ß (Aß) plaques and intracellular neurofibrillary tangles (NFTs), which serve as the disease's two hallmarks. Unfortunately, AD lacks a successful treatment strategy due to its multifaceted and complex pathology. Various clinical studies, both in vitro and in vivo, have been conducted to identify the various mechanisms by which erythropoietin exerts its neuroprotective effects. The results of clinical trials in patients with AD are also promising. Herein, it is summarized and reviews all such studies demonstrating erythropoietin's potential therapeutic benefits as a pleiotropic neuroprotective agent in the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Dapinder Kaur
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Neelam Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | | | - Syed Shams Ul Hassan
- School of Medicine and Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Mohammad Mehedi Hasan
- Department of Biochemistry and Molecular Biology, Faculty of Life Science, Mawlana Bhashani Science and Technology University, Tangail, Bangladesh
| | - Saurabh Bhatia
- Natural & Medical Sciences Research Centre, University of Nizwa, Nizwa, Oman
- Amity Institute of Pharmacy, Amity University, Noida, Haryana, India
| | - Ahmed Al-Harassi
- Natural & Medical Sciences Research Centre, University of Nizwa, Nizwa, Oman
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
18
|
Jurcau A, Simion A. Neuroinflammation in Cerebral Ischemia and Ischemia/Reperfusion Injuries: From Pathophysiology to Therapeutic Strategies. Int J Mol Sci 2021; 23:14. [PMID: 35008440 PMCID: PMC8744548 DOI: 10.3390/ijms23010014] [Citation(s) in RCA: 175] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 12/18/2021] [Accepted: 12/18/2021] [Indexed: 02/07/2023] Open
Abstract
Its increasing incidence has led stroke to be the second leading cause of death worldwide. Despite significant advances in recanalization strategies, patients are still at risk for ischemia/reperfusion injuries in this pathophysiology, in which neuroinflammation is significantly involved. Research has shown that in the acute phase, neuroinflammatory cascades lead to apoptosis, disruption of the blood-brain barrier, cerebral edema, and hemorrhagic transformation, while in later stages, these pathways support tissue repair and functional recovery. The present review discusses the various cell types and the mechanisms through which neuroinflammation contributes to parenchymal injury and tissue repair, as well as therapeutic attempts made in vitro, in animal experiments, and in clinical trials which target neuroinflammation, highlighting future therapeutic perspectives.
Collapse
Affiliation(s)
- Anamaria Jurcau
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania;
- Neurology Ward, Clinical Municipal Hospital “dr. G. Curteanu” Oradea, 410154 Oradea, Romania
| | - Aurel Simion
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania;
- Neurorehabilitation Ward, Clinical Municipal Hospital “dr. G. Curteanu” Oradea, 410154 Oradea, Romania
| |
Collapse
|
19
|
Four Matrix Metalloproteinase genes involved in murine breast cancer affected by ginger extract. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2021.101332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
20
|
Liu C, Bi X, Fan H, Ma L, Ge RL. Microcyst fluid promotes the migration and invasion of fibroblasts in the adventitial layer of alveolar echinococcosis. Acta Trop 2021; 223:106084. [PMID: 34389327 DOI: 10.1016/j.actatropica.2021.106084] [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: 12/09/2020] [Revised: 07/31/2021] [Accepted: 08/01/2021] [Indexed: 11/24/2022]
Abstract
Alveolar echinococcosis (AE) caused by Echinococcus multilocularis (E. multilocularis), characterized by lesions composed of an aggregate of microcysts embedded in a granulomatous host's reaction. The periphery of parasite granulomas often additionally displays fibrotic reactions of varying intensity, in which E. multilocularis microenvironment fibroblasts (EMFs) laid down collagen. However, the regulation of EMFs by the infiltration of E. multilocularis microcyst fluid (MF) into granulomas remains poorly defined. This study aimed to investigate the effect of MF on migration and invasion of primary isolated EMFs cells. A mouse model of secondary infection with AE was established, and the model construction was evaluated by HE staining. EMFs were cultured in primary by tissue block adherency method. The isolated cells were identified by qPCR, immunofluorescence and Western blot. Then CCK-8 assay, cell migration/invasion assay and flow cytometry were performed to detect the effects of MF on the proliferation, migration, invasion and cell cycle of EMFs, respectively. The expressions of MMP2 and MMP9 at mRNA and protein levels in EMFs were detected by RT-qPCR and Western blot. The effect of PI3K-Akt signal transduction pathway on regulating the expression of MMPs expression was assessed by Western blot. As indicated from the results, EMFs were successfully isolated from the E. multilocularis microenvironment and identified as myofibroblasts. MF significantly facilitated the proliferation and cell cycle progression of EMFs. In addition, MF significantly improved the migration and invasion of EMFs. MF was further confirmed to up-regulate mRNA and protein expressions of MMP2 and MMP9 in EMFs, which was related to the activation of the PI3K-Akt signaling pathway. The present study demonstrates that MF can promote the migration and invasion of EMFs cells significantly, which might be via activating PI3K-Akt signaling pathway.
Collapse
|
21
|
Xiao F, Zhang X, Ni P, Yu H, Gao Q, Li M, Huo P, Wei Z, Wang S, Zhang Y, Zhao R, Li A, Li Z, Li Y, Cheng H, Du L, Ren S, Yu Q, Liu Y, Zhao Y. Voltage-dependent potassium channel Kv4.2 alleviates the ischemic stroke impairments through activating neurogenesis. Neurochem Int 2021; 150:105155. [PMID: 34384853 DOI: 10.1016/j.neuint.2021.105155] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 07/29/2021] [Accepted: 08/07/2021] [Indexed: 10/20/2022]
Abstract
As well as their ion transportation function, the voltage-dependent potassium channels could act as the cell signal inducer in a variety of pathogenic processes. However, their roles in neurogenesis after stroke insults have not been clearly illustrated. In our preliminary study, the expressions of voltage-dependent potassium channels Kv4.2 was significantly decreased after stroke in cortex, striatum and hippocampus by real-time quantitative PCR assay. To underlie the neuroprotection of Kv4.2 in stroke rehabilitation, recombinant plasmids encoding the cDNAs of mouse Kv4.2 was constructed. Behavioral tests showed that the increased Kv4.2 could be beneficial to the recovery of the sensory, the motor functions and the cognitive deficits after stroke. Temozolomide (TMZ), an inhibitor of neurogenesis, could partially abolish the mentioned protections of Kv4.2. The immunocytochemical staining showed that Kv4.2 could promote the proliferations of neural stem cells and induce the neural stem cells to differentiate into neurons in vitro and in vivo. And Kv4.2 could up-regulate the expressions of ERK1/2, p-ERK1/2, p-STAT3, NGF, p-TrkA, and BDNF, CAMKII and the concentration of intracellular Ca2+. Namely, we concluded that Kv4.2 promoted neurogenesis through ERK1/2/STAT3, NGF/TrkA, Ca2+/CAMKII signal pathways and rescued the ischemic impairments. Kv4.2 might be a potential drug target for ischemic stroke intervention.
Collapse
Affiliation(s)
- Fuyao Xiao
- Department of Pharmacology, School of Basic Medical Science, Capital Medical University, Beijing, PR China; Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, PR China
| | - Xiaojie Zhang
- State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, PR China
| | - Pinfei Ni
- Department of Pharmacology, School of Basic Medical Science, Capital Medical University, Beijing, PR China; Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, PR China
| | - Haibo Yu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China
| | - Qiming Gao
- Department of Pharmacology, School of Basic Medical Science, Capital Medical University, Beijing, PR China
| | - Mengyao Li
- Department of Pharmacology, School of Basic Medical Science, Capital Medical University, Beijing, PR China; Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, PR China
| | - Peiyun Huo
- Department of Pharmacology, School of Basic Medical Science, Capital Medical University, Beijing, PR China
| | - Ziwei Wei
- Department of Pharmacology, School of Basic Medical Science, Capital Medical University, Beijing, PR China; Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, PR China
| | - Sihan Wang
- Department of Pharmacology, School of Basic Medical Science, Capital Medical University, Beijing, PR China
| | - Yi Zhang
- Department of Pharmacology, School of Basic Medical Science, Capital Medical University, Beijing, PR China; Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, PR China
| | - Rui Zhao
- Department of Pharmacology, School of Basic Medical Science, Capital Medical University, Beijing, PR China; Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, PR China
| | - Aixue Li
- Department of Pharmacology, School of Basic Medical Science, Capital Medical University, Beijing, PR China; Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, PR China
| | - Zhirui Li
- Department of Pharmacology, School of Basic Medical Science, Capital Medical University, Beijing, PR China
| | - Yuejia Li
- Department of Pharmacology, School of Basic Medical Science, Capital Medical University, Beijing, PR China
| | - Haixiao Cheng
- Department of Pharmacology, School of Basic Medical Science, Capital Medical University, Beijing, PR China
| | - Libo Du
- State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, PR China
| | - Suping Ren
- Beijing Institute of Transfusion Medicine, Beijing, 100850, PR China
| | - Qun Yu
- Beijing Institute of Transfusion Medicine, Beijing, 100850, PR China
| | - Yang Liu
- State Key Laboratory for Structural Chemistry of Unstable and Stable Species, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, PR China
| | - Yuming Zhao
- Department of Pharmacology, School of Basic Medical Science, Capital Medical University, Beijing, PR China.
| |
Collapse
|
22
|
Makkiyah F, Sadewo W, Nurrizka R. Comparative Dose of Intracarotid Autologous Bone Marrow Mononuclear Therapy in Chronic Ischemic Stroke in Rats. Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.5675] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Research on chronic ischemic stroke is limited. One of the more promising approaches showing positive effects in the acute stage is mononuclear bone marrow cell therapy. This research may be the first which presents data about the optimum dose of bone marrow mononuclear cells (BM-MNCs) for chronic ischemic stroke in rats and discusses factors influencing recovery in the chronic stage.
