1
|
Shi X. Research advances in cochlear pericytes and hearing loss. Hear Res 2023; 438:108877. [PMID: 37651921 PMCID: PMC10538405 DOI: 10.1016/j.heares.2023.108877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 08/03/2023] [Accepted: 08/18/2023] [Indexed: 09/02/2023]
Abstract
Pericytes are specialized mural cells surrounding endothelial cells in microvascular beds. They play a role in vascular development, blood flow regulation, maintenance of blood-tissue barrier integrity, and control of angiogenesis, tissue fibrosis, and wound healing. In recent decades, understanding of the critical role played by pericytes in retina, brain, lung, and kidney has seen significant progress. The cochlea contains a large population of pericytes. However, the role of cochlear pericytes in auditory pathophysiology is, by contrast, largely unknown. The present review discusses recent progress in identifying cochlear pericytes, mapping their distribution, and defining their role in regulating blood flow, controlling the blood-labyrinth barrier (BLB) and angiogenesis, and involvement in different types of hearing loss.
Collapse
Affiliation(s)
- Xiaorui Shi
- Department of Otolaryngology/Head & Neck Surgery, Oregon Hearing Research Center (NRC04), Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239-3098, USA.
| |
Collapse
|
2
|
Dabravolski SA, Markin AM, Andreeva ER, Eremin II, Orekhov AN, Melnichenko AA. Molecular Mechanisms Underlying Pathological and Therapeutic Roles of Pericytes in Atherosclerosis. Int J Mol Sci 2022; 23:11663. [PMID: 36232962 PMCID: PMC9570222 DOI: 10.3390/ijms231911663] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 11/17/2022] Open
Abstract
Pericytes are multipotent mesenchymal stromal cells playing an active role in angiogenesis, vessel stabilisation, maturation, remodelling, blood flow regulation and are able to trans-differentiate into other cells of the mesenchymal lineage. In this review, we summarised recent data demonstrating that pericytes play a key role in the pathogenesis and development of atherosclerosis (AS). Pericytes are involved in lipid accumulation, inflammation, growth, and vascularization of the atherosclerotic plaque. Decreased pericyte coverage, endothelial and pericyte dysfunction is associated with intraplaque angiogenesis and haemorrhage, calcification and cholesterol clefts deposition. At the same time, pericytes can be used as a novel therapeutic target to promote vessel maturity and stability, thus reducing plaque vulnerability. Finally, we discuss recent studies exploring effective AS treatments with pericyte-mediated anti-atherosclerotic, anti-inflammatory and anti-apoptotic effects.
Collapse
Affiliation(s)
- Siarhei A. Dabravolski
- Department of Biotechnology Engineering, ORT Braude College, Snunit 51, P.O. Box 78, Karmiel 2161002, Israel
| | - Alexander M. Markin
- Petrovsky National Research Center of Surgery, Abrikosovsky Lane, 2, 119991 Moscow, Russia
| | - Elena R. Andreeva
- Laboratory of Cell Physiology, Institute of Biomedical Problems, Russian Academy of Sciences, Khoroshevskoye Shosse, 76a, 123007 Moscow, Russia
| | - Ilya I. Eremin
- Petrovsky National Research Center of Surgery, Abrikosovsky Lane, 2, 119991 Moscow, Russia
| | - Alexander N. Orekhov
- Institute for Atherosclerosis Research, Osennyaya 4-1-207, 121609 Moscow, Russia
| | | |
Collapse
|
3
|
Heinrich UR, Meuser R, Ernst BP, Schmidtmann I, Dietrich D, Stauber RH, Strieth S. Regulation of Endothelial Nitric Oxide Synthase in the Reticular Lamina of the Organ of Corti by a Nitric Oxide Donor. J Histochem Cytochem 2021; 69:731-739. [PMID: 34666550 DOI: 10.1369/00221554211054642] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In the vertebrate cochlea, the reticular lamina seals the organ of Corti against the endolymph filled scala media. After noise exposure, fast alterations in the endothelial nitric oxide synthase (eNOS) expression level were identified in this cochlear structure. Minor amounts of nitric oxide (NO) produced by eNOS or applied by NO donors such as S-nitroso-N-acetyl-penicillamine (SNAP) might protect this vulnerable part of the organ of Corti, on the line of gap junctions of supporting cells and cochlear microcirculation. In n=5 anesthetized guinea pigs, SNAP was intravenously applied in two concentrations. Six untreated animals served as controls. The cochleae were removed and prepared for immunoelectron microscopy using specific gold-labeled anti-eNOS antibodies. The density of the gold particles was quantified for seven cellular regions in the reticular lamina at the ultrastructural level. Following SNAP application, a significant increase in eNOS expression (+176%) was detected compared with controls (p=0.012). The increase occurred mainly in actin-rich cuticular structures and the prominent microtubules bundles. Correlation analysis revealed three clear and five moderate cellular associations for controls, whereas only one clear and one moderate after SNAP application. Thus, application of the NO donor SNAP resulted in an increase in eNOS expression in distinct regions of the reticular lamina.
Collapse
Affiliation(s)
| | - Regina Meuser
- Institute for Medical Biometry, Epidemiology and Informatics
| | - Benjamin Philipp Ernst
- University Medical Center Mainz, Mainz, Germany, and Department of Otorhinolaryngology, University Medical Center Bonn (UKB), Bonn, Germany
| | | | - Dimo Dietrich
- University Medical Center Mainz, Mainz, Germany, and Department of Otorhinolaryngology, University Medical Center Bonn (UKB), Bonn, Germany
| | | | - Sebastian Strieth
- University Medical Center Mainz, Mainz, Germany, and Department of Otorhinolaryngology, University Medical Center Bonn (UKB), Bonn, Germany
| |
Collapse
|
4
|
Su H, Cantrell AC, Zeng H, Zhu SH, Chen JX. Emerging Role of Pericytes and Their Secretome in the Heart. Cells 2021; 10:548. [PMID: 33806335 PMCID: PMC8001346 DOI: 10.3390/cells10030548] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/25/2021] [Accepted: 02/27/2021] [Indexed: 12/11/2022] Open
Abstract
Pericytes, as mural cells covering microvascular capillaries, play an essential role in vascular remodeling and maintaining vascular functions and blood flow. Pericytes are crucial participants in the physiological and pathological processes of cardiovascular disease. They actively interact with endothelial cells, vascular smooth muscle cells (VSMCs), fibroblasts, and other cells via the mechanisms involved in the secretome. The secretome of pericytes, along with diverse molecules including proinflammatory cytokines, angiogenic growth factors, and the extracellular matrix (ECM), has great impacts on the formation, stabilization, and remodeling of vasculature, as well as on regenerative processes. Emerging evidence also indicates that pericytes work as mesenchymal cells or progenitor cells in cardiovascular regeneration. Their capacity for differentiation also contributes to vascular remodeling in different ways. Previous studies primarily focused on the roles of pericytes in organs such as the brain, retina, lung, and kidney; very few studies have focused on pericytes in the heart. In this review, following a brief introduction of the origin and fundamental characteristics of pericytes, we focus on pericyte functions and mechanisms with respect to heart disease, ending with the promising use of cardiac pericytes in the treatment of ischemic heart failure.
Collapse
Affiliation(s)
- Han Su
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Aubrey C Cantrell
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Heng Zeng
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Shai-Hong Zhu
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Jian-Xiong Chen
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
5
|
Hou Z, Neng L, Zhang J, Cai J, Wang X, Zhang Y, Lopez IA, Shi X. Acoustic Trauma Causes Cochlear Pericyte-to-Myofibroblast-Like Cell Transformation and Vascular Degeneration, and Transplantation of New Pericytes Prevents Vascular Atrophy. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:1943-1959. [PMID: 32562655 DOI: 10.1016/j.ajpath.2020.05.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/19/2020] [Accepted: 05/26/2020] [Indexed: 12/20/2022]
Abstract
Acoustic trauma disrupts cochlear blood flow and damages sensory hair cells. Damage and regression of capillaries after acoustic trauma have long been observed, but the underlying mechanism of pathology has not been understood. We show herein that loud sound causes change of phenotype from neural/glial antigen 2 positive/α-smooth muscle actin negative to neural/glial antigen 2 positive/α-smooth muscle actin positive in some pericytes (PCs) on strial capillaries that is strongly associated with up-regulation of transforming growth factor-β1. The acoustic trauma also reduced capillary density and increased deposition of matrix proteins, particularly in the vicinity of transformed PCs. In a newly established in vitro three-dimensional endothelial cell (EC) and PC co-culture model, transformed PCs induced thicker capillary-like branches in ECs and increased collagen IV and laminin expression. Transplantation of exogenous PCs derived from neonatal day 10 mouse cochleae to acoustic traumatized cochleae, however, significantly attenuated the decreased vascular density in the stria. Transplantation of PCs pretransfected with adeno-associated virus 1-vascular endothelial growth factor-A165 under control of a hypoxia-response element markedly promotes vascular volume and blood flow, increased proliferation of PCs and ECs, and attenuated loud sound-caused loss in endocochlear potential and hearing. Our results indicate that loud sound-triggered PC transformation contributes to capillary wall thickening and regression, and young PC transplantation effectively rehabilitates the vascular regression and improves hearing.
Collapse
Affiliation(s)
- Zhiqiang Hou
- Department of Otolaryngology/Head & Neck Surgery, Oregon Hearing Research Center, Oregon Health & Science University, Portland, Oregon
| | - Lingling Neng
- Department of Otolaryngology/Head & Neck Surgery, Oregon Hearing Research Center, Oregon Health & Science University, Portland, Oregon
| | - Jinhui Zhang
- Department of Otolaryngology/Head & Neck Surgery, Oregon Hearing Research Center, Oregon Health & Science University, Portland, Oregon
| | - Jing Cai
- Department of Otolaryngology/Head & Neck Surgery, Oregon Hearing Research Center, Oregon Health & Science University, Portland, Oregon
| | - Xiaohan Wang
- Department of Otolaryngology/Head & Neck Surgery, Oregon Hearing Research Center, Oregon Health & Science University, Portland, Oregon; Center for Life Sciences, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Yunpei Zhang
- Department of Otolaryngology/Head & Neck Surgery, Oregon Hearing Research Center, Oregon Health & Science University, Portland, Oregon
| | - Ivan A Lopez
- Cellular and Molecular Biology of the Inner Ear Laboratory, Department of Head and Neck Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Xiaorui Shi
- Department of Otolaryngology/Head & Neck Surgery, Oregon Hearing Research Center, Oregon Health & Science University, Portland, Oregon.
| |
Collapse
|
6
|
Burwood GWS, Dziennis S, Wilson T, Foster S, Zhang Y, Liu G, Yang J, Elkins S, Nuttall AL. The mechanoelectrical transducer channel is not required for regulation of cochlear blood flow during loud sound exposure in mice. Sci Rep 2020; 10:9229. [PMID: 32514013 PMCID: PMC7280509 DOI: 10.1038/s41598-020-66192-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 05/12/2020] [Indexed: 01/03/2023] Open
Abstract
The mammalian cochlea possesses unique acoustic sensitivity due to a mechanoelectrical ‘amplifier’, which requires the metabolic support of the cochlear lateral wall. Loud sound exposure sufficient to induce permanent hearing damage causes cochlear blood flow reduction, which may contribute to hearing loss. However, sensory epithelium involvement in the cochlear blood flow regulation pathway is not fully described. We hypothesize that genetic manipulation of the mechanoelectrical transducer complex will abolish sound induced cochlear blood flow regulation. We used salsa mice, a Chd23 mutant with no mechanoelectrical transduction, and deafness before p56. Using optical coherence tomography angiography, we measured the cochlear blood flow of salsa and wild-type mice in response to loud sound (120 dB SPL, 30 minutes low-pass filtered noise). An expected sound induced decrease in cochlear blood flow occurred in CBA/CaJ mice, but surprisingly the same sound protocol induced cochlear blood flow increases in salsa mice. Blood flow did not change in the contralateral ear. Disruption of the sympathetic nervous system partially abolished the observed wild-type blood flow decrease but not the salsa increase. Therefore sympathetic activation contributes to sound induced reduction of cochlear blood flow. Additionally a local, non-sensory pathway, potentially therapeutically targetable, must exist for cochlear blood flow regulation.
