1
|
Zedde M, Pascarella R. The Cerebrovascular Side of Plasticity: Microvascular Architecture across Health and Neurodegenerative and Vascular Diseases. Brain Sci 2024; 14:983. [PMID: 39451997 PMCID: PMC11506257 DOI: 10.3390/brainsci14100983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 09/24/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
The delivery of nutrients to the brain is provided by a 600 km network of capillaries and microvessels. Indeed, the brain is highly energy demanding and, among a total amount of 100 billion neurons, each neuron is located just 10-20 μm from a capillary. This vascular network also forms part of the blood-brain barrier (BBB), which maintains the brain's stable environment by regulating chemical balance, immune cell transport, and blocking toxins. Typically, brain microvascular endothelial cells (BMECs) have low turnover, indicating a stable cerebrovascular structure. However, this structure can adapt significantly due to development, aging, injury, or disease. Temporary neural activity changes are managed by the expansion or contraction of arterioles and capillaries. Hypoxia leads to significant remodeling of the cerebrovascular architecture and pathological changes have been documented in aging and in vascular and neurodegenerative conditions. These changes often involve BMEC proliferation and the remodeling of capillary segments, often linked with local neuronal changes and cognitive function. Cerebrovascular plasticity, especially in arterioles, capillaries, and venules, varies over different time scales in development, health, aging, and diseases. Rapid changes in cerebral blood flow (CBF) occur within seconds due to increased neural activity. Prolonged changes in vascular structure, influenced by consistent environmental factors, take weeks. Development and aging bring changes over months to years, with aging-associated plasticity often improved by exercise. Injuries cause rapid damage but can be repaired over weeks to months, while neurodegenerative diseases cause slow, varied changes over months to years. In addition, if animal models may provide useful and dynamic in vivo information about vascular plasticity, humans are more complex to investigate and the hypothesis of glymphatic system together with Magnetic Resonance Imaging (MRI) techniques could provide useful clues in the future.
Collapse
Affiliation(s)
- Marialuisa Zedde
- Neurology Unit, Stroke Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Viale Risorgimento 80, 42123 Reggio Emilia, Italy
| | - Rosario Pascarella
- Neuroradiology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Viale Risorgimento 80, 42123 Reggio Emilia, Italy;
| |
Collapse
|
2
|
Shigemoto-Mogami Y, Nakayama-Kitamura K, Sato K. The arrangements of the microvasculature and surrounding glial cells are linked to blood-brain barrier formation in the cerebral cortex. Front Neuroanat 2024; 18:1438190. [PMID: 39170850 PMCID: PMC11335649 DOI: 10.3389/fnana.2024.1438190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 07/29/2024] [Indexed: 08/23/2024] Open
Abstract
The blood-brain barrier (BBB) blocks harmful substances from entering the brain and dictates the central nervous system (CNS)-specific pharmacokinetics. Recent studies have shown that perivascular astrocytes and microglia also control BBB functions, however, information about the formation of BBB glial architecture remains scarce. We investigated the time course of the formation of BBB glial architecture in the rat brain cerebral cortex using Evans blue (EB) and tissue fixable biotin (Sulfo-NHS Biotin). The extent of the leakage into the brain parenchyma showed that the BBB was not formed at postnatal Day 4 (P4). The BBB gradually strengthened and reached a plateau at P15. We then investigated the changes in the configurations of blood vessels, astrocytes, and microglia with age by 3D image reconstruction of the immunohistochemical data. The endfeet of astrocytes covered the blood vessels, and the coverage rate rapidly increased after birth and reached a plateau at P15. Interestingly, microglia were also in contact with the capillaries, and the coverage rate was highest at P15 and stabilized at P30. It was also clarified that the microglial morphology changed from the amoeboid type to the ramified type, while the areas of the respective contact sites became smaller during P4 and P15. These results suggest that the perivascular glial architecture formation of the rat BBB occurs from P4 to P15 because the paracellular transport and the arrangements of perivascular glial cells at P15 are totally the same as those of P30. In addition, the contact style of perivascular microglia dramatically changed during P4-P15.
Collapse
Affiliation(s)
| | | | - Kaoru Sato
- Division of Pharmacology, Laboratory of Neuropharmacology, National Institute of Health Sciences, Kawasaki, Japan
| |
Collapse
|
3
|
Min W, Qin L, Zhang H, López-Giráldez F, Jiang N, Kim Y, Mohan VK, Su M, Murray KN, Grutzendler J, Zhou JH. mTORC1 Signaling in Brain Endothelial Progenitors Contributes to CCM Pathogenesis. Circ Res 2024; 135:e94-e113. [PMID: 38957991 PMCID: PMC11293987 DOI: 10.1161/circresaha.123.324015] [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: 11/18/2023] [Accepted: 06/21/2024] [Indexed: 07/04/2024]
Abstract
BACKGROUND Cerebral vascular malformations (CCMs) are primarily found within the brain, where they result in increased risk for stroke, seizures, and focal neurological deficits. The unique feature of the brain vasculature is the blood-brain barrier formed by the brain neurovascular unit. Recent studies suggest that loss of CCM genes causes disruptions of blood-brain barrier integrity as the inciting events for CCM development. CCM lesions are proposed to be initially derived from a single clonal expansion of a subset of angiogenic venous capillary endothelial cells (ECs) and respective resident endothelial progenitor cells (EPCs). However, the critical signaling events in the subclass of brain ECs/EPCs for CCM lesion initiation and progression are unclear. METHODS Brain EC-specific CCM3-deficient (Pdcd10BECKO) mice were generated by crossing Pdcd10fl/fl mice with Mfsd2a-CreERT2 mice. Single-cell RNA-sequencing analyses were performed by the chromium single-cell platform (10× genomics). Cell clusters were annotated into EC subtypes based on visual inspection and GO analyses. Cerebral vessels were visualized by 2-photon in vivo imaging and tissue immunofluorescence analyses. Regulation of mTOR (mechanistic target of rapamycin) signaling by CCM3 and Cav1 (caveolin-1) was performed by cell biology and biochemical approaches. RESULTS Single-cell RNA-sequencing analyses from P10 Pdcd10BECKO mice harboring visible CCM lesions identified upregulated CCM lesion signature and mitotic EC clusters but decreased blood-brain barrier-associated EC clusters. However, a unique EPC cluster with high expression levels of stem cell markers enriched with mTOR signaling was identified from early stages of the P6 Pdcd10BECKO brain. Indeed, mTOR signaling was upregulated in both mouse and human CCM lesions. Genetic deficiency of Raptor (regulatory-associated protein of mTOR), but not of Rictor (rapamycin-insensitive companion of mTOR), prevented CCM lesion formation in the Pdcd10BECKO model. Importantly, the mTORC1 (mTOR complex 1) pharmacological inhibitor rapamycin suppressed EPC proliferation and ameliorated CCM pathogenesis in Pdcd10BECKO mice. Mechanistic studies suggested that Cav1/caveolae increased in CCM3-depleted EPC-mediated intracellular trafficking and complex formation of the mTORC1 signaling proteins. CONCLUSIONS CCM3 is critical for maintaining blood-brain barrier integrity and CCM3 loss-induced mTORC1 signaling in brain EPCs initiates and facilitates CCM pathogenesis.
Collapse
Affiliation(s)
- Wang Min
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT
- Cardiovascular Medical Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Lingfeng Qin
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT
| | - Haifeng Zhang
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT
| | - Francesc López-Giráldez
- Yale Center for Genomic Analysis, Department of Genetics, Yale University School of Medicine, New Haven, CT
| | - Ning Jiang
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT
| | - Yeaji Kim
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT
| | - Varsha K. Mohan
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT
| | - Minhong Su
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT
| | - Katie N Murray
- Department of Neurobiology, Yale University School of Medicine, New Haven, CT
| | - Jaime Grutzendler
- Department of Neurobiology, Yale University School of Medicine, New Haven, CT
| | - Jenny Huanjiao Zhou
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
4
|
Harris S, Chinnery HR, Semple BD, Mychasiuk R. Shaking Up Our Approach: The Need for Characterization and Optimization of Pre-clinical Models of Infant Abusive Head Trauma. J Neurotrauma 2024; 41:1853-1870. [PMID: 38497766 DOI: 10.1089/neu.2023.0598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2024] Open
Abstract
Traumatic brain injuries (TBIs) are a large societal and individual burden. In the first year of life, the vast majority of these injuries are the result of inflicted abusive events by a trusted caregiver. Abusive head trauma (AHT) in infants, formerly known as shaken baby syndrome, is the leading cause of inflicted mortality and morbidity in this population. In this review we address clinical diagnosis, symptoms, prognosis, and neuropathology of AHT, emphasizing the burden of repetitive AHT. Next, we consider existing animal models of AHT, and we evaluate key features of an ideal model, highlighting important developmental milestones in children most vulnerable to AHT. We draw on insights from other injury models, such as repetitive, mild TBIs (RmTBIs), post-traumatic epilepsy (PTE), hypoxic-ischemic injuries, and maternal neglect, to speculate on key knowledge gaps and underline important new opportunities in pre-clinical AHT research. Finally, potential treatment options to facilitate healthy development in children following an AHT are considered. Together, this review aims to drive the field toward optimized, well-characterized animal models of AHT, which will allow for greater insight into the underlying neuropathological and neurobehavioral consequences of AHT.
Collapse
Affiliation(s)
- Sydney Harris
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Holly R Chinnery
- Department of Optometry and Vision Science, University of Melbourne, Parkville, Victoria, Australia
| | - Bridgette D Semple
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Neurology, Alfred Health, Prahran, Victoria, Australia
- Department of Medicine (Royal Melbourne Hospital), University of Melbourne, Parkville, Victoria, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Neurology, Alfred Health, Prahran, Victoria, Australia
| |
Collapse
|
5
|
Bennett HC, Zhang Q, Wu YT, Manjila SB, Chon U, Shin D, Vanselow DJ, Pi HJ, Drew PJ, Kim Y. Aging drives cerebrovascular network remodeling and functional changes in the mouse brain. Nat Commun 2024; 15:6398. [PMID: 39080289 PMCID: PMC11289283 DOI: 10.1038/s41467-024-50559-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 07/15/2024] [Indexed: 08/02/2024] Open
Abstract
Aging is frequently associated with compromised cerebrovasculature and pericytes. However, we do not know how normal aging differentially impacts vascular structure and function in different brain areas. Here we utilize mesoscale microscopy methods and in vivo imaging to determine detailed changes in aged murine cerebrovascular networks. Whole-brain vascular tracing shows an overall ~10% decrease in vascular length and branching density with ~7% increase in vascular radii in aged brains. Light sheet imaging with 3D immunolabeling reveals increased arteriole tortuosity of aged brains. Notably, vasculature and pericyte densities show selective and significant reductions in the deep cortical layers, hippocampal network, and basal forebrain areas. We find increased blood extravasation, implying compromised blood-brain barrier function in aged brains. Moreover, in vivo imaging in awake mice demonstrates reduced baseline and on-demand blood oxygenation despite relatively intact neurovascular coupling. Collectively, we uncover regional vulnerabilities of cerebrovascular network and physiological changes that can mediate cognitive decline in normal aging.
Collapse
Affiliation(s)
- Hannah C Bennett
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Qingguang Zhang
- Center for Neural Engineering, Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Physiology, Michigan State University, East Lansing, MI, 48824, USA
| | - Yuan-Ting Wu
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
- Department of Neurosurgery, Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Steffy B Manjila
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Uree Chon
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
- Neurosciences Graduate Program, Stanford University, Stanford, CA, 94305, USA
| | - Donghui Shin
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Daniel J Vanselow
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Hyun-Jae Pi
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA
| | - Patrick J Drew
- Center for Neural Engineering, Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Biomedical Engineering, Biology, and Neurosurgery, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Yongsoo Kim
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, PA, 17033, USA.
- Center for Neural Engineering, Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA, 16802, USA.
| |
Collapse
|
6
|
Tessier A, Ruze AJ, Varlet I, Laïb EMH, Royer E, Bernard M, Viola A, Perles-Barbacaru TA. Quantitative MRI of Gd-DOTA Accumulation in the Mouse Brain After Intraperitoneal Administration: Validation by Mass Spectrometry. J Magn Reson Imaging 2024; 60:316-324. [PMID: 37811700 DOI: 10.1002/jmri.29034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 09/19/2023] [Accepted: 09/19/2023] [Indexed: 10/10/2023] Open
Abstract
BACKGROUND In mice, intraperitoneal (ip) contrast agent (CA) administration is convenient for mapping microvascular parameters over a long-time window. However, continuous quantitative MRI of CA accumulation in brain over hours is still missing. PURPOSE To validate a quantitative time-resolved MRI technique for mapping the CA kinetics in brain upon ip administration. STUDY TYPE Prospective, animal model. SPECIMEN 25 C57Bl/6JRj mice underwent MRI. FIELD STRENGTH/SEQUENCE 7-T, gradient echo sequence. ASSESSMENT Gd-DOTA concentration was monitored by MRI (25 s/repetition) over 135 minutes with (N = 15) and without (N = 10) ip mannitol challenge (5 g/kg). After the final repetition, the brains were sampled to quantify gadolinium by mass spectrometry (MS). Upon manual brain segmentation, the average gadolinium concentration was compared with the MS quantification in transcardially perfused (N = 20) and unperfused (N = 5) mice. Precontrast T1-maps were acquired in 8 of 25 mice. STATISTICAL TESTS One-tailed Spearman and Pearson correlation between gadolinium quantification by MRI and by MS, D'Agostino-Pearson test for normal distribution, Bland-Altman analysis to evaluate the agreement between MRI and MS. Significance was set at P-value <0.05. RESULTS MRI showed that ip administered CA reached the blood compartment (>5 mM) within 10 minutes and accumulated continuously for 2 hours in cerebrospinal fluid (>1 mM) and in brain tissue. The MRI-derived concentration maps showed interindividual differences in CA accumulation (from 0.47 to 0.81 mM at 2 hours) with a consistent distribution resembling the pathways of the glymphatic system. The average in-vivo brain concentration 2 hours post-CA administration correlated significantly (r = 0.8206) with the brain gadolinium quantification by MS for N = 21 paired observations available. DATA CONCLUSION The presented experimental and imaging protocol may be convenient for monitoring the spatiotemporal pattern of CA uptake and clearance in the mouse brain over 2 hours. The quantification of the CA from the MRI signal in brain is corroborated by MS. EVIDENCE LEVEL N/A TECHNICAL EFFICACY: Stage 1.