We performed temporary middle cerebral artery occlusion (MCAO) procedures on the rats which were then randomly assigned to one of two experimental groups in which they were given either low or high doses of autologous BM-MNCs (5 million or 10 million cells per kg body weight).
Rat brains were fixed for HE, CD31, and doublecortin staining for analysis of the effects. Rat behavior was assessed weekly using the cylinder test and a modified neurological severity score (NSS) test.
In the four weeks prior to administration of BM-MNC, cylinder test scores improved to near normal, and NSS test scores improved moderately. The infarct zone decreased significantly (p <0,01), there was an improvement in angiogenesis (p = 0.1590) and a significant improvement in neurogenesis (p <0,01). Reduction of the infarct zone was associated with a higher dose whereas both higher and lower doses were found to have a similar effect on improving angiogenesis, and neurogenesis. Recovery was superior after twelve weeks compared with the recovery assessment at eight weeks.
In conclusion, a dose of 10 million cells was more effective than a dose of 5 million cells per kg body weight for reducing the infarct zone and ameliorating neurogenesis. There was an improvement of histopathological parameters associated with the longer infarct period.
Collapse
|
23
|
Winkelman MA, Koppes AN, Koppes RA, Dai G. Bioengineering the neurovascular niche to study the interaction of neural stem cells and endothelial cells. APL Bioeng 2021; 5:011507. [PMID: 33688617 PMCID: PMC7932757 DOI: 10.1063/5.0027211] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 02/15/2021] [Indexed: 12/13/2022] Open
Abstract
The ability of mammalian neural stem cells (NSCs) to self-renew and differentiate throughout adulthood has made them ideal to study neurogenesis and attractive candidates for neurodegenerative disease therapies. In the adult mammalian brain, NSCs are maintained in the neurovascular niche (NVN) where they are found near the specialized blood vessels, suggesting that brain endothelial cells (BECs) are prominent orchestrators of NSC fate. However, most of the current knowledge of the mammalian NVN has been deduced from nonhuman studies. To circumvent the challenges of in vivo studies, in vitro models have been developed to better understand the reciprocal cellular mechanisms of human NSCs and BECs. This review will cover the current understanding of mammalian NVN biology, the effects of endothelial cell-derived signals on NSC fate, and the in vitro models developed to study the interactions between NSCs and BECs.
Collapse
Affiliation(s)
- Max A Winkelman
- Department of Bioengineering, Northeastern University, Boston, Massachusetts 02115, USA
| | | | - Ryan A Koppes
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts 02115, USA
| | - Guohao Dai
- Department of Bioengineering, Northeastern University, Boston, Massachusetts 02115, USA
| |
Collapse
|
24
|
Yang J, Deng P, Qi Y, Feng X, Wen H, Chen F. MicroRNA-185 inhibits the proliferation and migration of HaCaT keratinocytes by targeting peroxisome proliferator-activated receptor β. Exp Ther Med 2021; 21:366. [PMID: 33732339 PMCID: PMC7903386 DOI: 10.3892/etm.2021.9797] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 12/12/2020] [Indexed: 11/06/2022] Open
Abstract
Proliferation and migration of keratinocytes are major processes of skin wound repair after injury. It has been indicated that microRNAs (miRNAs/miRs) are associated with the proliferation and migration of keratinocytes. However, the mechanism by which miR-185 affects these processes in keratinocytes remains unclear. In the present study, the expression level of miR-185 and peroxisome proliferator-activated receptor β (PPARβ) was examined by reverse transcription-quantitative PCR in HaCaT keratinocytes. Cell proliferation was evaluated using Cell Counting Kit-8 and colony formation assays. Western blot analysis was used to detect the levels of cell proliferation, migration and PI3K/AKT signaling pathway-associated proteins. In addition, the migratory capacity of the cells was determined using Transwell assay. The target gene of miR-185 was verified using dual-luciferase reporter assay. The results indicated that overexpression of miR-185 inhibited proliferation, migration and activation of the PI3K/AKT signaling pathway in HaCaT keratinocytes. PPARβ was indicated to be a target of miR-185 and its overexpression promoted the proliferation and migration of HaCaT keratinocytes, while its knockdown exhibited the adverse effects. Furthermore, PI3K inhibitor LY294002 inhibited activation of the PI3K/AKT signaling pathway and decreased the proliferation and migration of HaCaT keratinocytes. In addition, overexpressed PPARβ reversed the suppressive effects of miR-185 overexpression on proliferation, migration and activation of the PI3K/AKT signaling pathway. In conclusion, the results of the present study demonstrated that miR-185 suppressed activation of the PI3K/AKT signaling pathway via targeting PPARβ, thereby regulating proliferation and migration in HaCaT keratinocytes. The present study provided a novel theoretical basis for the use of miR-185 as a target in wound repair.
Collapse
Affiliation(s)
- Jingzhe Yang
- Department of Burn and Plastic Surgery, Affiliated Hospital of Chengde Medical University, South Wing Hospital of Chengde Medical College, Chengde, Hebei 067000, P.R. China
| | - Pingyang Deng
- Department of Burn and Plastic Surgery, Bayingol Mongolia Autonomous Prefecture People's Hospital, Urumqi, Xinjiang Uyghur Autonomous Region 841000, P.R. China
| | - Yonggang Qi
- Department of General Surgery, Bayingol Mongolia Autonomous Prefecture People's Hospital, Urumqi, Xinjiang Uyghur Autonomous Region 841000, P.R. China
| | - Xinshu Feng
- Department of Burn and Plastic Surgery, Affiliated Hospital of Chengde Medical University, South Wing Hospital of Chengde Medical College, Chengde, Hebei 067000, P.R. China
| | - Hailing Wen
- Department of Burn and Plastic Surgery, Affiliated Hospital of Chengde Medical University, South Wing Hospital of Chengde Medical College, Chengde, Hebei 067000, P.R. China
| | - Fengping Chen
- Department of Burn and Plastic Surgery, Affiliated Hospital of Chengde Medical University, South Wing Hospital of Chengde Medical College, Chengde, Hebei 067000, P.R. China
| |
Collapse
|
25
|
Rahman AA, Amruta N, Pinteaux E, Bix GJ. Neurogenesis After Stroke: A Therapeutic Perspective. Transl Stroke Res 2021; 12:1-14. [PMID: 32862401 PMCID: PMC7803692 DOI: 10.1007/s12975-020-00841-w] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/14/2020] [Accepted: 08/17/2020] [Indexed: 12/14/2022]
Abstract
Stroke is a major cause of death and disability worldwide. Yet therapeutic strategies available to treat stroke are very limited. There is an urgent need to develop novel therapeutics that can effectively facilitate functional recovery. The injury that results from stroke is known to induce neurogenesis in penumbra of the infarct region. There is considerable interest in harnessing this response for therapeutic purposes. This review summarizes what is currently known about stroke-induced neurogenesis and the factors that have been identified to regulate it. Additionally, some key studies in this field have been highlighted and their implications on future of stroke therapy have been discussed. There is a complex interplay between neuroinflammation and neurogenesis that dictates stroke outcome and possibly recovery. This highlights the need for a better understanding of the neuroinflammatory process and how it affects neurogenesis, as well as the need to identify new mechanisms and potential modulators. Neuroinflammatory processes and their impact on post-stroke repair have therefore also been discussed.
Collapse
Affiliation(s)
- Abir A Rahman
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, Room 1349, 131 S. Robertson, Ste 1300, New Orleans, LA, 70112, USA
| | - Narayanappa Amruta
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, Room 1349, 131 S. Robertson, Ste 1300, New Orleans, LA, 70112, USA
| | - Emmanuel Pinteaux
- Faculty of Biology, Medicine and Health, University of Manchester, A.V. Hill Building, Oxford Road, Manchester, M13 9PT, UK
| | - Gregory J Bix
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, Room 1349, 131 S. Robertson, Ste 1300, New Orleans, LA, 70112, USA.