Collapse
Affiliation(s)
- George W S Burwood
- Oregon Hearing Research Center, Dept. of Otolaryngology / HNS, Oregon Health & Science University, 3250S.W. Sam Jackson Park Rd., Portland, OR, 97239, USA
| | - Suzan Dziennis
- Oregon Hearing Research Center, Dept. of Otolaryngology / HNS, Oregon Health & Science University, 3250S.W. Sam Jackson Park Rd., Portland, OR, 97239, USA
| | - Teresa Wilson
- Oregon Hearing Research Center, Dept. of Otolaryngology / HNS, Oregon Health & Science University, 3250S.W. Sam Jackson Park Rd., Portland, OR, 97239, USA
| | - Sarah Foster
- Oregon Hearing Research Center, Dept. of Otolaryngology / HNS, Oregon Health & Science University, 3250S.W. Sam Jackson Park Rd., Portland, OR, 97239, USA
| | - Yuan Zhang
- Oregon Hearing Research Center, Dept. of Otolaryngology / HNS, Oregon Health & Science University, 3250S.W. Sam Jackson Park Rd., Portland, OR, 97239, USA
| | - Gangjun Liu
- Shenzhen Bay laboratory, 5F, No.9 Duxue Rd., Nanshan District, Shenzhen, Guangdong, China
| | - Jianlong Yang
- Ningbo Institute of Materials Technology and Engineering, No. 1219 Zhongguan West Road Zhenhai District, Ningbo City, Zhejiang Province, 315201, P.R. China
| | - Sean Elkins
- Oregon Hearing Research Center, Dept. of Otolaryngology / HNS, Oregon Health & Science University, 3250S.W. Sam Jackson Park Rd., Portland, OR, 97239, USA
| | - Alfred L Nuttall
- Oregon Hearing Research Center, Dept. of Otolaryngology / HNS, Oregon Health & Science University, 3250S.W. Sam Jackson Park Rd., Portland, OR, 97239, USA.
| |
Collapse
|
7
|
Nelson AR, Sagare MA, Wang Y, Kisler K, Zhao Z, Zlokovic BV. Channelrhodopsin Excitation Contracts Brain Pericytes and Reduces Blood Flow in the Aging Mouse Brain in vivo. Front Aging Neurosci 2020; 12:108. [PMID: 32410982 PMCID: PMC7201096 DOI: 10.3389/fnagi.2020.00108] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 03/30/2020] [Indexed: 01/02/2023] Open
Abstract
Brains depend on blood flow for the delivery of oxygen and nutrients essential for proper neuronal and synaptic functioning. French physiologist Rouget was the first to describe pericytes in 1873 as regularly arranged longitudinal amoeboid cells on capillaries that have a muscular coat, implying that these are contractile cells that regulate blood flow. Although there have been >30 publications from different groups, including our group, demonstrating that pericytes are contractile cells that can regulate hemodynamic responses in the brain, the role of pericytes in controlling cerebral blood flow (CBF) has not been confirmed by all studies. Moreover, recent studies using different optogenetic models to express light-sensitive channelrhodopsin-2 (ChR2) cation channels in pericytes were not conclusive; one, suggesting that pericytes expressing ChR2 do not contract after light stimulus, and the other, demonstrating contraction of pericytes expressing ChR2 after light stimulus. Since two-photon optogenetics provides a powerful tool to study mechanisms of blood flow regulation at the level of brain capillaries, we re-examined the contractility of brain pericytes in vivo using a new optogenetic model developed by crossing our new inducible pericyte-specific CreER mouse line with ChR2 mice. We induced expression of ChR2 in pericytes with tamoxifen, excited ChR2 by 488 nm light, and monitored pericyte contractility, brain capillary diameter changes, and red blood cell (RBC) velocity in aged mice by in vivo two-photon microscopy. Excitation of ChR2 resulted in pericyte contraction followed by constriction of the underlying capillary leading to approximately an 8% decrease (p = 0.006) in capillary diameter. ChR2 excitation in pericytes substantially reduced capillary RBC flow by 42% (p = 0.03) during the stimulation period compared to the velocity before stimulation. Our data suggests that pericytes contract in vivo and regulate capillary blood flow in the aging mouse brain. By extension, this might have implications for neurological disorders of the aging human brain associated with neurovascular dysfunction and pericyte loss such as stroke and Alzheimer's disease.
Collapse
Affiliation(s)
| | | | | | | | | | - Berislav V. Zlokovic
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
8
|
Zhang ZS, Zhou HN, He SS, Xue MY, Li T, Liu LM. Research advances in pericyte function and their roles in diseases. Chin J Traumatol 2020; 23:89-95. [PMID: 32192909 PMCID: PMC7156959 DOI: 10.1016/j.cjtee.2020.02.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 12/19/2019] [Accepted: 01/25/2020] [Indexed: 02/04/2023] Open
Abstract
Pericyte, a kind of pluripotent cell, may regulate the irrigation flow and permeability of microcirculation. Pericytes are similar to the smooth muscle cells, which express several kinds of contractile proteins and have contractility. The dysfunction of pericytes is related to many microvascular diseases, including hypoxia, hypertension, diabetic retinopathy, fibrosis, inflammation, Alzheimer's disease, multiple sclerosis, and tumor formation. For a long time, their existence and function have been neglected. The distribution, structure, biomarker, related signaling pathways as well as the roles of pericytes on vascular diseases will be introduced in this review.
Collapse
|
9
|
Nyberg S, Abbott NJ, Shi X, Steyger PS, Dabdoub A. Delivery of therapeutics to the inner ear: The challenge of the blood-labyrinth barrier. Sci Transl Med 2020; 11:11/482/eaao0935. [PMID: 30842313 DOI: 10.1126/scitranslmed.aao0935] [Citation(s) in RCA: 170] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 12/01/2017] [Accepted: 03/22/2018] [Indexed: 12/20/2022]
Abstract
Permanent hearing loss affects more than 5% of the world's population, yet there are no nondevice therapies that can protect or restore hearing. Delivery of therapeutics to the cochlea and vestibular system of the inner ear is complicated by their inaccessible location. Drug delivery to the inner ear via the vasculature is an attractive noninvasive strategy, yet the blood-labyrinth barrier at the luminal surface of inner ear capillaries restricts entry of most blood-borne compounds into inner ear tissues. Here, we compare the blood-labyrinth barrier to the blood-brain barrier, discuss invasive intratympanic and intracochlear drug delivery methods, and evaluate noninvasive strategies for drug delivery to the inner ear.
Collapse
Affiliation(s)
- Sophie Nyberg
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
| | - N Joan Abbott
- Institute of Pharmaceutical Science, King's College London, London SE1 9NH, UK
| | - Xiaorui Shi
- Oregon Hearing Research Center, Department of Otolaryngology, Head & Neck Surgery, Oregon Health & Science University, Portland, OR 97239, USA
| | - Peter S Steyger
- Oregon Hearing Research Center, Department of Otolaryngology, Head & Neck Surgery, Oregon Health & Science University, Portland, OR 97239, USA
| | - Alain Dabdoub
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada. .,Department of Otolaryngology-Head & Neck Surgery, University of Toronto, Toronto, ON M5G 2C4, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5G 2C4, Canada
| |
Collapse
|
10
|
Jiang Y, Zhang J, Rao Y, Chen J, Chen K, Tang Y. Lipopolysaccharide disrupts the cochlear blood-labyrinth barrier by activating perivascular resident macrophages and up-regulating MMP-9. Int J Pediatr Otorhinolaryngol 2019; 127:109656. [PMID: 31470202 DOI: 10.1016/j.ijporl.2019.109656] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 08/21/2019] [Accepted: 08/21/2019] [Indexed: 02/05/2023]
Abstract
OBJECTIVE To determine the distribution of perivascularresident macrophages (PVMs) in BLB and their relationship with capillaries, and to explore the possible mechanisms responsible for lipopolysaccharide (LPS)-induced activation of PVMs and the breakdown of BLB. METHODS Adult Balb/c mice were either trans-tympanically injected with LPS, or mock-treated. Auditory brainstem response was tested before and 48 h after treatments. Distribution of pericytes, PVMs and capillaries was analyzed by immunohistochemical staining, and BLB permeability was estimated by FITC-dextran leakage assay. Ultrastructure of stria vascularis was examined by transmission electron microscope. Protein and mRNA level of matrix metallopeptidase 9 (MMP-9), zona occludens-1 (ZO-1), interleukin-33 (IL-33) and its receptor suppression of tumorigenicity 2 (ST2) was measured by IHC and qRT-PCR. RESULTS Unlike pericytes that surround one capillary, PVMs branched to connect with more than one capillary. LPS caused hearing loss in mice. Following LPS challenge, cochleae showed vascular leakage in stria vascularis, and PVMs presented morphological changes including reduced contact with capillaries. TEM revealed a reduced number of tight junction contact points between endothelial cells and a wider space between PVMs, pericytes and endothelial cells. The mRNA and protein levels of MMP-9 and ST2 in stria vascularis were up-regulated, while ZO-1 were down-regulated after exposure to LPS. CONCLUSIONS Our results suggest that PVMs may play a more significant role than pericytes in maintaining the integrity of BLB. Our findings also reveal a possible mechanism contributing to LPS-induced activation of PVMs, breakdown of BLB and hearing loss.