Collapse
Affiliation(s)
- Anthony Tessier
- CNRS, Center for Magnetic Resonance Imaging in Biology and Medicine (CRMBM, UMR CNRS 7339), Aix-Marseille University, Marseille, France
- Department of Medical Imaging, Sainte-Anne Military Teaching Hospital (Hôpital d'Instruction des Armées), Toulon, France
| | - Anthony J Ruze
- CNRS, Center for Magnetic Resonance Imaging in Biology and Medicine (CRMBM, UMR CNRS 7339), Aix-Marseille University, Marseille, France
| | - Isabelle Varlet
- CNRS, Center for Magnetic Resonance Imaging in Biology and Medicine (CRMBM, UMR CNRS 7339), Aix-Marseille University, Marseille, France
| | - Estelle M H Laïb
- CNRS, Center for Magnetic Resonance Imaging in Biology and Medicine (CRMBM, UMR CNRS 7339), Aix-Marseille University, Marseille, France
| | - Emilien Royer
- CNRS, Center for Magnetic Resonance Imaging in Biology and Medicine (CRMBM, UMR CNRS 7339), Aix-Marseille University, Marseille, France
| | - Monique Bernard
- CNRS, Center for Magnetic Resonance Imaging in Biology and Medicine (CRMBM, UMR CNRS 7339), Aix-Marseille University, Marseille, France
| | - Angèle Viola
- CNRS, Center for Magnetic Resonance Imaging in Biology and Medicine (CRMBM, UMR CNRS 7339), Aix-Marseille University, Marseille, France
| | - Teodora-Adriana Perles-Barbacaru
- CNRS, Center for Magnetic Resonance Imaging in Biology and Medicine (CRMBM, UMR CNRS 7339), Aix-Marseille University, Marseille, France
| |
Collapse
|
7
|
Srivastava T, Nguyen H, Haden G, Diba P, Sowa S, LaNguyen N, Reed-Dustin W, Zhu W, Gong X, Harris EN, Baltan S, Back SA. TSG-6-Mediated Extracellular Matrix Modifications Regulate Hypoxic-Ischemic Brain Injury. J Neurosci 2024; 44:e2215232024. [PMID: 38569926 PMCID: PMC11112645 DOI: 10.1523/jneurosci.2215-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/24/2024] [Accepted: 03/27/2024] [Indexed: 04/05/2024] Open
Abstract
Proteoglycans containing link domains modify the extracellular matrix (ECM) to regulate cellular homeostasis and can also sensitize tissues/organs to injury and stress. Hypoxic-ischemic (H-I) injury disrupts cellular homeostasis by activating inflammation and attenuating regeneration and repair pathways. In the brain, the main component of the ECM is the glycosaminoglycan hyaluronic acid (HA), but whether HA modifications of the ECM regulate cellular homeostasis and response to H-I injury is not known. In this report, employing both male and female mice, we demonstrate that link-domain-containing proteoglycan, TNFα-stimulated gene-6 (TSG-6), is active in the brain from birth onward and differentially modifies ECM HA during discrete neurodevelopmental windows. ECM HA modification by TSG-6 enables it to serve as a developmental switch to regulate the activity of the Hippo pathway effector protein, yes-associated protein 1 (YAP1), in the maturing brain and in response to H-I injury. Mice that lack TSG-6 expression display dysregulated expression of YAP1 targets, excitatory amino acid transporter 1 (EAAT1; glutamate-aspartate transporter) and 2 (EAAT2; glutamate transporter-1). Dysregulation of YAP1 activation in TSG-6-/- mice coincides with age- and sex-dependent sensitization of the brain to H-I injury such that 1-week-old neonates display an anti-inflammatory response in contrast to an enhanced proinflammatory injury reaction in 3-month-old adult males but not females. Our findings thus support that a key regulator of age- and sex-dependent H-I injury response in the mouse brain is modulation of the Hippo-YAP1 pathway by TSG-6-dependent ECM modifications.
Collapse
Affiliation(s)
- Taasin Srivastava
- Department of Pediatrics, Oregon Health and Science University (OHSU), Portland, Oregon 97239
| | - Hung Nguyen
- Division of Anesthesiology and Perioperative Medicine (APOM), Oregon Health and Science University (OHSU), Portland, Oregon 97239
| | - Gage Haden
- Department of Pediatrics, Oregon Health and Science University (OHSU), Portland, Oregon 97239
| | - Parham Diba
- Department of Pediatrics, Oregon Health and Science University (OHSU), Portland, Oregon 97239
| | - Steven Sowa
- Department of Pediatrics, Oregon Health and Science University (OHSU), Portland, Oregon 97239
| | - Norah LaNguyen
- Department of Pediatrics, Oregon Health and Science University (OHSU), Portland, Oregon 97239
| | - William Reed-Dustin
- Department of Pediatrics, Oregon Health and Science University (OHSU), Portland, Oregon 97239
| | - Wenbin Zhu
- Division of Anesthesiology and Perioperative Medicine (APOM), Oregon Health and Science University (OHSU), Portland, Oregon 97239
| | - Xi Gong
- Department of Pediatrics, Oregon Health and Science University (OHSU), Portland, Oregon 97239
| | - Edward N Harris
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, Nebraska 68588
| | - Selva Baltan
- Division of Anesthesiology and Perioperative Medicine (APOM), Oregon Health and Science University (OHSU), Portland, Oregon 97239
| | - Stephen A Back
- Department of Pediatrics, Oregon Health and Science University (OHSU), Portland, Oregon 97239
- Department of Neurology, Oregon Health and Science University (OHSU), Portland, Oregon 97239
| |
Collapse
|
8
|
Pastor-Alonso D, Berg M, Boyer F, Fomin-Thunemann N, Quintard M, Davit Y, Lorthois S. Modeling oxygen transport in the brain: An efficient coarse-grid approach to capture perivascular gradients in the parenchyma. PLoS Comput Biol 2024; 20:e1011973. [PMID: 38781253 PMCID: PMC11257410 DOI: 10.1371/journal.pcbi.1011973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 07/18/2024] [Accepted: 03/05/2024] [Indexed: 05/25/2024] Open
Abstract
Recent progresses in intravital imaging have enabled highly-resolved measurements of periarteriolar oxygen gradients (POGs) within the brain parenchyma. POGs are increasingly used as proxies to estimate the local baseline oxygen consumption, which is a hallmark of cell activity. However, the oxygen profile around a given arteriole arises from an interplay between oxygen consumption and delivery, not only by this arteriole but also by distant capillaries. Integrating such interactions across scales while accounting for the complex architecture of the microvascular network remains a challenge from a modelling perspective. This limits our ability to interpret the experimental oxygen maps and constitutes a key bottleneck toward the inverse determination of metabolic rates of oxygen. We revisit the problem of parenchymal oxygen transport and metabolism and introduce a simple, conservative, accurate and scalable direct numerical method going beyond canonical Krogh-type models and their associated geometrical simplifications. We focus on a two-dimensional formulation, and introduce the concepts needed to combine an operator-splitting and a Green's function approach. Oxygen concentration is decomposed into a slowly-varying contribution, discretized by Finite Volumes over a coarse cartesian grid, and a rapidly-varying contribution, approximated analytically in grid-cells surrounding each vessel. Starting with simple test cases, we thoroughly analyze the resulting errors by comparison with highly-resolved simulations of the original transport problem, showing considerable improvement of the computational-cost/accuracy balance compared to previous work. We then demonstrate the model ability to flexibly generate synthetic data reproducing the spatial dynamics of oxygen in the brain parenchyma, with sub-grid resolution. Based on these synthetic data, we show that capillaries distant from the arteriole cannot be overlooked when interpreting POGs, thus reconciling recent measurements of POGs across cortical layers with the fundamental idea that variations of vascular density within the depth of the cortex may reveal underlying differences in neuronal organization and metabolic load.
Collapse
Affiliation(s)
- David Pastor-Alonso
- Institut de Mécanique des Fluides de Toulouse (IMFT), UMR 5502, Université de Toulouse, CNRS, Toulouse, France
| | - Maxime Berg
- Institut de Mécanique des Fluides de Toulouse (IMFT), UMR 5502, Université de Toulouse, CNRS, Toulouse, France
- Department of Mechanical Engineering, University College London, London, United Kingdom
| | - Franck Boyer
- Institut de Mathématiques de Toulouse (IMT), UMR 5219, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Natalie Fomin-Thunemann
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, United States of America
| | - Michel Quintard
- Institut de Mécanique des Fluides de Toulouse (IMFT), UMR 5502, Université de Toulouse, CNRS, Toulouse, France
| | - Yohan Davit
- Institut de Mécanique des Fluides de Toulouse (IMFT), UMR 5502, Université de Toulouse, CNRS, Toulouse, France
| | - Sylvie Lorthois
- Institut de Mécanique des Fluides de Toulouse (IMFT), UMR 5502, Université de Toulouse, CNRS, Toulouse, France
| |
Collapse
|
9
|
Di Martino E, Rayasam A, Vexler ZS. Brain Maturation as a Fundamental Factor in Immune-Neurovascular Interactions in Stroke. Transl Stroke Res 2024; 15:69-86. [PMID: 36705821 PMCID: PMC10796425 DOI: 10.1007/s12975-022-01111-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 01/28/2023]
Abstract
Injuries in the developing brain cause significant long-term neurological deficits. Emerging clinical and preclinical data have demonstrated that the pathophysiology of neonatal and childhood stroke share similar mechanisms that regulate brain damage, but also have distinct molecular signatures and cellular pathways. The focus of this review is on two different diseases-neonatal and childhood stroke-with emphasis on similarities and distinctions identified thus far in rodent models of these diseases. This includes the susceptibility of distinct cell types to brain injury with particular emphasis on the role of resident and peripheral immune populations in modulating stroke outcome. Furthermore, we discuss some of the most recent and relevant findings in relation to the immune-neurovascular crosstalk and how the influence of inflammatory mediators is dependent on specific brain maturation stages. Finally, we comment on the current state of treatments geared toward inducing neuroprotection and promoting brain repair after injury and highlight that future prophylactic and therapeutic strategies for stroke should be age-specific and consider gender differences in order to achieve optimal translational success.
Collapse
Affiliation(s)
- Elena Di Martino
- Department of Neurology, University California San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158-0663, USA
| | - Aditya Rayasam
- Department of Neurology, University California San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158-0663, USA
| | - Zinaida S Vexler
- Department of Neurology, University California San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158-0663, USA.
| |
Collapse
|
10
|
Harry GJ. Developmental Associations between Neurovascularization and Microglia Colonization. Int J Mol Sci 2024; 25:1281. [PMID: 38279280 PMCID: PMC10816009 DOI: 10.3390/ijms25021281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/15/2024] [Accepted: 01/16/2024] [Indexed: 01/28/2024] Open
Abstract
The temporal and spatial pattern of microglia colonization and vascular infiltration of the nervous system implies critical associated roles in early stages of nervous system development. Adding to existing reviews that cover a broad spectrum of the various roles of microglia during brain development, the current review will focus on the developmental ontogeny and interdependency between the colonization of the nervous system with yolk sac derived macrophages and vascularization. Gaining a better understanding of the timing and the interdependency of these two processes will significantly contribute to the interpretation of data generated regarding alterations in either process during early development. Additionally, such knowledge should provide a framework for understanding the influence of the early gestational environmental and the impact of genetics, disease, disorders, or exposures on the early developing nervous system and the potential for long-term and life-time effects.
Collapse
Affiliation(s)
- G Jean Harry
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute Environmental Health Sciences, 111 T.W. Alexander Drive, Research Triangle Park, Durham, NC 27709, USA
| |
Collapse
|
11
|
Harry GJ. Microglia Colonization Associated with Angiogenesis and Neural Cell Development. ADVANCES IN NEUROBIOLOGY 2024; 37:163-178. [PMID: 39207692 DOI: 10.1007/978-3-031-55529-9_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
The temporal and spatial pattern of microglia colonization of the nervous system implies a role in early stages of organ development including cell proliferation, differentiation, and neurovascularization. As microglia colonize and establish within the developing nervous system, they assume a neural-specific identity and contribute to key developmental events. Their association around blood vessels implicates them in development of the vascular system or vice versa. A similar association has been reported for neural cell proliferation and associated phenotypic shifts and for cell fate differentiation to neuronal or glial phenotypes. These processes are accomplished by phagocytic activities, cell-cell contact relationships, and secretion of various factors. This chapter will present data currently available from studies evaluating the dynamic and interactive nature of these processes throughout the progression of nervous system development.
Collapse
Affiliation(s)
- G Jean Harry
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute Environmental Health Sciences, Research Triangle Park, NC, USA.
| |
Collapse
|
12
|
Jeong JY, Lee HJ, Kim N, Li Y, Rah JC, Oh WJ. Impaired neuronal activity as a potential factor contributing to the underdeveloped cerebrovasculature in a young Parkinson's disease mouse model. Sci Rep 2023; 13:22613. [PMID: 38114623 PMCID: PMC10730707 DOI: 10.1038/s41598-023-49900-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 12/13/2023] [Indexed: 12/21/2023] Open
Abstract
Misfolding of α-synuclein (α-Syn) in the brain causes cellular dysfunction, leading to cell death in a group of neurons, and consequently causes the progression of Parkinson's disease (PD). Although many studies have demonstrated the pathological connections between vascular dysfunction and neurodegenerative diseases, it remains unclear how neuronal accumulation of α-Syn affects the structural and functional aspects of the cerebrovasculature to accelerate early disease progression. Here, we demonstrated the effect of aberrant α-Syn expression on the brain vasculature using a PD mouse model expressing a familial mutant form of human α-Syn selectively in neuronal cells. We showed that young PD mice have an underdeveloped cerebrovasculature without significant α-Syn accumulation in the vasculature. During the early phase of PD, toxic α-Syn was selectively increased in neuronal cells, while endothelial cell proliferation was decreased in the absence of vascular cell death or neuroinflammation. Instead, we observed altered neuronal activation and minor changes in the activity-dependent gene expression in brain endothelial cells (ECs) in young PD mice. These findings demonstrated that neuronal expression of mutant α-Syn in the early stage of PD induces abnormal neuronal activity and contributes to vascular patterning defects, which could be associated with a reduced angiogenic potential of ECs.