- Tulane Brain Institute, Tulane University, New Orleans, LA, 70112, USA.
| |
Collapse
|
26
|
Kaur G, Poljak A, Ali SA, Zhong L, Raftery MJ, Sachdev P. Extending the Depth of Human Plasma Proteome Coverage Using Simple Fractionation Techniques. J Proteome Res 2021; 20:1261-1279. [PMID: 33471535 DOI: 10.1021/acs.jproteome.0c00670] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Human plasma is one of the most widely used tissues in clinical analysis, and plasma-based biomarkers are used for monitoring patient health status and/or response to medical treatment to avoid unnecessary invasive biopsy. Data-driven plasma proteomics has suffered from a lack of throughput and detection sensitivity, largely due to the complexity of the plasma proteome and in particular the enormous quantitative dynamic range, estimated to be between 9 and 13 orders of magnitude between the lowest and the highest abundance protein. A major challenge is to identify workflows that can achieve depth of plasma proteome coverage while minimizing the complexity of the sample workup and maximizing the sample throughput. In this study, we have performed intensive depletion of high-abundant plasma proteins or enrichment of low-abundant proteins using the Agilent multiple affinity removal liquid chromatography (LC) column-Human 6 (Hu6), the Agilent multiple affinity removal LC column-Human 14 (Hu14), and ProteoMiner followed by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS PAGE) and C18 prefractionation techniques. We compared the performance of each of these fractionation approaches to identify the method that satisfies requirements for analysis of clinical samples and to include good plasma proteome coverage in combination with reasonable sample output. In this study, we report that one-dimensional (1D) gel-based prefractionation allows parallel sample processing and no loss of proteome coverage, compared with serial chromatographic separation, and significantly accelerates analysis time, particularly important for large clinical projects. Furthermore, we show that a variety of methodologies can achieve similarly high plasma proteome coverage, allowing flexibility in method selection based on project-specific needs. These considerations are important in the effort to accelerate plasma proteomics research so as to provide efficient, reliable, and accurate diagnoses, population-based health screening, clinical research studies, and other clinical work.
Collapse
Affiliation(s)
- Gurjeet Kaur
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW 2052, Australia.,Bioanalytical Mass Spectrometry Facility, Mark Wainwright Analytical Centre, University of New South Wales, Wallace Wurth Building (C27), Sydney, NSW 2052, Australia
| | - Anne Poljak
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW 2052, Australia.,Bioanalytical Mass Spectrometry Facility, Mark Wainwright Analytical Centre, University of New South Wales, Wallace Wurth Building (C27), Sydney, NSW 2052, Australia
| | - Syed Azmal Ali
- Cell Biology and Proteomics Lab, National Dairy Research Institute, Karnal, Haryana 132001, India
| | - Ling Zhong
- Bioanalytical Mass Spectrometry Facility, Mark Wainwright Analytical Centre, University of New South Wales, Wallace Wurth Building (C27), Sydney, NSW 2052, Australia
| | - Mark J Raftery
- Bioanalytical Mass Spectrometry Facility, Mark Wainwright Analytical Centre, University of New South Wales, Wallace Wurth Building (C27), Sydney, NSW 2052, Australia
| | - Perminder Sachdev
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW 2052, Australia.,Neuropsychiatric Institute, Euroa Centre, Prince of Wales Hospital, Sydney, NSW 2052, Australia
| |
Collapse
|
27
|
Matta R, Yousafzai MS, Murrell M, Gonzalez AL. Endothelial cell secreted metalloproteinase-2 enhances neural stem cell N-cadherin expression, clustering, and migration. FASEB J 2021; 35:e21311. [PMID: 33417253 DOI: 10.1096/fj.202002302rr] [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: 10/12/2020] [Revised: 12/09/2020] [Accepted: 12/11/2020] [Indexed: 11/11/2022]
Abstract
Neuroblasts have a clustered phenotype critical for their unidirectional migration, which in part is dependent on signaling from microvascular endothelial cells (EC) and pericytes (PC). Diffusible signals secreted by vascular cells have been demonstrated to increase survival, proliferation, and differentiation of subventricular zone resident neural stem cells (NSC); however, the signals that promote the necessary initiating step of NSC clustering are undefined. To investigate the role of vascular cells in promoting NSC clustering and directing migration, we created a 3-D hydrogel that mimics the biomechanics, biochemistry, and architectural complexity of brain tissue. We demonstrate that EC, and not PC, have a crucial role in NSC clustering and migration, further verified through microfluidic chamber systems and traction force microscopy. Ablation of the extended NSC aggregate arm halts aggregate movement, suggesting that clustering is a prerequisite for migration. When cultured with EC, NSC clustering occurs and NSC coincidentally increase their expression of N-cadherin, as compared to NSC cultured alone. NSC-presented N-cadherin expression was increased following exposure to EC secreted metalloproteinase-2 (MMP2). We demonstrate that inhibition of MMP2 prevented NSC N-cadherin surface expression and subsequent NSC clustering, even when NSC were in direct contact with EC. Furthermore, with exogenous activation of EGFR, which serves as a downstream activator of N-cadherin cleavage, NSC form clusters. Our results suggest that EC secretion of MMP2 promotes NSC clustering through N-cadherin expression. The insight gained about the mechanisms by which EC promote NSC migration may enhance NSC therapeutic response to sites of injury.
Collapse
Affiliation(s)
- Rita Matta
- Biomedical Engineering Department, Yale University, New Haven, CT, USA
| | - Muhammad Sulaiman Yousafzai
- Biomedical Engineering Department, Yale University, New Haven, CT, USA.,Systems Biology Institute, Yale University, West Haven, CT, USA
| | - Michael Murrell
- Biomedical Engineering Department, Yale University, New Haven, CT, USA.,Systems Biology Institute, Yale University, West Haven, CT, USA.,Physics Department, Yale University, New Haven, CT, USA
| | | |
Collapse
|
28
|
Impaired capillary tube formation induced by elevated secretion of IL8 involves altered signaling via the CXCR1/PI3K/MMP2 pathway. Mol Biol Rep 2021; 48:601-610. [PMID: 33411234 DOI: 10.1007/s11033-020-06104-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 12/18/2020] [Indexed: 10/22/2022]
Abstract
Angiogenesis is a multistep process requiring endothelial cell activation, migration, proliferation and tube formation. We recently reported that elevated secretion of interlukin 8 (IL8) by myotubes (MT) from subjects with Type-2 Diabetes (T2D) reduced angiogenesis by human umbilical vein endothelial cells (HUVEC) and human skeletal muscle explants. This lower vascularization was mediated through impaired activation of the phosphatidylinositol 3-kinase (PI3K)-pathway. We sought to investigate additional signaling elements that might mediate reduced angiogenesis. HUVEC were exposed to levels of IL8 equal to those secreted by MT from non-diabetic (ND) and T2D subjects and the involvement of components in the angiogenic response pathway examined. Cellular content of reactive oxygen species and Nitrate secretion were similar after treatment with [ND-IL8] and [T2D-IL8]. CXCR1 protein was down-regulated after treatment with [T2D-IL8] (p < 0.01 vs [ND-IL8] treatment); CXCR2 expression was unaltered. Addition of neutralizing antibodies against CXCR1 and CXCR2 to HUVEC treated with IL8 confirmed that CXCR1 alone mediated the angiogenic response to IL8. A key modulator of angiogenesis is matrix metalloproteinase-2 (MMP2). MMP2 secretion was higher after treatment with [ND-IL8] vs [T2D-IL8] (p < 0.01). MMP2 inhibition reduced tube formation to greater extent with [ND-IL8] than with [T2D-IL8] (p < 0.005). The PI3K-pathway inhibitor LY294002 reduced IL8-induced MMP2 release. IL8 regulation of MMP2 release was CXCR1 dependent, as anti-CXCR1 significantly reduced MMP2 release (p < 0.05). These results suggest that high levels of IL8 secreted by T2D MT trigger reduced capillarization via lower activation of a CXCR1-PI3K pathway, followed by impaired release and activity of MMP2.
Collapse
|
29
|
Li Y, Hoffman MD, Benoit DSW. Matrix metalloproteinase (MMP)-degradable tissue engineered periosteum coordinates allograft healing via early stage recruitment and support of host neurovasculature. Biomaterials 2021; 268:120535. [PMID: 33271450 PMCID: PMC8110201 DOI: 10.1016/j.biomaterials.2020.120535] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 10/17/2020] [Accepted: 11/06/2020] [Indexed: 12/15/2022]
Abstract
Despite serving as the clinical "gold standard" treatment for critical size bone defects, decellularized allografts suffer from long-term failure rates of ~60% due to the absence of the periosteum. Stem and osteoprogenitor cells within the periosteum orchestrate autograft healing through host cell recruitment, which initiates the regenerative process. To emulate periosteum-mediated healing, tissue engineering approaches have been utilized with mixed outcomes. While vascularization has been widely established as critical for bone regeneration, innervation was recently identified to be spatiotemporally regulated together with vascularization and similarly indispensable to bone healing. Notwithstanding, there are no known approaches that have focused on periosteal matrix cues to coordinate host vessel and/or axon recruitment. Here, we investigated the influence of hydrogel degradation mechanism, i.e. hydrolytic or enzymatic (cell-dictated), on tissue engineered periosteum (TEP)-modified allograft healing, especially host vessel/nerve recruitment and integration. Matrix metalloproteinase (MMP)-degradable hydrogels supported endothelial cell migration from encapsulated spheroids whereas no migration was observed in hydrolytically degradable hydrogels in vitro, which correlated with increased neurovascularization in vivo. Specifically, ~2.45 and 1.84-fold, and ~3.48 and 2.58-fold greater vessel and nerve densities with high levels of vessel and nerve co-localization was observed using MMP degradable TEP (MMP-TEP) -modified allografts versus unmodified and hydrolytically degradable TEP (Hydro-TEP)-modified allografts, respectively, at 3 weeks post-surgery. MMP-TEP-modified allografts exhibited greater longitudinal graft-localized vascularization and endochondral ossification, along with 4-fold and 2-fold greater maximum torques versus unmodified and Hydro-TEP-modified allografts after 9 weeks, respectively, which was comparable to that of autografts. In summary, our results demonstrated that the MMP-TEP coordinated allograft healing via early stage recruitment and support of host neurovasculature.