Collapse
Affiliation(s)
- Ying Jiang
- Department of Otorhinolaryngology Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Jie Zhang
- Key Laboratory of Transplant Engineering and Immunology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, China
| | - Yufang Rao
- Department of Otorhinolaryngology Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Junhong Chen
- Department of Otorhinolaryngology Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Kai Chen
- Department of Otorhinolaryngology Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yuedi Tang
- Department of Otorhinolaryngology Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
11
|
Shin SA, Lyu AR, Jeong SH, Kim TH, Park MJ, Park YH. Acoustic Trauma Modulates Cochlear Blood Flow and Vasoactive Factors in a Rodent Model of Noise-Induced Hearing Loss. Int J Mol Sci 2019; 20:ijms20215316. [PMID: 31731459 PMCID: PMC6862585 DOI: 10.3390/ijms20215316] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 10/22/2019] [Accepted: 10/23/2019] [Indexed: 12/12/2022] Open
Abstract
Noise exposure affects the organ of Corti and the lateral wall of the cochlea, including the stria vascularis and spiral ligament. Although the inner ear vasculature and spiral ligament fibrocytes in the lateral wall consist of a significant proportion of cells in the cochlea, relatively little is known regarding their functional significance. In this study, 6-week-old male C57BL/6 mice were exposed to noise trauma to induce transient hearing threshold shift (TTS) or permanent hearing threshold shift (PTS). Compared to mice with TTS, mice with PTS exhibited lower cochlear blood flow and lower vessel diameter in the stria vascularis, accompanied by reduced expression levels of genes involved in vasodilation and increased expression levels of genes related to vasoconstriction. Ultrastructural analyses by transmission electron microscopy revealed that the stria vascularis and spiral ligament fibrocytes were more damaged by PTS than by TTS. Moreover, mice with PTS expressed significantly higher levels of proinflammatory cytokines in the cochlea (e.g., IL-1β, IL-6, and TNF-α). Overall, our findings suggest that cochlear microcirculation and lateral wall pathologies are differentially modulated by the severity of acoustic trauma and are associated with changes in vasoactive factors and inflammatory responses in the cochlea.
Collapse
Affiliation(s)
- Sun-Ae Shin
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Chungnam National University, Daejeon 35015, Korea; (S.-A.S.); (A.-R.L.)
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea;
| | - Ah-Ra Lyu
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Chungnam National University, Daejeon 35015, Korea; (S.-A.S.); (A.-R.L.)
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea;
| | - Seong-Hun Jeong
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea;
| | - Tae Hwan Kim
- Biomedical Convergence Research Center, Chungnam National University Hospital, Daejeon 35015, Korea;
| | - Min Jung Park
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Chungnam National University, Daejeon 35015, Korea; (S.-A.S.); (A.-R.L.)
- Brain Research Institute, College of Medicine, Chungnam National University, Daejeon 35015, Korea
- Correspondence: (M.J.P.); (Y.-H.P.); Tel.: +82-42-280-7697(M.J.P.); Fax: +82-42-253-4059 (M.J.P.)
| | - Yong-Ho Park
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Chungnam National University, Daejeon 35015, Korea; (S.-A.S.); (A.-R.L.)
- Brain Research Institute, College of Medicine, Chungnam National University, Daejeon 35015, Korea
- Correspondence: (M.J.P.); (Y.-H.P.); Tel.: +82-42-280-7697(M.J.P.); Fax: +82-42-253-4059 (M.J.P.)
| |
Collapse
|
12
|
Alarcon-Martinez L, Yilmaz-Ozcan S, Yemisci M, Schallek J, Kılıç K, Villafranca-Baughman D, Can A, Di Polo A, Dalkara T. Retinal ischemia induces α-SMA-mediated capillary pericyte contraction coincident with perivascular glycogen depletion. Acta Neuropathol Commun 2019; 7:134. [PMID: 31429795 PMCID: PMC6701129 DOI: 10.1186/s40478-019-0761-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 06/26/2019] [Indexed: 12/21/2022] Open
Abstract
Increasing evidence indicates that pericytes are vulnerable cells, playing pathophysiological roles in various neurodegenerative processes. Microvascular pericytes contract during cerebral and coronary ischemia and do not relax after re-opening of the occluded artery, causing incomplete reperfusion. However, the cellular mechanisms underlying ischemia-induced pericyte contraction, its delayed emergence, and whether it is pharmacologically reversible are unclear. Here, we investigate i) whether ischemia-induced pericyte contractions are mediated by alpha-smooth muscle actin (α-SMA), ii) the sources of calcium rise in ischemic pericytes, and iii) if peri-microvascular glycogen can support pericyte metabolism during ischemia. Thus, we examined pericyte contractility in response to retinal ischemia both in vivo, using adaptive optics scanning light ophthalmoscopy and, ex vivo, using an unbiased stereological approach. We found that microvascular constrictions were associated with increased calcium in pericytes as detected by a genetically encoded calcium indicator (NG2-GCaMP6) or a fluoroprobe (Fluo-4). Knocking down α-SMA expression with RNA interference or fixing F-actin with phalloidin or calcium antagonist amlodipine prevented constrictions, suggesting that constrictions resulted from calcium- and α-SMA-mediated pericyte contractions. Carbenoxolone or a Cx43-selective peptide blocker also reduced calcium rise, consistent with involvement of gap junction-mediated mechanisms in addition to voltage-gated calcium channels. Pericyte calcium increase and capillary constrictions became significant after 1 h of ischemia and were coincident with depletion of peri-microvascular glycogen, suggesting that glucose derived from glycogen granules could support pericyte metabolism and delay ischemia-induced microvascular dysfunction. Indeed, capillary constrictions emerged earlier when glycogen breakdown was pharmacologically inhibited. Constrictions persisted despite recanalization but were reversible with pericyte-relaxant adenosine administered during recanalization. Our study demonstrates that retinal ischemia, a common cause of blindness, induces α-SMA- and calcium-mediated persistent pericyte contraction, which can be delayed by glucose driven from peri-microvascular glycogen. These findings clarify the contractile nature of capillary pericytes and identify a novel metabolic collaboration between peri-microvascular end-feet and pericytes.
Collapse
Affiliation(s)
- Luis Alarcon-Martinez
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Sihhiye, 06100, Ankara, Turkey
- Department of Neuroscience and Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Université de Montréal, Montréal, Québec, Canada
| | - Sinem Yilmaz-Ozcan
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Sihhiye, 06100, Ankara, Turkey
| | - Muge Yemisci
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Sihhiye, 06100, Ankara, Turkey.
- Department of Neurology, Faculty of Medicine, Hacettepe University, Ankara, Turkey.
| | - Jesse Schallek
- Flaum Eye Institute and Center for Visual Science, University of Rochester, Rochester, New York, USA
| | - Kıvılcım Kılıç
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Sihhiye, 06100, Ankara, Turkey
| | - Deborah Villafranca-Baughman
- Department of Neuroscience and Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Université de Montréal, Montréal, Québec, Canada
| | - Alp Can
- Department of Histology and Embryology, School of Medicine, Ankara University, Ankara, Turkey
| | - Adriana Di Polo
- Department of Neuroscience and Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Université de Montréal, Montréal, Québec, Canada
| | - Turgay Dalkara
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Sihhiye, 06100, Ankara, Turkey.
- Department of Neurology, Faculty of Medicine, Hacettepe University, Ankara, Turkey.
| |
Collapse
|
13
|
Bielefeld EC, Kobel MJ. Advances and Challenges in Pharmaceutical Therapies to Prevent and Repair Cochlear Injuries From Noise. Front Cell Neurosci 2019; 13:285. [PMID: 31297051 PMCID: PMC6607696 DOI: 10.3389/fncel.2019.00285] [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: 04/02/2019] [Accepted: 06/13/2019] [Indexed: 12/20/2022] Open
Abstract
Noise induces a broad spectrum of pathological injuries to the cochlea, reflecting both mechanical damage to the delicate architecture of the structures of the organ of Corti and metabolic damage within the organ of Corti and lateral wall tissues. Unlike ototoxic medications, the blood-labyrinth barrier does not offer protection against noise injury. The blood-labyrinth barrier is a target of noise injury, and can be weakened as part of the metabolic pathologies in the cochlea. However, it also offers a potential for therapeutic intervention with oto-protective compounds. Because the blood-labyrinth barrier is weakened by noise, penetration of blood-borne oto-protective compounds could be higher. However, systemic dosing for cochlear protection from noise offers other significant challenges. An alternative option to systemic dosing is local administration to the cochlea through the round window membrane using a variety of drug delivery techniques. The review will discuss noise-induced cochlear pathology, including alterations to the blood-labyrinth barrier, and then transition into discussing approaches for delivery of oto-protective compounds to reduce cochlear injury from noise.
Collapse
Affiliation(s)
- Eric C Bielefeld
- Department of Speech and Hearing Science, The Ohio State University, Columbus, OH, United States
| | - Megan J Kobel
- Department of Speech and Hearing Science, The Ohio State University, Columbus, OH, United States.,Department of Otolaryngology-Head & Neck Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
14
|
Methner C, Mishra A, Golgotiu K, Li Y, Wei W, Yanez ND, Zlokovic B, Wang RK, Alkayed NJ, Kaul S, Iliff JJ. Pericyte constriction underlies capillary derecruitment during hyperemia in the setting of arterial stenosis. Am J Physiol Heart Circ Physiol 2019; 317:H255-H263. [PMID: 31125259 DOI: 10.1152/ajpheart.00097.2019] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Capillary derecruitment distal to a coronary stenosis is implicated as the mechanism of reversible perfusion defect and potential myocardial ischemia during coronary hyperemia; however, the underlying mechanisms are not defined. We tested whether pericyte constriction underlies capillary derecruitment during hyperemia under conditions of stenosis. In vivo two-photon microscopy (2PM) and optical microangiography (OMAG) were used to measure hyperemia-induced changes in capillary diameter and perfusion in wild-type and pericyte-depleted mice with femoral artery stenosis. OMAG demonstrated that hyperemic challenge under stenosis produced capillary derecruitment associated with decreased RBC flux. 2PM demonstrated that hyperemia under control conditions induces 26 ± 5% of capillaries to dilate and 19 ± 3% to constrict. After stenosis, the proportion of capillaries dilating to hyperemia decreased to 14 ± 4% (P = 0.05), whereas proportion of constricting capillaries increased to 32 ± 4% (P = 0.05). Hyperemia-induced changes in capillary diameter occurred preferentially in capillary segments invested with pericytes. In a transgenic mouse model featuring partial pericyte depletion, only 14 ± 3% of capillaries constricted to hyperemic challenge after stenosis, a significant reduction from 33 ± 4% in wild-type littermate controls (P = 0.04). These results provide for the first time direct visualization of hyperemia-induced capillary derecruitment distal to arterial stenosis and demonstrate that pericyte constriction underlies this phenomenon in vivo. These results could have important therapeutic implications in the treatment of exercise-induced ischemia. NEW & NOTEWORTHY In the setting of coronary arterial stenosis, hyperemia produces a reversible perfusion defect resulting from capillary derecruitment that is believed to underlie cardiac ischemia under hyperemic conditions. We use optical microangiography and in vivo two-photon microscopy to visualize capillary derecruitment distal to a femoral arterial stenosis with cellular resolution. We demonstrate that capillary constriction in response to hyperemia in the setting of stenosis is dependent on pericytes, contractile mural cells investing the microcirculation.