Collapse
Affiliation(s)
- Jin-Young Jeong
- Neurovascular Biology Laboratory, Neurovascular Unit Research Group, Korea Brain Research Institute, Daegu, 41062, South Korea
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, 42988, South Korea
| | - Hyun Jung Lee
- Sensory and Motor System Research Group, Korea Brain Research Institute, Daegu, 41062, South Korea
| | - Namsuk Kim
- Neurovascular Biology Laboratory, Neurovascular Unit Research Group, Korea Brain Research Institute, Daegu, 41062, South Korea
| | - Yan Li
- Neurovascular Biology Laboratory, Neurovascular Unit Research Group, Korea Brain Research Institute, Daegu, 41062, South Korea
| | - Jong-Cheol Rah
- Sensory and Motor System Research Group, Korea Brain Research Institute, Daegu, 41062, South Korea
| | - Won-Jong Oh
- Neurovascular Biology Laboratory, Neurovascular Unit Research Group, Korea Brain Research Institute, Daegu, 41062, South Korea.
| |
Collapse
|
13
|
Henneicke S, Meuth SG, Schreiber S. [Cerebral Small Vessel Disease: Advances in Understanding its Pathophysiology]. FORTSCHRITTE DER NEUROLOGIE-PSYCHIATRIE 2023; 91:494-502. [PMID: 38081163 DOI: 10.1055/a-2190-8957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Sporadic cerebral small vessel disease determines age- and vascular-risk-factor-related processes of the small brain vasculature. The underlying pathology develops in a stage-dependent manner - probably over decades - often already starting in midlife. Endothelial and pericyte activation precedes blood-brain barrier leaks, extracellular matrix remodeling and neuroinflammation, which ultimately result in bleeds, synaptic and neural dysfunction. Hemodynamic compromise of the small vessel walls promotes perivascular drainage failure and accumulation of neurotoxic waste products in the brain. Clinical diagnosis is mainly based on magnetic resonance imaging according to the Standards for Reporting Vascular Changes on Neuroimaging 2. Cerebral amyloid angiopathy is particularly stratified according to the Boston v2.0 criteria. Small vessel disease of the brain could be clinically silent, or manifested through a heterogeneous spectrum of diseases, where cognitive decline and stroke-related symptoms are the most common ones. Prevention and therapy are centered around vascular risk factor control, physically and cognitively enriched life style and, presumably, maintenance of a good sleep quality, which promotes sufficient perivascular drainage. Prevention of ischemic stroke through anticoagulation that carries at the same time an increased risk for large brain hemorrhages - particularly in the presence of disseminated cortical superficial siderosis - remains one of the main challenges. The cerebral small vessel disease field is rapidly evolving, focusing on the establishment of early disease stage imaging and biofluid biomarkers of neurovascular unit remodeling and the compromise of perivascular drainage. New prevention and therapy strategies will correspondingly center around the dedicated targeting of, e. g., cellular small vessel wall and perivascular tissue structures. Growing knowledge about brain microvasculature bridging neuroimmunological, neurovascular and neurodegenerative fields might lead to a rethink about apparently separate disease entities and the development of overarching concepts for a common line of prevention and treatment for several diseases.
Collapse
Affiliation(s)
- Solveig Henneicke
- Neurologie, Otto-von-Guericke-Universität Magdeburg Medizinische Fakultät, Magdeburg, Germany
| | | | - Stefanie Schreiber
- Neurologie, Otto-von-Guericke-Universität Magdeburg Medizinische Fakultät, Magdeburg, Germany
| |
Collapse
|
14
|
Troncoso F, Sandoval H, Ibañez B, López-Espíndola D, Bustos F, Tapia JC, Sandaña P, Escudero-Guevara E, Nualart F, Ramírez E, Powers R, Vatish M, Mistry HD, Kurlak LO, Acurio J, Escudero C. Reduced Brain Cortex Angiogenesis in the Offspring of the Preeclampsia-Like Syndrome. Hypertension 2023; 80:2559-2571. [PMID: 37767691 DOI: 10.1161/hypertensionaha.123.21756] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/17/2023] [Indexed: 09/29/2023]
Abstract
BACKGROUND Children from pregnancies affected by preeclampsia have an increased risk of cognitive and behavioral alterations via unknown pathophysiology. We tested the hypothesis that preeclampsia generated reduced brain cortex angiogenesis in the offspring. METHODS The preeclampsia-like syndrome (PELS) mouse model was generated by administering the nitric oxide inhibitor NG-nitroarginine methyl ester hydrochloride. Confirmatory experiments were done using 2 additional PELS models. While in vitro analysis used mice and human brain endothelial cells exposed to serum of postnatal day 5 pups or umbilical plasma from preeclamptic pregnancies, respectively. RESULTS We report significant reduction in the area occupied by blood vessels in the motor and somatosensory brain cortex of offspring (postnatal day 5) from PELS compared with uncomplicated control offspring. These data were confirmed using 2 additional PELS models. Furthermore, circulating levels of critical proangiogenic factors, VEGF (vascular endothelial growth factor), and PlGF (placental growth factor) were lower in postnatal day 5 PELS. Also we found lower VEGF receptor 2 (KDR [kinase insert domain-containing receptor]) levels in mice and human endothelial cells exposed to the serum of postnatal day 5 PELS or fetal plasma of preeclamptic pregnancies, respectively. These changes were associated with lower in vitro angiogenic capacity, diminished cell migration, larger F-actin filaments, lower number of filopodia, and lower protein levels of F-actin polymerization regulators in brain endothelial cells exposed to serum or fetal plasma of offspring from preeclampsia. CONCLUSIONS Offspring from preeclampsia exhibited diminished brain cortex angiogenesis, associated with lower circulating VEGF/PlGF/KDR protein levels, impaired brain endothelial migration, and dysfunctional assembly of F-actin filaments. These alterations may predispose to structural and functional alterations in long-term brain development.
Collapse
Affiliation(s)
- Felipe Troncoso
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (F.T., H.S., B.I., E.E.-G., J.A., C.E.)
| | - Hermes Sandoval
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (F.T., H.S., B.I., E.E.-G., J.A., C.E.)
| | - Belén Ibañez
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (F.T., H.S., B.I., E.E.-G., J.A., C.E.)
| | - Daniela López-Espíndola
- Escuela de Tecnología Médica, Facultad de Medicina, Universidad de Valparaíso, Chile (D.L.-E., F.B.)
- Group of Research and Innovation in Vascular Health, Chillan, Chile (D.L.-E., C.E.)
| | - Francisca Bustos
- Escuela de Tecnología Médica, Facultad de Medicina, Universidad de Valparaíso, Chile (D.L.-E., F.B.)
| | - Juan Carlos Tapia
- Stem Cells and Neuroscience Center, School of Medicine, University of Talca, Chile (J.C.T.)
| | - Pedro Sandaña
- Anatomopatholy Unit, Hospital Clinico Herminda Martin, Chillan, Chile (P.S.)
| | - Esthefanny Escudero-Guevara
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (F.T., H.S., B.I., E.E.-G., J.A., C.E.)
| | - Francisco Nualart
- Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy CMA Bio-Bio, Faculty of Biological Sciences, University of Concepcion, Chile (F.N., E.R.)
- Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Chile (F.N.)
| | - Eder Ramírez
- Laboratory of Neurobiology and Stem Cells NeuroCellT, Department of Cellular Biology, Center for Advanced Microscopy CMA Bio-Bio, Faculty of Biological Sciences, University of Concepcion, Chile (F.N., E.R.)
| | - Robert Powers
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, PA (R.P.)
| | - Manu Vatish
- Nuffield Department of Women's Health and Reproductive Research, University of Oxford, England (M.V.)
| | - Hiten D Mistry
- Division of Women and Children's Health, School of Life Course and Population Sciences, King's College London, United Kingdom (H.D.M.)
| | - Lesia O Kurlak
- Stroke Trials Unit, School of Medicine, University of Nottingham, United Kingdom (L.O.K.)
| | - Jesenia Acurio
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (F.T., H.S., B.I., E.E.-G., J.A., C.E.)
| | - Carlos Escudero
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (F.T., H.S., B.I., E.E.-G., J.A., C.E.)
- Group of Research and Innovation in Vascular Health, Chillan, Chile (D.L.-E., C.E.)
| |
Collapse
|
15
|
Han C, Nguyen CL, Scherschinski L, Schriber TD, Arthur HM, Lawton MT, Oh SP. VEGFR2 Expression Correlates with Postnatal Development of Brain Arteriovenous Malformations in a Mouse Model of Type I Hereditary Hemorrhagic Telangiectasia. Biomedicines 2023; 11:3153. [PMID: 38137374 PMCID: PMC10740421 DOI: 10.3390/biomedicines11123153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/20/2023] [Accepted: 11/25/2023] [Indexed: 12/24/2023] Open
Abstract
Brain arteriovenous malformations (BAVMs) are a critical concern in hereditary hemorrhagic telangiectasia (HHT) patients, carrying the risk of life-threatening intracranial hemorrhage. While traditionally seen as congenital, the debate continues due to documented de novo cases. Our primary goal was to identify the precise postnatal window in which deletion of the HHT gene Endoglin (Eng) triggers BAVM development. We employed SclCreER(+);Eng2f/2f mice, enabling timed Eng gene deletion in endothelial cells via tamoxifen. Tamoxifen was given during four postnatal periods: P1-3, P8-10, P15-17, and P22-24. BAVM development was assessed at 2-3 months using latex dye perfusion. We examined the angiogenic activity by assessing vascular endothelial growth factor receptor 2 (VEGFR2) expression via Western blotting and Flk1-LacZ reporter mice. Longitudinal magnetic resonance angiography (MRA) was conducted up to 9 months. BAVMs emerged in 88% (P1-3), 86% (P8-10), and 55% (P15-17) of cases, with varying localization. Notably, the P22-24 group did not develop BAVMs but exhibited skin AVMs. VEGFR2 expression peaked in the initial 2 postnatal weeks, coinciding with BAVM onset. These findings support the "second hit" theory, highlighting the role of early postnatal angiogenesis in initiating BAVM development in HHT type I mice.
Collapse
Affiliation(s)
- Chul Han
- Barrow Aneurysm and AVM Research Center, Department of Translational Neuroscience, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA; (C.H.); (C.L.N.); (L.S.); (M.T.L.)
| | - Candice L. Nguyen
- Barrow Aneurysm and AVM Research Center, Department of Translational Neuroscience, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA; (C.H.); (C.L.N.); (L.S.); (M.T.L.)
| | - Lea Scherschinski
- Barrow Aneurysm and AVM Research Center, Department of Translational Neuroscience, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA; (C.H.); (C.L.N.); (L.S.); (M.T.L.)
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA
- Department of Neurosurgery, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| | - Tyler D. Schriber
- Barrow Aneurysm and AVM Research Center, Department of Translational Neuroscience, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA; (C.H.); (C.L.N.); (L.S.); (M.T.L.)
| | - Helen M. Arthur
- Biosciences Institute, Newcastle University, Newcastle NE1 7RU, UK;
| | - Michael T. Lawton
- Barrow Aneurysm and AVM Research Center, Department of Translational Neuroscience, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA; (C.H.); (C.L.N.); (L.S.); (M.T.L.)
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA
| | - Suk Paul Oh
- Barrow Aneurysm and AVM Research Center, Department of Translational Neuroscience, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ 85013, USA; (C.H.); (C.L.N.); (L.S.); (M.T.L.)
| |
Collapse
|
16
|
Travasso RDM, Coelho-Santos V. Image-based angio-adaptation modelling: a playground to study cerebrovascular development. Front Physiol 2023; 14:1223308. [PMID: 37565149 PMCID: PMC10411953 DOI: 10.3389/fphys.2023.1223308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 07/12/2023] [Indexed: 08/12/2023] Open
Affiliation(s)
- Rui D. M. Travasso
- Department of Physics, Center for Physics of the University of Coimbra (CFisUC), University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, Coimbra, Portugal
| | - Vanessa Coelho-Santos
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, Coimbra, Portugal
- Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
| |
Collapse
|
17
|
Gheres KW, Ünsal HS, Han X, Zhang Q, Turner KL, Zhang N, Drew PJ. Arousal state transitions occlude sensory-evoked neurovascular coupling in neonatal mice. Commun Biol 2023; 6:738. [PMID: 37460780 PMCID: PMC10352318 DOI: 10.1038/s42003-023-05121-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 07/07/2023] [Indexed: 07/20/2023] Open
Abstract
In the adult sensory cortex, increases in neural activity elicited by sensory stimulation usually drive vasodilation mediated by neurovascular coupling. However, whether neurovascular coupling is the same in neonatal animals as adults is controversial, as both canonical and inverted responses have been observed. We investigated the nature of neurovascular coupling in unanesthetized neonatal mice using optical imaging, electrophysiology, and BOLD fMRI. We find in neonatal (postnatal day 15, P15) mice, sensory stimulation induces a small increase in blood volume/BOLD signal, often followed by a large decrease in blood volume. An examination of arousal state of the mice revealed that neonatal mice were asleep a substantial fraction of the time, and that stimulation caused the animal to awaken. As cortical blood volume is much higher during REM and NREM sleep than the awake state, awakening occludes any sensory-evoked neurovascular coupling. When neonatal mice are stimulated during an awake period, they showed relatively normal (but slowed) neurovascular coupling, showing that that the typically observed constriction is due to arousal state changes. These result show that sleep-related vascular changes dominate over any sensory-evoked changes, and hemodynamic measures need to be considered in the context of arousal state changes.
Collapse
Affiliation(s)
- Kyle W Gheres
- Molecular Cellular and Integrative Bioscience program, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Hayreddin S Ünsal
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Electrical and Electronics Engineering, Abdullah Gul University, Kayseri, Türkiye
| | - Xu Han
- Molecular Cellular and Integrative Bioscience program, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Qingguang Zhang
- Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Kevin L Turner
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Nanyin Zhang
- Molecular Cellular and Integrative Bioscience program, The Pennsylvania State University, University Park, PA, 16802, USA
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
- Center for Neurotechnology in Mental Health Research, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Patrick J Drew
- Molecular Cellular and Integrative Bioscience program, The Pennsylvania State University, University Park, PA, 16802, USA.
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA.
- Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA, 16802, USA.
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA, 16802, USA.
- Center for Neurotechnology in Mental Health Research, The Pennsylvania State University, University Park, PA, 16802, USA.