Collapse
Affiliation(s)
- Yiming Li
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA; Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.
| | - Michael D Hoffman
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA; Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.
| | - Danielle S W Benoit
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA; Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA; Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA; Materials Science Program, University of Rochester, Rochester, NY, USA; Department of Chemical Engineering, University of Rochester, Rochester, NY, USA; Department of Biomedical Genetics and Center for Oral Biology, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
30
|
Ding X, Yan Y, Zhang C, Xu X, Yang F, Liu Y, Wang G, Qin Y. OCT4 regulated neointimal formation in injured mouse arteries by matrix metalloproteinase 2-mediated smooth muscle cells proliferation and migration. J Cell Physiol 2020; 236:5421-5431. [PMID: 33372301 DOI: 10.1002/jcp.30248] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 11/09/2022]
Abstract
The excessive proliferation and migration of vascular smooth muscle cells (VSMCs) play vital roles in neointimal hyperplasia and vascular restenosis. In the present study, we aimed to investigate the function and mechanism of octamer-binding transcription factor 4 (OCT4, a key transcription factor for maintaining stem cells in de-differentiated state) on neointima formation in response to vascular injury. Quantitative reverse-transcription polymerase chain reaction and western blot results displayed a significant increase of OCT4 levels in injured carotid arteries. Immunohistochemistry and immunofluorescence assays confirmed that the increased OCT4 expression was primarily localized in α-SMA-positive VSMCs from neointima, and colocalized with PCNA in the nuclei of VSMCs. Adenovirus-mediated OCT4 overexpression in injured carotid arteries exacerbated intimal thickening, while OCT4 knockdown significantly inhibited intimal thickening. In-vitro experiments confirmed that the increased OCT4 expression in VMSCs could be induced by platelet-derived growth factor-BB (PDGF-BB) in a time-dependent manner. Overexpression of OCT4 greatly promoted VSMCs proliferation and migration, while OCT4 knockdown significantly retarded the PDGF-BB-induced excessive proliferation and migration of VSMCs. Bioinformatics analysis, dual-luciferase reporter assay, and chromatin immunoprecipitation assay confirmed that OCT4 could upregulate matrix metalloproteinases 2 (MMP2) expression through promoting its transcription. Moreover, knockdown of MMP2 significantly attenuated OCT4-mediated VSMCs proliferation and migration. These results indicated that OCT4 facilitated neointimal formation in response to vascular injury by MMP2-mediated VSMCs proliferation and migration, and targeting OCT4 in VSMCs might be a novel therapeutic strategy for vascular restenosis.
Collapse
Affiliation(s)
- Xueyan Ding
- Department of Cardiology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China.,Department of Cardiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yan Yan
- Department of Cardiovascular Surgery, Institute of Cardiac Surgery, Changhai Hospital, Naval Medical University, Shanghai, China.,Department of Cardiothoracic Surgery, No. 903 Hospital of Chinese People's Liberation Army, Hangzhou, Zhejiang, China
| | - Chengke Zhang
- Department of Cardiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xudong Xu
- Department of Cardiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Fan Yang
- Department of Cardiovascular Surgery, Institute of Cardiac Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yang Liu
- Department of Cardiovascular Surgery, Institute of Cardiac Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Guokun Wang
- Department of Cardiovascular Surgery, Institute of Cardiac Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yongwen Qin
- Department of Cardiology, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
31
|
Pang R, Avdic-Belltheus A, Meehan C, Martinello K, Mutshiya T, Yang Q, Sokolska M, Torrealdea F, Hristova M, Bainbridge A, Golay X, Juul SE, Robertson NJ. Melatonin and/or erythropoietin combined with hypothermia in a piglet model of perinatal asphyxia. Brain Commun 2020; 3:fcaa211. [PMID: 33604569 PMCID: PMC7876304 DOI: 10.1093/braincomms/fcaa211] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/06/2020] [Accepted: 11/10/2020] [Indexed: 12/11/2022] Open
Abstract
As therapeutic hypothermia is only partially protective for neonatal encephalopathy, safe and effective adjunct therapies are urgently needed. Melatonin and erythropoietin show promise as safe and effective neuroprotective therapies. We hypothesized that melatonin and erythropoietin individually augment 12-h hypothermia (double therapies) and hypothermia + melatonin + erythropoietin (triple therapy) leads to optimal brain protection. Following carotid artery occlusion and hypoxia, 49 male piglets (<48 h old) were randomized to: (i) hypothermia + vehicle (n = 12), (ii) hypothermia + melatonin (20 mg/kg over 2 h) (n = 12), (iii) hypothermia + erythropoietin (3000 U/kg bolus) (n = 13) or (iv) tripletherapy (n = 12). Melatonin, erythropoietin or vehicle were given at 1, 24 and 48 h after hypoxia–ischaemia. Hypoxia–ischaemia severity was similar across groups. Therapeutic levels were achieved 3 hours after hypoxia–ischaemia for melatonin (15–30 mg/l) and within 30 min of erythropoietin administration (maximum concentration 10 000 mU/ml). Compared to hypothermia + vehicle, we observed faster amplitude-integrated EEG recovery from 25 to 30 h with hypothermia + melatonin (P = 0.02) and hypothermia + erythropoietin (P = 0.033) and from 55 to 60 h with tripletherapy (P = 0.042). Magnetic resonance spectroscopy lactate/N-acetyl aspartate peak ratio was lower at 66 h in hypothermia + melatonin (P = 0.012) and tripletherapy (P = 0.032). With hypothermia + melatonin, terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labelled-positive cells were reduced in sensorimotor cortex (P = 0.017) and oligodendrocyte transcription factor 2 labelled-positive counts increased in hippocampus (P = 0.014) and periventricular white matter (P = 0.039). There was no reduction in terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labelled-positive cells with hypothermia + erythropoietin, but increased oligodendrocyte transcription factor 2 labelled-positive cells in 5 of 8 brain regions (P < 0.05). Overall, melatonin and erythropoietin were safe and effective adjunct therapies to hypothermia. Hypothermia + melatonin double therapy led to faster amplitude-integrated EEG recovery, amelioration of lactate/N-acetyl aspartate rise and reduction in terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labelled-positive cells in the sensorimotor cortex. Hypothermia + erythropoietin doubletherapy was in association with EEG recovery and was most effective in promoting oligodendrocyte survival. Tripletherapy provided no added benefit over the double therapies in this 72-h study. Melatonin and erythropoietin influenced cell death and oligodendrocyte survival differently, reflecting distinct neuroprotective mechanisms which may become more visible with longer-term studies. Staggering the administration of therapies with early melatonin and later erythropoietin (after hypothermia) may provide better protection; each therapy has complementary actions which may be time critical during the neurotoxic cascade after hypoxia–ischaemia.
Collapse
Affiliation(s)
- Raymand Pang
- Department of Neonatology, Institute for Women's Health, University College London, London, UK
| | - Adnan Avdic-Belltheus
- Department of Neonatology, Institute for Women's Health, University College London, London, UK
| | - Christopher Meehan
- Department of Neonatology, Institute for Women's Health, University College London, London, UK
| | - Kathryn Martinello
- Department of Neonatology, Institute for Women's Health, University College London, London, UK
| | - Tatenda Mutshiya
- Department of Neonatology, Institute for Women's Health, University College London, London, UK
| | - Qin Yang
- Department of Neonatology, Institute for Women's Health, University College London, London, UK
| | - Magdalena Sokolska
- Department of Medical Physics and Biomedical Engineering, University College London Hospitals, London, UK
| | - Francisco Torrealdea
- Department of Medical Physics and Biomedical Engineering, University College London Hospitals, London, UK
| | - Mariya Hristova
- Department of Neonatology, Institute for Women's Health, University College London, London, UK
| | - Alan Bainbridge
- Department of Medical Physics and Biomedical Engineering, University College London Hospitals, London, UK
| | - Xavier Golay
- Department of Brain Repair and Rehabilitation, Institute of Neurology, Queen's Square, University College London, London, UK
| | - Sandra E Juul
- Department of Pediatrics, University of Washington, Seattle, Washington, DC, USA
| | - Nicola J Robertson
- Department of Neonatology, Institute for Women's Health, University College London, London, UK
| |
Collapse
|
32
|
Bongaarts A, de Jong JM, Broekaart DWM, van Scheppingen J, Anink JJ, Mijnsbergen C, Jansen FE, Spliet WGM, den Dunnen WFA, Gruber VE, Scholl T, Hainfellner JA, Feucht M, Borkowska J, Kotulska K, Jozwiak S, Grajkowska W, Buccoliero AM, Caporalini C, Giordano F, Genitori L, Scicluna BP, Schouten-van Meeteren AYN, van Vliet EA, Mühlebner A, Mills JD, Aronica E. Dysregulation of the MMP/TIMP Proteolytic System in Subependymal Giant Cell Astrocytomas in Patients With Tuberous Sclerosis Complex: Modulation of MMP by MicroRNA-320d In Vitro. J Neuropathol Exp Neurol 2020; 79:777-790. [PMID: 32472129 PMCID: PMC7304985 DOI: 10.1093/jnen/nlaa040] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/11/2020] [Accepted: 04/26/2020] [Indexed: 12/15/2022] Open
Abstract
Tuberous sclerosis complex (TSC), a rare genetic disorder caused by a mutation in the TSC1 or TSC2 gene, is characterized by the growth of hamartomas in several organs. This includes the growth of low-grade brain tumors, known as subependymal giant cell astrocytomas (SEGA). Previous studies have shown differential expression of genes related to the extracellular matrix in SEGA. Matrix metalloproteinases (MMPs), and their tissue inhibitors (TIMPs) are responsible for remodeling the extracellular matrix and are associated with tumorigenesis. This study aimed to investigate the MMP/TIMP proteolytic system in SEGA and the regulation of MMPs by microRNAs, which are important post-transcriptional regulators of gene expression. We investigated the expression of MMPs and TIMPs using previously produced RNA-Sequencing data, real-time quantitative PCR and immunohistochemistry in TSC-SEGA samples and controls. We found altered expression of several MMPs and TIMPs in SEGA compared to controls. We identified the lowly expressed miR-320d in SEGA as a potential regulator of MMPs, which can decrease MMP2 expression in human fetal astrocyte cultures. This study provides evidence of a dysregulated MMP/TIMP proteolytic system in SEGA of which MMP2 could be rescued by microRNA-320d. Therefore, further elucidating microRNA-mediated MMP regulation may provide insights into SEGA pathogenesis and identify novel therapeutic targets.