Collapse
Affiliation(s)
- Carmen Methner
- Knight Cardiovascular Institute, Oregon Health & Science University , Portland, Oregon
| | - Anusha Mishra
- Knight Cardiovascular Institute, Oregon Health & Science University , Portland, Oregon
| | - Kirsti Golgotiu
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University , Portland, Oregon
| | - Yuandong Li
- Department of Bioengineering, University of Washington , Seattle, Washington
| | - Wei Wei
- Department of Bioengineering, University of Washington , Seattle, Washington
| | - N David Yanez
- Division of Biostatistics, School of Public Health, Oregon Health & Science University , Portland, Oregon
| | - Berislav Zlokovic
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Ruikang K Wang
- Department of Bioengineering, University of Washington , Seattle, Washington
| | - Nabil J Alkayed
- Knight Cardiovascular Institute, Oregon Health & Science University , Portland, Oregon.,Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University , Portland, Oregon
| | - Sanjiv Kaul
- Knight Cardiovascular Institute, Oregon Health & Science University , Portland, Oregon
| | - Jeffrey J Iliff
- Knight Cardiovascular Institute, Oregon Health & Science University , Portland, Oregon.,Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University , Portland, Oregon
| |
Collapse
|
15
|
Burwood GWS, Fridberger A, Wang RK, Nuttall AL. Revealing the morphology and function of the cochlea and middle ear with optical coherence tomography. Quant Imaging Med Surg 2019; 9:858-881. [PMID: 31281781 PMCID: PMC6571188 DOI: 10.21037/qims.2019.05.10] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 05/09/2019] [Indexed: 01/17/2023]
Abstract
Optical coherence tomography (OCT) has revolutionized physiological studies of the hearing organ, the vibration and morphology of which can now be measured without opening the surrounding bone. In this review, we provide an overview of OCT as used in the otological research, describing advances and different techniques in vibrometry, angiography, and structural imaging.
Collapse
Affiliation(s)
- George W. S. Burwood
- Department of Otolaryngology, Oregon Hearing Research Center/HNS, Oregon Health & Science University, Portland, OR, USA
| | - Anders Fridberger
- Department of Otolaryngology, Oregon Hearing Research Center/HNS, Oregon Health & Science University, Portland, OR, USA
- Department of Clinical and Experimental Medicine, Section for Neurobiology, Linköping University, Linköping, Sweden
| | - Ruikang K. Wang
- Department of Bioengineering and Department of Ophthalmology, University of Washington, Seattle, WA, USA
| | - Alfred L. Nuttall
- Department of Otolaryngology, Oregon Hearing Research Center/HNS, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
16
|
Kim YY, Chao JR, Kim C, Kang TC, Park HS, Chang J, Suh JG, Lee JH. Applicability of vital staining and tissue clearing to vascular anatomy and melanocytes' evaluation of temporal bone in six laboratory species. Anat Histol Embryol 2019; 48:296-305. [PMID: 30916435 DOI: 10.1111/ahe.12440] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 02/11/2019] [Accepted: 02/26/2019] [Indexed: 12/14/2022]
Abstract
The purpose of the present study was to define the applicability of tissue clearing to the field of otology. We combined tissue clearing with vital staining perfusion via a pumping system to examine the vascular anatomy of temporal bones in laboratory animals. We used six different types of species including Korean wild mouse, mouse, Mongolian gerbil, hamsters and Guinea pigs. A mixture of Alcian blue reagent and 4% paraformaldehyde was circulated throughout the entire circulatory system of the animal via a perfusion pump system. Transparency images were obtained from the temporal bones according to the protocol of the SunHyun 3D Imaging Kit. In examining the inner surface of the tympanic membrane, flaccid part (pars flaccida) was positioned along the entire marginal area in Guinea pig. In the Guinea pig, unlike the other species, the cortical bone of the mastoid (bullae) was easily removed using cold instruments, allowing a direct approach to the enclosed structures. The distribution and pattern of cochlea melanocytes were compared among the species. "Mobius strip"-like accumulated melanocytes in vestibules were shown in both the Korean wild mouse and mouse. The collateral blood supply to the cochlea in six different species was checked in various pattern. Combining dye infusion with tissue-clearing techniques, we documented the middle ear and transparent inner ear structures in six different species. The information and associated images will help other researchers to develop hypotheses and design experimental investigations.
Collapse
Affiliation(s)
- Yoo Yeon Kim
- Department of Medical Genetics, College of Medicine, Hallym University, Chuncheon, Korea
| | - Janet Ren Chao
- Division of Otolaryngology, Department of Surgery, Yale School of Medicine, New Haven, Connecticut
| | - Chulho Kim
- Department of Neurology, Chuncheon Sacred Heart Hospital, Hallym University, Chuncheon, Korea
| | - Tae-Cheon Kang
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chuncheon, Korea
| | - Hae Sang Park
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon, Korea.,Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Hallym University, Chuncheon, Korea
| | - Jiwon Chang
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Hallym University, Chuncheon, Korea
| | - Jun-Gyo Suh
- Department of Medical Genetics, College of Medicine, Hallym University, Chuncheon, Korea
| | - Jun Ho Lee
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon, Korea.,Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Hallym University, Chuncheon, Korea
| |
Collapse
|
17
|
Canis M, Bertlich M. Cochlear Capillary Pericytes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1122:115-123. [PMID: 30937866 DOI: 10.1007/978-3-030-11093-2_7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Capillary pericytes in the cochlea of mammals are-compared to pericytes in other tissues, like the CNS-relatively poorly researched. To begin with, there is still a considerable debate as to whether the very last precapillary arterioles should-due to their contractile properties-may be considered to be pericytes.However, cochlear capillary pericytes have shifted into the center of attention in the past decade. Most mammals show a considerable number of pericytes in the stria vascularis of the cochlea-up to 1300 in a mouse alone. This high number may be explained by the observation that cochlear capillary pericytes may be differentiated into different subgroups, depending on the immune markers that are expressed by them. Corresponding with these subpopulations, cochlear pericytes fulfill three core functions in the physiology of the cochlea: Formation of the intrastrial blood-fluid barrier-Pericytes monitor the ion, fluid, and nutrient household and aid in the homeostasis thereof. Regulation of cochlear blood flow-By contraction on relaxation, pericytes contribute to the regulation of cochlear blood flow, a paramount function parameter of the cochlea. Immune response-Pericytes actually contribute to the immune response in inflammation of the cochlea. Due to these central roles in the physiology of the cochlea, pericytes actually play a major role in numerous cochlear pathologies, including, but not limited to, sudden sensorineural hearing loss, acoustic trauma, and inflammation of the cochlea.
Collapse
Affiliation(s)
- Martin Canis
- The Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital, Munich, Germany
| | - Mattis Bertlich
- The Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital, Munich, Germany.
| |
Collapse
|
18
|
Giurdanella G, Montalbano G, Gennuso F, Brancati S, Lo Furno D, Augello A, Bucolo C, Drago F, Salomone S. Isolation, cultivation, and characterization of primary bovine cochlear pericytes: A new in vitro model of stria vascularis. J Cell Physiol 2018; 234:1978-1986. [PMID: 30317595 DOI: 10.1002/jcp.27545] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 09/14/2018] [Indexed: 12/14/2022]
Abstract
The study of strial pericytes has gained great interest as they are pivotal for the physiology of stria vascularis. To provide an easily accessible in vitro model, here we described a growth medium-based approach to obtain and cultivate primary bovine cochlear pericytes (BCP) from the stria vascularis of explanted bovine cochleae. We obtained high-quality pericytes in 8-10 days with a > 90% purity after the second passage. Immunocytochemical analysis showed a homogeneous population of cells expressing typical pericyte markers, such as neural/glial antigen 2 (NG2), platelet-derived growth factor receptorβ (PDGFRβ), α-smooth muscle actin (α-SMA), and negative for the endothelial marker von Willebrand factor. When challenged with tumor necrosis factor or lipopolysaccharide, BCP changed their shape, similarly to human retinal pericytes (HRPC). The sensitivity of BCP to ototoxic drugs was evaluated by challenging with cisplatin or gentamicin for 48 hr. Compared to human retinal endothelial cells and HRPC, cell viability of BCP was significantly lower ( p < 0.05) after the treatment with gentamicin or cisplatin. These data indicate that our protocol provides a simple and reliable method to obtain highly pure strial BCP. Furthermore, BCP are suitable to assess the safety profile of molecules which supposedly exert ototoxic activity, and may represent a valid alternative to in vivo tests.
Collapse
Affiliation(s)
- Giovanni Giurdanella
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Giuseppe Montalbano
- Department of Veterinary Sciences and Zebrafish Neuromorphology Lab, University of Messina, Messina, Italia
| | - Florinda Gennuso
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Serena Brancati
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Debora Lo Furno
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Antonio Augello
- ASP Catania Dipartimento di Prevenzione Veterinaria, Servizio Igiene degli Alimenti di Origine Animale (SIAOA), Catania, Italy
| | - Claudio Bucolo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Filippo Drago
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Salvatore Salomone
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| |
Collapse
|
19
|
Khennouf L, Gesslein B, Brazhe A, Octeau JC, Kutuzov N, Khakh BS, Lauritzen M. Active role of capillary pericytes during stimulation-induced activity and spreading depolarization. Brain 2018; 141:2032-2046. [PMID: 30053174 PMCID: PMC6022680 DOI: 10.1093/brain/awy143] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 04/05/2018] [Accepted: 04/07/2018] [Indexed: 12/27/2022] Open
Abstract
Spreading depolarization is assumed to be the mechanism of migraine with aura, which is accompanied by an initial predominant hyperaemic response followed by persistent vasoconstriction. Cerebral blood flow responses are impaired in patients and in experimental animals after spreading depolarization. Understanding the regulation of cortical blood vessels during and after spreading depolarization could help patients with migraine attacks, but our knowledge of these vascular mechanisms is still incomplete. Recent findings show that control of cerebral blood flow does not only occur at the arteriole level but also at capillaries. Pericytes are vascular mural cells that can constrict or relax around capillaries, mediating local cerebral blood flow control. They participate in the constriction observed during brain ischaemia and might be involved the disruption of the microcirculation during spreading depolarization. To further understand the regulation of cerebral blood flow in spreading depolarization, we examined penetrating arterioles and capillaries with respect to vascular branching order, pericyte location and pericyte calcium responses during somatosensory stimulation and spreading depolarization. Mice expressing a red fluorescent indicator and intravenous injections of FITC-dextran were used to visualize pericytes and vessels, respectively, under two-photon microscopy. By engineering a genetically encoded calcium indicator we could record calcium changes in both pericytes around capillaries and vascular smooth muscle cells around arterioles. We show that somatosensory stimulation evoked a decrease in cytosolic calcium in pericytes located on dilating capillaries, up to the second order capillaries. Furthermore, we show that prolonged vasoconstriction following spreading depolarization is strongest in first order capillaries, with a persistent increase in pericyte calcium. We suggest that the persistence of the 'spreading cortical oligaemia' in migraine could be caused by this constriction of cortical capillaries. After spreading depolarization, somatosensory stimulation no longer evoked changes in capillary diameter and pericyte calcium. Thus, calcium changes in pericytes located on first order capillaries may be a key determinant in local blood flow control and a novel vascular mechanism in migraine. We suggest that prevention or treatment of capillary constriction in migraine with aura, which is an independent risk factor for stroke, may be clinically useful.