- Departments of Neurosurgery and Biology, The Pennsylvania State University, University Park, PA, 16802, USA.
| |
Collapse
|
18
|
Vittani M, Knak PAG, Fukuda M, Nagao M, Wang X, Kjaerby C, Konno A, Hirai H, Nedergaard M, Hirase H. Virally induced CRISPR/Cas9-based knock-in of fluorescent albumin allows long-term visualization of cerebral circulation in infant and adult mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.10.548084. [PMID: 37503027 PMCID: PMC10369863 DOI: 10.1101/2023.07.10.548084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Albumin, a protein produced by liver hepatocytes, represents the most abundant protein in blood plasma. We have previously engineered a liver-targeting adeno-associated viral vector (AAV) that expresses fluorescent protein-tagged albumin to visualize blood plasma in mice. While this approach is versatile for imaging in adult mice, transgene expression vanishes when AAV is administered in neonates due to dilution of the episomal AAV genome in the rapidly growing liver. Here, we use CRISPR/Cas9 genome editing to insert the fluorescent protein mNeonGreen (mNG) gene into the albumin (Alb) locus of hepatocytes to produce fluorescently labeled albumin (Alb-mNG). We constructed a CRISPR AAV that includes ∼1 kb homologous arms around Alb exon 14 to express Alb-mNG. Subcutaneous injection of this AAV with AAV-CMV-Cas9 in postnatal day 3 mice resulted in two-photon visualization of the cerebral cortex vasculature within ten days. The expression levels of Alb-mNG were persistent for at least three months and were so robust that vasomotion and capillary blood flow could be assessed transcranially in early postnatal mice. This knock-in approach provides powerful means for micro- and macroscopic imaging of cerebral vascular dynamics in postnatal and adult mice.
Collapse
|
19
|
Coelho-Santos V, Cruz AJN, Shih AY. Does Perinatal Intermittent Hypoxia Affect Cerebrovascular Network Development? Dev Neurosci 2023; 46:44-54. [PMID: 37231864 DOI: 10.1159/000530957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 05/02/2023] [Indexed: 05/27/2023] Open
Abstract
Perinatal hypoxia is an inadequate delivery of oxygen to the fetus in the period immediately before, during, or after the birth process. The most frequent form of hypoxia occurring in human development is chronic intermittent hypoxia (CIH) due to sleep-disordered breathing (apnea) or bradycardia events. CIH incidence is particularly high with premature infants. During CIH, repetitive cycles of hypoxia and reoxygenation initiate oxidative stress and inflammatory cascades in the brain. A dense microvascular network of arterioles, capillaries, and venules is required to support the constant metabolic demands of the adult brain. The development and refinement of this microvasculature is orchestrated throughout gestation and in the initial weeks after birth, at a critical juncture when CIH can occur. There is little knowledge on how CIH affects the development of the cerebrovasculature. However, since CIH (and its treatments) can cause profound abnormalities in tissue oxygen content and neural activity, there is reason to believe that it can induce lasting abnormalities in vascular structure and function at the microvascular level contributing to neurodevelopmental disorders. This mini-review discusses the hypothesis that CIH induces a positive feedback loop to perpetuate metabolic insufficiency through derailment of normal cerebrovascular development, leading to long-term deficiencies in cerebrovascular function.
Collapse
Affiliation(s)
- Vanessa Coelho-Santos
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, Coimbra, Portugal
- Institute of Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
| | - Anne-Jolene N Cruz
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Andy Y Shih
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
| |
Collapse
|
20
|
Stamenkovic S, Li Y, Waters J, Shih A. Deep Imaging to Dissect Microvascular Contributions to White Matter Degeneration in Rodent Models of Dementia. Stroke 2023; 54:1403-1415. [PMID: 37094035 PMCID: PMC10460612 DOI: 10.1161/strokeaha.122.037156] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2023]
Abstract
The increasing socio-economic burden of Alzheimer disease (AD) and AD-related dementias has created a pressing need to define targets for therapeutic intervention. Deficits in cerebral blood flow and neurovascular function have emerged as early contributors to disease progression. However, the cause, progression, and consequence of small vessel disease in AD/AD-related dementias remains poorly understood, making therapeutic targets difficult to pinpoint. Animal models that recapitulate features of AD/AD-related dementias may provide mechanistic insight because microvascular pathology can be studied as it develops in vivo. Recent advances in in vivo optical and ultrasound-based imaging of the rodent brain facilitate this goal by providing access to deeper brain structures, including white matter and hippocampus, which are more vulnerable to injury during cerebrovascular disease. Here, we highlight these novel imaging approaches and discuss their potential for improving our understanding of vascular contributions to AD/AD-related dementias.
Collapse
Affiliation(s)
- Stefan Stamenkovic
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA, USA
| | - Yuandong Li
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA, USA
| | - Jack Waters
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Andy Shih
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
- Allen Institute for Brain Science, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| |
Collapse
|
21
|
Huo R, Yang Y, Sun Y, Zhou Q, Zhao S, Mo Z, Xu H, Wang J, Weng J, Jiao Y, Zhang J, He Q, Wang S, Zhao J, Wang J, Cao Y. Endothelial hyperactivation of mutant MAP3K3 induces cerebral cavernous malformation enhanced by PIK3CA GOF mutation. Angiogenesis 2023; 26:295-312. [PMID: 36719480 DOI: 10.1007/s10456-023-09866-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 01/18/2023] [Indexed: 02/01/2023]
Abstract
Cerebral cavernous malformations (CCMs) refer to a common vascular abnormality that affects up to 0.5% of the population. A somatic gain-of-function mutation in MAP3K3 (p.I441M) was recently reported in sporadic CCMs, frequently accompanied by somatic activating PIK3CA mutations in diseased endothelium. However, the molecular mechanisms of these driver genes remain elusive. In this study, we performed whole-exome sequencing and droplet digital polymerase chain reaction to analyze CCM lesions and the matched blood from sporadic patients. 44 of 94 cases harbored mutations in KRIT1/CCM2 or MAP3K3, of which 75% were accompanied by PIK3CA mutations (P = 0.006). AAV-BR1-mediated brain endothelial-specific MAP3K3I441M overexpression induced CCM-like lesions throughout the brain and spinal cord in adolescent mice. Interestingly, over half of lesions disappeared at adulthood. Single-cell RNA sequencing found significant enrichment of the apoptosis pathway in a subset of brain endothelial cells in MAP3K3I441M mice compared to controls. We then demonstrated that MAP3K3I441M overexpression activated p38 signaling that is associated with the apoptosis of endothelial cells in vitro and in vivo. In contrast, the mice simultaneously overexpressing PIK3CA and MAP3K3 mutations had an increased number of CCM-like lesions and maintained these lesions for a longer time compared to those with only MAP3K3I441M. Further in vitro and in vivo experiments showed that activating PI3K signaling increased proliferation and alleviated apoptosis of endothelial cells. By using AAV-BR1, we found that MAP3K3I441M mutation can provoke CCM-like lesions in mice and the activation of PI3K signaling significantly enhances and maintains these lesions, providing a preclinical model for the further mechanistic and therapeutic study of CCMs.
Collapse
Affiliation(s)
- Ran Huo
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, 119S Fourth Ring Rd W, Fengtai District, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yingxi Yang
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Yingfan Sun
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, 119S Fourth Ring Rd W, Fengtai District, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Qiuxia Zhou
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Shaozhi Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, 119S Fourth Ring Rd W, Fengtai District, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Zongchao Mo
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Hongyuan Xu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, 119S Fourth Ring Rd W, Fengtai District, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Jie Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, 119S Fourth Ring Rd W, Fengtai District, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Jiancong Weng
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, 119S Fourth Ring Rd W, Fengtai District, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yuming Jiao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, 119S Fourth Ring Rd W, Fengtai District, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Junze Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, 119S Fourth Ring Rd W, Fengtai District, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Qiheng He
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, 119S Fourth Ring Rd W, Fengtai District, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Shuo Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, 119S Fourth Ring Rd W, Fengtai District, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Jizong Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, 119S Fourth Ring Rd W, Fengtai District, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Jiguang Wang
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China.
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China.
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong SAR, China.
| | - Yong Cao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, 119S Fourth Ring Rd W, Fengtai District, Beijing, China.
- China National Clinical Research Center for Neurological Diseases, Beijing, China.
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
- Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China.
| |
Collapse
|
22
|
Gheres KW, Ãœnsal HS, Han X, Zhang Q, Turner KL, Zhang N, Drew PJ. Arousal state transitions occlude sensory-evoked neurovascular coupling in neonatal mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.18.529057. [PMID: 36824895 PMCID: PMC9949139 DOI: 10.1101/2023.02.18.529057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2023]
Abstract
In the adult sensory cortex, increases in neural activity elicited by sensory stimulation usually drives vasodilation mediated by neurovascular coupling. However, whether neurovascular coupling is the same in neonatal animals as adults is controversial, as both canonical and inverted responses have been observed. We investigated the nature of neurovascular coupling in unanesthetized neonatal mice using optical imaging, electrophysiology, and BOLD fMRI. We find in neonatal (postnatal day 15, P15) mice, sensory stimulation induces a small increase in blood volume/BOLD signal, often followed by a large decrease in blood volume. An examination of arousal state of the mice revealed that neonatal mice were asleep a substantial fraction of the time, and that stimulation caused the animal to awaken. As cortical blood volume is much higher during REM and NREM sleep than the awake state, awakening occludes any sensory-evoked neurovascular coupling. When neonatal mice are stimulated during an awake period, they showed relatively normal (but slowed) neurovascular coupling, showing that that the typically observed constriction is due to arousal state changes. These result show that sleep-related vascular changes dominate over any sensory-evoked changes, and hemodynamic measures need to be considered in the context of arousal state changes. Significance Statement In the adult brain, increases in neural activity are often followed by vasodilation, allowing activity to be monitored using optical or magnetic resonance imaging. However, in neonates, sensory stimulation can drive vasoconstriction, whose origin was not understood. We used optical and magnetic resonance imaging approaches to investigate hemodynamics in neonatal mice. We found that sensory-induced vasoconstriction occurred when the mice were asleep, as sleep is associated with dilation of the vasculature of the brain relative to the awake state. The stimulus awakens the mice, causing a constriction due to the arousal state change. Our study shows the importance of monitoring arousal state, particularly when investigating subjects that may sleep, and the dominance arousal effects on brain hemodynamics.
Collapse
|
23
|
Significance of Pulmonary Endothelial Injury and the Role of Cyclooxygenase-2 and Prostanoid Signaling. BIOENGINEERING (BASEL, SWITZERLAND) 2023; 10:bioengineering10010117. [PMID: 36671689 PMCID: PMC9855370 DOI: 10.3390/bioengineering10010117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023]
Abstract
The endothelium plays a key role in the dynamic balance of hemodynamic, humoral and inflammatory processes in the human body. Its central importance and the resulting therapeutic concepts are the subject of ongoing research efforts and form the basis for the treatment of numerous diseases. The pulmonary endothelium is an essential component for the gas exchange in humans. Pulmonary endothelial dysfunction has serious consequences for the oxygenation and the gas exchange in humans with the potential of consecutive multiple organ failure. Therefore, in this review, the dysfunction of the pulmonary endothel due to viral, bacterial, and fungal infections, ventilator-related injury, and aspiration is presented in a medical context. Selected aspects of the interaction of endothelial cells with primarily alveolar macrophages are reviewed in more detail. Elucidation of underlying causes and mechanisms of damage and repair may lead to new therapeutic approaches. Specific emphasis is placed on the processes leading to the induction of cyclooxygenase-2 and downstream prostanoid-based signaling pathways associated with this enzyme.
Collapse
|
24
|
Salehi A, Salari S, Jullienne A, Daglian J, Chen K, Baram TZ, Obenaus A. Vascular topology and blood flow are acutely impacted by experimental febrile status epilepticus. J Cereb Blood Flow Metab 2023; 43:84-98. [PMID: 35912523 PMCID: PMC9875348 DOI: 10.1177/0271678x221117625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Febrile status epilepticus (FSE) is an important risk factor for temporal lobe epilepsy and early identification of those at high risk for epilepsy is vital. In a rat model of FSE, we identified an acute (2 hrs) novel MRI signal where reduced T2 relaxation values in the basolateral amygdala (BLA) predicted epilepsy in adulthood; this T2 signal remains incompletely understood and we hypothesized that it may be influenced by vascular topology. Experimental FSE induced in rat pups reduced blood vessel density of the cortical vasculature in a lateralized manner at 2 hrs post FSE. Middle cerebral artery (MCA) exhibited abnormal topology in FSE pups but not in controls. In the BLA, significant vessel junction reductions and decreased vessel diameter were observed, together with a strong trend for reduced vessel length. Perfusion weighted MRI (PWI) was acutely increased cerebral blood flow (CBF) in cortex, amygdala and hippocampus of FSE pups that correlated to decreased T2 relaxation values compared to controls. This is consistent with increased levels of deoxyhemoglobin associated with increased metabolic demand. In summary, FSE acutely modifies vascular topological and CBF in cortex and BLA that may underlie acute MRI signal changes that predict progression to future epilepsy.
Collapse
Affiliation(s)
- Arjang Salehi
- Department of Anatomy and Neurobiology, School of Medicine, University of California Irvine, Irvine CA, USA
| | - Sirus Salari
- Department of Pediatrics, School of Medicine, University of California Irvine, Irvine CA, USA
| | - Amandine Jullienne
- Department of Pediatrics, School of Medicine, University of California Irvine, Irvine CA, USA
| | - Jennifer Daglian
- Department of Anatomy and Neurobiology, School of Medicine, University of California Irvine, Irvine CA, USA
| | - Kevin Chen
- Department of Anatomy and Neurobiology, School of Medicine, University of California Irvine, Irvine CA, USA
| | - Tallie Z Baram
- Department of Anatomy and Neurobiology, School of Medicine, University of California Irvine, Irvine CA, USA.,Department of Pediatrics, School of Medicine, University of California Irvine, Irvine CA, USA
| | - Andre Obenaus
- Department of Anatomy and Neurobiology, School of Medicine, University of California Irvine, Irvine CA, USA.,Department of Pediatrics, School of Medicine, University of California Irvine, Irvine CA, USA
| |
Collapse
|
25
|
Kurki SN, Uvarov P, Pospelov AS, Trontti K, Hübner AK, Srinivasan R, Watanabe M, Hovatta I, Hübner CA, Kaila K, Virtanen MA. Expression patterns of NKCC1 in neurons and non-neuronal cells during cortico-hippocampal development. Cereb Cortex 2022; 33:5906-5923. [PMID: 36573432 PMCID: PMC10183754 DOI: 10.1093/cercor/bhac470] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/05/2022] [Accepted: 11/06/2022] [Indexed: 12/28/2022] Open
Abstract
Abstract
The Na-K-2Cl cotransporter NKCC1 is widely expressed in cells within and outside the brain. However, our understanding of its roles in brain functions throughout development, as well as in neuropsychiatric and neurological disorders, has been severely hindered by the lack of reliable data on its developmental and (sub)cellular expression patterns. We provide here the first properly controlled analysis of NKCC1 protein expression in various cell types of the mouse brain using custom-made antibodies and an NKCC1 knock-out validated immunohistochemical procedure, with parallel data based on advanced mRNA approaches. NKCC1 protein and mRNA are expressed at remarkably high levels in oligodendrocytes. In immature neurons, NKCC1 protein was located in the somata, whereas in adult neurons, only NKCC1 mRNA could be clearly detected. NKCC1 immunoreactivity is also seen in microglia, astrocytes, developing pericytes, and in progenitor cells of the dentate gyrus. Finally, a differential expression of NKCC1 splice variants was observed, with NKCC1a predominating in non-neuronal cells and NKCC1b in neurons. Taken together, our data provide a cellular basis for understanding NKCC1 functions in the brain and enable the identification of major limitations and promises in the development of neuron-targeting NKCC1-blockers.