Collapse
Affiliation(s)
- Anika Bongaarts
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Jody M de Jong
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Diede W M Broekaart
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Jackelien van Scheppingen
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Jasper J Anink
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Caroline Mijnsbergen
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Floor E Jansen
- Department of Pediatric Neurology, University Medical Center Utrecht Brain Center, Utrecht, The Netherlands
| | - Wim G M Spliet
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands (WGMS); Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands (WFAdD)
| | | | - Victoria E Gruber
- Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Theresa Scholl
- Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | | | - Martha Feucht
- Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Julita Borkowska
- Department of Neurology and Epileptology, Children's Memorial Health Institute, Warsaw, Poland
| | - Katarzyna Kotulska
- Department of Neurology and Epileptology, Children's Memorial Health Institute, Warsaw, Poland
| | - Sergiusz Jozwiak
- Department of Neurology and Epileptology, Children's Memorial Health Institute, Warsaw, Poland.,Department of Child Neurology, Medical University of Warsaw, Warsaw, Poland
| | - Wieslawa Grajkowska
- Department of Pathology, Children's Memorial Health Institute, Warsaw, Poland
| | | | | | - Flavio Giordano
- Department of Neurosurgery, Anna Meyer Children's Hospital, Florence, Italy
| | - Lorenzo Genitori
- Department of Neurosurgery, Anna Meyer Children's Hospital, Florence, Italy
| | - Brendon P Scicluna
- Department of Clinical Epidemiology, Biostatistics & Bioinformatics, Center for Experimental & Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam
| | - Antoinette Y N Schouten-van Meeteren
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.,Department of Pediatric Oncology, Emma Children's Hospital, Amsterdam UMC, Amsterdam, The Netherlands
| | - Erwin A van Vliet
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands.,Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
| | - Angelika Mühlebner
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - James D Mills
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands.,Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, The Netherlands
| |
Collapse
|
33
|
Zinc in the Brain: Friend or Foe? Int J Mol Sci 2020; 21:ijms21238941. [PMID: 33255662 PMCID: PMC7728061 DOI: 10.3390/ijms21238941] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/22/2020] [Accepted: 11/24/2020] [Indexed: 12/12/2022] Open
Abstract
Zinc is a trace metal ion in the central nervous system that plays important biological roles, such as in catalysis, structure, and regulation. It contributes to antioxidant function and the proper functioning of the immune system. In view of these characteristics of zinc, it plays an important role in neurophysiology, which leads to cell growth and cell proliferation. However, after brain disease, excessively released and accumulated zinc ions cause neurotoxic damage to postsynaptic neurons. On the other hand, zinc deficiency induces degeneration and cognitive decline disorders, such as increased neuronal death and decreased learning and memory. Given the importance of balance in this context, zinc is a biological component that plays an important physiological role in the central nervous system, but a pathophysiological role in major neurological disorders. In this review, we focus on the multiple roles of zinc in the brain.
Collapse
|
34
|
Zhang B, Zhang J, Xia L, Luo J, Zhang L, Xu Y, Zhu X, Chen G. Inhibition of CDC42 reduces macrophage recruitment and suppresses lung tumorigenesis in vivo. J Recept Signal Transduct Res 2020; 41:504-510. [PMID: 32998602 DOI: 10.1080/10799893.2020.1828916] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
BACKGROUND Cell division control (CDC) 42 has been involved in the regulation of diverse cancers. Macrophage recruitment plays an important role in the pathogenesis and development of tumor. However, it remains unclear whether CDC42 contributes to macrophage recruitment and lung tumorigenesis in vivo. METHODS Small interference RNA (siRNA) was used to knock down CDC42 in the Lewis lung carcinoma (LLC)1. The invasion capability of CDC42 knockdown LLC1 cells was evaluated. LLC1 cells with CDC42 targeted small hairpin RNA (shRNA) were inoculated into C57BL/6 mice to establish the tumor-bearing animal model Tumor size and metastasis related proteins were measured. In addition, the invasion of macrophages in the tumor site as well as macrophage chemokine were also determined in the model. RESULTS The capacity of invasion and metastasis of LLC1 cells significantly decreased when CDC42 was knocked down. When inoculated with CDC42 knockdown LLC1 cells in vivo, the tumor size and metastasis related proteins levels both decreased. The invasion capacity of macrophages and the associated macrophage chemokine were also significantly down-regulated. CONCLUSION Our data suggest that the inhibition of CDC42 expression in lung cancer cells can significantly prevent the pathogenesis and development of tumor in an allograft tumor model in vivo, which might provide a novel therapeutic target and potential strategy for lung cancer treatment in the future.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Thoracic Surgery, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, China
| | - Jian Zhang
- Department of Thoracic Surgery, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, China
| | - Lilong Xia
- Department of Thoracic Surgery, Zhejiang Hospital, Hangzhou, China
| | - Jing Luo
- Department of Thoracic Surgery, Zhejiang Hospital, Hangzhou, China
| | - Lei Zhang
- Department of Thoracic Surgery, Zhejiang Hospital, Hangzhou, China
| | - Yanhui Xu
- Department of Thoracic Surgery, Zhejiang Hospital, Hangzhou, China
| | - Xinhai Zhu
- Department of Thoracic Surgery, Zhejiang Hospital, Hangzhou, China
| | - Guoping Chen
- Department of Thoracic Surgery, Zhejiang Hospital, Hangzhou, China
| |
Collapse
|
35
|
Samson F, He W, Sripathi SR, Patrick AT, Madu J, Chung H, Frost MC, Jee D, Gutsaeva DR, Jahng WJ. Dual Switch Mechanism of Erythropoietin as an Antiapoptotic and Pro-Angiogenic Determinant in the Retina. ACS OMEGA 2020; 5:21113-21126. [PMID: 32875248 PMCID: PMC7450639 DOI: 10.1021/acsomega.0c02763] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 07/28/2020] [Indexed: 05/07/2023]
Abstract
Constant or intense light degenerates the retina and retinal pigment epithelial cells. Light generates reactive oxygen species and nitric oxide leading to initial reactions of retinal degeneration. Apoptosis is the primary mechanism of abnormal death of photoreceptors, retinal ganglion cells, or retinal pigment epithelium (RPE) in degenerative retinal diseases, including diabetic retinopathy and age-related macular degeneration. The current study evaluated the function of erythropoietin (EPO) on angiogenesis and apoptosis in the retina and RPE under oxidative stress. We determined the pro-angiogenic and antiapoptotic mechanism of EPO under stress conditions using a conditional EPO knockdown model using siRNA, EPO addition, proteomics, immunocytochemistry, and bioinformatic analysis. Our studies verified that EPO protected retinal cells from light-, hypoxia-, hyperoxia-, and hydrogen peroxide-induced apoptosis through caspase inhibition, whereas up-regulated angiogenic reactions through vascular endothelial growth factor (VEGF) and angiotensin pathway. We demonstrated that the EPO expression in the retina and subsequent serine/threonine/tyrosine kinase phosphorylations might be linked to oxidative stress response tightly to determining angiogenesis and apoptosis. Neuroprotective roles of EPO may involve the balance between antiapoptotic and pro-angiogenic signaling molecules, including BCL-xL, c-FOS, caspase-3, nitric oxide, angiotensin, and VEGF receptor. Our data indicate a new therapeutic application of EPO toward retinal degeneration based on the dual roles in apoptosis and angiogenesis at the molecular level under oxidative stress.