Collapse
Affiliation(s)
- Lila Khennouf
- Department of Neuroscience and Center for Healthy Aging, University of Copenhagen, Copenhagen N, Denmark
| | - Bodil Gesslein
- Department of Neuroscience and Center for Healthy Aging, University of Copenhagen, Copenhagen N, Denmark
| | - Alexey Brazhe
- Department of Neuroscience and Center for Healthy Aging, University of Copenhagen, Copenhagen N, Denmark
- Department of Biophysics, Faculty of Biology, Moscow State University, Moscow, Russia
| | - J Christopher Octeau
- Department of Physiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Nikolay Kutuzov
- Department of Neuroscience and Center for Healthy Aging, University of Copenhagen, Copenhagen N, Denmark
| | - Baljit S Khakh
- Department of Physiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Martin Lauritzen
- Department of Neuroscience and Center for Healthy Aging, University of Copenhagen, Copenhagen N, Denmark
- Department of Clinical Neurophysiology, Rigshospitalet, Glostrup, Denmark
| |
Collapse
|
20
|
Smyth LCD, Rustenhoven J, Scotter EL, Schweder P, Faull RLM, Park TIH, Dragunow M. Markers for human brain pericytes and smooth muscle cells. J Chem Neuroanat 2018; 92:48-60. [PMID: 29885791 DOI: 10.1016/j.jchemneu.2018.06.001] [Citation(s) in RCA: 150] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 04/20/2018] [Accepted: 06/06/2018] [Indexed: 01/24/2023]
Abstract
Brain pericytes and vascular smooth muscle cells (vSMCs) are a critical component of the neurovascular unit and are important in regulating cerebral blood flow and blood-brain barrier integrity. Identification of subtypes of mural cells in tissue and in vitro is important to any study of their function, therefore we identified distinct mural cell morphologies in neurologically normal post-mortem human brain. Further, the distribution of mural cell markers platelet-derived growth factor receptor-β (PDGFRβ), α-smooth muscle actin (αSMA), CD13, neural/glial antigen-2 (NG2), CD146 and desmin was examined. We determined that PDGFRβ, NG2, CD13, and CD146 were expressed in capillary-associated pericytes. NG2, and CD13 were also present on vSMCs in large vessels, however abundant CD146 and desmin staining was also detected in vSMCs on large vessels, co-labelling with αSMA. To determine whether cultures recapitulated observations from tissue, primary human brain pericytes derived from neurologically normal autopsies were analysed for the presence of pericyte markers by immunocytochemistry, western blotting and qPCR. The proteins observed in brain pericytes in tissue (PDGFRβ, αSMA, desmin, CD146, CD13, and NG2) were present in vitro, validating a panel of proteins that can be used to label brain pericytes and vSMCs in tissue and in vitro. Finally, we showed that the proteins CD146 and desmin that are expressed on large vessels in situ, are also selective markers of a smooth muscle cell phenotype in vitro.
Collapse
Affiliation(s)
- Leon C D Smyth
- Department of Pharmacology and Clinical Pharmacology, Auckland, New Zealand; Centre for Brain Research, Auckland, New Zealand
| | - Justin Rustenhoven
- Department of Pharmacology and Clinical Pharmacology, Auckland, New Zealand; Centre for Brain Research, Auckland, New Zealand
| | - Emma L Scotter
- Department of Pharmacology and Clinical Pharmacology, Auckland, New Zealand; Centre for Brain Research, Auckland, New Zealand
| | - Patrick Schweder
- Centre for Brain Research, Auckland, New Zealand; Auckland City Hospital, Auckland, New Zealand
| | - Richard L M Faull
- Centre for Brain Research, Auckland, New Zealand; Department of Anatomy and Medical Imaging, Auckland, New Zealand
| | - Thomas I H Park
- Department of Pharmacology and Clinical Pharmacology, Auckland, New Zealand; Centre for Brain Research, Auckland, New Zealand; Department of Anatomy and Medical Imaging, Auckland, New Zealand
| | - Mike Dragunow
- Department of Pharmacology and Clinical Pharmacology, Auckland, New Zealand; Centre for Brain Research, Auckland, New Zealand.
| |
Collapse
|
21
|
Bertlich M, Ihler F, Weiss BG, Freytag S, Strupp M, Canis M. Cochlear Pericytes Are Capable of Reversibly Decreasing Capillary Diameter In Vivo After Tumor Necrosis Factor Exposure. Otol Neurotol 2018; 38:e545-e550. [PMID: 29135875 DOI: 10.1097/mao.0000000000001523] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVE The aim of this work was to evaluate the effect of tumor necrosis factor (TNF) and its neutralization with etanercept on the capability of cochlear pericytes to alter capillary diameter in the stria vascularis. METHODS Twelve Dunkin-Hartley guinea pigs were randomly assigned to one of three groups. Each group was treated either with placebo and then placebo, TNF and then placebo, or TNF and then etanercept. Cochlear pericytes were visualized using diaminofluorescein-2-diacetate and intravasal blood flow by fluorescein-dextrane. Vessel diameter at sites of pericyte somas and downstream controls were quantified by specialized software. Values were obtained before treatment, after first treatment with tumor necrosis factor or placebo and after second treatment with etanercept or placebo. RESULTS Overall, 199 pericytes in 12 animals were visualized. After initial treatment with TNF, a significant decrease in vessel diameter at sites of pericyte somas (3.6 ±4.3%, n = 141) compared with placebo and downstream controls was observed. After initial treatment with TNF, the application of etanercept caused a significant increase (3.3 ±5.5%, n = 59) in vessel diameter at the sites of pericyte somata compared with placebo and downstream controls. CONCLUSION We have been able to show that cochlear pericytes are capable of reducing capillary diameter after exposition to TNF. Moreover, the reduction in capillary diameter observed after the application of TNF is revertible after neutralization of tumor necrosis factor by the application of etanercept. It seems that contraction of cochlear pericytes contributes to the regulation of cochlear blood flow.
Collapse
Affiliation(s)
- Mattis Bertlich
- *Department of Otorhinolaryngology-Head and Neck Surgery, University Medical Center Göttingen, Göttingen, Germany †Population Health and Immunity Division, Walter and Eliza Hall Institute ‡Department of Medical Biology, University of Melbourne, Parkville, Australia §Department of Neurology, Munich University Hospital, Munich, Germany
| | | | | | | | | | | |
Collapse
|
22
|
Ghelfi E, Grondin Y, Millet EJ, Bartos A, Bortoni M, Oliveira Gomes Dos Santos C, Trevino-Villarreal HJ, Sepulveda R, Rogers R. In vitro gentamicin exposure alters caveolae protein profile in cochlear spiral ligament pericytes. Proteome Sci 2018; 16:7. [PMID: 29760588 PMCID: PMC5938607 DOI: 10.1186/s12953-018-0132-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 02/04/2018] [Indexed: 12/20/2022] Open
Abstract
Background The aminoglycoside antibiotic gentamicin is an ototoxic drug and has been used experimentally to investigate cochlear damage induced by noise.We have investigated the changes in the protein profile associated with caveolae in gentamicin treated and untreated spiral ligament (SL) pericytes, specialized cells in the blood labyrinth barrier of the inner ear microvasculature. Pericytes from various microvascular beds express caveolae, protein and cholesterol rich microdomains, which can undergo endocytosis and transcytosis to transport small molecules in and out the cells. A different protein profile in transport-specialized caveolae may induce pathological changes affecting the integrity of the blood labyrinth barrier and ultimately contributing to hearing loss. Method Caveolae isolation from treated and untreated cells is achieved through ultracentrifugation of the lysates in discontinuous gradients. Mass spectrometry (LC-MS/MS) analysis identifies the proteins in the two groups. Proteins segregating with caveolae isolated from untreated SL pericytes are then compared to caveolae isolated from SL pericytes treated with the gentamicin for 24 h. Data are analyzed using bioinformatic tools. Results The caveolae proteome in gentamicin treated cells shows that 40% of total proteins are uniquely associated with caveolae during the treatment, and 15% of the proteins normally associated with caveolae in untreated cell are suppressed. Bioinformatic analysis of the data shows a decreased expression of proteins involved in genetic information processing, and an increase in proteins involved in metabolism, vesicular transport and signal transduction in gentamicin treated cells. Several Rab GTPases proteins, ubiquitous transporters, uniquely segregate with caveolae and are significantly enriched in gentamicin treated cells. Conclusion We report that gentamicin exposure modifies protein profile of caveolae from SL pericytes. We identified a pool of proteins which are uniquely segregating with caveolae during the treatment, mainly participating in metabolic and biosynthetic pathways, in transport pathways and in genetic information processing. Finally, we show for the first time proteins associated with caveolae SL pericytes linked to nonsyndromic hearing loss.
Collapse
Affiliation(s)
- Elisa Ghelfi
- 1Harvard T.H. Chan School of Public Health, Department of Environmental Health, MIPS Program, Boston, MA USA
| | - Yohann Grondin
- 1Harvard T.H. Chan School of Public Health, Department of Environmental Health, MIPS Program, Boston, MA USA
| | - Emil J Millet
- 1Harvard T.H. Chan School of Public Health, Department of Environmental Health, MIPS Program, Boston, MA USA
| | - Adam Bartos
- 1Harvard T.H. Chan School of Public Health, Department of Environmental Health, MIPS Program, Boston, MA USA
| | - Magda Bortoni
- 1Harvard T.H. Chan School of Public Health, Department of Environmental Health, MIPS Program, Boston, MA USA
| | - Clara Oliveira Gomes Dos Santos
- 1Harvard T.H. Chan School of Public Health, Department of Environmental Health, MIPS Program, Boston, MA USA.,2Universidade de Sao Paulo, Faculdade de Medicina, Sao Paulo, Brazil
| | | | - Rosalinda Sepulveda
- 1Harvard T.H. Chan School of Public Health, Department of Environmental Health, MIPS Program, Boston, MA USA.,4Universidad Autónoma de Nuevo León, Facultad de Medicina, Monterrey, Mexico
| | - Rick Rogers
- 1Harvard T.H. Chan School of Public Health, Department of Environmental Health, MIPS Program, Boston, MA USA
| |
Collapse
|
23
|
Liu YH, Zhang ZP, Wang Y, Song J, Ma KT, Si JQ, Li L. Electrophysiological properties of strial pericytes and the effect of aspirin on pericyte K+ channels. Mol Med Rep 2017; 17:2861-2868. [PMID: 29257229 PMCID: PMC5783500 DOI: 10.3892/mmr.2017.8194] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 10/06/2017] [Indexed: 01/08/2023] Open
Abstract
The present study was designed to investigate the electrophysiological properties of strial pericytes and the effect of aspirin on pericyte K+ channels. Pericytes were identified by determining their morphological characteristics and using pericyte-associated immunofluorescence techniques. The electrophysiological properties of strial pericytes were observed with a whole-cell patch-clamp technique. Alterations in the outward current of cochlear pericytes in the stria vascularis of guinea pigs were examined following the application of K+ channel retardants. The effects of aspirin on pericyte K+ channels were also evaluated with the whole-cell patch-clamp technique. The results demonstrated that pericytes were desmin positive, and their nuclei were large and surrounded by a small proportion of the cytoplasm. Cytoplasmic processes gradually declined in size as branches grew parallel to the capillary axis. Thus, capillaries were surrounded by tips. The electrophysiological properties of the cochlear pericytes in the stria vascularis of guinea pigs were also determined. The membrane capacitance of the pericytes was 5.9±0.3 pF, while the membrane resistance and resting potential were 2.2±0.3 GΩ and −30.9±1.2 mV, respectively. The current densities of the pericytes (pA/pF) were 3.2±0.7, 10.6±1.0, 15.7±0.9 and 21.3±1.2 at command voltages of 0, +20, +40, and +60 mV, respectively. The K+ channels were activated when the pericytes were within the range of −20 mV to +20 mV, particularly at 0 mV. The inhibition rates of the outward current of cochlear pericytes in the stria vascularis of the guinea pigs were determined by administering iberiotoxin (IBTX) and IBTX + 4-aminopyridine. Once the background leakage current was removed, the following inhibition rates were obtained with 3, 10, 30, 300 and 1,000 µmol/l aspirin: 20.8±4.8, 34.1±6.9, 48.2±6.7, 63.6±7.1 and 65.7±8.1%, respectively. The outward current of the cochlear pericytes in the stria vascularis was inhibited by aspirin with a half maximal inhibitory concentration of 24.5±4.5 µmol/l. The membranes of the pericytes in the stria vascularis are characterized by high-conductance calcium-activated K+ (BKCa) and voltage-dependent K+ (KV) channels. The outward current of the cochlear pericytes in the stria vascularis of guinea pigs was inhibited by aspirin in a concentration-dependent manner. In addition, BKCa and KV channels were inhibited by aspirin.