Collapse
Affiliation(s)
- Samu N Kurki
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| | - Pavel Uvarov
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| | - Alexey S Pospelov
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| | - Kalevi Trontti
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
- University of Helsinki SleepWell Research Program, Faculty of Medicine, , 00014 Helsinki , Finland
- University of Helsinki Department of Psychology and Logopedics, , 00014 Helsinki , Finland
| | - Antje K Hübner
- Jena University Hospital, Friedrich Schiller Universität Institute of Human Genetics, , 07747 Jena , Germany
| | - Rakenduvadhana Srinivasan
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| | - Masahiko Watanabe
- Hokkaido University Department of Anatomy, Faculty of Medicine, , Sapporo 060–8638 , Japan
| | - Iiris Hovatta
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
- University of Helsinki SleepWell Research Program, Faculty of Medicine, , 00014 Helsinki , Finland
- University of Helsinki Department of Psychology and Logopedics, , 00014 Helsinki , Finland
| | - Christian A Hübner
- Jena University Hospital, Friedrich Schiller Universität Institute of Human Genetics, , 07747 Jena , Germany
| | - Kai Kaila
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| | - Mari A Virtanen
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| |
Collapse
|
26
|
Lara E, Rivera N, González-Bernal A, Rojas D, López-Espíndola D, Rodríguez A, Escudero C. Abnormal cerebral microvascular perfusion and reactivity in female offspring of reduced uterine perfusion pressure (RUPP) mice model. J Cereb Blood Flow Metab 2022; 42:2318-2332. [PMID: 36008921 PMCID: PMC9670000 DOI: 10.1177/0271678x221121872] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 07/19/2022] [Accepted: 07/24/2022] [Indexed: 12/14/2022]
Abstract
Children born from women with preeclampsia have alterations in cerebral neurovascular development and a high risk for developing cognitive alterations. Because cerebral blood vessels are critical components in cerebrovascular development, we evaluated the brain microvascular perfusion and microvascular reactivity (exposed to external stimuli of warm and cold) in pups born to preeclampsia-like syndrome based on the reduction of uterine perfusion (RUPP). Also, we evaluate the angiogenic proteomic profile in those brains. Pregnant mice showed a reduction in uterine flow after RUPP surgery (-40 to 50%) associated with unfavorable perinatal results compared to sham mice. Furthermore, offspring of the RUPP mice exhibited reduced brain microvascular perfusion at postnatal day 5 (P5) compared with offspring from sham mice. This reduction was preferentially observed in females. Also, brain microvascular reactivity to external stimuli (warm and cold) was reduced in pups of RUPP mice. Furthermore, a differential expression of the angiogenic profile associated with inflammation, extrinsic apoptotic, cancer, and cellular senescence processes as the primary signaling impaired process was found in the brains of RUPP-offspring. Then, offspring (P5) from preeclampsia-like syndrome exhibit impaired brain perfusion and microvascular reactivity, particularly in female mice, associated with differential expression of angiogenic proteins in the brain tissue.
Collapse
Affiliation(s)
- Evelyn Lara
- Vascular Physiology Laboratory, Department of Basic Sciences,
Universidad del Bio-Bio, Chillán, Chile
| | - Nathaly Rivera
- Vascular Physiology Laboratory, Department of Basic Sciences,
Universidad del Bio-Bio, Chillán, Chile
| | - Alejandro González-Bernal
- Department of Clinical Sciences, Faculty of Veterinary Sciences,
Universidad de Concepción, Chillán, Chile
| | - Daniela Rojas
- Department of Pathology, Faculty of Veterinary Sciences,
Universidad de Concepción, Chillán, Chile
| | - Daniela López-Espíndola
- School of Medical Technology and Biomedical Research Center,
Faculty of Medicine, Universidad de Valparaíso
- Group of Research and Innovation in Vascular Health (GRIVAS
Health), Chillán, Chile
| | - Andrés Rodríguez
- Vascular Physiology Laboratory, Department of Basic Sciences,
Universidad del Bio-Bio, Chillán, Chile
- Group of Research and Innovation in Vascular Health (GRIVAS
Health), Chillán, Chile
| | - Carlos Escudero
- Vascular Physiology Laboratory, Department of Basic Sciences,
Universidad del Bio-Bio, Chillán, Chile
- Group of Research and Innovation in Vascular Health (GRIVAS
Health), Chillán, Chile
| |
Collapse
|
27
|
Drew PJ. Neurovascular coupling: motive unknown. Trends Neurosci 2022; 45:809-819. [PMID: 35995628 PMCID: PMC9768528 DOI: 10.1016/j.tins.2022.08.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/01/2022] [Accepted: 08/05/2022] [Indexed: 12/13/2022]
Abstract
In the brain, increases in neural activity drive changes in local blood flow via neurovascular coupling. The common explanation for increased blood flow (known as functional hyperemia) is that it supplies the metabolic needs of active neurons. However, there is a large body of evidence that is inconsistent with this idea. Baseline blood flow is adequate to supply oxygen needs even with elevated neural activity. Neurovascular coupling is irregular, absent, or inverted in many brain regions, behavioral states, and conditions. Increases in respiration can increase brain oxygenation without flow changes. Simulations show that given the architecture of the brain vasculature, areas of low blood flow are inescapable and cannot be removed by functional hyperemia. As discussed in this article, potential alternative functions of neurovascular coupling include supplying oxygen for neuromodulator synthesis, brain temperature regulation, signaling to neurons, stabilizing and optimizing the cerebral vascular structure, accommodating the non-Newtonian nature of blood, and driving the production and circulation of cerebrospinal fluid (CSF).
Collapse
Affiliation(s)
- Patrick J Drew
- Center for Neural Engineering, Departments of Engineering Science and Mechanics, Neurosurgery, Biology, and Biomedical Engineering, The Pennsylvania State University, W-317 Millennium Science Complex, University Park, PA 16802, USA.
| |
Collapse
|
28
|
Kovtanyuk A, Turova V, Sidorenko I, Chebotarev A, Lampe R. Modeling of the cerebral blood circulation in a capillary network accounting for the influence of the endothelial surface layer. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2022; 224:107008. [PMID: 35901640 DOI: 10.1016/j.cmpb.2022.107008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 06/22/2022] [Accepted: 07/05/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND AND OBJECTIVE The paper describes a mathematical model of blood flow in capillaries with accounting for the endothelial surface layer (ESL). METHOD The influence of ESL is modeled by a boundary layer with zero flow velocity. Finite element modeling and an analytical approach based on the homogenization of the core region of blood flow occupied by erythrocytes are developed to describe the resistance of a capillary. The reliability of the results obtained is verified for different values of the discharge hematocrit and vessel diameter using known in vivo data. RESULTS The proposed approach is applied to the numerical simulation of blood circulation in a capillary network of the germinal matrix of infants born at 25 gestational weeks. The influence of the hematocrit level and effective thickness of ESL on the resistance of the capillary network of the germinal matrix of preterm infants is studied. It was found that a decrease in the effective thickness of ESL in the capillary network (and/or a decrease in the hematocrit) leads to reducing the resistance of the capillary network. CONCLUSION A decrease in the effective thickness of ESL in the capillary network leads to an increase in the pressure drop in arterioles, which may be considered as an additional risk factor for hemorrhages in fragile blood vessels within the germinal matrix.
Collapse
Affiliation(s)
- Andrey Kovtanyuk
- Klinikum rechts der Isar, Technische Universität München, Ismaningerstr. 22, München, 81675, Germany.
| | - Varvara Turova
- Klinikum rechts der Isar, Technische Universität München, Ismaningerstr. 22, München, 81675, Germany.
| | - Irina Sidorenko
- Fakultät für Mathematik, Technische Universität München, Boltzmannstr. 3, Garching bei München, 85747, Germany.
| | - Alexander Chebotarev
- Far Eastern Federal University, Far Eastern Center for Research and Education in Mathematics, Ajax Bay 10, Russky Island, Vladivostok, 690922, Russia.
| | - Renée Lampe
- Klinikum rechts der Isar, Technische Universität München, Ismaningerstr. 22, München, 81675, Germany.
| |
Collapse
|
29
|
Sundaram SM, Arrulo Pereira A, Müller-Fielitz H, Köpke H, De Angelis M, Müller TD, Heuer H, Körbelin J, Krohn M, Mittag J, Nogueiras R, Prevot V, Schwaninger M. Gene therapy targeting the blood-brain barrier improves neurological symptoms in a model of genetic MCT8 deficiency. Brain 2022; 145:4264-4274. [PMID: 35929549 DOI: 10.1093/brain/awac243] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 06/03/2022] [Accepted: 06/22/2022] [Indexed: 12/27/2022] Open
Abstract
A genetic deficiency of the solute carrier monocarboxylate transporter 8 (MCT8), termed Allan-Herndon-Dudley syndrome, is an important cause of X-linked intellectual and motor disability. MCT8 transports thyroid hormones across cell membranes. While thyroid hormone analogues improve peripheral changes of MCT8 deficiency, no treatment of the neurological symptoms is available so far. Therefore, we tested a gene replacement therapy in Mct8- and Oatp1c1-deficient mice as a well-established model of the disease. Here, we report that targeting brain endothelial cells for Mct8 expression by intravenously injecting the vector AAV-BR1-Mct8 increased tri-iodothyronine (T3) levels in the brain and ameliorated morphological and functional parameters associated with the disease. Importantly, the therapy resulted in a long-lasting improvement in motor coordination. Thus, the data support the concept that MCT8 mediates the transport of thyroid hormones into the brain and indicate that a readily accessible vascular target can help overcome the consequences of the severe disability associated with MCT8 deficiency.
Collapse
Affiliation(s)
- Sivaraj M Sundaram
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism, University of Lübeck, 23562 Lübeck, Germany
| | - Adriana Arrulo Pereira
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism, University of Lübeck, 23562 Lübeck, Germany
| | - Helge Müller-Fielitz
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism, University of Lübeck, 23562 Lübeck, Germany
| | - Hannes Köpke
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism, University of Lübeck, 23562 Lübeck, Germany
| | - Meri De Angelis
- Institute for Diabetes and Obesity, Helmholtz Zentrum Munich, Munich, and German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany.,Institute of Experimental Genetics, Helmholtz Zentrum Munich, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Zentrum Munich, Munich, and German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Heike Heuer
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
| | - Jakob Körbelin
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism, University of Lübeck, 23562 Lübeck, Germany.,Department of Oncology, Hematology and Bone Marrow Transplantation, UKE Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Markus Krohn
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism, University of Lübeck, 23562 Lübeck, Germany
| | - Jens Mittag
- Institute for Endocrinology and Diabetes, Center of Brain, Behavior and Metabolism, University of Lübeck, 23562 Lübeck, Germany
| | - Ruben Nogueiras
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782 Santiago de Compostela, Spain
| | - Vincent Prevot
- Université Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S 1172, European Genomic Institute for Diabetes (EGID), 59045 Lille Cedex, France
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism, University of Lübeck, 23562 Lübeck, Germany.,DZHK (German Research Centre for Cardiovascular Research), Hamburg-Lübeck-Kiel, Germany
| |
Collapse
|
30
|
Cumsille P, Lara E, Verdugo-Hernández P, Acurio J, Escudero C. A robust quantitative approach for laser speckle contrast imaging perfusion analysis revealed anomalies in the brain blood flow in offspring mice of preeclampsia. Microvasc Res 2022; 144:104418. [PMID: 35931124 DOI: 10.1016/j.mvr.2022.104418] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 07/27/2022] [Accepted: 07/31/2022] [Indexed: 02/01/2023]
Abstract
Microcirculation analysis of the brain cortex is challenging because surface perfusion varies rapidly in small space-time regions and is bone protected. The laser speckle contrast imaging (LSCI) technique allows analyzing in vivo brain vascular perfusion generating a large amount of data that requires sophisticated data analytics, making researchers invest much effort in processing. Our research question was whether the reduced placental perfusion model (RUPP) of preeclampsia (PE) was associated with impaired blood perfusion in the offspring's brains. We aimed to develop a robust numerical approach that mainly consisted of applying a signal-processing tool for calculating optimal segmentation and piece-wise fits of the offspring's brain perfusion signals obtained from the LSCI technique. We combined this tool with the usual statistical analysis, implementing both in Matlab software. We performed brain perfusion measurements from offspring (five days postnatal, P5) of control pregnant dams (sham, n = 13) and of RUPP dams (RUPP, n = 7) using the Pericam® PSI-HR system at a basal condition and after thermal stimuli (warm and cold). We found that pups of RUPP mice exhibited significant differences in perfusion and vascular response to thermal stimuli compared to the sham mice. These differences were associated with high data variability in the Sham group, while in the RUPP group, perfusion looks "stiffer." Data also suggest sex-dimorphism in the vascular response since female pups in the Sham group but not male pups showed statistically significant differences in response to the warm stimulus. Again, this sex-related difference was absent in pups of RUPP mice. In conclusion, we present a robust quantitative approach for LSCI measurements that revealed anomalies in the brain blood flow in offspring of the RUPP model of PE.