Collapse
Affiliation(s)
| | - Weilue He
- Department
of Biomedical Engineering, Michigan Technological
University, Houghton 49931, United States
| | - Srinivas R. Sripathi
- Department
of Ophthalmology, Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Ambrose Teru Patrick
- Department
of Petroleum Chemistry, American University
of Nigeria, Yola 640101, Nigeria
| | - Joshua Madu
- Department
of Petroleum Chemistry, American University
of Nigeria, Yola 640101, Nigeria
| | - Hyewon Chung
- Department
of Ophthalmology, School of Medicine, Konkuk
University, Seoul 05030, Korea
| | - Megan C. Frost
- Department
of Biomedical Engineering, Michigan Technological
University, Houghton 49931, United States
| | - Donghyun Jee
- Division
of Vitreous and Retina, Department of Ophthalmology, St. Vincent’s
Hospital, College of Medicine, The Catholic
University of Korea, Suwon 16247, Korea
| | - Diana R. Gutsaeva
- Department
of Ophthalmology, Augusta University, Augusta, Georgia 30912, United States
| | - Wan Jin Jahng
- Department
of Petroleum Chemistry, American University
of Nigeria, Yola 640101, Nigeria
| |
Collapse
|
36
|
Rouleau N, Murugan NJ, Rusk W, Koester C, Kaplan DL. Matrix Deformation with Ectopic Cells Induced by Rotational Motion in Bioengineered Neural Tissues. Ann Biomed Eng 2020; 48:2192-2203. [PMID: 32671625 PMCID: PMC7405955 DOI: 10.1007/s10439-020-02561-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 06/29/2020] [Indexed: 11/30/2022]
Abstract
The brain's extracellular matrix (ECM) is a dynamic protein-based scaffold within which neural networks can form, self-maintain, and re-model. When the brain incurs injuries, microscopic tissue tears and active ECM re-modelling give way to abnormal brain structure and function including the presence of ectopic cells. Post-mortem and neuroimaging data suggest that the brains of jet pilots and astronauts, who are exposed to rotational forces, accelerations, and microgravity, display brain anomalies which could be indicative of a mechanodisruptive pathology. Here we present a model of non-impact-based brain injury induced by matrix deformation following mechanical shaking. Using a bioengineered 3D neural tissue platform, we designed a repetitive shaking paradigm to simulate subtle rotational acceleration. Our results indicate shaking induced ectopic cell clustering that could be inhibited by physically restraining tissue movement. Imaging revealed that the collagen substrate surrounding cells was deformed following shaking. Applied to neonatal rat brains, shaking induced deformation of extracellular spaces within the cerebral cortices and reduced the number of cell bodies at higher accelerations. We hypothesize that ECM deformation may represent a more significant role in brain injury progression than previously assumed and that the present model system contributes to its understanding as a phenomenon.
Collapse
Affiliation(s)
- Nicolas Rouleau
- Department of Biomedical Engineering, Science & Technology Center, Tufts University, Medford, MA, USA
- Initiative for Neural Science, Disease, and Engineering (INSciDE), Tufts University, Medford, USA
- The Allen Discovery Center, Tufts University, Medford, USA
| | - Nirosha J Murugan
- Department of Biomedical Engineering, Science & Technology Center, Tufts University, Medford, MA, USA
- The Allen Discovery Center, Tufts University, Medford, USA
- Department of Biology, Tufts University, Medford, USA
| | - William Rusk
- Department of Biomedical Engineering, Science & Technology Center, Tufts University, Medford, MA, USA
| | - Cole Koester
- Department of Biomedical Engineering, Science & Technology Center, Tufts University, Medford, MA, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Science & Technology Center, Tufts University, Medford, MA, USA.
- Initiative for Neural Science, Disease, and Engineering (INSciDE), Tufts University, Medford, USA.
- The Allen Discovery Center, Tufts University, Medford, USA.
| |
Collapse
|
37
|
Ulbrich P, Khoshneviszadeh M, Jandke S, Schreiber S, Dityatev A. Interplay between perivascular and perineuronal extracellular matrix remodelling in neurological and psychiatric diseases. Eur J Neurosci 2020; 53:3811-3830. [DOI: 10.1111/ejn.14887] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 05/29/2020] [Accepted: 06/18/2020] [Indexed: 12/31/2022]
Affiliation(s)
- Philipp Ulbrich
- German Center for Neurodegenerative Diseases (DZNE) Magdeburg Germany
- Department of Neurology Otto‐von‐Guericke University Magdeburg Germany
| | - Mahsima Khoshneviszadeh
- German Center for Neurodegenerative Diseases (DZNE) Magdeburg Germany
- Department of Neurology Otto‐von‐Guericke University Magdeburg Germany
| | - Solveig Jandke
- German Center for Neurodegenerative Diseases (DZNE) Magdeburg Germany
- Department of Neurology Otto‐von‐Guericke University Magdeburg Germany
| | - Stefanie Schreiber
- German Center for Neurodegenerative Diseases (DZNE) Magdeburg Germany
- Department of Neurology Otto‐von‐Guericke University Magdeburg Germany
- Center for Behavioral Brain Sciences (CBBS) Magdeburg Germany
| | - Alexander Dityatev
- German Center for Neurodegenerative Diseases (DZNE) Magdeburg Germany
- Center for Behavioral Brain Sciences (CBBS) Magdeburg Germany
- Medical Faculty Otto‐von‐Guericke University Magdeburg Germany
| |
Collapse
|
38
|
Dey S, Cui Z, Gavrilova O, Zhang X, Gassmann M, Noguchi CT. Sex-specific brain erythropoietin regulation of mouse metabolism and hypothalamic inflammation. JCI Insight 2020; 5:134061. [PMID: 32078583 DOI: 10.1172/jci.insight.134061] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 02/12/2020] [Indexed: 12/17/2022] Open
Abstract
The blood hormone erythropoietin (EPO), upon binding to its receptor (EpoR), modulates high-fat diet-induced (HFD-induced) obesity in mice, improves glucose tolerance, and prevents white adipose tissue inflammation. Transgenic mice with constitutive overexpression of human EPO solely in the brain (Tg21) were used to assess the neuroendocrine EPO effect without increasing the hematocrit. Male Tg21 mice resisted HFD-induced weight gain; showed lower serum adrenocorticotropic hormone, corticosterone, and C-reactive protein levels; and prevented myeloid cell recruitment to the hypothalamus compared with WT male mice. HFD-induced hypothalamic inflammation (HI) and microglial activation were higher in male mice, and Tg21 male mice exhibited a lower increase in HI than WT male mice. Physiological EPO function in the brain also showed sexual dimorphism in regulating HFD response. Female estrogen production blocked reduced weight gain and HI. Targeted deletion of EpoR gene expression in neuronal cells worsened HFD-induced glucose intolerance in both male and female mice but increased weight gain and HI in the hypothalamus in male mice only. Both male and female Tg21 mice kept on normal chow and HFD showed significantly improved glycemic control. Our data indicate that cerebral EPO regulates weight gain and HI in a sex-dependent response, distinct from EPO regulation of glycemic control, and independent of erythropoietic EPO response.
Collapse
Affiliation(s)
| | | | | | - Xiaojie Zhang
- Laboratory of Animal Sciences Section, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Max Gassmann
- Institute of Veterinary Physiology and Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.,Universidad Peruana Cayetano Heredia, Lima, Peru
| | | |
Collapse
|
39
|
Meng NN, Zhang RR, Liu C, Wang Q, Wang XK, Guo X, Wang PP, Sun JY. PDB-1 from Potentilla discolor Bunge suppresses lung cancer cell migration and invasion via FAK/Src and MAPK signaling pathways. Med Chem Res 2020. [DOI: 10.1007/s00044-020-02527-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
40
|
Zhang Y, Weng Q, Han J, Chen J. Alantolactone suppresses human osteosarcoma through the PI3K/AKT signaling pathway. Mol Med Rep 2019; 21:675-684. [PMID: 31974628 PMCID: PMC6947914 DOI: 10.3892/mmr.2019.10882] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 11/18/2019] [Indexed: 12/24/2022] Open
Abstract
Osteosarcoma is the most common type of malignant bone cancer and results in cancer-related deaths among adolescents. Alantolactone (ALT) demonstrates antitumor properties in various diseases; however, its potential role in osteosarcoma is relatively unclear. The aim of the present study was to evaluate the effect of ALT on osteosarcoma. ALT significantly decreased the viability of U2OS and HOS osteosarcoma cell lines. Cells flow cytometry assay and Hoechst 33258 staining assay revealed that ALT significantly increased the proportion of apoptotic U2OS cells. In addition, wound healing and Transwell invasion assays demonstrated that the invasion and migration of osteosarcoma were markedly reduced upon ALT treatment. It was hypothesized that the antitumor functions of ALT are mediated through inhibition of the PI3K/AKT signaling pathway. In conclusion, the results of the present study confirmed the inhibition of ALT on osteosarcoma cells via downregulation of PI3K/AKT signaling pathways, suggesting ALT as a potential therapeutic candidate for osteosarcoma.