Collapse
Affiliation(s)
- Yan-Hui Liu
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Zhi-Ping Zhang
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Yang Wang
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Jia Song
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Ke-Tao Ma
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Jun-Qiang Si
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Li Li
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| |
Collapse
|
24
|
Bertlich M, Ihler F, Weiss BG, Freytag S, Strupp M, Jakob M, Canis M. Role of capillary pericytes and precapillary arterioles in the vascular mechanism of betahistine in a guinea pig inner ear model. Life Sci 2017; 187:17-21. [DOI: 10.1016/j.lfs.2017.08.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 08/03/2017] [Accepted: 08/14/2017] [Indexed: 10/19/2022]
|
25
|
Tumor Necrosis Factor-induced Decrease of Cochlear Blood Flow Can Be Reversed by Etanercept or JTE-013. Otol Neurotol 2017; 37:e203-8. [PMID: 27295443 DOI: 10.1097/mao.0000000000001095] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
HYPOTHESIS This study aimed to quantify the effects of tumor necrosis factor (TNF) inhibitor Etanercept and sphingosine-1-phosphate receptor 2 antagonist JTE-013 on cochlear blood flow in guinea pigs after TNF-induced decrease. BACKGROUND Sudden sensorineural hearing loss is a common cause for disability and reduced quality of life. Good understanding of the pathophysiology and strong evidence-based therapy concepts are still missing. In various inner ear disorders, inflammation and impairment of cochlear blood flow (CBF) have been considered factors in the pathophysiology. A central mediator of inflammation and microcirculation in the cochlea is TNF. S1P acts downstream in one TNF pathway. METHODS Cochlea lateral wall vessels were exposed surgically and assessed by intravital microscopy in guinea pigs in vivo. Twenty-eight animals were randomly distributed into four groups of seven each. Exposed vessels were superfused by TNF (5.0 ng/ml) and afterward repeatedly either by Etanercept (1.0 μg/ml), JTE-013 (10 μmol/L), or vehicle (0.9 % NaCl solution or ethanol: phosphate-buffered saline buffer, respectively). RESULTS After decreasing CBF with TNF (p <0.001, two-way RM ANOVA), both treatments reversed CBF, compared with vehicle (p <0.001, two-way RM ANOVA). The comparison of the vehicle groups showed no difference (p = 0.969, two-way RM ANOVA), while there was also no difference between the treatment groups (p = 0.850, two-way RM ANOVA). CONCLUSION Both Etanercept and JTE-013 reverse the decreasing effect of TNF on cochlear blood flow and, therefore, TNF and the S1P-signalling pathway might be targets for treatment of microcirculation-related hearing loss.
Collapse
|
26
|
Sweeney MD, Ayyadurai S, Zlokovic BV. Pericytes of the neurovascular unit: key functions and signaling pathways. Nat Neurosci 2017; 19:771-83. [PMID: 27227366 DOI: 10.1038/nn.4288] [Citation(s) in RCA: 731] [Impact Index Per Article: 104.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 02/29/2016] [Indexed: 12/12/2022]
Abstract
Pericytes are vascular mural cells embedded in the basement membrane of blood microvessels. They extend their processes along capillaries, pre-capillary arterioles and post-capillary venules. CNS pericytes are uniquely positioned in the neurovascular unit between endothelial cells, astrocytes and neurons. They integrate, coordinate and process signals from their neighboring cells to generate diverse functional responses that are critical for CNS functions in health and disease, including regulation of the blood-brain barrier permeability, angiogenesis, clearance of toxic metabolites, capillary hemodynamic responses, neuroinflammation and stem cell activity. Here we examine the key signaling pathways between pericytes and their neighboring endothelial cells, astrocytes and neurons that control neurovascular functions. We also review the role of pericytes in CNS disorders including rare monogenic diseases and complex neurological disorders such as Alzheimer's disease and brain tumors. Finally, we discuss directions for future studies.
Collapse
Affiliation(s)
- Melanie D Sweeney
- Department of Physiology and Biophysics, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA.,Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Shiva Ayyadurai
- Systems Biology Group, CytoSolve Research Division, Cambridge, Massachusetts, USA
| | - Berislav V Zlokovic
- Department of Physiology and Biophysics, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA.,Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| |
Collapse
|
27
|
Kisler K, Nelson AR, Montagne A, Zlokovic BV. Cerebral blood flow regulation and neurovascular dysfunction in Alzheimer disease. Nat Rev Neurosci 2017; 18:419-434. [PMID: 28515434 PMCID: PMC5759779 DOI: 10.1038/nrn.2017.48] [Citation(s) in RCA: 774] [Impact Index Per Article: 110.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cerebral blood flow (CBF) regulation is essential for normal brain function. The mammalian brain has evolved a unique mechanism for CBF control known as neurovascular coupling. This mechanism ensures a rapid increase in the rate of CBF and oxygen delivery to activated brain structures. The neurovascular unit is composed of astrocytes, mural vascular smooth muscle cells and pericytes, and endothelia, and regulates neurovascular coupling. This Review article examines the cellular and molecular mechanisms within the neurovascular unit that contribute to CBF control, and neurovascular dysfunction in neurodegenerative disorders such as Alzheimer disease.
Collapse
Affiliation(s)
- Kassandra Kisler
- Zilkha Neurogenetic Institute, 1501 San Pablo Street, Los Angeles, California 90089, USA
| | - Amy R Nelson
- Zilkha Neurogenetic Institute, 1501 San Pablo Street, Los Angeles, California 90089, USA
| | - Axel Montagne
- Zilkha Neurogenetic Institute, 1501 San Pablo Street, Los Angeles, California 90089, USA
| | - Berislav V Zlokovic
- Zilkha Neurogenetic Institute, 1501 San Pablo Street, Los Angeles, California 90089, USA
| |
Collapse
|
28
|
Pericyte degeneration leads to neurovascular uncoupling and limits oxygen supply to brain. Nat Neurosci 2017; 20:406-416. [PMID: 28135240 PMCID: PMC5323291 DOI: 10.1038/nn.4489] [Citation(s) in RCA: 344] [Impact Index Per Article: 49.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Accepted: 12/30/2016] [Indexed: 12/11/2022]
Abstract
Pericytes are perivascular mural cells of brain capillaries that are positioned centrally within the neurovascular unit between endothelial cells, astrocytes and neurons. This unique position allows them to play a major role in regulating key neurovascular functions of the brain. The role of pericytes in the regulation of cerebral blood flow (CBF) and neurovascular coupling remains, however, debatable. Using loss-of-function pericyte-deficient mice, here we show that pericyte degeneration diminishes global and individual capillary CBF responses to neuronal stimulus resulting in neurovascular uncoupling, reduced oxygen supply to brain and metabolic stress. We show that these neurovascular deficits lead over time to impaired neuronal excitability and neurodegenerative changes. Thus, pericyte degeneration as seen in neurological disorders such as Alzheimer’s disease may contribute to neurovascular dysfunction and neurodegeneration associated with human disease.
Collapse
|
29
|
Two-photon microscopy allows imaging and characterization of cochlear microvasculature in vivo. BIOMED RESEARCH INTERNATIONAL 2015; 2015:154272. [PMID: 25883941 PMCID: PMC4390612 DOI: 10.1155/2015/154272] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 02/26/2015] [Accepted: 03/02/2015] [Indexed: 11/18/2022]
Abstract
Impairment of cochlear blood flow has been discussed as factor in the pathophysiology of various inner ear disorders. However, the microscopic study of cochlear microcirculation is limited due to small scale and anatomical constraints. Here, two-photon fluorescence microscopy is applied to visualize cochlear microvessels. Guinea pigs were injected with Fluorescein isothiocyanate- or Texas red-dextrane as plasma marker. Intravital microscopy was performed in four animals and explanted cochleae from four animals were studied. The vascular architecture of the cochlea was visualized up to a depth of 90.0±22.7 μm. Imaging yielded a mean contrast-to-noise ratio (CNR) of 3.3±1.7. Mean diameter in vivo was 16.5±6.0 μm for arterioles and 8.0±2.4 μm for capillaries. In explanted cochleae, the diameter of radiating arterioles and capillaries was measured with 12.2±1.6 μm and 6.6±1.0 μm, respectively. The difference between capillaries and arterioles was statistically significant in both experimental setups (P<0.001 and P=0.022, two-way ANOVA). Measured vessel diameters in vivo and ex vivo were in agreement with published data. We conclude that two-photon fluorescence microscopy allows the investigation of cochlear microvessels and is potentially a valuable tool for inner ear research.
Collapse
|
30
|
Shi X, Zhang F, Urdang Z, Dai M, Neng L, Zhang J, Chen S, Ramamoorthy S, Nuttall AL. Thin and open vessel windows for intra-vital fluorescence imaging of murine cochlear blood flow. Hear Res 2014; 313:38-46. [PMID: 24780131 DOI: 10.1016/j.heares.2014.04.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Revised: 03/07/2014] [Accepted: 04/15/2014] [Indexed: 12/20/2022]
Abstract
Normal microvessel structure and function in the cochlea is essential for maintaining the ionic and metabolic homeostasis required for hearing function. Abnormal cochlear microcirculation has long been considered an etiologic factor in hearing disorders. A better understanding of cochlear blood flow (CoBF) will enable more effective amelioration of hearing disorders that result from aberrant blood flow. However, establishing the direct relationship between CoBF and other cellular events in the lateral wall and response to physio-pathological stress remains a challenge due to the lack of feasible interrogation methods and difficulty in accessing the inner ear. Here we report on new methods for studying the CoBF in a mouse model using a thin or open vessel-window in combination with fluorescence intra-vital microscopy (IVM). An open vessel-window enables investigation of vascular cell biology and blood flow permeability, including pericyte (PC) contractility, bone marrow cell migration, and endothelial barrier leakage, in wild type and fluorescent protein-labeled transgenic mouse models with high spatial and temporal resolution. Alternatively, the thin vessel-window method minimizes disruption of the homeostatic balance in the lateral wall and enables study CoBF under relatively intact physiological conditions. A thin vessel-window method can also be used for time-based studies of physiological and pathological processes. Although the small size of the mouse cochlea makes surgery difficult, the methods are sufficiently developed for studying the structural and functional changes in CoBF under normal and pathological conditions.