Collapse
Affiliation(s)
- Patricio Cumsille
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Campus Fernando May, Av. Andrés Bello 720, Casilla 447, Chillán, Chile; Centre for Biotechnology and Bioengineering (CeBiB), University of Chile, Santiago, Chile.
| | - Evelyn Lara
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Campus Fernando May, Av. Andrés Bello 720, Casilla 447, Chillán, Chile; Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile
| | - Paula Verdugo-Hernández
- Escuela de Pedagogía en Ciencias Naturales y Exactas, Facultad de Ciencias de la Educación, Universidad de Talca, Chile
| | - Jesenia Acurio
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Campus Fernando May, Av. Andrés Bello 720, Casilla 447, Chillán, Chile; Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile
| | - Carlos Escudero
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Campus Fernando May, Av. Andrés Bello 720, Casilla 447, Chillán, Chile; Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile.
| |
Collapse
|
31
|
Morphogenesis of vascular and neuronal networks and the relationships between their remodeling processes. Brain Res Bull 2022; 186:62-69. [DOI: 10.1016/j.brainresbull.2022.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/18/2022] [Accepted: 05/29/2022] [Indexed: 11/21/2022]
|
32
|
Critical Role of Neuronal Vps35 in Blood Vessel Branching and Maturation in Developing Mouse Brain. Biomedicines 2022; 10:biomedicines10071653. [PMID: 35884959 PMCID: PMC9313219 DOI: 10.3390/biomedicines10071653] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/05/2022] [Accepted: 07/07/2022] [Indexed: 11/17/2022] Open
Abstract
Vps35 (vacuolar protein sorting 35), a key component of retromer, plays a crucial role in selective retrieval of transmembrane proteins from endosomes to trans-Golgi networks. Dysfunctional Vps35/retromer is a risk factor for the development of neurodegenerative diseases. Vps35 is highly expressed in developing pyramidal neurons, both in the mouse neocortex and hippocampus, Although embryonic neuronal Vps35’s function in promoting neuronal terminal differentiation and survival is evident, it remains unclear whether and how neuronal Vps35 communicates with other types of brain cells, such as blood vessels (BVs), which are essential for supplying nutrients to neurons. Dysfunctional BVs contribute to the pathogenesis of various neurodegenerative disorders. Here, we provide evidence for embryonic neuronal Vps35 as critical for BV branching and maturation in the developing mouse brain. Selectively knocking out (KO) Vps35 in mouse embryonic, not postnatal, neurons results in reductions in BV branching and density, arteriole diameter, and BV-associated pericytes and microglia but an increase in BV-associated reactive astrocytes. Deletion of microglia by PLX3397 enhances these BV deficits in mutant mice. These results reveal the function of neuronal Vps35 in neurovascular coupling in the developing mouse brain and implicate BV-associated microglia as underlying this event.
Collapse
|
33
|
Coelho-Santos V, Tieu T, Shih AY. Reinforced thinned-skull window for repeated imaging of the neonatal mouse brain. NEUROPHOTONICS 2022; 9:031918. [PMID: 35673538 PMCID: PMC9163199 DOI: 10.1117/1.nph.9.3.031918] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 05/03/2022] [Indexed: 06/15/2023]
Abstract
Significance: Two-photon microscopy is a powerful tool for in vivo imaging of the mammalian brain at cellular to subcellular resolution. However, resources that describe methods for imaging live newborn mice have remained sparse. Aim: We describe a non-invasive cranial window procedure for longitudinal imaging of neonatal mice. Approach: We demonstrate construction of the cranial window by iterative shaving of the calvarium of P0 to P12 mouse pups. We use the edge of a syringe needle and scalpel blades to thin the bone to ∼ 15 - μ m thickness. The window is then reinforced with cyanoacrylate glue and a coverslip to promote stability and optical access for at least a week. The head cap also includes a light-weight aluminum flange for head-fixation during imaging. Results: The resulting chronic thinned-skull window enables in vivo imaging to a typical cortical depth of ∼ 200 μ m without disruption of the intracranial environment. We highlight techniques to measure vascular structure and blood flow during development, including use of intravenous tracers and transgenic mice to label the blood plasma and vascular cell types, respectively. Conclusions: This protocol enables direct visualization of the developing neurogliovascular unit in the live neonatal brain during both normal and pathological states.
Collapse
Affiliation(s)
- Vanessa Coelho-Santos
- Seattle Children’s Research Institute, Center for Developmental Biology and Regenerative Medicine, Seattle, Washington, United States
- University of Washington, Department of Pediatrics, Seattle, Washington, United States
| | - Taryn Tieu
- Seattle Children’s Research Institute, Center for Developmental Biology and Regenerative Medicine, Seattle, Washington, United States
| | - Andy Y. Shih
- Seattle Children’s Research Institute, Center for Developmental Biology and Regenerative Medicine, Seattle, Washington, United States
- University of Washington, Department of Pediatrics, Seattle, Washington, United States
- University of Washington, Department of Bioengineering, Seattle, Washington, United States
| |
Collapse
|
34
|
Brosolo M, Lecointre M, Laquerrière A, Janin F, Genty D, Lebon A, Lesueur C, Vivien D, Marret S, Marguet F, Gonzalez BJ. In utero alcohol exposure impairs vessel-associated positioning and differentiation of oligodendrocytes in the developing neocortex. Neurobiol Dis 2022; 171:105791. [PMID: 35760273 DOI: 10.1016/j.nbd.2022.105791] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/30/2022] [Accepted: 06/10/2022] [Indexed: 11/16/2022] Open
Abstract
Prenatal alcohol exposure (PAE) is a major cause of nongenetic mental retardation and can lead to fetal alcohol syndrome (FAS), the most severe manifestation of fetal alcohol spectrum disorder (FASD). FASD infants present behavioral disabilities resulting from neurodevelopmental defects. Both grey and white matter lesions have been characterized and are associated with apoptotic death and/or ectopic migration profiles. In the last decade, it was shown that PAE impairs brain angiogenesis, and the radial organization of cortical microvessels is lost. Concurrently, several studies have reported that tangential migration of oligodendrocyte precursors (OPCs) originating from ganglionic eminences is vascular associated. Because numerous migrating oligodendrocytes enter the developing neocortex, the present study aimed to determine whether migrating OPCs interacted with radial cortical microvessels and whether alcohol-induced vascular impairments were associated with altered positioning and differentiation of cortical oligodendrocytes. Using a 3D morphometric analysis, the results revealed that in both human and mouse cortices, 15 to 40% of Olig2-positive cells were in close association with radial cortical microvessels, respectively. Despite perinatal vascular disorganization, PAE did not modify the vessel association of Olig2-positive cells but impaired their positioning between deep and superficial cortical layers. At the molecular level, PAE markedly but transiently reduced the expression of CNPase and MBP, two differentiation markers of immature and mature oligodendrocytes. In particular, PAE inverted their distribution profiles in cortical layers V and VI and reduced the thickness of the myelin sheath of efferent axons. These perinatal oligo-vascular defects were associated with motor disabilities that persisted in adults. Altogether, the present study provides the first evidence that Olig2-positive cells entering the neocortex are associated with radial microvessels. PAE disorganized the cortical microvasculature and delayed the positioning and differentiation of oligodendrocytes. Although most of these oligovascular defects occurred in perinatal life, the offspring developed long-term motor troubles. Altogether, these data suggest that alcohol-induced oligo-vascular impairments contribute to the neurodevelopmental issues described in FASD.
Collapse
Affiliation(s)
- M Brosolo
- Normandie Univ, UNIROUEN, INSERM U1245, Normandy Centre for Genomic and Personalized Medicine, F 76000 Rouen, France
| | - M Lecointre
- Normandie Univ, UNIROUEN, INSERM U1245, Normandy Centre for Genomic and Personalized Medicine, F 76000 Rouen, France
| | - A Laquerrière
- Normandie Univ, UNIROUEN, INSERM U1245, Normandy Centre for Genomic and Personalized Medicine, F 76000 Rouen, France; Department of Pathology, Rouen University Hospital, 76000 Rouen, France
| | - F Janin
- Normandie Univ, UNIROUEN, INSERM U1245, Normandy Centre for Genomic and Personalized Medicine, F 76000 Rouen, France
| | - D Genty
- Department of Pathology, Rouen University Hospital, 76000 Rouen, France
| | - A Lebon
- Normandie Univ, UNIROUEN, INSERM US 51, CNRS UAR 2026, HeRacLeS-PRIMACEN, 76000 Rouen, France
| | - C Lesueur
- Normandie Univ, UNIROUEN, INSERM U1245, Normandy Centre for Genomic and Personalized Medicine, F 76000 Rouen, France
| | - D Vivien
- Normandie Univ, UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), 14000 Caen, France; Department of Clinical Research, Caen-Normandie University Hospital, CHU, Avenue de la côte de Nacre, Caen, France
| | - S Marret
- Normandie Univ, UNIROUEN, INSERM U1245, Normandy Centre for Genomic and Personalized Medicine, F 76000 Rouen, France; Department of Neonatal Pediatrics and Intensive Care, Rouen University Hospital, 76000 Rouen, France
| | - F Marguet
- Normandie Univ, UNIROUEN, INSERM U1245, Normandy Centre for Genomic and Personalized Medicine, F 76000 Rouen, France; Department of Pathology, Rouen University Hospital, 76000 Rouen, France
| | - B J Gonzalez
- Normandie Univ, UNIROUEN, INSERM U1245, Normandy Centre for Genomic and Personalized Medicine, F 76000 Rouen, France.
| |
Collapse
|
35
|
Shen J, Miao X, Vu C, Xu B, González-Zacarías C, Nederveen AJ, Wood JC. Anemia Increases Oxygen Extraction Fraction in Deep Brain Structures but Not in the Cerebral Cortex. Front Physiol 2022; 13:896006. [PMID: 35784894 PMCID: PMC9248375 DOI: 10.3389/fphys.2022.896006] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 05/19/2022] [Indexed: 01/26/2023] Open
Abstract
Sickle cell disease (SCD) is caused by a single amino acid mutation in hemoglobin, causing chronic anemia and neurovascular complications. However, the effects of chronic anemia on oxygen extraction fraction (OEF), especially in deep brain structures, are less well understood. Conflicting OEF values have been reported in SCD patients, but have largely attributed to different measurement techniques, faulty calibration, and different locations of measurement. Thus, in this study, we investigated the reliability and agreement of two susceptibility-based methods, quantitative susceptibility mapping (QSM) and complex image summation around a spherical or a cylindrical object (CISSCO), for OEF measurements in internal cerebral vein (ICV), reflecting oxygen saturation in deep brain structures. Both methods revealed that SCD patients and non-sickle anemia patients (ACTL) have increased OEF in ICV (42.6% ± 5.6% and 30.5% ± 3.6% in SCD by CISSCO and QSM respectively, 37.0% ± 4.1% and 28.5% ± 2.3% in ACTL) compared with controls (33.0% ± 2.3% and 26.8% ± 1.8%). OEF in ICV varied reciprocally with hematocrit (r 2 = 0.92, 0.53) and oxygen content (r 2 = 0.86, 0.53) respectively. However, an opposite relationship was observed for OEF measurements in sagittal sinus (SS) with the widely used T2-based oximetry, T2-Relaxation-Under-Spin-Tagging (TRUST), in the same cohorts (31.2% ± 6.6% in SCD, 33.3% ± 5.9% in ACTL and 36.8% ± 5.6% in CTL). Importantly, we demonstrated that hemoglobin F and other fast moving hemoglobins decreased OEF by TRUST and explained group differences in sagittal sinus OEF between anemic and control subjects. These data demonstrate that anemia causes deep brain hypoxia in anemia subjects with concomitant preservation of cortical oxygenation, as well as the key interaction of the hemoglobin dissociation curve and cortical oxygen extraction.
Collapse
Affiliation(s)
- Jian Shen
- Biomedical Engineering, University of Southern California, Los Angeles, Los Angeles, CA, United States
| | - Xin Miao
- Siemens, Boston, MA, United States
| | - Chau Vu
- Biomedical Engineering, University of Southern California, Los Angeles, Los Angeles, CA, United States
| | - Botian Xu
- Biomedical Engineering, University of Southern California, Los Angeles, Los Angeles, CA, United States
| | - Clio González-Zacarías
- Neuroscience Graduate Program, University of Southern California, Los Angeles, Los Angeles, CA, United States
| | - Aart J. Nederveen
- Amsterdam UMC, Radiology and Nuclear Medicine, University of Amsterdam, Amsterdam, Netherlands
| | - John C. Wood
- Biomedical Engineering, University of Southern California, Los Angeles, Los Angeles, CA, United States,Department of Pediatrics and Radiology, Children’s Hospital Los Angeles, Los Angeles, CA, United States,*Correspondence: John C. Wood,
| |
Collapse
|
36
|
Blondel S, Strazielle N, Amara A, Guy R, Bain C, Rose A, Guibaud L, Tiribelli C, Gazzin S, Ghersi-Egea JF. Vascular network expansion, integrity of blood-brain interfaces, and cerebrospinal fluid cytokine concentration during postnatal development in the normal and jaundiced rat. Fluids Barriers CNS 2022; 19:47. [PMID: 35672829 PMCID: PMC9172137 DOI: 10.1186/s12987-022-00332-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 04/20/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Severe neonatal jaundice resulting from elevated levels of unconjugated bilirubin in the blood induces dramatic neurological impairment. Central oxidative stress and an inflammatory response have been associated with the pathophysiological mechanism. Cells forming the blood-brain barrier and the choroidal blood-CSF barrier are the first CNS cells exposed to increased plasma levels of unconjugated bilirubin. These barriers are key regulators of brain homeostasis and require active oxidative metabolism to fulfill their protective functions. The choroid plexus-CSF system is involved in neuroinflammatory processes. In this paper, we address the impact of neonatal hyperbilirubinemia on some aspects of brain barriers. We describe physiological changes in the neurovascular network, blood-brain/CSF barriers integrities, and CSF cytokine levels during the postnatal period in normobilirubinemic animals, and analyze these parameters in parallel in Gunn rats that are deficient in bilirubin catabolism and develop postnatal hyperbilirubinemia. METHODS Gunn rats bearing a mutation in UGT1a genes were used. The neurovascular network was analyzed by immunofluorescence stereomicroscopy. The integrity of the barriers was evaluated by [14C]-sucrose permeability measurement. CSF cytokine levels were measured by multiplex immunoassay. The choroid plexus-CSF system response to an inflammatory challenge was assessed by enumerating CSF leukocytes. RESULTS In normobilirubinemic animals, the neurovascular network expands postnatally and displays stage-specific regional variations in its complexity. Network expansion is not affected by hyperbilirubinemia. Permeability of the blood-brain and blood-CSF barriers to sucrose decreases between one- and 9-day-old animals, and does not differ between normobilirubinemic and hyperbilirubinemic rats. Cytokine profiles differ between CSF and plasma in all 1-, 9-, and 18-day-old animals. The CSF cytokine profile in 1-day-old animals is markedly different from that established in older animals. Hyperbilirubinemia perturbs these cytokine profiles only to a very limited extent, and reduces CSF immune cell infiltration triggered by systemic exposure to a bacterial lipopeptide. CONCLUSION The data highlight developmental specificities of the blood-brain barrier organization and of CSF cytokine content. They also indicate that a direct effect of bilirubin on the vascular system organization, brain barriers morphological integrity, and inflammatory response of the choroid plexus-CSF system is not involved in the alteration of brain functions induced by severe neonatal jaundice.