Collapse
Affiliation(s)
- Yong Zhang
- Department of Trauma Orthopedics, Ningbo No. 6 Hospital, Ningbo, Zhejiang 315000, P.R. China
| | - Qiuyan Weng
- Department of Neurology, The Affiliated Hospital of Medical School of Ningbo University, Ningbo, Zhejiang 315000, P.R. China
| | - Jinming Han
- Department of Spinal Surgery, Ningbo No. 6 Hospital, Ningbo, Zhejiang 315000, P.R. China
| | - Jianming Chen
- Department of Trauma Orthopedics, Ningbo No. 6 Hospital, Ningbo, Zhejiang 315000, P.R. China
| |
Collapse
|
41
|
Chiu H, Lee H, Lee K, Zhao Y, Hsu CY, Shyu W. Mechanisms of ischaemic neural progenitor proliferation: a regulatory role of the HIF‐1α‐CBX7 pathway. Neuropathol Appl Neurobiol 2019; 46:391-405. [PMID: 31630421 DOI: 10.1111/nan.12585] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 10/08/2019] [Indexed: 01/14/2023]
Affiliation(s)
- H.‐Y. Chiu
- Children’s Hospital China Medical University and Hospital Taichung Taiwan
| | - H.‐T. Lee
- Department of Neurosurgery Taichung Veterans General Hospital Taichung Taiwan
- Graduate Institute of Medical Sciences National Defense Medical Center Taipei Taiwan
| | - K.‐H. Lee
- Eshelman School of Pharmacy University of North Carolina Chapel Hill NC USA
| | - Y. Zhao
- Eshelman School of Pharmacy University of North Carolina Chapel Hill NC USA
| | - C. Y. Hsu
- Graduate Institute of Biomedical Science and Drug Development Center China Medical University Taichung Taiwan
| | - W.C. Shyu
- Graduate Institute of Biomedical Science and Drug Development Center China Medical University Taichung Taiwan
- Translational Medicine Research Center China Medical University & Hospital Taichung Taiwan
- Department of Neurology China Medical University & Hospital Taichung Taiwan
- Department of Occupational Therapy Asia University Taichung Taiwan
| |
Collapse
|
42
|
Kośla K, Płuciennik E, Styczeń-Binkowska E, Nowakowska M, Orzechowska M, Bednarek AK. The WWOX Gene Influences Cellular Pathways in the Neuronal Differentiation of Human Neural Progenitor Cells. Front Cell Neurosci 2019; 13:391. [PMID: 31543760 PMCID: PMC6730490 DOI: 10.3389/fncel.2019.00391] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 08/08/2019] [Indexed: 11/13/2022] Open
Abstract
The brain is the most functionally organized structure of all organs. It manages behavior, perception and higher cognitive functions. The WWOX gene is non-classical tumor suppressor gene, which has been shown to have an impact on proliferation, apoptosis and migration processes. Moreover, genetic aberrations in WWOX induce severe neuropathological phenotypes in humans and rodents. The aim of the present study was to investigate in detail the impact of WWOX on human neural progenitor cell (hNPC) maintenance and how depletion of WWOX disturbs signaling pathways playing a pivotal role in neuronal differentiation and central nervous system (CNS) organogenesis. hNPC with a silenced WWOX gene exhibited lowered mitochondrial redox potential, enhanced adhesion to fibronectin and extracellular matrix protein mixture, downregulation of MMP2/9 expression and impaired 3D growth. Global transcriptome analysis using cap analysis of gene expression (CAGE) found that WWOX downregulation significantly changes the expression of multiple genes engaged in cytoskeleton organization, adhesion, cell signaling and chromatin remodeling. The massive changes in gene expression caused by WWOX silencing may strongly affect the differentiation and migration of neurons in organogenesis, brain injury, cancerogenesis or neurodifferentiation. WWOX gene appears to be an important regulator of neural tissue architecture and function.
Collapse
Affiliation(s)
- Katarzyna Kośla
- Department of Molecular Carcinogenesis, Medical University of Łódź, Łódź, Poland
| | - Elżbieta Płuciennik
- Department of Molecular Carcinogenesis, Medical University of Łódź, Łódź, Poland
| | | | - Magdalena Nowakowska
- Department of Molecular Carcinogenesis, Medical University of Łódź, Łódź, Poland
| | | | - Andrzej K Bednarek
- Department of Molecular Carcinogenesis, Medical University of Łódź, Łódź, Poland
| |
Collapse
|
43
|
Ceci M, Mariano V, Romano N. Zebrafish as a translational regeneration model to study the activation of neural stem cells and role of their environment. Rev Neurosci 2019; 30:45-66. [PMID: 30067512 DOI: 10.1515/revneuro-2018-0020] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 04/27/2018] [Indexed: 02/07/2023]
Abstract
The review is an overview of the current knowledge of neuronal regeneration properties in mammals and fish. The ability to regenerate the damaged parts of the nervous tissue has been demonstrated in all vertebrates. Notably, fish and amphibians have the highest capacity for neurogenesis, whereas reptiles and birds are able to only regenerate specific regions of the brain, while mammals have reduced capacity for neurogenesis. Zebrafish (Danio rerio) is a promising model of study because lesions in the brain or complete cross-section of the spinal cord are followed by an effective neuro-regeneration that successfully restores the motor function. In the brain and the spinal cord of zebrafish, stem cell activity is always able to re-activate the molecular programs required for central nervous system regeneration. In mammals, traumatic brain injuries are followed by reduced neurogenesis and poor axonal regeneration, often insufficient to functionally restore the nervous tissue, while spinal injuries are not repaired at all. The environment that surrounds the stem cell niche constituted by connective tissue and stimulating factors, including pro-inflammation molecules, seems to be a determinant in triggering stem cell proliferation and/or the trans-differentiation of connective elements (mainly fibroblasts). Investigating and comparing the neuronal regeneration in zebrafish and mammals may lead to a better understanding of the mechanisms behind neurogenesis, and the failure of the regenerative response in mammals, first of all, the role of inflammation, considered the main inhibitor of the neuronal regeneration.
Collapse
Affiliation(s)
- Marcello Ceci
- Department of Ecological and Biological Sciences, University of Tuscia, largo dell'Università, I-01100 Viterbo, Italy
| | - Vittoria Mariano
- Department of Fundamental Neurosciences, University of Lausanne, CH-1005 Lausanne, Switzerland
| | - Nicla Romano
- Department of Ecological and Biological Sciences, University of Tuscia, largo dell'Università, I-01100 Viterbo, Italy
| |
Collapse
|
44
|
Montaner J, Ramiro L, Simats A, Hernández-Guillamon M, Delgado P, Bustamante A, Rosell A. Matrix metalloproteinases and ADAMs in stroke. Cell Mol Life Sci 2019; 76:3117-3140. [PMID: 31165904 PMCID: PMC11105215 DOI: 10.1007/s00018-019-03175-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 05/22/2019] [Accepted: 05/29/2019] [Indexed: 12/27/2022]
Abstract
Stroke is a leading cause of death and disability worldwide. However, after years of in-depth research, the pathophysiology of stroke is still not fully understood. Increasing evidence shows that matrix metalloproteinases (MMPs) and "a disintegrin and metalloproteinase" (ADAMs) participate in the neuro-inflammatory cascade that is triggered during stroke but also in recovery phases of the disease. This review covers the involvement of these proteins in brain injury following cerebral ischemia which has been widely studied in recent years, with efforts to modulate this group of proteins in neuroprotective therapies, together with their implication in neurorepair mechanisms. Moreover, the review also discusses the role of these proteins in specific forms of neurovascular disease, such as small vessel diseases and intracerebral hemorrhage. Finally, the potential use of MMPs and ADAMs as guiding biomarkers of brain injury and repair for decision-making in cases of stroke is also discussed.
Collapse
Affiliation(s)
- Joan Montaner
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research, Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron, 119-129, 08035, Barcelona, Spain.
| | - Laura Ramiro
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research, Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron, 119-129, 08035, Barcelona, Spain
| | - Alba Simats
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research, Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron, 119-129, 08035, Barcelona, Spain
| | - Mar Hernández-Guillamon
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research, Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron, 119-129, 08035, Barcelona, Spain
| | - Pilar Delgado
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research, Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron, 119-129, 08035, Barcelona, Spain
| | - Alejandro Bustamante
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research, Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron, 119-129, 08035, Barcelona, Spain
| | - Anna Rosell
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research, Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron, 119-129, 08035, Barcelona, Spain
| |
Collapse
|
45
|
Muri L, Leppert D, Grandgirard D, Leib SL. MMPs and ADAMs in neurological infectious diseases and multiple sclerosis. Cell Mol Life Sci 2019; 76:3097-3116. [PMID: 31172218 PMCID: PMC7079810 DOI: 10.1007/s00018-019-03174-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 05/23/2019] [Accepted: 05/29/2019] [Indexed: 12/24/2022]
Abstract
Metalloproteinases-such as matrix metalloproteinases (MMPs) and a disintegrin and metalloproteinases (ADAMs)-are involved in various diseases of the nervous system but also contribute to nervous system development, synaptic plasticity and neuroregeneration upon injury. MMPs and ADAMs proteolytically cleave many substrates including extracellular matrix components but also signaling molecules and receptors. During neuroinfectious disease with associated neuroinflammation, MMPs and ADAMs regulate blood-brain barrier breakdown, bacterial invasion, neutrophil infiltration and cytokine signaling. Specific and broad-spectrum inhibitors for MMPs and ADAMs have experimentally been shown to decrease neuroinflammation and brain damage in diseases with excessive neuroinflammation as a common denominator, such as pneumococcal meningitis and multiple sclerosis, thereby improving the disease outcome. Timing of metalloproteinase inhibition appears to be critical to effectively target the cascade of pathophysiological processes leading to brain damage without inhibiting the neuroregenerative effects of metalloproteinases. As the critical role of metalloproteinases in neuronal repair mechanisms and regeneration was only lately recognized, the original idea of chronic MMP inhibition needs to be conceptually revised. Recently accumulated research urges for a second chance of metalloproteinase inhibitors, which-when correctly applied and dosed-harbor the potential to improve the outcome of different neuroinflammatory diseases.