Collapse
Affiliation(s)
- Xiaorui Shi
- Oregon Hearing Research Center, Department of Otolaryngology/Head & Neck Surgery, Oregon Health & Science University, Portland, OR, USA.
| | - Fei Zhang
- Oregon Hearing Research Center, Department of Otolaryngology/Head & Neck Surgery, Oregon Health & Science University, Portland, OR, USA
| | - Zachary Urdang
- Oregon Hearing Research Center, Department of Otolaryngology/Head & Neck Surgery, Oregon Health & Science University, Portland, OR, USA
| | - Min Dai
- Oregon Hearing Research Center, Department of Otolaryngology/Head & Neck Surgery, Oregon Health & Science University, Portland, OR, USA
| | - Lingling Neng
- Oregon Hearing Research Center, Department of Otolaryngology/Head & Neck Surgery, Oregon Health & Science University, Portland, OR, USA
| | - Jinhui Zhang
- Oregon Hearing Research Center, Department of Otolaryngology/Head & Neck Surgery, Oregon Health & Science University, Portland, OR, USA
| | - Songlin Chen
- Oregon Hearing Research Center, Department of Otolaryngology/Head & Neck Surgery, Oregon Health & Science University, Portland, OR, USA
| | - Sripriya Ramamoorthy
- Oregon Hearing Research Center, Department of Otolaryngology/Head & Neck Surgery, Oregon Health & Science University, Portland, OR, USA
| | - Alfred L Nuttall
- Oregon Hearing Research Center, Department of Otolaryngology/Head & Neck Surgery, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
31
|
Ihler F, Sharaf K, Bertlich M, Strieth S, Reichel CA, Berghaus A, Canis M. Etanercept Prevents Decrease of Cochlear Blood Flow Dose-Dependently Caused by Tumor Necrosis Factor Alpha. Ann Otol Rhinol Laryngol 2013; 122:468-73. [DOI: 10.1177/000348941312200711] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Objectives: Tumor necrosis factor alpha (TNF-alpha) is a mediator of inflammation and microcirculation in the cochlea. This study aimed to quantify the effect of a local increase of TNF-alpha and study the effect of its interaction with etanercept on cochlear microcirculation. Methods: Cochlear lateral wall vessels were exposed surgically and assessed by intravital microscopy in guinea pigs in vivo. First, 24 animals were randomly distributed into 4 groups of 6 each. Exposed vessels were superfused repeatedly either with 1 of 3 different concentrations of TNF-alpha (5.0, 0.5, and 0.05 ng/mL) or with placebo (0.9% saline solution). Second, 12 animals were randomly distributed into 2 groups of 6 each. Vessels were pretreated with etanercept (1.0 μg/mL) or placebo (0.9% saline solution), and then treated by repeated superfusion with TNF-alpha (5.0 ng/mL). Results: TNF-alpha was shown to be effective in decreasing cochlear blood flow at a dose of 5.0 ng/mL (p < 0.01, analysis of variance on ranks). Lower concentrations or placebo treatment did not lead to significant changes. After pretreatment with etanercept, TNF-alpha at a dose of 5.0 ng/mL no longer led to a change in cochlear blood flow. Conclusions: The decreasing effect that TNF-alpha has on cochlear blood flow is dose-dependent. Etanercept abrogates this effect.
Collapse
|
32
|
Arpornchayanon W, Canis M, Ihler F, Settevendemie C, Strieth S. TNF-α inhibition using etanercept prevents noise-induced hearing loss by improvement of cochlear blood flow in vivo. Int J Audiol 2013; 52:545-52. [PMID: 23786392 DOI: 10.3109/14992027.2013.790564] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Exposure to loud noise can impair cochlear microcirculation and cause noise-induced hearing loss (NIHL). TNF-α signaling has been shown to be activated in NIHL and to control spiral modiolar artery vasoconstriction that regulates cochlear microcirculation. It was the aim of this experimental study to analyse the effects of the TNF-α inhibitor etanercept on cochlear microcirculation and hearing threshold shift in NIHL in vivo. DESIGN After assessment of normacusis using ABR, loud noise (106 dB SPL, 30 minutes) was applied on both ears in guinea pigs. Etanercept was administered systemically after loud noise exposure while control animals received a saline solution. In vivo fluorescence microscopy of strial capillaries was performed after surgical exposure of the cochlea for microcirculatory analysis. ABR measurements were derived from the contralateral ear. STUDY SAMPLE Guinea pigs (n = 6, per group). RESULTS Compared to controls, cochlear blood flow in strial capillary segments was significantly increased in etanercept-treated animals. Additionally, hearing threshold was preserved in animals receiving the TNF-α inhibitor in contrast to a significant threshold raising in controls. CONCLUSIONS TNF-α inhibition using etanercept improves cochlear microcirculation and protects hearing levels after loud noise exposure and appears as a promising treatment strategy for human NIHL.
Collapse
|
33
|
Heinrich UR, Helling K. Nitric oxide--a versatile key player in cochlear function and hearing disorders. Nitric Oxide 2012; 27:106-16. [PMID: 22659013 DOI: 10.1016/j.niox.2012.05.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Revised: 05/10/2012] [Accepted: 05/24/2012] [Indexed: 12/13/2022]
Abstract
Nitric oxide (NO) is a signaling molecule which can generally be formed by three nitric oxide synthases (NOS). Two of them, the endothelial nitric oxide synthase (eNOS) and the neural nitric oxide synthase (nNOS), are calcium/calmodulin-dependent and constitutively expressed in many cell types. Both isoforms are found in the vertebrate cochlea. The inducible nitric oxide synthase (iNOS) is independent of calcium and normally not detectable in the un-stimulated cochlea. In the inner ear, as in other tissues, NO was identified as a multitask molecule involved in various processes such as neurotransmission and neuromodulation. In addition, increasing evidence demonstrates that the NO-dependent processes of cell protection or, alternatively, cell destruction seem to depend, among other things, on changes in the local cochlear NO-concentration. These alterations can occur at the cellular level or within a distinct cell population both leading to an NO-imbalance within the hearing organ. This dysfunction can result in hearing loss or even in deafness. In cases of cochlear malfunction, regulatory systems such as the gap junction system, the blood vessels or the synaptic region might be affected temporarily or permanently by an altered NO-level. This review discusses potential cellular mechanisms how NO might contribute to different forms of hearing disorders. Approaches of NO-reduction are evaluated and the transfer of results obtained from experimental animal models to human medication is discussed.
Collapse
Affiliation(s)
- Ulf-Rüdiger Heinrich
- Department of Otorhinolaryngology, Head and Neck Surgery, University Medical Center of The Johannes Gutenberg-University Mainz, Germany.
| | | |
Collapse
|
34
|
Abstract
According to current knowledge, it must be assumed that temporary idiopathic hearing loss and its spontaneous remission are based on mechanical and/or pathological alterations in the inner ear. The causal mechanisms might be based on inter-individual variations. Induced by dose-dependent activators, temporary as well as permanent damage might occur. Sudden hearing loss may be initiated by an increase in the local nitric oxide (NO) concentration. Spontaneous remission, i.e. functional restoration, can be explained by a local decrease in the NO concentration. In this context, regulatory systems such as the gap-junction system, blood vessels or synapses might be affected. In addition, alterations in the hormone level of estrogen and mineralocorticoids, as well as cellular glutathione and vitamin levels, might lead to temporary alterations in the inner ear. Recent experimental findings indicate a role for the shuttle protein Survivin in the spontaneous remission of sudden hearing loss.
Collapse
|
35
|
Abstract
Normal blood supply to the cochlea is critically important for establishing the endocochlear potential and sustaining production of endolymph. Abnormal cochlear microcirculation has long been considered an etiologic factor in noise-induced hearing loss, age-related hearing loss (presbycusis), sudden hearing loss or vestibular function, and Meniere's disease. Knowledge of the mechanisms underlying the pathophysiology of cochlear microcirculation is of fundamental clinical importance. A better understanding of cochlear blood flow (CoBF) will enable more effective management of hearing disorders resulting from aberrant blood flow. This review focuses on recent discoveries and findings related to the physiopathology of the cochlear microvasculature.
Collapse
Affiliation(s)
- Xiaorui Shi
- Oregon Hearing Research Center (NRC04), Department of Otolaryngology/Head & Neck Surgery, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA.
| |
Collapse
|
36
|
Armulik A, Genové G, Betsholtz C. Pericytes: developmental, physiological, and pathological perspectives, problems, and promises. Dev Cell 2011; 21:193-215. [PMID: 21839917 DOI: 10.1016/j.devcel.2011.07.001] [Citation(s) in RCA: 1865] [Impact Index Per Article: 143.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Pericytes, the mural cells of blood microvessels, have recently come into focus as regulators of vascular morphogenesis and function during development, cardiovascular homeostasis, and disease. Pericytes are implicated in the development of diabetic retinopathy and tissue fibrosis, and they are potential stromal targets for cancer therapy. Some pericytes are probably mesenchymal stem or progenitor cells, which give rise to adipocytes, cartilage, bone, and muscle. However, there is still confusion about the identity, ontogeny, and progeny of pericytes. Here, we review the history of these investigations, indicate emerging concepts, and point out problems and promise in the field of pericyte biology.
Collapse
Affiliation(s)
- Annika Armulik
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, SE-171 77 Stockholm, Sweden
| | | | | |
Collapse
|
37
|
Dai M, Yang Y, Shi X. Lactate dilates cochlear capillaries via type V fibrocyte-vessel coupling signaled by nNOS. Am J Physiol Heart Circ Physiol 2011; 301:H1248-54. [PMID: 21856924 DOI: 10.1152/ajpheart.00315.2011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Transduction of sound in the inner ear demands tight control over delivery of oxygen and glucose. However, the mechanisms underlying the control of regional blood flow are not yet fully understood. In this study, we report a novel local control mechanism that regulates cochlear blood flow to the stria vascularis, a high energy-consuming region of the inner ear. We found that extracellular lactate had a vasodilatory effect on the capillaries of the spiral ligament under both in vitro and in vivo conditions. The lactate, acting through monocarboxylate transporter 1 (MCT1), initiated neuronal nitric oxide (NO) synthase (nNOS) and catalyzed production of NO for the vasodilation. Blocking MCT1 with the MCT blocker, α-cyano-4-hydroxycinnamate (CHC), or a suppressing NO production with either the nonspecific inhibitor of NO synthase, N(G)-nitro-L-arginine methyl ester (L-NAME), or either of two selective nNOS inhibitors, 3-bromo-7-nitroindazole or (4S)-N-(4-amino-5[aminoethyl]aminopentyl)-N'-nitroguanidine (TFA), totally abolished the lactate-induced vasodilation. Pretreatment with the selective endothelial NO synthase inhibitor, L-N(5)-(1-iminoethyl)ornithine (L-NIO), eliminated the inhibition of lactate-induced vessel dilation. With immunohistochemical labeling, we found the expression of MCT1 and nNOS in capillary-coupled type V fibrocytes. The data suggest that type V fibrocytes are the source of the lactate-induced NO. Cochlear microvessel tone, regulated by lactate, is mediated by an NO-signaled coupling of fibrocytes and capillaries.