Collapse
Affiliation(s)
| | - Nathalie Strazielle
- Brain-i, Lyon, France
- Fluid Team Lyon Neurosciences Research Center, INSERM U1028, CNRS UMR5292, Lyon University, Bron, France
| | - Amel Amara
- Fluid Team Lyon Neurosciences Research Center, INSERM U1028, CNRS UMR5292, Lyon University, Bron, France
| | - Rainui Guy
- BIP Facility, Lyon Neurosciences Research Center, Bron, France
| | | | | | - Laurent Guibaud
- Fluid Team Lyon Neurosciences Research Center, INSERM U1028, CNRS UMR5292, Lyon University, Bron, France
| | - Claudio Tiribelli
- Fondazione Italiana Fegato-Onlus, AREA Science Park, Basovizza, Trieste, Italy
| | - Silvia Gazzin
- Fondazione Italiana Fegato-Onlus, AREA Science Park, Basovizza, Trieste, Italy
| | - Jean-François Ghersi-Egea
- BIP Facility, Lyon Neurosciences Research Center, Bron, France.
- Fluid Team Lyon Neurosciences Research Center, INSERM U1028, CNRS UMR5292, Lyon University, Bron, France.
| |
Collapse
|
37
|
Bonney SK, Sullivan LT, Cherry TJ, Daneman R, Shih AY. Distinct features of brain perivascular fibroblasts and mural cells revealed by in vivo two-photon imaging. J Cereb Blood Flow Metab 2022; 42:966-978. [PMID: 34929105 PMCID: PMC9125487 DOI: 10.1177/0271678x211068528] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 11/11/2021] [Accepted: 11/19/2021] [Indexed: 12/04/2022]
Abstract
Perivascular fibroblasts (PVFs) are recognized for their pro-fibrotic role in many central nervous system disorders. Like mural cells, PVFs surround blood vessels and express Pdgfrβ. However, these shared attributes hinder the ability to distinguish PVFs from mural cells. We used in vivo two-photon imaging and transgenic mice with PVF-targeting promoters (Col1a1 or Col1a2) to compare the structure and distribution of PVFs and mural cells in cerebral cortex of healthy, adult mice. We show that PVFs localize to all cortical penetrating arterioles and their offshoots (arteriole-capillary transition zone), as well as the main trunk of only larger ascending venules. However, the capillary zone is devoid of PVF coverage. PVFs display short-range mobility along the vessel wall and exhibit distinct structural features (flattened somata and thin ruffled processes) not seen with smooth muscle cells or pericytes. These findings clarify that PVFs and mural cells are distinct cell types coexisting in a similar perivascular niche.
Collapse
Affiliation(s)
- Stephanie K Bonney
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA, USA
| | - Liam T Sullivan
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA, USA
| | - Timothy J Cherry
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA, USA
| | - Richard Daneman
- Departments of Neurosciences and Pharmacology, University of California San Diego, La Jolla, CA, USA
| | - Andy Y Shih
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| |
Collapse
|
38
|
Reeson P, Schager B, Van Sprengel M, Brown CE. Behavioral and Neural Activity-Dependent Recanalization of Plugged Capillaries in the Brain of Adult and Aged Mice. Front Cell Neurosci 2022; 16:876746. [PMID: 35722620 PMCID: PMC9204343 DOI: 10.3389/fncel.2022.876746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/12/2022] [Indexed: 11/13/2022] Open
Abstract
The capillaries of the brain, owing to their small diameter and low perfusion pressure, are vulnerable to interruptions in blood flow. These tiny occlusions can have outsized consequences on angioarchitecture and brain function; especially when exacerbated by disease states or accumulate with aging. A distinctive feature of the brain’s microvasculature is the ability for active neurons to recruit local blood flow. The coupling of neural activity to blood flow could play an important role in recanalizing obstructed capillaries. To investigate this idea, we experimentally induced capillary obstructions in mice by injecting fluorescent microspheres and then manipulated neural activity levels though behavioral or pharmacologic approaches. We show that engaging adult and aged mice with 12 h exposure to an enriched environment (group housing, novel objects, exercise wheels) was sufficient to significantly reduce the density of obstructed capillaries throughout the forebrain. In order to more directly manipulate neural activity, we pharmacologically suppressed or increased neuronal activity in the somatosensory cortex. When we suppressed cortical activity, recanalization was impaired given the density of obstructed capillaries was significantly increased. Conversely, increasing cortical activity improved capillary recanalization. Since systemic cardiovascular factors (changes in heart rate, blood pressure) could explain these effects on recanalization, we demonstrate that unilateral manipulations of neural activity through whisker trimming or injection of muscimol, still had significant and hemisphere specific effects on recanalization, even in mice exposed to enrichment where cardiovascular effects would be evident in both hemispheres. In summary, our studies reveal that neural activity bi-directionally regulates the recanalization of obstructed capillaries. Further, we show that stimulating brain activity through behavioral engagement (i.e., environmental enrichment) can promote vascular health throughout the lifespan.
Collapse
Affiliation(s)
- Patrick Reeson
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Ben Schager
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | | | - Craig E. Brown
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
- *Correspondence: Craig E. Brown,
| |
Collapse
|
39
|
Lin X, Chen L, Jullienne A, Zhang H, Salehi A, Hamer M, C. Holmes T, Obenaus A, Xu X. Longitudinal dynamics of microvascular recovery after acquired cortical injury. Acta Neuropathol Commun 2022; 10:59. [PMID: 35468870 PMCID: PMC9036719 DOI: 10.1186/s40478-022-01361-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 04/05/2022] [Indexed: 01/04/2023] Open
Abstract
Acquired brain injuries due to trauma damage the cortical vasculature, which in turn impairs blood flow to injured tissues. There are reports of vascular morphological recovery following traumatic brain injury, but the remodeling process has not been examined longitudinally in detail after injury in vivo. Understanding the dynamic processes that influence recovery is thus critically important. We evaluated the longitudinal and dynamic microvascular recovery and remodeling up to 2 months post injury using live brain miniscope and 2-photon microscopic imaging. The new imaging approaches captured dynamic morphological and functional recovery processes at high spatial and temporal resolution in vivo. Vessel painting documented the initial loss and subsequent temporal morphological vascular recovery at the injury site. Miniscopes were used to longitudinally image the temporal dynamics of vascular repair in vivo after brain injury in individual mice across each cohort. We observe near-immediate nascent growth of new vessels in and adjacent to the injury site that peaks between 14 and 21 days post injury. 2-photon microscopy confirms new vascular growth and further demonstrates differences between cortical layers after cortical injury: large vessels persist in deeper cortical layers (> 200 μm), while superficial layers exhibit a dense plexus of fine (and often non-perfused) vessels displaying regrowth. Functionally, blood flow increases mirror increasing vascular density. Filopodia development and endothelial sprouting is measurable within 3 days post injury that rapidly transforms regions devoid of vessels to dense vascular plexus in which new vessels become increasingly perfused. Within 7 days post injury, blood flow is observed in these nascent vessels. Behavioral analysis reveals improved vascular modulation after 9 days post injury, consistent with vascular regrowth. We conclude that morphological recovery events are closely linked to functional recovery of blood flow to the compromised tissues, which subsequently leads to improved behavioral outcomes.
Collapse
|
40
|
Taïb S, Lamandé N, Martin S, Coulpier F, Topilko P, Brunet I. Myelinating Schwann cells and Netrin-1 control intra-nervous vascularization of the developing mouse sciatic nerve. eLife 2022; 11:64773. [PMID: 35019839 PMCID: PMC8782568 DOI: 10.7554/elife.64773] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 01/11/2022] [Indexed: 11/13/2022] Open
Abstract
Peripheral nerves are vascularized by a dense network of blood vessels to guarantee their complex function. Despite the crucial role of vascularization to ensure nerve homeostasis and regeneration, the mechanisms governing nerve invasion by blood vessels remain poorly understood. We found, in mice, that the sciatic nerve invasion by blood vessels begins around embryonic day 16 and continues until birth. Interestingly, intra-nervous blood vessel density significantly decreases during post-natal period, starting from P10. We show that, while the axon guidance molecule Netrin-1 promotes nerve invasion by blood vessels via the endothelial receptor UNC5B during embryogenesis, myelinated Schwann cells negatively control intra-nervous vascularization during post-natal period.
Collapse
Affiliation(s)
- Sonia Taïb
- Center for Interdisciplinary Research in Biology, Collège de France, Paris, France
| | - Noël Lamandé
- Center for Interdisciplinary Research in Biology, Collège de France, Paris, France
| | - Sabrina Martin
- Center for Interdisciplinary Research in Biology, Collège de France, Paris, France
| | - Fanny Coulpier
- UMR U955 INSERM UPEC, Institut Mondor de Recherche Biomédicale, Créteil, France
| | - Piotr Topilko
- UMR U955 INSERM UPEC, Institut Mondor de Recherche Biomédicale, Créteil, France
| | - Isabelle Brunet
- Center for Interdisciplinary Research in Biology, Collège de France, Paris, France
| |
Collapse
|
41
|
Guo F, Zhang S. Hypoxia inducible factor and diffuse white matter injury in the premature brain: perspectives from genetic studies in mice. Neural Regen Res 2022; 17:105-107. [PMID: 34100442 PMCID: PMC8451551 DOI: 10.4103/1673-5374.314301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Fuzheng Guo
- Department of Neurology, School of Medicine, the University of California, Davis; Institute for Pediatric Regenerative Medicine (IPRM), Shriners Hospitals for Children, Northern California, Sacramento, CA, USA
| | - Sheng Zhang
- Department of Neurology, School of Medicine, the University of California, Davis; Institute for Pediatric Regenerative Medicine (IPRM), Shriners Hospitals for Children, Northern California, Sacramento, CA, USA
| |
Collapse
|
42
|
Korom M, Camacho MC, Filippi CA, Licandro R, Moore LA, Dufford A, Zöllei L, Graham AM, Spann M, Howell B, Shultz S, Scheinost D. Dear reviewers: Responses to common reviewer critiques about infant neuroimaging studies. Dev Cogn Neurosci 2021; 53:101055. [PMID: 34974250 PMCID: PMC8733260 DOI: 10.1016/j.dcn.2021.101055] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 11/28/2021] [Accepted: 12/26/2021] [Indexed: 01/07/2023] Open
Abstract
The field of adult neuroimaging relies on well-established principles in research design, imaging sequences, processing pipelines, as well as safety and data collection protocols. The field of infant magnetic resonance imaging, by comparison, is a young field with tremendous scientific potential but continuously evolving standards. The present article aims to initiate a constructive dialog between researchers who grapple with the challenges and inherent limitations of a nascent field and reviewers who evaluate their work. We address 20 questions that researchers commonly receive from research ethics boards, grant, and manuscript reviewers related to infant neuroimaging data collection, safety protocols, study planning, imaging sequences, decisions related to software and hardware, and data processing and sharing, while acknowledging both the accomplishments of the field and areas of much needed future advancements. This article reflects the cumulative knowledge of experts in the FIT’NG community and can act as a resource for both researchers and reviewers alike seeking a deeper understanding of the standards and tradeoffs involved in infant neuroimaging. The field of infant MRI is young with evolving standards. We address 20 questions that researchers commonly receive reviewers. These come from research ethics boards, grant, and manuscript reviewers. This article reflects the cumulative knowledge of experts in the FIT’NG community.
Collapse
Affiliation(s)
- Marta Korom
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, USA.
| | - M Catalina Camacho
- Division of Biology and Biomedical Sciences (Neurosciences), Washington University School of Medicine, St. Louis, MO, USA.
| | - Courtney A Filippi
- Emotion and Development Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - Roxane Licandro
- Institute of Visual Computing and Human-Centered Technology, Computer Vision Lab, TU Wien, Vienna, Austria; Department of Biomedical Imaging and Image-guided Therapy, Computational Imaging Research, Medical University of Vienna, Vienna, Austria
| | - Lucille A Moore
- Department of Psychiatry, Oregon Health and Science University, Portland, OR, USA
| | - Alexander Dufford
- Department of Radiology & Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Lilla Zöllei
- A.A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Alice M Graham
- Department of Psychiatry, Oregon Health and Science University, Portland, OR, USA
| | - Marisa Spann
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Brittany Howell
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Department of Human Development and Family Science, Virginia Polytechnic Institute and State University, Roanoke, VA, USA
| | | | - Sarah Shultz
- Division of Autism & Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA; Marcus Autism Center, Children's Healthcare of Atlanta, Atlanta, GA, USA.
| | - Dustin Scheinost
- Department of Radiology & Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
43
|
Seker FB, Fan Z, Gesierich B, Gaubert M, Sienel RI, Plesnila N. Neurovascular Reactivity in the Aging Mouse Brain Assessed by Laser Speckle Contrast Imaging and 2-Photon Microscopy: Quantification by an Investigator-Independent Analysis Tool. Front Neurol 2021; 12:745770. [PMID: 34858312 PMCID: PMC8631776 DOI: 10.3389/fneur.2021.745770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 10/08/2021] [Indexed: 11/13/2022] Open
Abstract
The brain has a high energy demand but little to no energy stores. Therefore, proper brain function relies on the delivery of glucose and oxygen by the cerebral vasculature. The regulation of cerebral blood flow (CBF) occurs at the level of the cerebral capillaries and is driven by a fast and efficient crosstalk between neurons and vessels, a process termed neurovascular coupling (NVC). Experimentally NVC is mainly triggered by sensory stimulation and assessed by measuring either CBF by laser Doppler fluxmetry, laser speckle contrast imaging (LSCI), intrinsic optical imaging, BOLD fMRI, near infrared spectroscopy (NIRS) or functional ultrasound imaging (fUS). Since these techniques have relatively low spatial resolution, diameters of cerebral vessels are mainly assessed by 2-photon microscopy (2-PM). Results of studies on NVC rely on stable animal physiology, high-quality data acquisition, and unbiased data analysis, criteria, which are not easy to achieve. In the current study, we assessed NVC using two different imaging modalities, i.e., LSCI and 2-PM, and analyzed our data using an investigator-independent Matlab-based analysis tool, after manually defining the area of analysis in LSCI and vessels to measure in 2-PM. By investigating NVC in 6–8 weeks, 1-, and 2-year-old mice, we found that NVC was maximal in 1-year old mice and was significantly reduced in aged mice. These findings suggest that NVC is differently affected during the aging process. Most interestingly, specifically pial arterioles, seem to be distinctly affected by the aging. The main finding of our study is that the automated analysis tool works very efficiently in terms of time and accuracy. In fact, the tool reduces the analysis time of one animal from approximately 23 h to about 2 s while basically making no mistakes. In summary, we developed an experimental workflow, which allows us to reliably measure NVC with high spatial and temporal resolution in young and aged mice and to analyze these data in an investigator-independent manner.