Collapse
Affiliation(s)
- Lukas Muri
- Neuroinfection Laboratory, Institute for Infectious Diseases, University of Bern, Friedbühlstrasse 51, 3001, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Freiestrasse 1, 3012, Bern, Switzerland
| | - David Leppert
- Department of Neurology, University Hospital Basel, Petersgraben 4, 4031, Basel, Switzerland
| | - Denis Grandgirard
- Neuroinfection Laboratory, Institute for Infectious Diseases, University of Bern, Friedbühlstrasse 51, 3001, Bern, Switzerland
| | - Stephen L Leib
- Neuroinfection Laboratory, Institute for Infectious Diseases, University of Bern, Friedbühlstrasse 51, 3001, Bern, Switzerland.
| |
Collapse
|
46
|
He C, Shan N, Xu P, Ge H, Yuan Y, Liu Y, Zhang P, Wen L, Zhang F, Xiong L, Peng C, Qi H, Tong C, Baker PN. Hypoxia-induced Downregulation of SRC-3 Suppresses Trophoblastic Invasion and Migration Through Inhibition of the AKT/mTOR Pathway: Implications for the Pathogenesis of Preeclampsia. Sci Rep 2019; 9:10349. [PMID: 31316078 PMCID: PMC6637123 DOI: 10.1038/s41598-019-46699-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 06/29/2019] [Indexed: 01/14/2023] Open
Abstract
Preeclampsia (PE) is characterized by poor placentation, consequent on aberrant extravillous trophoblast (EVT) cell function during placental development. The SRC family of proteins is important during pregnancy, especially SRC-3, which regulates placental morphogenesis and embryo survival. Although SRC-3 expression in mouse trophoblast giant cells has been documented, its role in the functional regulation of extravillous trophoblasts and the development of PE remains unknown. This study found that SRC-3 expression was significantly lower in placentas from PE pregnancies as compared to uncomplicated pregnancies. Additionally, both CoCl2-mimicked hypoxia and suppression of endogenous SRC-3 expression by lentivirus short hairpin RNA attenuated the migration and invasion abilities of HTR-8/SVneo cells. Moreover, we demonstrated that SRC-3 physically interacts with AKT to regulate the migration and invasion of HTR-8 cells, via the AKT/mTOR pathway. We also found that the inhibition of HTR-8 cell migration and invasion by CoCl2-mimicked hypoxia was through the SRC-3/AKT/mTOR axis. Our findings indicate that, in early gestation, accumulation of HIF-1α inhibits the expression of SRC-3, which impairs extravillous trophoblastic invasion and migration by directly interacting with AKT. This potentially leads to insufficient uterine spiral artery remodeling and placental hypoperfusion, and thus the development of PE.
Collapse
Affiliation(s)
- Chengjin He
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.,International Collaborative Joint Laboratory of Reproduction and Development, Ministry of Education of China, Chongqing Medical University, Chongqing, 400016, China.,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Nan Shan
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.,International Collaborative Joint Laboratory of Reproduction and Development, Ministry of Education of China, Chongqing Medical University, Chongqing, 400016, China.,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Ping Xu
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.,International Collaborative Joint Laboratory of Reproduction and Development, Ministry of Education of China, Chongqing Medical University, Chongqing, 400016, China.,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Huisheng Ge
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.,International Collaborative Joint Laboratory of Reproduction and Development, Ministry of Education of China, Chongqing Medical University, Chongqing, 400016, China.,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yu Yuan
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.,International Collaborative Joint Laboratory of Reproduction and Development, Ministry of Education of China, Chongqing Medical University, Chongqing, 400016, China.,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yangming Liu
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.,International Collaborative Joint Laboratory of Reproduction and Development, Ministry of Education of China, Chongqing Medical University, Chongqing, 400016, China.,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Pu Zhang
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Li Wen
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.,International Collaborative Joint Laboratory of Reproduction and Development, Ministry of Education of China, Chongqing Medical University, Chongqing, 400016, China.,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Fumei Zhang
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.,International Collaborative Joint Laboratory of Reproduction and Development, Ministry of Education of China, Chongqing Medical University, Chongqing, 400016, China.,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Liling Xiong
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.,International Collaborative Joint Laboratory of Reproduction and Development, Ministry of Education of China, Chongqing Medical University, Chongqing, 400016, China.,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Chuan Peng
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Hongbo Qi
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China. .,International Collaborative Joint Laboratory of Reproduction and Development, Ministry of Education of China, Chongqing Medical University, Chongqing, 400016, China. .,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Chao Tong
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China. .,International Collaborative Joint Laboratory of Reproduction and Development, Ministry of Education of China, Chongqing Medical University, Chongqing, 400016, China. .,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Philip N Baker
- International Collaborative Joint Laboratory of Reproduction and Development, Ministry of Education of China, Chongqing Medical University, Chongqing, 400016, China.,Liggins Institute, University of Auckland, Auckland, 1142, New Zealand.,College of Life Sciences, University of Leicester, Leicester, LE1 7RH, UK
| |
Collapse
|
47
|
Adult Neurogenesis in the Subventricular Zone and Its Regulation After Ischemic Stroke: Implications for Therapeutic Approaches. Transl Stroke Res 2019; 11:60-79. [DOI: 10.1007/s12975-019-00717-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 06/13/2019] [Accepted: 06/27/2019] [Indexed: 12/21/2022]
|
48
|
Dief AE, Hassan PS, Hartmut O, Jirikowski GF. Neuronal and glial regeneration after focal cerebral ischemia in rat, an immunohistochemical and electron microscopical study. ALEXANDRIA JOURNAL OF MEDICINE 2019. [DOI: 10.1016/j.ajme.2018.02.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Abeer E. Dief
- Department of Physiology, University of Alexandria, Egypt
| | | | | | | |
Collapse
|
49
|
Chen Z, Wei H, Zhao X, Xin X, Peng L, Ning Y, Wang Y, Lan Y, Zhang Q. Metformin treatment alleviates polycystic ovary syndrome by decreasing the expression of MMP‐2 and MMP‐9 via H19/miR‐29b‐3p and AKT/mTOR/autophagy signaling pathways. J Cell Physiol 2019; 234:19964-19976. [PMID: 30989649 DOI: 10.1002/jcp.28594] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 03/01/2019] [Accepted: 03/06/2019] [Indexed: 12/27/2022]
Affiliation(s)
- Zhilan Chen
- Department of Obstetrics and Gynecology The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Huafang Wei
- Department of Obstetrics and Gynecology Central Theater General Hospital of PLA Wuhan China
| | - Xiaoling Zhao
- Department of Obstetrics and Gynecology Central Theater General Hospital of PLA Wuhan China
| | - Xin Xin
- Department of Obstetrics and Gynecology The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Ling Peng
- Department of Obstetrics and Gynecology The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Yang Ning
- Department of Obstetrics and Gynecology The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Yapei Wang
- Department of Obstetrics and Gynecology The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Yanli Lan
- Department of Obstetrics and Gynecology Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science Xiangyang China
| | - Qinghua Zhang
- Department of Obstetrics and Gynecology The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| |
Collapse
|
50
|
Mulens-Arias V, Balfourier A, Nicolás-Boluda A, Carn F, Gazeau F. Disturbance of adhesomes by gold nanoparticles reveals a size- and cell type-bias. Biomater Sci 2019; 7:389-408. [PMID: 30484789 DOI: 10.1039/c8bm01267a] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Gold nanoparticles (AuNP) have been thoroughly studied as multifunctional theranosis agents for cell imaging and cancer therapy as well as sensors due to their tunable physical and chemical properties. Although AuNP have proved to be safe in a wide concentration range, yet other important biological effects can arise in the sublethal window of treatment. This is especially pivotal to understand how AuNP can affect cell biology when labeling steps are needed for cell tracking in vivo, as nanoparticle loading can affect cell migratory/invasion ability, a function mediated by filamentous actin-rich nanometric structures collectively called adhesomes. It is noteworthy that, although numerous research studies have addressed the cell response to AuNP loading, yet none of them focuses on adhesome dynamics as a target of intracellular pathways affected by AuNP. We intend to study the collective dynamics of adhesive F-actin rich structures upon AuNP treatment as an approach to understand the complex AuNP-triggered modulation of migration/invasion related cellular functions. We demonstrated that citrate-coated spherical AuNP of different sizes (3, 11, 16, 30 and 40 nm) disturbed podosome-forming rosettes and the resulting extracellular matrix (ECM) degradation in a murine macrophage model depending on core size. This phenomenon was accompanied by a reduction in metalloproteinase MMP2 and an increment in metalloproteinase inhibitors, TIMP-1/2 and SerpinE1. We also found that AuNP treatment has opposite effects on focal adhesions (FA) in endothelial and mesenchymal stem cells. While endothelial cells reduced their mature FA number and ECM degradation rate upon AuNP treatment, mouse mesenchymal stem cells increased the number and size of mature FA and, therefore, the ECM degradation rate. Overall, AuNP appear to disturb adhesive structures and therefore migratory/invasive cell functions measured as ECM degradation ability, providing new insights into AuNP-cell interaction depending on cell type.
Collapse
Affiliation(s)
- Vladimir Mulens-Arias
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7075, CNRS and Université Paris Diderot, Université Sorbonne Paris Cité (USPC), 10 Rue Alice Domon et Léonie Duquet, 75205 Paris Cedex 13, France.
| | | | | | | | | |
Collapse
|