Collapse
Affiliation(s)
- Min Dai
- Department of Otolaryngology/Head and Neck Surgery, Oregon Hearing Research Center, Oregon Health and Science University, Portland, Oregon 97239-3098, USA
| | | | | |
Collapse
|
38
|
Dai M, Shi X. Fibro-vascular coupling in the control of cochlear blood flow. PLoS One 2011; 6:e20652. [PMID: 21673815 PMCID: PMC3106013 DOI: 10.1371/journal.pone.0020652] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2010] [Accepted: 05/06/2011] [Indexed: 12/20/2022] Open
Abstract
Background Transduction of sound in the cochlea is metabolically demanding. The lateral
wall and hair cells are critically vulnerable to hypoxia, especially at high
sound levels, and tight control over cochlear blood flow (CBF) is a
physiological necessity. Yet despite the importance of CBF for hearing,
consensus on what mechanisms are involved has not been obtained. Methodology/Principal Findings We report on a local control mechanism for regulating inner ear blood flow
involving fibrocyte signaling. Fibrocytes in the super-strial region are
spatially distributed near pre-capillaries of the spiral ligament of the
albino guinea pig cochlear lateral wall, as demonstrably shown in
transmission electron microscope and confocal images. Immunohistochemical
techniques reveal the inter-connected fibrocytes to be positive for
Na+/K+ ATPase β1 and S100. The connected fibrocytes display
more Ca2+ signaling than other cells in the cochlear lateral
wall as indicated by fluorescence of a Ca2+ sensor, fluo-4.
Elevation of Ca2+ in fibrocytes, induced by photolytic
uncaging of the divalent ion chelator o-nitrophenyl EGTA,
results in propagation of a Ca2+ signal to neighboring
vascular cells and vasodilation in capillaries. Of more physiological
significance, fibrocyte to vascular cell coupled signaling was found to
mediate the sound stimulated increase in cochlear blood flow (CBF).
Cyclooxygenase-1 (COX-1) was required for capillary dilation. Conclusions/Significance The findings provide the first evidence that signaling between fibrocytes and
vascular cells modulates CBF and is a key mechanism for meeting the cellular
metabolic demand of increased sound activity.
Collapse
Affiliation(s)
- Min Dai
- Oregon Hearing Research Center, Department of Otolaryngology/Head and
Neck Surgery, Oregon Health & Science University, Portland, Oregon, United
States of America
| | - Xiaorui Shi
- Oregon Hearing Research Center, Department of Otolaryngology/Head and
Neck Surgery, Oregon Health & Science University, Portland, Oregon, United
States of America
- The Institute of Microcirculation, Chinese Academy of Medical Sciences
and Peking Union Medical College, Beijing, China
- Department of Otolaryngology, Renji Hospital, Shanghai Jiao Tong
University, Shanghai, China
- * E-mail:
| |
Collapse
|
39
|
Wu T, Dai M, Shi XR, Jiang ZG, Nuttall AL. Functional expression of P2X4 receptor in capillary endothelial cells of the cochlear spiral ligament and its role in regulating the capillary diameter. Am J Physiol Heart Circ Physiol 2011; 301:H69-78. [PMID: 21460192 DOI: 10.1152/ajpheart.01035.2010] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The cochlear lateral wall generates the endocochlear potential (EP), which creates a driving force for the hair cell transduction current and is essential for normal hearing. Blood flow at the cochlear lateral wall is critically important for maintaining the EP. The vulnerability of the EP to hypoxia suggests that the blood flow in the cochlear lateral wall is dynamically and precisely regulated to meet the changing metabolic needs of the cochlear lateral wall. It has been reported that ATP, an important extracellular signaling molecule, plays an essential role in regulating cochlear blood flow. However, the cellular mechanism underlying ATP-induced regional blood flow changes has not been investigated. In the current study, we demonstrate that 1) the P2X4 receptor is expressed in endothelial cells (ECs) of spiral ligament (SL) capillaries. 2) ATP elicits a characteristic current through P2X4 on ECs in a dose-dependent manner (EC(50) = 0.16 mM). The ATP current has a reversal potential at ∼0 mV; is inhibited by 5-(3-bromophenyl)-1,3-dihydro-2H-benzofuro[3,2-e]-1,4-diazepin-2-one (5-BDBD), LaCl(3), pyridoxal phosphate-6-azo(benzene-2,4-disulfonic acid) tetrasodium salt hydrate (PPADS), and extracellular acidosis; and is less sensitive to α,β-methyleneadenosine 5'-triphosphate (α,β-MeATP) and 2'- and 3'-O-(4-benzoyl-benzoyl) adenosine 5'-triphosphate (BzATP). 3) ATP elicits a transient increase of intracellular Ca(2+) in ECs. 4) In accordance with the above in vitro findings, perilymphatic ATP (1 mM) caused dilation in SL capillaries in vivo by 11.5%. N(ω)-nitro-l-arginine methyl ester hydrochloride (l-NAME), a nonselective inhibitor of nitric oxide synthase, or 5-BDBD, the specific P2X4 inhibitor, significantly blocked the dilation. These findings support our hypothesis that extracellular ATP regulates cochlear lateral blood flow through P2X4 activation in ECs.
Collapse
Affiliation(s)
- T Wu
- Oregon Hearing Research Center, NRC04, Oregon Health & Science Univ., 3181 S.W. Sam Jackson Park Rd., Portland, OR 97239, USA
| | | | | | | | | |
Collapse
|
40
|
Slice cultures as a model to study neurovascular coupling and blood brain barrier in vitro. Cardiovasc Psychiatry Neurol 2011; 2011:646958. [PMID: 21350722 PMCID: PMC3042620 DOI: 10.1155/2011/646958] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Accepted: 12/24/2010] [Indexed: 11/18/2022] Open
Abstract
Proper neuronal functioning depends on a strictly regulated interstitial environment and tight coupling of neuronal and metabolic activity involving adequate vascular responses. These functions take place at the blood brain barrier (BBB) composed of endothelial cells, basal lamina covered with pericytes, and the endfeet of perivascular astrocytes. In conventional in vitro models of the BBB, some of these components are missing.
Here we describe a new model system for studying BBB and neurovascular coupling by using confocal microscopy and fluorescence staining protocols in organotypic hippocampal slice cultures.
An elaborated network of vessels is retained in culture in spite of the absence of blood flow. Application of calcein-AM either from the interstitial or from the luminal side resulted in different staining patterns indicating the maintenance of a barrier. By contrast, the ethidium derivative MitoSox penetrated perivascular basal lamina and revealed free radical formation in contractile cells embracing the vessels, likely pericytes.
Collapse
|
41
|
Scherer EQ, Yang J, Canis M, Reimann K, Ivanov K, Diehl CD, Backx PH, Wier WG, Strieth S, Wangemann P, Voigtlaender-Bolz J, Lidington D, Bolz SS. Tumor necrosis factor-α enhances microvascular tone and reduces blood flow in the cochlea via enhanced sphingosine-1-phosphate signaling. Stroke 2010; 41:2618-24. [PMID: 20930159 DOI: 10.1161/strokeaha.110.593327] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND PURPOSE We sought to demonstrate that tumor necrosis factor (TNF)-α, via sphingosine-1-phosphate signaling, has the potential to alter cochlear blood flow and thus, cause ischemic hearing loss. METHODS We performed intravital fluorescence microscopy to measure blood flow and capillary diameter in anesthetized guinea pigs. To measure capillary diameter ex vivo, capillary beds from the gerbil spiral ligament were isolated from the cochlear lateral wall and maintained in an organ bath. Isolated gerbil spiral modiolar arteries, maintained and transfected in organ culture, were used to measure calcium sensitivity (calcium-tone relationship). In a clinical study, a total of 12 adult patients presenting with typical symptoms of sudden hearing loss who were not responsive or only partially responsive to prednisolone treatment were identified and selected for etanercept treatment. Etanercept (25 mg s.c.) was self-administered twice a week for 12 weeks. RESULTS TNF-α induced a proconstrictive state throughout the cochlear microvasculature, which reduced capillary diameter and cochlear blood flow in vivo. In vitro isolated preparations of the spiral modiolar artery and spiral ligament capillaries confirmed these observations. Antagonizing sphingosine-1-phosphate receptor 2 subtype signaling (by 1 μmol/L JTE013) attenuated the effects of TNF-α in all models. TNF-α activated sphingosine kinase 1 (Sk1) and induced its translocation to the smooth muscle cell membrane. Expression of a dominant-negative Sk1 mutant (Sk1(G82D)) eliminated both baseline spiral modiolar artery calcium sensitivity and TNF-α effects, whereas a nonphosphorylatable Sk1 mutant (Sk1(S225A)) blocked the effects of TNF-α only. A small group of etanercept-treated, hearing loss patients recovered according to a 1-phase exponential decay (half-life=1.56 ± 0.20 weeks), which matched the kinetics predicted for a vascular origin. CONCLUSIONS TNF-α indeed reduces cochlear blood flow via activation of vascular sphingosine-1-phosphate signaling. This integrates hearing loss into the family of ischemic microvascular pathologies, with implications for risk stratification, diagnosis, and treatment.
Collapse
Affiliation(s)
- Elias Q Scherer
- Hals-Nasen-Ohrenklinik und Poliklinik, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
PURPOSE OF REVIEW To describe ion and water homeostatic mechanisms in the inner ear, how they are compromised in hearing disorders, and what treatments are employed to restore auditory function. RECENT FINDINGS The ion and water transport functions in the inner ear help maintain the proper endolymph K concentration required for hair cell function. Gene defects and idiopathic alterations in these transport functions cause hearing loss, but often the underlying cause is unknown. Current therapies largely involve glucocorticoid treatment, although the mechanisms of restoration are often undeterminable. Recent studies of these ion homeostatic functions in the ear are characterizing their cellular and molecular control. It is anticipated that future management of these hearing disorders will be more targeted to the cellular processes involved and improve the likelihood of hearing recovery. SUMMARY A better understanding of the ion homeostatic processes in the ear will permit more effective management of their associated hearing disorders. Sufficient insight into many homeostatic hearing disorders has now been attained to usher in a new era of better therapies and improved clinical outcomes.
Collapse
Affiliation(s)
- Dennis R Trune
- Oregon Hearing Research Center, Department of Otolaryngology Head Neck Surgery, Oregon Health & Science University, Portland, Oregon 97239-3098, USA.
| |
Collapse
|
43
|
Current Opinion in Otolaryngology & Head and Neck Surgery. Current world literature. Curr Opin Otolaryngol Head Neck Surg 2010; 18:466-74. [PMID: 20827086 DOI: 10.1097/moo.0b013e32833f3865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|