Collapse
Affiliation(s)
- Fatma Burcu Seker
- Institute for Stroke and Dementia Research, Munich University Hospital and University of Munich, Munich, Germany
| | - Ziyu Fan
- Institute for Stroke and Dementia Research, Munich University Hospital and University of Munich, Munich, Germany
| | - Benno Gesierich
- Institute for Stroke and Dementia Research, Munich University Hospital and University of Munich, Munich, Germany
| | - Malo Gaubert
- Institute for Stroke and Dementia Research, Munich University Hospital and University of Munich, Munich, Germany
| | - Rebecca Isabella Sienel
- Institute for Stroke and Dementia Research, Munich University Hospital and University of Munich, Munich, Germany
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research, Munich University Hospital and University of Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
44
|
Kim S, Lee S, Lim J, Choi H, Kang H, Jeon NL, Son Y. Human bone marrow-derived mesenchymal stem cells play a role as a vascular pericyte in the reconstruction of human BBB on the angiogenesis microfluidic chip. Biomaterials 2021; 279:121210. [PMID: 34710793 DOI: 10.1016/j.biomaterials.2021.121210] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 10/07/2021] [Accepted: 10/20/2021] [Indexed: 12/16/2022]
Abstract
A blood-brain barrier (BBB) on a chip similar to the in vivo BBB is important for evaluating the efficacy of reparative cell therapeutics for ischemic stroke in vitro. In this study, we established human BBB-like microvasculature on an angiogenesis microfluidic chip and analyzed the role of human pericytes (hPCs) and human astrocytes (hACs) on the architecture of human brain microvascular endothelial cells (hBMEC)-derived microvasculature on a chip. We found that human bone marrow mesenchymal stem cells (hBM-MSCs) play a role as perivascular pericytes in tight BBB reformation with a better vessel-constrictive capacity than that of hPCs, providing evidence of reparative stem cells on BBB repair rather than a paracrine effect. We also demonstrated that pericytes play an important role in vessel constriction, and astrocytes may induce the maturation of a capillary network. Higher expression of VEGF, SDF-1α, PDGFRβ, N-cadherin, and α-SMA in hBM-MSCs than in hPCs and their subsequent downregulation with hBMEC co-culture suggest that hBM-MSCs may be better recruited and engaged in the BBB-microvasculature than hPCs. Collectively, the human BBB on a chip may be adopted as an alternative to evaluate in vitro cellular behavior and the engagement of cell therapeutics in BBB regeneration and may also be used for studying stroke.
Collapse
Affiliation(s)
- Sumin Kim
- Department of Genetic Biotechnology, College of Life Science and Graduate School of Biotechnology, Kyung Hee University, Yong in, 17104, South Korea
| | - Somin Lee
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, South Korea
| | - Jungeun Lim
- Department of Mechanical Engineering, Seoul National University, Seoul, 08826, South Korea
| | - Hyeri Choi
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, South Korea
| | - Habin Kang
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, South Korea
| | - Noo Li Jeon
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, South Korea; Department of Mechanical Engineering, Seoul National University, Seoul, 08826, South Korea
| | - Youngsook Son
- Department of Genetic Biotechnology, College of Life Science and Graduate School of Biotechnology, Kyung Hee University, Yong in, 17104, South Korea; Kyung Hee Institute of Regenerative Medicine (KIRM), Medical Science Research Institute, Kyung Hee University Hospital, Seoul, 02447, South Korea.
| |
Collapse
|
45
|
Tang M, Monani UR. Glut1 deficiency syndrome: New and emerging insights into a prototypical brain energy failure disorder. Neurosci Insights 2021; 16:26331055211011507. [PMID: 34589708 PMCID: PMC8474335 DOI: 10.1177/26331055211011507] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/01/2021] [Indexed: 11/16/2022] Open
Abstract
Considering its small size relative to the rest of the body, the
mammalian brain has a disproportionately high energy requirement. This
energy is supplied to the brain mainly in the form of glucose through
the principal cerebral glucose transporter, Glut1. Inactivation of
even a single copy of the Glut1 gene, SLC2A1, has
dire consequences for the brain, starving cerebral neurons of energy
and triggering the debilitating neurodevelopmental disorder, Glut1
deficiency syndrome (Glut1 DS). Considering the monogenic nature of
Glut1 DS, the disease serves as an excellent paradigm to study the
larger family of brain energy failure syndromes. Here we review how
studies of Glut1 DS are proving instructive to the brain’s energy
needs, focusing first on the requirements, both spatial and temporal
of the transporter, second, on proposed mechanisms linking low Glut1
to brain dysfunction and, finally on efforts to treat the disease and
thus restore nutritional support to the brain. These studies promise
not only to inform mechanisms and treatments for the relatively rare
Glut1 DS but also the myriad other conditions involving the Glut1
protein.
Collapse
Affiliation(s)
- Maoxue Tang
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA.,Center for Motor Neuron Biology and Disease, Columbia University Irving Medical Center, New York, NY, USA
| | - Umrao R Monani
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA.,Center for Motor Neuron Biology and Disease, Columbia University Irving Medical Center, New York, NY, USA.,Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
46
|
Wälchli T, Bisschop J, Miettinen A, Ulmann-Schuler A, Hintermüller C, Meyer EP, Krucker T, Wälchli R, Monnier PP, Carmeliet P, Vogel J, Stampanoni M. Hierarchical imaging and computational analysis of three-dimensional vascular network architecture in the entire postnatal and adult mouse brain. Nat Protoc 2021; 16:4564-4610. [PMID: 34480130 DOI: 10.1038/s41596-021-00587-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 06/08/2021] [Indexed: 02/08/2023]
Abstract
The formation of new blood vessels and the establishment of vascular networks are crucial during brain development, in the adult healthy brain, as well as in various diseases of the central nervous system. Here, we describe a step-by-step protocol for our recently developed method that enables hierarchical imaging and computational analysis of vascular networks in postnatal and adult mouse brains. The different stages of the procedure include resin-based vascular corrosion casting, scanning electron microscopy, synchrotron radiation and desktop microcomputed tomography imaging, and computational network analysis. Combining these methods enables detailed visualization and quantification of the 3D brain vasculature. Network features such as vascular volume fraction, branch point density, vessel diameter, length, tortuosity and directionality as well as extravascular distance can be obtained at any developmental stage from the early postnatal to the adult brain. This approach can be used to provide a detailed morphological atlas of the entire mouse brain vasculature at both the postnatal and the adult stage of development. Our protocol allows the characterization of brain vascular networks separately for capillaries and noncapillaries. The entire protocol, from mouse perfusion to vessel network analysis, takes ~10 d.
Collapse
Affiliation(s)
- Thomas Wälchli
- Group of CNS Angiogenesis and Neurovascular Link, Neuroscience Center Zurich, and Division of Neurosurgery, University and University Hospital Zurich, Zurich, Switzerland.
- Division of Neurosurgery, University Hospital Zurich, Zurich, Switzerland.
- Group Brain Vasculature and Perivascular Niche, Division of Experimental and Translational Neuroscience, Krembil Brain Institute, Krembil Research Institute, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada.
- Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, University of Toronto, Toronto, Ontario, Canada.
| | - Jeroen Bisschop
- Group of CNS Angiogenesis and Neurovascular Link, Neuroscience Center Zurich, and Division of Neurosurgery, University and University Hospital Zurich, Zurich, Switzerland
- Division of Neurosurgery, University Hospital Zurich, Zurich, Switzerland
- Group Brain Vasculature and Perivascular Niche, Division of Experimental and Translational Neuroscience, Krembil Brain Institute, Krembil Research Institute, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
- Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, University of Toronto, Toronto, Ontario, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Arttu Miettinen
- Swiss Light Source, Paul Scherrer Institute, Villigen, Switzerland
- Institute for Biomedical Engineering, University and ETH Zurich, Zurich, Switzerland
- Department of Physics, University of Jyväskylä, Jyväskylä, Finland
| | | | | | - Eric P Meyer
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Thomas Krucker
- Novartis Institutes for BioMedical Research Inc, Emeryville, CA, USA
| | - Regula Wälchli
- Department of Dermatology, Pediatric Skin Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Philippe P Monnier
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Krembil Research Institute, Vision Division, Krembil Discovery Tower, Toronto, Ontario, Canada
- Department of Ophthalmology and Vision Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, VIB Center for Cancer Biology, VIB, Leuven, Belgium
| | - Johannes Vogel
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Marco Stampanoni
- Swiss Light Source, Paul Scherrer Institute, Villigen, Switzerland
- Institute for Biomedical Engineering, University and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
47
|
Not just a 'drain': venules sprout brain capillaries. Trends Neurosci 2021; 44:849-851. [PMID: 34452754 DOI: 10.1016/j.tins.2021.08.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 08/11/2021] [Indexed: 11/21/2022]
Abstract
The cerebral cortex requires a dense, highly organized network of vasculature that ensures high-volume and continuous oxygen delivery to metabolically active neuronal circuits. In a recent paper, Coelho-Santos et al. used in vivo two-photon microscopy to reveal how this precise network is constructed during a short window of mouse postnatal development.
Collapse
|
48
|
Abstract
The CNS vasculature tightly regulates the passage of circulating molecules and leukocytes into the CNS. In the neuroinflammatory disease multiple sclerosis (MS), these regulatory mechanisms fail, and autoreactive T cells invade the CNS via blood vessels, leading to neurological deficits depending on where the lesions are located. The region-specific mechanisms directing the development of such lesions are not well understood. In this study, we investigated whether pericytes regulate CNS endothelial cell permissiveness toward leukocyte trafficking into the brain parenchyma. By using a pericyte-deficient mouse model, we show that intrinsic changes in the brain vasculature due to absence of pericytes facilitate the neuroinflammatory cascade and can influence the localization of the neuroinflammatory lesions. Pericytes regulate the development of organ-specific characteristics of the brain vasculature such as the blood–brain barrier (BBB) and astrocytic end-feet. Whether pericytes are involved in the control of leukocyte trafficking in the adult central nervous system (CNS), a process tightly regulated by CNS vasculature, remains elusive. Using adult pericyte-deficient mice (Pdgfbret/ret), we show that pericytes limit leukocyte infiltration into the CNS during homeostasis and autoimmune neuroinflammation. The permissiveness of the vasculature toward leukocyte trafficking in Pdgfbret/ret mice inversely correlates with vessel pericyte coverage. Upon induction of experimental autoimmune encephalomyelitis (EAE), pericyte-deficient mice die of severe atypical EAE, which can be reversed with fingolimod, indicating that the mortality is due to the massive influx of immune cells into the brain. Additionally, administration of anti-VCAM-1 and anti–ICAM-1 antibodies reduces leukocyte infiltration and diminishes the severity of atypical EAE symptoms of Pdgfbret/ret mice, indicating that the proinflammatory endothelium due to absence of pericytes facilitates exaggerated neuroinflammation. Furthermore, we show that the presence of myelin peptide-specific peripheral T cells in Pdgfbret/ret;2D2tg mice leads to the development of spontaneous neurological symptoms paralleled by the massive influx of leukocytes into the brain. These findings indicate that intrinsic changes within brain vasculature can promote the development of a neuroinflammatory disorder.
Collapse
|
49
|
Poppe T, Willers Moore J, Arichi T. Individual focused studies of functional brain development in early human infancy. Curr Opin Behav Sci 2021. [DOI: 10.1016/j.cobeha.2021.04.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
50
|
Abstract
The distribution of blood throughout the brain is facilitated by highly interconnected capillary networks. However, the steps involved in the construction of these networks has remained unclear. We used in vivo two-photon imaging through noninvasive cranial windows to study the engineering of capillary networks in the cerebral cortex of mouse neonates. We find that angiogenic activity originates at ascending venules, which undergo a burst of sprouting in the second postnatal week. This sprouting activity first establishes long paths to connect venules to blood input from neighboring arterioles, and then expands capillary interconnectivity with a multitude of short-range connections. Our study provides an experimental foundation to understand how capillary networks are shaped in the living mammalian brain during postnatal development. Capillary networks are essential for distribution of blood flow through the brain, and numerous other homeostatic functions, including neurovascular signal conduction and blood–brain barrier integrity. Accordingly, the impairment of capillary architecture and function lies at the root of many brain diseases. Visualizing how brain capillary networks develop in vivo can reveal innate programs for cerebrovascular growth and repair. Here, we use longitudinal two-photon imaging through noninvasive thinned skull windows to study a burst of angiogenic activity during cerebrovascular development in mouse neonates. We find that angiogenesis leading to the formation of capillary networks originated exclusively from cortical ascending venules. Two angiogenic sprouting activities were observed: 1) early, long-range sprouts that directly connected venules to upstream arteriolar input, establishing the backbone of the capillary bed, and 2) short-range sprouts that contributed to expansion of anastomotic connectivity within the capillary bed. All nascent sprouts were prefabricated with an intact endothelial lumen and pericyte coverage, ensuring their immediate perfusion and stability upon connection to their target vessels. The bulk of this capillary expansion spanned only 2 to 3 d and contributed to an increase of blood flow during a critical period in cortical development.
Collapse
|